Summary:
Immune responses must be tightly regulated to ensure both optimal protective immunity and tolerance. Costimulatory pathways within the B7-CD28 family provide essential signals for optimal T cell activation and clonal expansion. They provide crucial inhibitory signals that maintain immune homeostasis, control resolution of inflammation, regulate host defense and promote tolerance to prevent autoimmunity. Tumors and chronic pathogens can exploit these pathways to evade eradication by the immune system. Advances in understanding B7-CD28 pathways have ushered in a new era of immunotherapy with effective drugs to treat cancer, autoimmune diseases, infectious diseases, and transplant rejection. Here we discuss current understanding of the mechanisms underlying the coinhibitory functions of CTLA-4, PD-1, PD-L1—B7-1 and PD-L2—RGMb interactions, and less studied B7 family members, including HHLA2, VISTA, BTNL2, and BTN3A1, as well as their overlapping and unique roles in regulating immune responses, and the therapeutic potential of these insights.
ETOC blurb:
Tight regulation of immune responses is critical. In this review, Sharpe and colleagues review inhibitory members of the B7-CD28 family and their roles in regulating immune responses with an emphasis on CTLA-4, PD-1 and the increasingly complex network of cis and trans interactions with their ligands in lymphoid organs and the periphery with implications for infection, autoimmunity and anti-tumor immunity.
Introduction
The two-signal model for T cell activation1,2 catalyzed research that has defined mechanisms regulating T cell responses. This model posits activation of naïve T cells requires two distinct signals: signal 1, delivered by interaction of the T cell receptor (TCR) with antigen-MHC complex on antigen presenting cells (APC), conferring specificity to the response, and signal 2 (co-signal or costimulatory signal) provided by interactions of molecules on APC with their receptors on T cells.
While the two-signal model was proposed for the activation of naïve T cells, we now appreciate that second signals can also promote (costimulate) or attenuate (coinhibit) signals through the TCR of antigen-experienced cells such as effector, memory, and regulatory T cells. The balance between costimulatory and coinhibitory pathways is critical for regulating T cell activation, T cell tolerance and T cell exhaustion, and maintaining immune homeostasis. Dysregulation of these pathways can contribute to autoimmune disorders3,4, cancer5,6, and infectious diseases7,8. This mechanistic understanding has provided a basis for developing immunotherapies for treating cancer, autoimmune and infectious diseases, and transplant rejection.
The complementary roles of the costimulatory receptor, CD28, and coinhibitory receptor, CTLA-4, which both bind B7-1 (CD80) or B7-2 (CD86)9,10 provided the initial paradigm for what we now see as a broader role of costimulation and coinhibition within the B7:CD28 family. CD28 family members share common structural motifs, including an extracellular Immunoglobulin V-set (IgV), stalk, transmembrane domain, and a cytoplasmic domain with tyrosine signaling motifs. B7-2 provides the major signal for CD28 costimulation of naïve T cells11. B7 binding to CD28 leads to phosphorylation of tyrosine motifs, attracting adaptor proteins with Src Homology 2 (SH2) or Src homology 3 (SH3) domains to initiate downstream signaling and recruitment of the transcription factors NFAT, AP-1, and NFκb. This drives T cell proliferation, differentiation, and cytokine production9 (Table 1). CD28 and other costimulatory receptors in this family, such as ICOS, have been extensively reviewed elsewhere9,12–14.
Table 1:
Characteristics of CD28/CD28-like and B7 family members
CD28 family or B7 family | % Identity to CD28 or B7-1 EC domain | Regulation | Cytoplasmic signaling motifs/modifications | Expression | |||
---|---|---|---|---|---|---|---|
Stimuli to drive expression | Transcriptional regulators | Hematopoietic | Non-hematopoietic | Receptor/Ligand | |||
CD28 | 100 | Constitutive, TCR | Alpha and beta motifs in promoter region (negative) | PI3K motif, PP2A | T cell, plasma cell, NK, NKT | Unknown | B7-1, B7-2, ICOSL (In humans) |
CTLA-4 (CD152) | 26 | TCR/CD28, BCR | NFAT, FOXP3 | PI3K motif, PP2A, SHP2, PKC-n ubiquitination | T cell, B cell, NK, NKT, DC | Unknown | B7-1, B7-2, RGMb (soluble CTLA-4 only) |
ICOS (CD278) | 23 | TCR/CD28, IL-6/12/23 | NF-κB, AP-1, FOXO1, STAT3 | PI3K motif | T cell, NK, NKT | Unknown | ICOSL |
TMIGD2 (CD28-H, IGPR1) *Human only | 23 | TCR (negative) | NF-κB, AP-1, FOXO1, RFX1 | Y192, Y222 | T cells, NK cells, DC, ILC | Endothelial, epithelial, Constitutive (naive T cells), downregulated (activated T cells), NK, monocytes | HHLA2 |
PD-1 (CD279) | 16 | TCR, BCR, Type I IFN, IL-1/6/7/12/21/33, Notch signaling, TLR2, TLR4, TLR9, NOD signaling (in DC) | NFAT, AP-1, STAT3, STAT4, NF-κB, Blimp1, T-bet, Eomes, TOX, TOX2, FOXO1 | ITIM, ITSM, SHP1, SHP2, ubiquitination | T cell, B cell, NK cell, DC, myeloid cells, ILC | Rare: Tumor (melanoma) | PD-L1, PD-L2 |
B7-1 (CD80) | 100 | BCR, type I and type II IFN, GM-CSF, cAMP, MHC II, CD40, LPS, TLR, | IRF7, NF-κB, Jun N-terminal kinase, PU.1 | Unknown | B cells, NK cells, DC, macrophages | Rare: Tumor (follicular lymphoma) | CD28, CTLA-4, PD-L1 |
B7-2 (CD86) | 26 | BCR, CD40, LPS, type I and type II IFN, GM-CSF | NF-κB, STAT1, IRF1, AP-1, PU.1 | Unknown | B cells, NK cells, DC, macrophages | Tumor (hematopoietic) | CD28, CTLA-4 |
B7-H3 (CD276) | 25 | IFN-γ, IL-4 (negative) | NF-κB, STAT3, mTORC1, PI3K-AKT | Unknown | B cells, DC, macrophages (mice), T cell, NK, monocyte (human) | Tumor, Rare: Epithelia, fibroblast, osteoblast, | Unknown |
ICOSL (CD275, B7-H2, B7h, B7RP-1) | 24 | Type II interferon, TNF-α, LPS, IL-10 (negative) | NF-κB, STAT3, IRF1, AP-1, BAFF | Unknown | T cell, B cell, monocytes, macrophages mast cells | Endothelial, epithelial, fibroblast, osteoblast | ICOS, CD28 (human) |
BTNL2 (BTL-II) | 23 | TNF-α, IL-1β | NF-κB, IRF1, AP-1 | Unknown | T, B cells, macrophages | Stomach, small intestine, lung, tumor | Unknown but expressed on activated T and B cells |
PD-L2 (CD273, B7-DC) | 22 | IL-4, Type I and II IFN, common γ chain cytokines, TNF, GM-CSF, complement C5a, hypoxia | NF-κB, STAT3, AP-1, IRF1, PI3K-AKT, mTOR, TFEB, GATA2 | Unknown | T, B cells, macrophages, DC, macrophages, BM-mast cells | Endothelial cells, nasal epithelial cells, tumor | PD-1, RGMb |
PD-L1 (CD274, B7-H1) | 21 | Type I and type II IFN, common γ chain cytokines, TNF-α, IL-6/10/12/17/25/27, TGF-β, EGFR, Hypoxia, complement C5a, GM-CSF | STAT1, STAT3, IRF1, FOXOA1, NF-κB, AP-1, RAS/RAF/MEK, MYC, HIF-1α, HIF-2α, MAPK, PI3K-AKT, TFEB, GATA2, HSF1, ATF3, FOXP3 | Ubiquitination | B cells, NK cells, DC, macrophages | Endothelial cells, epithelial cells, keratinocytes, mesenchymal stem cells, fibroblast, pancreas islet, placenta, tumor | PD-1, B7-1 |
B7-H4 (B7x, B7s1, Vtcn1) | 21 | IL-6, IL-10, TGF-β, hypoxia, GM-CSF (negative), IL-4 (negative) | PKC, ATM, STAT3, AP-1, NF-κB, HIF-1α, PI3K-AKT | Unknown | DC, monocytes, macrophages | Mesenchymal stem cells, stroma, tumor | Unknown |
HHLA2 (B7-H5, B7-H7) *Human only | 21 | Type II IFN, IL-10, LPS, BMP-4 | METLL3, IRF1, NF-κB, STAT3 | Unknown | B cells, DC, monocytes | Endothelial cells, epithelial cells, kidney, colon, small intestine, lung, placenta, tumor | KIR3DL3, TMIGD2 |
BTN3A1 (CD277, BTN3.1, BTF5) | 19 | Type I IFN, mevalonate pathway, OXPHOS (negative) | AMPK, RUNXI1 (negative), CtBP1 (negative) | B30.2 (PRYSPRY) | T cells, B cells, NK cells, DC, macrophages, neutrophils | Endothelial cells, tumor | Mevalonate pathway phospho-antigens (intracellular) |
BTN2A1 (BTN2.1, BTF1) | 19 | Mevalonate pathway, OXPHOS (negative) | AMPK, RUNX1 (negative) | B30.2 (PRYSPRY) | T cells, B cells, NK cells, DC, macrophages, neutrophils | Epithelial cells, tumor | Mevalonate pathway phospho-antigens (intracellular) |
VISTA (VSIR, PD1-H) | 16 | Hypoxia, TGF-β, VEGF | FOXD3, NF-κB, STAT3, VEGF, HIF-1α, PI3K-AKT | Casein kinase 2, phospho-kinase C sites | T cells, DC, monocytes | Lung, tumor | VSIG-3, VSIG8, PSGL-1, LRIG, Synecdan-2 |
RGMb (DRAGON) | Not related | Curcumin (negative) | Unknown | Unknown (GPI anchor) | Macrophages | Neural tissues, epithelial cells, organs | PD-L2, Neogenin, BMP2, BMP4 |
Abbreviations are: CD, cluster of differentiation; CTLA-4, cytotoxic T-lymphocyte associated protein-4; ICOS, inducible T cell costimulator; TMIGD2, transmembrane and immunoglobulin domain containing 2; PD-1, programmed death-1; B7-H3, B7 homolog 3; B7-H4, B7 homolog 4; ICOSL, inducible T cell costimulator ligand; PD-L1, programmed death ligand-1; VISTA, V-domain immunoglobulin suppressor of T cell activation; BTNL2, butyrophilin like 2; PD-L2, programmed death ligand-2; BTN3A1, butyrophilin subfamily 3; BTN2A1, butyrophilin subfamily 2; HHLA2, HERV–HLTR-associating protein 2; RGMb, repulsive guidance molecule BMP co-receptor B; LRIG, leucine Rich Repeats and Immunoglobulin Like Domains 1; PSGL-1, P-selectin glycoprotein ligand-1; VSIG3, V-set and Immunoglobulin containing 3; VSIG-8, V-set and Immunoglobulin containing 8; KIR3DL3, Killer cell immunoglobulin-like receptor 3DL3; BMP, bone morphogenetic protein; TCR, T cell receptor; BCR, B cell receptor; IL, interleukin; TLR, Toll like receptor; NOD, Nucleotide oligomerization domain-like receptor; IFN, interferon; GM-CSF, granulocyte-macrophage colony stimulating factor; cAMP, cyclic adenosine monophosphate; MHC, major histocompatibility complex II, LPS, lipopolysaccharide; TNF, tumor necrosis factor; TGF, tumor growth factor; EGFR, epidermal growth factor receptor; VEGF, vascular endothelial growth factor; OXPHOS, oxidative phosphorylation; NK, natural killer cell; NKT, natural killer T cell; DC, dendritic cell; ILC, innate lymphoid cell; BM, bone marrow
This review builds on prior reviews10,15–17 and focuses on recent advances in our understanding of coinhibitory pathways in the B7-CD28 family with an emphasis on Cytotoxic T-lymphocyte associated protein 4 (CTLA-4), Programmed Cell Death Protein 1 (PD-1) and its ligands Programmed death-ligand 1 (PD-L1) and Programmed cell death 1 ligand 2 (PD-L2) (Figure 1). We also discuss select B7 family members: V-domain Ig suppressor of T cell activation (VISTA), Human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2), and butyrophilins, including Butyrophilin subfamily 3 member A1 (BTN3A1) and Butyrophilin like 2 (BTNL2). We review how these pathways regulate differentiation and function of naïve, effector, memory and regulatory T cells as well as activity of other cell types in different anatomical locations and disease context. In addition, we examine interactions between these pathways, in particular how interaction of PD-L1 with B7-1 creates a bridge between the PD-1 pathway and the B7-CD28/CTLA-4 pathway and how interaction of PD-L2 with Repulsive Guidance Molecule BMP Co-Receptor B (RGMb) creates a bridge between the PD-1 pathway and the RGMb/BMP2/BMP4 and the RGMb/neogenin complexes. Finally, we consider how this knowledge is refining our view of T cell costimulation, strategies for therapeutic modulation, and key areas for future investigation.
Figure 1. Costimulatory and coinhibitory receptor-ligand interactions in the B7-CD28 family.
The TCR is activated by recognition of peptide antigen presented by MHC and this gives specificity to the T cell response. Optimal T cell activation requires a second, costimulatory signal. B7-1 or B7-2 on APC bind to CD28 on T cells and provide a costimulatory signal. T cell activation is modulated by further stimulatory or inhibitory pathways between APC and T cells. These pathways regulate the fate of T cells, their differentiation into effector, memory, and regulatory T cells and their function. Some of the receptors/ligands are expressed on both APC and T cells as described in Table 1 and the text. Tumor cells and other cells can also express some of these receptors/ligands as described in Table 1 and the text.
CTLA-4
The gene for cytotoxic T-lymphocyte associated protein 4 (CTLA-4) encompasses a signal sequence, IgV-like extracellular domain containing the MYPPPY motif that binds its B7 ligands, stalk, transmembrane domain, and cytoplasmic domain. A juxtamembrane cysteine enforces homodimerization. A splice variant lacking the transmembrane domain encodes a soluble form, while a splice variant lacking the IgV domain encodes a ligand-independent form. Both have biological activity and are reviewed elsewhere10.
CTLA-4 expression is controlled at transcriptional and post-transcriptional levels (Table 1). Within T cells, both TCR activation18 through NFAT19 and forkhead box P3 (Foxp3) drive Ctla4 expression. CTLA-4 is upregulated on CD4+ Foxp3- and CD8+ T cells following initial activation. In contrast, CTLA-4 is constitutively expressed on regulatory T cells (Treg) due to Foxp3. CTLA-4 mRNA stability in the cytoplasm is regulated through the 3’ UTR20. MicroRNAs can directly (e.g. miR-145 and miR-155), or indirectly, through modulation of Foxp321, regulate CTLA-4 translation.
Cell surface expression of CTLA-4 protein is controlled at multiple levels. Following export from the Golgi apparatus, CTLA-4 is contained within intracellular vesicles that continuously cycle between the plasma membrane, cytoplasm, and/or lysosome. This process is at least partially mediated by the adaptor proteins AP-1 and AP-2 associating with the YVKM motif in the CTLA-4 cytoplasmic domain. Within endosomes, unphosphorylated CTLA-4 bound to AP-1 trafficks from the trans-Golgi network to the lysosome for subsequent degradation. Unphosphorylated CTLA-4 bound to AP-2 favors rapid internalization from the plasma membrane into endosomes in the cytoplasm. In contrast, when the YVKM motif is phosphorylated by the tyrosine kinases Lck or Fyn, CTLA-4, in conjunction with LPS responsive beige-like anchor protein (LRBA) and Rab GTPases, exhibits delayed internalization and may be recycled to the cell surface depending on the ligand bound, as discussed below.
The critical role of CTLA-4 in regulating tolerance was first demonstrated in mice, where germline deletion of Ctla4 led to systemic autoimmunity characterized by hyperproliferation of lymphocytes, multi-organ tissue destruction, elevated immunoglobulin levels, and death within 2–4 weeks of age22,23. Similarly, in humans, haploinsufficiency of CTLA4 causes a syndrome marked by immune dysregulation, including lymphoproliferation, autoimmunity and inflammation manifesting as multi-organ lymphocytic infiltrates and cytopenia24,25. Mutations in the cell machinery involved with trafficking CTLA-4-containing endosomes to the cell surface, such as LRBA3,26 or associated scaffold-binding proteins, such as DEF, phenocopy germline CTLA4 haploinsufficiency, causing hypogammaglobulinemia, B cell lymphopenia, enteropathies, and often autoimmune-related cytopenias (e.g. autoimmune hemolytic anemia) 26,27. In addition, many polymorphisms in CTLA4 in the regulatory or promoter regions, particularly the A49G polymorphism, have been associated with numerous autoimmune conditions, including Hashimoto’s and Graves’ Disease28. These polymorphisms suggest subtle modulation of CTLA-4 may predispose to autoimmunity.
CTLA-4 inhibits T cell responses through both cell-intrinsic and extrinsic mechanisms. A major mechanism by which CTLA-4 modulates T cell responses is by regulating surface availability of its ligands, B7-1 (CD80) and B7-2 (CD86). CTLA-4 has a higher affinity for these ligands than CD28 and acts as a competitive inhibitor for CD28. Disease in CTLA-4 deficient mice can be abrogated by genetic deletion of both B7-1 and B7-229 or genetic modification of CD28 to disrupt the Lck phosphorylation site30, suggesting that disease arises from unopposed costimulation through CD28.
One mechanism by which CTLA-4 can exert cell-extrinsic effects is through transendocytosis, a process by which it binds to and removes B7-1 and B7-2 from the surface of APCs, including dendritic cells (DC) and B cells. Because B7-1 is a non-covalent homodimer and CTLA-4 is a disulfide linked homodimer, CTLA-4—B7-1 interactions can be multimeric with low and high avidity. In contrast, B7-2 is a monomer and engages with CTLA-4 as a low avidity interaction31,32. These different affinity receptor-ligand interactions further regulate CTLA-4. The high affinity CTLA-4—B7-1 interaction leads to CTLA-4 ubiquitination and shunting of the complex toward lysosomes for subsequent degradation33. In contrast, CTLA-4 molecules bound to B7-2 do not trigger ubiquitination, enabling CTLA-4 to disengage from B7-2 and for CTLA-4 to recycle to the cell surface. Through these mechanisms, CTLA-4 engagement with B7-1 or B7-2 decreases their availability for CD28, thereby diminishing costimulation and T cell activation. In this way, Treg, which express CTLA-4 constitutively and at high levels, control CD28-dependent activation of naïve T cells by limiting their access to B7-1 and B7-2. Migratory DC, both conventional dendritic cell (cDC) cDC1 and cDC2 subsets, are a major target of CTLA-4 - mediated transendocytosis in the lymph node34. Within the tumor microenvironment, CTLA-4 - expressing Treg also interact with B7-1 or B7-2 - expressing DC35. Transendocytosis of B7-1 by CTLA-4 on Treg also can increase the availability of B7-1’s other binding partner, programmed death-ligand 1 (PD-L1), on APCs36, as will be discussed further in the PD-L1 section of this review. This provides evidence that modulation of CTLA-4 expression can indirectly influence the PD-1/PD-L1 pathway.
CTLA-4 also exerts cell-extrinsic effects by inducing expression of indoleamine 2, 3-dioxygenase (IDO) in DC, which in turn contributes to the inhibitory effects of CTLA-4 on T cell responses37. CTLA-4 may also inhibit T cell responses through cell-intrinsic mechanisms. Following ligand binding, CTLA-4 is phosphorylated, which leads to recruitment of the phosphatases SHP-2 and protein phosphatase 2A (PP2A) 38. These phosphatases, in turn, directly inhibit intrinsic signaling through the TCR and CD28, attenuating T cell proliferation and interleukin-2 (IL-2) production.
CTLA-4 is a key mediator of Treg cell suppressive function and when co-expressed with other co-inhibitory molecules such as programmed cell death protein 1 (PD-1), lymphocyte-activation gene 3 (LAG-3), or T cell immunoreceptor with immunoglobulin and ITM domain (TIGIT), these Treg cells display enhanced suppressive function39–42. Studies of antigen-specific Ctla4 deficient Treg cells, generated to examine the consequences of CTLA-4 deficiency without lymphoproliferation, revealed that Ctla4 deletion abrogates Treg function in vivo43. Constitutive conditional loss of Ctla4 on Treg cells led to a lethal autoimmune syndrome akin to germline knockout, albeit with delayed kinetics44. However, when Ctla4 is deleted inducibly on Treg cells in adult mice, these mice do not develop systemic autoinflammation, suggesting an important function for CTLA-4 during the perinatal period in thymic selection and central tolerance45. Inducible loss of Ctla4 in adulthood on all cells or only on Treg cells protected mice from experimental autoimmune encephalomyelitis (EAE), and these Treg cells suppressed proliferation of T conventional cells. Massive expansion of Treg cells in the draining lymph node (LN) and central nervous system (CNS), together with upregulation of IL-10 and other coinhibitory receptors, including PD-1 and LAG-3, may explain the protection from EAE. Effector T cells from adult mice in which Ctla4 was deleted can become pathogenic in some settings but do not appear to contribute to EAE resistance of these mice45,46. These studies demonstrate the role of CTLA-4 in limiting peripheral Treg cell expansion and activation, and capacity to control T conventional cells.
CTLA-4 also regulates specialized populations of Foxp3+ CD4+ T cells including T follicular regulatory (Tfr) cells47,48 that suppress germinal center (GC) reactions and antibody responses, as well as T follicular helper (Tfh) cells, a CD4+ T cell subset that provides T cell help to B cells and is essential for GC formation and high affinity antibody production. Inducible Ctla4 gene deletion studies in adult mice revealed multifaceted roles for CTLA-4 in Tfr and Tfh cells. These studies show CTLA-4 suppresses Tfr cell differentiation and/or maintenance but are essential for the inhibitory function of differentiated Tfr cells. CTLA-4 also inhibits Tfh cell differentiation and capacity of differentiated Tfh cells to stimulate B cell responses. The transcription factors Tcf1 and Lef1 are critical to restrain Ctla4 expression in Tfh cells, an effect mediated by epigenetic modification through histone deacetylation of the Ctla4 locus in a model of vaccination49. Further research is warranted to elucidate the potential of modulating CTLA-4 in Tfh, Tfr or Treg cells to augment vaccination efficacy and pathogen eradication.
While initial rationale for anti-CTLA-4 therapy focused on unfettering CD28 costimulation, it was later recognized that anti-CTLA-4 could also act through Fc-mediated depletion of high CTLA-4 - expressing cells, namely Treg cells50–52. Subsequent studies demonstrated that Treg cells remained present in the tumor microenvironment of patients treated with anti-CTLA-453, suggesting that its clinical efficacy likely stems from a combination of Fc-mediated antibody-dependent cellular cytotoxicity (ADCC) and blockade of the CTLA-4—B7-1/2 axis in both the lymph node and the tumor35,54. The balance of expression of activating and inhibitory Fc receptors differs between human and mouse, which may explain this difference55,56. Recent evidence further suggests that anti-CTLA-4 may destabilize Treg cell function by altering glycolysis57, at least in part due to the high linkage between CTLA-4 and FoxP3 function.
Within conventional CD4+ T cell populations, costimulation and coinhibition can modulate the strength of TCR signaling, favoring specific lineages of CD4+ T cell subsets. Germline loss of Ctla4 or antibody blockade promotes the generation of Th2 cells58 in a STAT6 - dependent manner and the generation/induction of Th17 cells. The generation of Th2 cells provides a compelling explanation for the pronounced autoantibody production observed in CTLA-4 haploinsufficiency syndromes which display profound autoantibody generation, possibly by modulating the strength of TCR signal and interleukin-4 (IL-4) production.
In addition to regulating T cells, CTLA-4 also has functions in B cells. B cells from CTLA-4 germline deficient mice exhibit a more activated phenotype with elevated antibody titers compared to their WT counterparts22,23. Initially, this was attributed to CTLA-4 expression on T cells, particularly Tfh and Tfr cells. However, CTLA-4 is present on both canonical B cells in the follicle, termed B2 cells, and a non-canonical, more innate-like, B1 cell population, namely B-1a cells59.
On canonical B cells CTLA-4 expression is induced by T cell help60. Blockade of CTLA-4 during in vitro stimulation of purified B cells with IL-4 in combination with CD40 or LPS led to decreased class switching to IgG1 and IgE-expressing B cells and IL-8, IL-10, and TNFα production via an NF-κb or STAT6-dependent pathway61. Although not tested in vivo, this suggests that CTLA-4 has a cell-intrinsic effect on B cells to modulate class switching and cytokine production.
CTLA-4 plays a critical role in B-1a cells, a distinct lineage of mature CD5+ B cells derived during fetal development and differing from follicular B cells62. B-1a cells persist as a small minority in the spleen but are the predominant B cell population in the peritoneal cavity. B-1a cells reside outside the GC and uniquely produce natural antibodies (often of low affinity) secreted through T cell independent mechanisms. CTLA-4 expression is constitutive on B-1a cells in both the spleen and peritoneal cavity. B-1a cells possess self-renewal capacity, and loss of CTLA-4 in this population enhances their proliferation and interaction with follicular dendritic cells (FDC). Intriguingly, B-1a cells express high levels of both B7-1 and B7-259. This raises intriguing questions about whether CTLA-4 in this cell population functions either in cis or trans with other B-1a cells or FDC to establish an additional layer of negative feedback. Furthermore, B-1a cells from mice lacking Ctla4 specifically in B cells exhibit enhanced activation and ability to differentiate into APCs and induce Tfh cells and GC formation when transferred to congenically distinct hosts59.
Although a direct parallel between murine B-1a cells and their human counterparts has yet to be established, CTLA-4 expression has been observed in multiple human hematologic malignancies, including acute myeloid leukemia63, non-Hodgkin’s lymphoma, and chronic lymphocytic leukemia (CLL). In CLL, a generally indolent disease characterized by clonal expansion of a mature CD5+CD23+ B cell population, the presence of CTLA-4 may mark an additional immunosuppressive mechanism.
PD-1
PD-1 has signal, IgV-like, stalk, transmembrane, and a cytoplasmic domain containing ITIM and ITSM motifs. A number of splice variants have been identified, as well as numerous polymorphisms linked to increased susceptibility to autoimmune diseases including lupus and multiple sclerosis and alterations in viral setpoint for chronic hepatitis B infection10.
Expression of Pdcd1 transcripts reflects a dynamic interplay between epigenetic modifiers (e.g. DNA methylation, histone modification) for chromatin availability and transcription factor binding to regulate gene expression, which varies according to cell state. PD-1 is not expressed on naïve CD4+ or CD8+ T cells, but rapidly upregulated in a TCR-dependent manner. TCR signaling is the primary driver of Pdcd1 expression64 (Table 1) and is accompanied by demethylation of the Pdcd1 locus65 and binding of the transcription factors NFATc1 and AP-166. Cell surface protein expression emerges within 24 hours or less and even prior to the initial cell division64. Other pathways that can induce Pdcd1 expression include histone methyltransferases such as H3K4m3 to mediate NF-κB p50/p50 homodimer binding to the Pdcd1 promoter67; cytokine signaling through IFNα68, IL-6/STAT369 or IL-12/STAT-4; and Notch signaling70. Following resolution of infection, CD8+ memory T cells demonstrate signs of Pdcd1 locus remethylation65, partly through the transcription factors BLIMP-1, Tbet, and Eomes and recruitment of lysine-specific demethylase LSD1 via BLIMP-171, to reduce Pdcd1 expression.
PD-1 levels may remain high on select populations, including CD8+ exhausted T cells, Tfh cells, and tissue resident memory cells (Trm) cells. Chronic TCR engagement, as occurs during chronic viral infection, drives a state of T cell dysfunction, known as T cell exhaustion, in which T cells exhibit limited proliferative potential and effector function. During T cell exhaustion, the Pdcd1 promoter undergoes remodeling and demethylation65, marked by distinct epigenetic changes in a 23kb region upstream of the transcriptional start site72,73. Under these conditions, NFAT signaling is repressed74, while other transcription factors are expressed, including FOXO175, contributing to Pdcd1 transcription. Interestingly, in chronic LCMV infection, BLIMP-1 remains bound to the Pdcd1 locus. Emerging evidence suggests that IL-6 and subsequent STAT3 binding do not displace BLIMP-1 binding but rather overcome its inhibitory effects through increased histone acetylation69. This implies that prolonged cytokine signaling can induce additional changes in chromatin availability, potentially contributing to sustained PD-1 expression.
Epigenetic remodeling of the Pdcd1 locus during chronic infection or tumor growth is at least partly due to the transcription factor TOX, as genetic deletion studies of TOX have demonstrated reduction or abolished PD-1 expression76–78. However, even after antigen is cleared, as has been elegantly shown following antiviral treatment or spontaneous clearance of hepatitis C viral (HCV) infection79,80, PD-1 surface levels decrease but its locus retains persistent alterations in chromatin accessibility. Similarly, both TOX and PD-1 expression and epigenetic remodeling persist in mouse CD8+ T cells exposed to iterative stimulation and rest cycles between stimulations81 or in humans at higher levels in effector memory CD8+ T cells recognizing epitopes from latent EBV and CMV viral infections than in influenza-specific CD8+ T cells82. These EBV and CMV-specific cells retain both proliferative capacity and cytokine production, suggesting a potential protective role for TOX and PD-1 expression following repeated stimulation. These findings suggest that both the persistent presence of antigen and recurrent, limited stimulation over time may modulate chromatin accessibility and PD-1 expression in overlapping but distinct ways.
Several studies have demonstrated that PD-1 expression can be further regulated at the post-translational level. Fucosylation at four N-glycan sites mediated by FUT8 is required for proper PD-1 localization at the plasma membrane83. Moreover, cell surface expression of PD-1 is additionally regulated at the level of translation through polyubiquitination by the E3 ligase FBXO3884. Finally, in a model of hepatocellular carcinoma, TOX stabilizes PD-1 in the cytoplasm, promoting its translocation to the cell surface85. These findings warrant further investigation of posttranslational regulation of PD-1 expression.
PD-1 has two ligands, PD-L1 and programmed cell death protein 1 ligand 2 (PD-L2) (Figure 1, Table 1)86,87. PD-1 engagement with its ligands opposes TCR and CD28 signaling88,89, resulting in inhibition of T cell proliferation, differentiation and effector functions including IFNγ, TNFα, and IL-2 production. Upon binding either ligand, PD-1 undergoes phosphorylation of its immunoreceptor tyrosine-based switch motif (ITSM) and immunoreceptor tyrosine-based inhibitory motif (ITIM) by Fyn and Lck in T cells90. This phosphorylation recruits phosphatases, particularly src homology 2-domain containing phosphatase 2 (SHP-2) but also SHP-1, which dephosphorylate key signaling molecules including the zeta chain of CD3 (CD3z), ZAP-70, and PKCθ91 and antagonizes the effects of CD2892 through dephosphorylation CD28 signaling components88. Consequently, AKT, the PI3K pathway, PLCγ1, Ras-MEK-ERK signaling cascade are inhibited, leading to suppression of T cell responses. Ligand binding by PD-1 on B cells also triggers phosphorylation of both tyrosines of PD-1, followed by recruitment of SHP2 and downstream dephosphorylation of BCR - associated signaling proteins, including Ig beta, Syk, phospholipase C-γ2 (PLC-γ2) and ERK1/293. However, SHP-2 may not be the only mediator of PD-1 signaling. Mice with T cell selective deletion of Ptpn11 (gene encoding SHP-2) do not completely recapitulate the phenotype of Pdcd1 deficient mice during chronic viral infection94, suggesting there may be other mechanisms downstream of PD-1, redundant to SHP-2. Alternatively, the effects of PD-1 may differ by cell type. For example, cell-specific effects of SHP-2 or PD-1-SHP2 signaling in T cells versus myeloid cells may provide one possible explanation95.
PD-1 signaling also alters cell metabolism. T cells undergo a profound metabolic shift toward glycolysis following activation. PD-1 in T cells inhibits this switch by suppressing glucose and glutamine metabolism and promoting lipolysis and fatty acid oxidation16,17,96. PD-1 deletion in myeloid progenitor cells can shift cells toward intermediates of glycolysis and the TCA cycle and elevated cholesterol97. This increased reliance on alternate carbon sources may provide therapeutic opportunities.
Like CTLA-4, PD-1 may exert a push-pull relationship, influencing initial activation of naïve T cells, differentiation into effector or memory cells, and the balance between effector and suppressive cell populations in both the lymphoid organs and the periphery. Loss of PD-1, through genetic deletion or antibody blockade, leads to enhanced T cell activation and effector function16,98, as has been shown in numerous disease contexts, including acute infection99,100, autoimmunity101,102, chronic viral infection7,8,103,104 and malignancy5,105. In acute infection models such as intranasal vaccina and influenza, the absence of PD-1 leads to more rapid and robust expansion of effector T cells100. However, this effector T cells expansion during acute infection may come at the cost of diminished number and function of memory CD8+ T cells99. Similarly, in mouse models of autoimmune disease, loss of PD-1 signaling, accelerates disease onset with increased numbers and function of pathogenic self-reactive T cells.
Early seminal work in the LCMV Clone 13 chronic infection model8 and HIV7 demonstrated a key role for PD-1 in T cell exhaustion. Virus-specific, exhausted CD8+ T cells highly expressed PD-1 and antibody blockade of PD-1 or PD-L1 led to improved proliferative capacity, increased production of cytokines such as IFNγ, as well as cytolytic function7,8,106. This strategy of PD-1 or PD-L1 blockade has revolutionized cancer treatment, with FDA approval of PD-1 inhibitors for over 25 types of cancer and two tumor-agnostic indications: tumor mutational burden >10 mutations/megabase and microsatellite instable (MSIhigh)107.
Exhausted CD8+ T cells are heterogenous: stem-like progenitor exhausted cells retain the capacity to differentiate and give rise to effector progeny, while a more terminally differentiated population expressing higher levels of PD-1 in concert with other co-inhibitory molecules has the most impaired proliferative capacity and the most limited cytokine production108. These populations of cells have been identified in lymph organs104,109, and target organs during chronic viral infections, and tumors5,105,110. PD-1 blockade increases proliferation and differentiation of the stem-like exhausted CD8+ T cells (defined by Tcf1+PD-1+) into effector-like cells. Using either LCMV clone 13 or a tumor model, two groups suggest that this occurs in part due to asymmetric cell division111,112. CD28 is necessary for response of these CD8+ T cells92,113 to anti-PD-1 blockade. Future work will likely delineate how PD-1 may coordinate these effects. Stem-like T cells in the lymph node respond to anti-PD-1. Recent work in tumor models shows that stem-like T cells further differentiate into effector cells in the tumor microenvironment in the presence of additional costimulation114. These data suggest that strategies to augment costimulatory signals in the tumor microenvironment may be needed to promote effector T cell differentiation and function.
PD-1 also is expressed on tissue resident memory cells (Trm) in mice following resolution of viral infection115 and on Trm-like cells in inflammatory lesions in brain parenchyma in mice and multiple sclerosis patients and tumors116,117. Multiple studies link the presence of Trm-signatures or Trm-like cells to response to anti-PD-1 cancer immunotherapy118,119 and in anti-PD-1 related colitis120. One challenge with this nomenclature is that canonical Trm cells survive in the absence of antigen with a gene expression program related to retention in the tissues. Chronically stimulated cells may have gene programs overlapping with Trm due to their localization in tissue while simultaneously encountering antigen. Although PD-1 is expressed on both Trm cells and chronically stimulated exhausted cells, it remains to be discovered whether PD-1 drives a similar program in each of these situations. However, given the wide distribution of Trm cells across the human body, an intriguing question is how PD-1 pathway blockade interacts with signaling cues, such as the cytokine milieu, to reactivate select Trm-like populations. Further open questions include understanding how modulation of the PD-1 pathway influences the differentiation of different types of memory cells (e.g. central memory where PD-1 is not expressed vs Trm cells where PD-1 is expressed), the maintenance of these memory populations, and characteristics of memory T cells in the context of chronic inflammation. Interestingly, PD-1+ CD8+ T cells bearing a tissue resident signature have been identified in proximity to PD-L1+ macrophages in healthy human pancreatic donor tissue121, suggesting an important role for ongoing receptor-ligand signaling. Understanding the role of PD-1 in memory T cell biology could lead to more effective strategies for treating chronic infections and cancer.
Selective loss of PD-1 signaling on Treg cells enhances their suppressive capacity in autoimmune diseases122, infection123, and tumors124,125. In murine models of EAE and type I diabetes122, absence of PD-1 on Treg cells leads to increased suppression and protection against disease. Mechanistically, loss of PD-1 signaling reduced signaling through the PI3K-AKT pathway, contributing to enhanced Treg cell suppressive capacity122 as enhanced AKT signaling has been associated with decreased Treg cell suppressive capacity126. Similarly, in a model of toxoplasmosis, PD-1 deficient Treg cells play a critical role in suppressing cDC1 function, thereby limiting pathogenesis. Loss of PD-1 on Treg cells in this model results in increased effector Treg, elevated IL-10, and increased immunopathology123.
In cancer, PD-1 blockade has been linked to cases of tumor hyperprogression, characterized by rapid or explosive tumor growth. One hypothesis suggests hyperprogression arises from the enhanced suppressive capacity and abundance of Treg cells, enabling them to counteract anti-tumor effects of PD-1 blockade124,125. In addition, PD-1 expression on Treg cells has clinical implications. In metastatic clear cell renal cell carcinoma (ccRCC), patients with a high PD-1+ Treg/CD8+ T cell ratio experience shorter progression-free survival, overall survival and lower overall response rate, suggesting the PD-1+ Treg/CD8+ T cell ratio may have predictive value127. These findings further suggest that the ratio of PD-1+ Treg cells to PD-1+ CD8+ T cells at the beginning of therapy may influence treatment response. Therefore, strategies to deplete Treg cells or impair Treg cell function in combination with PD-1 blockade may improve treatment outcomes.
Like CTLA-4, PD-1 can be expressed on Tfh cells, Tfr cells128 and B cells within the GC and outside the follicle to regulate both T cell dependent and T cell independent (TI) processes. PD-1 and PD-L1 inhibit the differentiation and suppressive function of Tfr cells. Vaccination of Pdcd1-/- mice led to four-fold greater expansion of Tfr cells relative to wildtype controls without a change in the relative percent of Tfh cells, and these PD-1 deficient Tfr cells have more potential inhibitory function128. PD-1 also can inhibit Tfh cell expansion, differentiation and help for IgG class switching129. PD-1—PDL1/L2 interactions play an important role, as the PD-L1/L2 expressed on B cells can bind to and signal into PD-1 on Tfh cells, leading to reduced plasma cell numbers130. However, as PD-1 may also be expressed on GC B cells, the PD-1 pathway may have context-dependent effects in regulating humoral immune responses. In one study, a subset of PD-1high B cells impaired anti-tumor CD8+ T cell responses through an IL-10 dependent mechanism in a mouse hepatoma model, suggesting a way in which PD-1 signaling in B cells may inhibit anti-tumor immunity and prevent tumor growth control131.
High expression of PD-1 characterizes a CD4+ CXCR5- T cell population found in peripheral blood and synovial fluid of patients with rheumatoid arthritis. This population, termed T peripheral helper cells (Tph), promotes plasma cell expansion via CXCL13 and IL-21132. Similar Tph cells have been identified in other autoimmune conditions including juvenile rheumatoid arthritis. These findings raise the question of whether PD-1 agonism may be a viable strategy to target T-B interactions in select autoimmune conditions. Indeed, recent findings show that PD-1 agonistic antibodies that target the lower juxtamembrane region of PD-1 and have enhanced Fc receptor binding activity can ameliorate disease in two autoimmune mouse models133. A phase II clinical trial suggests PD-1 agonists have therapeutic potential in rheumatoid arthritis134.
It is increasingly clear that PD-1 also functions on non-T cell populations. This is particularly evident in B1 cells residing in the peritoneum, NK cells, macrophages, and innate lymphoid cells (ILC) inhabiting the respiratory and mucosal surfaces. Initial studies of Pdcd1 deficient mice revealed heightened T cell-independent B cell responses, marked by elevated IgG3 production in response to DNP-Ficoll, a TI antigen135. This was accompanied by enhanced expansion of CD5- B-1b cells, suggesting a cell-intrinsic role for PD-1 signaling on B cells. Subsequent mechanistic studies demonstrated that PD-1 inhibited formation and reactivation of TI B cell memory cells, an interaction mediated, at least in part, by PD-L1 or PD-L2 on non-antigen specific B cells136. Immunization with 2,4,6 trinitrophenol (TNP)-Ficoll selectively activated B-1b cells, inducing their expansion, isotype switching, and plasmablast/plasma cell differentiation. Likewise, exposure of Pdcd1 deficient mice or wild-type mice treated with anti-PD-1 to sublethal pneumococcal infection or vaccination with the polysaccharide S. pneumoniae-capsule enhanced isotype switching and IgG production by pneumococcal specific B cells, processes that are T cell independent137. Further research is warranted to elucidate the mechanisms of these PD-1 -mediated B cell intrinsic pathways. Given that cancers may display recurrent and aberrant glycosylation patterns, exploring how the PD-1 pathway can be harnessed to engineer T cell independent anti-tumor responses holds promise for novel therapeutic approaches.
PD-1 expression has been observed on NK cells both intrinsically and through transendocytosis138–140. ILCs also express PD-1. Particular attention has focused on IL-33 signaling in group 2 innate lymphoid cells (ILC2), which regulate immunity, including nosocomial and commensal pathogens, through antigen-independent mechanisms. Induction of IL-33 upregulates PD-1 to limit ILC2 responses in models of airway hyperreactivity141, cancer142, and within adipose tissue of mice fed a high fat diet 143. PD-1 blockade increased ILC2 expansion and production of cytokines such as GM-CSF142.
Likewise, PD-1 also can be expressed on myeloid cells within the tumor microenvironment or during infection. PD-1 expression was higher on tumor-infiltrating macrophages with an immunosuppressive “M2-like” phenotype, exhibiting a foamy morphology with impaired phagocytosis144. Within the tumor microenvironment, PD-L1 on tumor cells can induce negative signals in PD-1-expressing myeloid cells through SHP2, blocking type I IFN/STAT1/CXCL9 signaling to suppress cytotoxic T cell recruitment in lung metastases145. In a model of listeria monocytogenes infection, PD-1 was selectively induced on DCs through engagement of TLR2, TLR3, TLR4, and nucleotide-binding oligomerization domain (NOD) proteins, inhibiting IL-12 or TNFα production146. Collectively, these studies identify non-T cell mediated mechanisms by which PD-1 tempers inflammatory responses.
Finally, PD-1 has also been shown to be expressed on neurons. Both Pdcd1 deficient and selective deletion of Pdcd1 on CA1 hippocampal neurons increased CA1 neuron excitability through increased long-term potentiation, improved learning and memory behavioral test. Interestingly, PD-L1, which is expressed on neurons and microglia, was upregulated. PD-L2 was not examined147. This study identifies a previously unappreciated role for PD-1 pathway in a non-hematopoietic tissue (neuron) and points to the need to better characterize the effects of coinhibitory pathways on neurological function.
There is crosstalk between PD-1 and CTLA-4 on immune cell populations. Activation through the TCR will result in upregulation of both CTLA-4 and PD-1. Moreover, loss of expression of CTLA-4 is accompanied by upregulation of PD-1 and vice-versa, as well as the increased expression of other costimulatory and co-inhibitory receptors148. Blockade of either anti-CTLA-4 or anti-PD-1 can lead to inflammatory syndromes that mirror autoimmunity and are known as immune-related adverse events (irAE). For example, one study in metastatic melanoma reported rates of severe irAE (clinically grade 3 or higher) as 27% for anti-CTLA-4, 16% for anti-PD-1 and 55% for combination anti-CTLA-4/anti-PD-1149, with other studies showing a dose-dependent effect of anti-CTLA-4 therapy150. Although an active area of investigation, mechanisms to explain this higher rate are limited. A mouse model of myocarditis using Pdcd1-/-Ctla4+/- mice151 suggests that the myocardial pathology requires CD8+ T cells152. Given the distinct expression levels of these co-inhibitory receptors, with higher expression of CTLA-4 on Treg, enhanced CTLA-4 and PD-1 on effector Treg and elevated PD-1 on exhausted CD8+ T cells, further work is warranted to understand how PD-1 and CTLA-4 interact on different cell types in autoimmunity, infection and cancer.
PD-L1 and PD-L2
The PD-1 ligands, PD-L1 and PD-L2, have 36% sequence identity, with PD-L2 having ~3-fold greater affinity for PD-1 than PD-L187,153. Both are expressed in lymphoid and non-lymphoid tissues, including tumors (Table 1). Pro-inflammatory cytokines induce PD-L1 and PD-L2 expression, potentially as a physiologic negative feedback mechanism to attenuate T cell responses, restore homeostasis and limit immune-mediated tissue damage, but enabling tumor cell evasion of the immune system. PD-L1 and PD-L2 each have second unique binding partners: PD-L1 with B7-1153 and PD-L2 with RGMb154. In this section, we discuss current understanding of these interactions.
The PD-L1–B7-1 interaction serves as a point of intersection between the CTLA-4–B7-1 and PD-L1–PD-1 inhibitory pathways (Figure 2). The orientation of the PD-L1 molecule and presence of a stalk domain allows it to establish an interaction with B7-1 that can only occur in cis on the same cell surface but not in trans across two different cells155–158. Co-expression of B7-1 and PD-L1 within DC or tumor infiltrating myeloid cells supports a functional relevance of this interaction155,156. PD-1 and B7-1 share a common binding region on the IgV domain of PD-L1 and can competitively inhibit each other’s binding to PD-L1153,155. Using DC from knock-in mice with mutations in the IgV domains of PD-L1 or B7-1 that abrogated the cis-PD-L1–B7-1 interaction, Sugiura et al. showed these mutant DC promoted sustained PD-1 signaling. In turn, this led to diminished anti-tumor and self-reactive T cell responses. These findings indicate that the cis interaction of PD-L1 and B7-1 on APC blocks PD-L1 binding to PD-1 and thereby impairs inhibition of T cell responses156. The similar affinities of PD-L1 for the trans interaction with PD-1 (Kd 0.77 mM) and the cis interaction of PD-L1 with B7-1 (Kd 1.4 mM) suggest a competition which may be dominated by the high local concentrations of PD-L1 and B7-1 on a cell surface155.
Figure 2. Novel interactions among B7-1, PD-L1, CD28, CTLA-4, and PD-1.
B7-1 can form a back-to-back homodimer (interaction surface denoted as dark green on B7-1 IgV domain). A single B7-1 can interact with CD28 to deliver a co-stimulatory signal for T cell activation (interaction surface denoted as orange on B7-1 IgV domain). The B7-1 homodimer can interact with CTLA-4 in a multimeric fashion and CTLA-4 can remove and internalize the B7-1 by trogocytosis (interaction surface denoted as orange on B7-1 IgV domain), resulting in loss of B7-1 costimulatory activity. PD-L1 can interact with PD-1 (interaction surface denoted as light green on PD-L1 IgV domain), leading to phosphorylation of the PD-1 ITIM and ITSM tyrosines, recruitment and activation of SHP-2, and inhibition of T cell activation. B7-1 can interact with PD-L1 to form a heterodimer in which the B7-1 can still engage CD28 or CTLA-4 but the PD-L1 cannot engage PD-1. The CD28 signal remains active but the PD-1 signal is diminished. The CTLA-4 interaction is unimolecular since the B7-1 is monomeric but the CTLA-4 can still trogocytose the B7-1, resulting in loss of B7-1 costimulatory activity and free PD-L1 that can engage PD-1 to deliver an inhibitory signal.
B7-1 is a back-to-back, non-covalent homodimer and surprisingly, PD-L1 interacts with this dimer interface on B7-1, reducing B7-1 to a monomer156–158. This still allows the interaction of B7-1 with CD28 and CTLA-4. Co-culture of T cells with APCs that co-express B7-1 and PD-L1 showed that the B7-1—CD28 trans interaction efficiently engaged and resulted in formation of CD28 microclusters on T cells157. However, the functional consequences of PD-L1—B7-1 interactions on CTLA-4—B7-1 interactions remain an area of active investigation. Zhao157 proposed that the PD-L1—B7-1 interaction, by reducing B7-1 to a monomer, would prevent the formation of a CTLA-4—B7-1 multimeric structure. Nevertheless, Tegkuc et al36 show that CTLA-4 transendocytoses the B7-1 from a PD-L1—B7-1 heterodimer, thereby freeing PD-L1 to engage PD-1, increasing coinhibitory activity and also decreasing B7-1—CD28 signaling. Thus, reduced B7-1 on APCs has dual suppressive effects on T cells responses by limiting B7-1 co-stimulation and increasing free PD-L1 to interact with PD-1.
Whether PD-L1 interacts with B7-1 in cis on other cell types may also matter. A recent study suggests that membrane invagination can enable either B7-1 and B7-2 molecules expressed on CD8+ T cells to interact with CD28 in cis and in turn, this cis interaction can drive migration and cytokine production in vitro and mediate anti-tumor immunity159. The authors posit that this may enable further cell survival in peripheral tissues where APCs are scarce. However, the frequency of this occurrence relative to a competing mechanism for cis-interactions of B7-1 with PD-L1 expression on a CD8+ T cell remains to be determined.
Anti-B7-1 antibodies that target the B7-1 interface and selectively block the PD-L1—B7-1 interaction but not the B7-1—CD28 or CTLA-4 interaction can reduce disease severity in the autoimmune mouses models of experimental autoimmune encephalitis (akin to multiple sclerosis), arthritis, and Sjogren’s160. Anti-B7-1 antibodies that target the PD-L1 interface, would liberate PD-L1 to more effectively deliver PD-1-mediated inhibitory signals. In comparison, anti-PD-L1 antibodies that selectively block the PD-L1–B7-1 interaction but still allow the interaction with PD-1 accelerated type 1 diabetes in NOD mice, particularly self-reactive effector T cells161,162, and impaired the induction and maintenance of oral tolerance163. Thus, selective blocking of the B7-1 interface in the PD-L1—B7-1 interaction offers exciting therapeutic directions.
Of note, almost all PD-L1 therapeutic and phenotyping antibodies target the PD-1 binding region of PD-L1. Since this region is masked by the PD-L1—B7-1 interaction, this can result in the underestimation of PD-L1 levels in flow cytometry and immunohistochemistry.
Extracellular PD-L1 exists in both secreted and exosomal forms, with elevated circulating levels identified in malignancy, pregnancy, and infection164. Secreted PD-L1 can result from splice variants or cleavage from the cell surface by metalloproteinases in tumor cells165,166 or activated DC in an inflammatory milieu166–168. Some splice variants of PD-L1 contain a stop codon before the transmembrane domain, leading to a secreted form with both monomer and covalent homodimers identified165,166. The homodimeric form can inhibit T lymphocytes, as PD-1 inhibition requires multimeric crosslinking of PD-1 with the TCR. Another secreted form of PD-L1 shed through proteolytic cleavage by metalloproteinase has unknown function168.
Exosomes are extracellular vesicles shed from eukaryotic cells that can communicate with the extracellular environment via their target protein, DNA, or RNA169,170. Both tumor and myeloid cells may produce exosomal PD-L1171,172. Exosomal PD-L1 may not always correlate with cell surface PD-L1169,173. High IFN-γ can increase both secreted and exosomal PD-L1164,166, both of which have been proposed to act as a decoy for anti-PD-L1 antibody by sequestering the anti-PD-L1 antibody or attenuating anti-tumor immunity through inhibition of T cell function170,174,175.
High level of exosomal PD-L1 in head and neck cancer176, gastric cancer177, renal cell carcinoma167 and soluble PD-L1 in DLBCL178 was associated with worse prognosis, at least in part due to its association with larger tumors or more advanced disease. Notably, data on soluble PD-L1 as a biomarker for response to immunotherapy are mixed166,173,179,180. The specific type of extracellular PD-L1 may matter, as exosomal PD-L1 stratifies patients with metastatic melanoma on pembrolizumab (anti-PD-1) therapy into responders or non-responders while the other secreted forms of PD-L1 do not170. More work is needed to stratify extracellular PD-L1 forms as a therapeutic biomarker and whether in select cases, targeting exosomal secretion of PD-L1174,176 is a viable therapeutic approach.
Nuclear expression of PD-L1 has been reported in multiple cancers including renal cell, hepatocellular, esophageal, lung, colorectal and prostate carcinoma181,182 and following doxorubicin chemotherapy in breast cancer183. PD-L1 may translocate from the plasma membrane to the nucleus, where it can bind DNA and regulate gene expression, and sister chromatid cohesion and segregation in cancer cells184. Three mechanisms for this translocation from the cell surface to the nucleus have been proposed, including via the nuclear import protein KPNB1 in non-small lung carcinoma cells185, interaction with vimentin and importin proteins to enter the nucleus following deacetylation of its C-terminus by HDAC2 and promotion of clathrin-dependent endocytosis186, or hypoxia and gasdermin C through pSTAT3 interaction with the PD-L1 cytoplasmic domain187. Du et al. showed that once in the nucleus, PD-L1 can interact with the transcription factor Sp1 to stimulate Gas6 transcription and cell proliferation185. Gao et al. showed that nuclear PD-L1 can stimulate expression of NF-κB pathway and immunoregulatory genes including VISTA and B7-H3186. Together, nuclear PD-L1 has the potential to regulate tumor cell proliferation and immune evasion in multiple ways.
The PD-L2–RGMb interaction serves as a point of intersection between the PD-L2–PD-1 pathway and RGMb–BMP2–BMP4–BMPR and RGMb–neogenin–netrin complexes188,189 (Figure 3). RGMb is expressed on both hematopoietic and non-hematopoietic cells (Table 1). RGMb is a glycosylphosphatidylinositol (GPI)-anchored protein that can be expressed on the cell surface but is mainly localized intracellularly190. In addition to PD-L2, RGMb binds to bone morphogenic proteins 2 and 4 (BMP) and neogenin. RGMb does not directly signal but serves as a co-receptor with BMP for signaling through type 1 and type II BMP receptors or alternatively a RGMb dimer can dimerize neogenin191. RGMb and PD-1 bind to PD-L2 with a similar binding affinity (Kd 48.5 for PD-1-PD-L2 and Kd 58.8 for RGMb-PD-L2) and on distinct sites of PD-L2154. RGMb can simultaneously bind to PD-L2 and BMP, as they recognize distinct binding sites on RGMb154. Thus, PD-L2 may be able to engage in a tripartite complex binding both PD-1 and RGMb, as well as the RGMb supercomplex in trans to regulate signaling downstream. When on the same cell, RGMb can bring BMP and neogenin together wherein BMP acts as a bridge between two neogenin and RGMb dimers191 and activates signals downstream. RGMb also can be cleaved from the cell surface by phospholipases, resulting in a soluble form. RGMb, when shed from the cell surface, can bind to neogenin and facilitate neogenin dimerization. In addition, RGMb can bind to the soluble monomeric form of CTLA-4, but not the dimeric membrane form of CTLA-4. RGMb increases binding of soluble CTLA-4 to B7-1, enabling it to block B7-1 interaction with CD28 more effectively192. How this sequestration of B7-1 alters known interactions with PD-L1 remains to be investigated.
Figure 3: Novel interactions of PD-L2 with PD-1 or the BMPR–BMP2/4–RGMb–neogenin complex.
PD-L2 can interact with PD-1, leading to phosphorylation of the PD-1 ITIM and ITSM tyrosines, recruitment and activation of SHP-2, and inhibition of T cell activation. RGMb serves as a co-receptor for BMPs for the type I and type II BMP receptors. The BMPR–BMP2/4–RGMb interaction results in phosphorylation of the BMP receptors and activation of the MAPK, ERK, and SMAD1/5/8 pathways. An RGMb dimer can bridge two Neogenin molecules, forming a RGMb-neogenin complex. RGMb bridging of neogenin engages Rho pathway signaling and cytoskeletal rearrangement. PD-L2 can also interact with RGMb in the BMPR-BMP2-RGMb-neogenin supercomplex and this interaction results in respiratory and mucosal tolerance.
Although RGMb was initially identified in the context of neuronal cell differentiation during brain development, its immunoregulatory roles are now appreciated. RGMb-BMP interactions inhibit IL-6 secretion by macrophages via p38 and MAPK signaling193. In models of airway hyperreactivity, RGMb interactions with either neogenin or PD-L2 regulate respiratory tolerance. Blockade of PD-L2 interactions on lung DC with RGMb+ macrophages and eosinophils using an anti-PD-L2 mAb that selectively inhibits the PD-L2-RGMb interaction (but not PD-1—PD-L2) abrogated respiratory tolerance induced by exposure to ovalbumin prior to T cell priming154. Likewise, a soluble mutated form of PD-L2 capable of only binding to RGMb but not PD-1 reduced airway hyperreactivity and lung inflammation194. However, a separate role for RGMb-neogenin interactions independent of PD-L2 has also been shown. Anti-RGMb mAb inhibited the expansion of RGMb+ interstitial macrophages and eosinophils and reduced IL-25 production from RGMb+ macrophages and eosinophils in a neogenin-dependent and PD-L2 - independent manner190. These findings identify RGMb—neogenin interaction as an attractive therapeutic target for IL-25 driven diseases, including allergen or rhinovirus-induced airway hyperreactivity. Furthermore, the expression of neogenin on RGMb+ macrophages and surrounding epithelium suggests that interactions between RGMb and neogenin may occur in both cis and trans.
RGMb is also expressed in gut epithelium. An anti-RGMb antibody that blocked RGMb interaction with BMP2/4 and neogenin reduced GVHD and colitis195. In contrast, blockade of the RGMb binding site on PD-L2 worsened GVHD. RGMb deficient mice showed increased severity of colitis196, perhaps due to the elimination of RGMb interaction with its binding partners. In addition, the gut microbiota influences PD-L2 and RGMb expression. Blockade of PD-L2—RGMb interactions can overcome microbiota-dependent resistance to anti-PD-L1 therapy in tumor-bearing germ-free, antibiotic treated mice, or mice reconstituted with microbiota from patients who did not respond to anti-PD-1 treatment. Intriguingly, this study identified a novel function for RGMb on T cells and showed that interaction in trans between PD-L2 on APC and RGMb on T cells inhibited CD8+ T function197. Altogether, these findings define mechanisms by which RGMb regulates gut inflammation or anti-tumor immunity through interaction with ligands on hematopoietic and non-hematopoietic cells. Further work is needed to understand the functional consequences of RGMb interactions with its respective binding partners and how to optimize strategies for designing RGMb therapeutics.
We next describe other B7 molecules that have a co-stimulatory or co-inhibitory function (Table 1). Here we focus on HHLA2, VISTA, BTNL2 and BTN3A1 and refer the reader to recent reviews on ICOSL198, B7-H3199 , and B7-H4200.
HHLA2
HHLA2 (Human endogenous retrovirus-H long terminal repeat-associating 2) is longer than other B7 family members having the usual IgV-IgC but followed by an extra IgC and transmembrane and cytoplasmic domains201. HHLA2 and its ligand orthologs are present in humans, monkeys, and many other mammals, but not in rodents202,203. HHLA2 is found in various tissues, cancer cells and immune cells (Table 1)201–205. PD-L1 and HHLA2 are differentially regulated, with interferons and hypoxia being important for PD-L1 upregulation, but not HHLA2. A moderate percentage of lung, renal, cholangiocarcinoma, and prostate tumors express PD-L1 but a higher percentage express HHLA2. Few tumors co-express both, suggesting non-redundant pathways of immune evasion205–210. HHLA2 deficiency inhibits tumorigenic pathways such as EGFR/MAPK/ERK pathway in NSCLC211 and JAK/STAT in hepatocellular cancer212. HHLA2 has been defined as a prognostic marker in multiple cancers213–216.
HHLA2 can deliver a dominant inhibitory signal that impedes proliferation and cytokine secretion in activated T cells201,217 through KIR3DL3 (killer cell immunoglobulin-like receptor, three immunoglobulin domains and long cytoplasmic tail 3)209,218. KIR3DL3 contains three IgV domains, a transmembrane domain, and a cytoplasmic tail with a single ITIM motif. The KIR3DL3-HHLA2 pathway delivers a co-inhibitory signal to NK cells and T cells. KIR3DL3+ CD8+ T cells and KIR3DL3+ NK cells had reduced cytotoxicity for HHLA2+ targets209,218. Recruitment of SHP1 and SHP2 to the KIR3DL3 ITIM motif mediated the inhibitory effect by reducing ERK1/2 protein kinase and NF-κB signaling218. Antibody blockade of KIR3DL3 on NK cells reduced tumor burden in a lung metastasis model218.
TMIGD2 (Transmembrane and Immunoglobulin Domain Containing 2), a member of the CD28 family, is the co-stimulatory receptor for HHLA2202,203. TMIGD2 has a single IgV, transmembrane, and cytoplasmic domain but lacks the juxta-membrane disulfide that dimerizes CD28 and CTLA-4203. TMIGD2 is expressed on naive T cells and many NK cells but expression declines on activated and memory T cells202. Engagement of TMIGD2 by HHLA2 transmits a costimulatory signal to T202 and NK cells219 via AKT-dependent signaling. Further work is needed to understand contexts when TMIGD2 costimulation and KIR3DL3 coinhibition are most important.
VISTA
VISTA contains a single large IgV domain with homology to PD-L1 but no IgC domain. VISTA has several unique features including an extended C-C’ loop and an extra Beta-strand, H, with a disulfide bond that clamps the structure so the histidine-rich CDR-like loops are positioned facing sideways rather than the more usual upper position220,221. The cytoplasmic domain of VISTA contains multiple casein kinase 2 and phospho-kinase C sites but no tyrosine signaling motifs, suggesting that it can signal into VISTA expressing cells222. VISTA is most abundantly expressed on myeloid and DC, but relatively less on T cells (Table 1).
The abundance of histidine residues in its extracellular domain allows VISTA to have pH-dependent interactions with its ligands, since the histidines become charged as the extracellular pH declines from pH 7.4 to pH 6. Such a low pH is found in the tumor microenvironment, areas of the lymph node, and in healing wounds221. VISTA has five reported ligands, PSGL-1, Syndecan-2, LRIG-1 , VSIG8, and VSIG3, which have a ratio of binding at pH 6.0 to pH 7.4 of 125, 78, 9, 9, and 3, respectively223–227. PSGL-1 is expressed on T and B cells, myeloid cells, and dendritic cells228. PSGL-1 signaling can inhibit T cell proliferation and push them towards an exhausted phenotype229, although the interaction between VISTA and PSGL-1 was not explicitly shown to mediate this effect. VSIG3-VISTA and VSIG8-VISTA interactions inhibit T cell activation and effector function223,225.
VISTA can function as both ligand or receptor. VISTA is expressed at high levels in naive T cells to enforce quiescence and tolerance230. Expression of VISTA declines on activated T cell populations at later time points231. Higher expression of VISTA is found on tumor infiltrating macrophages, MDSC232–236 and tumor cells230. In synovial sarcoma, secretory factors from cancer cells such as VEGF can upregulate VISTA on endothelial cells237.
VISTA fusion protein inhibits proliferation and cytokine production by activated T cells during early stages of activation. Mice lacking VISTA develop arthritis and lupus-like autoimmune disease, due its function in tolerance231,235,238,239. VISTA induces Treg formation from human CD4+ T cells240. VISTA promotes suppression in myeloid cells and tolerogenic DC through inhibition of MAPK and NF-κB pathways upon TLR signaling241. In an imiquimod (IMQ)-induced mouse model of psoriasis, VISTA deficiency exacerbated disease due to increased IL-17 and IL-23 from DC242. In addition, VISTA agonist antibodies ameliorated autoimmunity, asthma, and GVHD in mouse models222,239,243,244.
Early studies with rat anti-mouse VISTA antibodies combined with anti PD-1 or anti-PD-L1 showed efficacy in multiple mouse tumor models233–236. Combined blockade resulted in increased inflammatory signatures in myeloid derived suppressor cells241, reduced differentiation to Tregs and increased T cell effector function234–236. Newer VISTA antibodies that block the interaction with PSGL-1 in a pH-selective fashion at pH 6.0 but not pH 7.4 had an added benefit with anti-PD-1 in mouse tumor models226,240,245. Such antibodies are concentrated in the acidic tumor microenvironment and not in major VISTA-expressing sites like the spleen. These pH-selective VISTA antibodies had an improved safety profile relative to non-pH selective antibodies in non-human primates227. Clinical development of pH-selective VISTA and other antibodies offers a novel strategy in cancer.
BTN3A1 and BTN2A1
BTN3A1 and BTN2A1 are members of the B7-like family of butyrophilins. Like other members of the butyrophilin family, they contain one IgV domain, one IgC domain, a transmembrane domain, a juxta transmembrane domain and a cytoplasmic domain containing a B30.2 (PRYSPRY) domain. BTN3A1 has three isoforms and is expressed on immune cells, tumor cells and endothelial cells246,247 (Table 1). BTN3A1 and BTN2A1 are a ligand for the γ9Vδ2 TCR in human γδ T cells. This interaction triggers activation of these T cells in response to transformed or stressed target cells248. Activated γ9Vδ2 T cells are cytotoxic and critical in immune responses in cancer, tuberculosis, or other diseases in an MHC-independent manner249,250. The B30.2 domains of BTN3A1 and BTN2A1 comprise an intracellular binding pocket for phosphorylated intermediates of the mevalonate pathway such as isopentyl pyrophosphate251,252. The mevalonate pathway feeds into cholesterol and other biosynthetic pathways and is overactive in cancer cells. This interaction is mediated through positively-charged amino acids in the B30.2 domain and negatively-charged amino acids in these phosphoantigens253. A recent study showed that these phosphoantigens “glue” B30.2 domains of BTN3A1 and BTN2A1 together, resulting in a conformational change that transmits an “inside-out” signal to the extracellular domains of the butyrophilins allowing them to engage the γ9Vδ2 TCR254,255 . Activation of the AMPK pathway can upregulate BTN3A1 and BTN2A1 cell surface expression in tumor cells, increasing γ9Vδ2 T cell cytotoxicity256. ICT101, a humanized agonistic antibody to BTN3A1/2/3 is in clinical trials in combination with anti-PD-1 for treatment of solid and hematologic malignancies257. Small molecule analogs of these phosphoantigens also have been developed for therapeutic use254,256.
BTNL2
BTNL2 belongs to the butyrophilin family and contains 4 Ig domains in an IgV-IgC-IgV-IgC arrangement followed by transmembrane and cytoplasmic domains258,259. Unlike other butyrophilin family members, BTNL2 does not contain a B30.2 domain in its cytoplasmic domain260. BTNL2 is expressed in the gut particularly on small intestinal epithelial cells as well as immune cells (Figure 1)258,259,261. The receptor of BTNL2 is not known, but studies with BTNL2-Fc inferred its expression on activated T cells and B cells259 (Table 1). BTNL2-Fc inhibited T cell proliferation, IL-2 production258,259,261 and promoted induced Treg development262. BTNL2 polymorphisms have been reported and confirmed to be associated with multiple autoimmune diseases including inflammatory bowel disease258,259, sarcoidosis263,264, rheumatoid arthritis265 and colon or prostate cancer266,267. Mice reconstituted with BTNL2 deficient bone marrow have lower survival in a mouse model of cerebral malaria268. BTNL2 has an important role in maintaining intestinal homeostasis. BTNL2 deficient mice had severe colitis as BTNL2 regulates the response of γδ T cells in intestinal epithelial lymphocytes269. BTNL2-Fc promotes secretion of IL-17 from γδ T cells or IL-22 from γδ T cells, CD4 and ILCs in the colon270. BTNL2 monoclonal antibody reduced tumor growth in mouse models of lung cancer, colon cancer and lymphoma261. In contrast, BTNL2-Fc imparted a protective effect in colitis while BTNL2 antibody reduced tumor growth in colitis induced colon tumorigenesis270. Higher levels of BTNL2 mRNA expression were associated with worse survival in patients with lung and colon adenocarcinoma261. While human BTNL2 has been detected with polyclonal antibodies in western blots and immunohistochemistry, no human mAbs have been reported, perhaps due to the difficulty of expressing human BTNL2, whose amino terminal sequence differs greatly from mouse.
Outstanding questions and concluding remarks
In summary, coinhibitory pathways in the B7—CD28 family play a crucial role in maintaining immune balance, protecting against infection, and preventing autoimmunity. These pathways not only regulate the activation of naïve T cells, but also control T cell fate decisions and function of effector, memory, and regulatory T cells. Coinhibitory signals are essential for tolerance and resolving inflammation, but their overactivity can contribute to the immunosuppressive environment found in tumors and chronic viral infections. Although our understanding of the immunoregulatory roles of coinhibitory pathways in the B7—CD28 family has advanced, these insights raise questions of fundamental and translational importance:
First, what are the ramifications of PD-L1—B7-1 and PD-L2—RGMb interactions? PD-1 and its ligands comprise a network rather than a linear pathway. PD-L1 bridges to the B7-1/B7-2/CD28 pathway and PD-L2 bridges to the RGMb/BMP2/BMP4/neogenin pathway. This knowledge opens new mechanistic insights into the efficacy of current immunotherapies as well as opportunities for designing new treatment strategies for cancer, autoimmunity, allergies and transplant rejection.
Second, how can the multifaceted roles of coinhibitory receptors in regulating different types of T cells be exploited for therapy? For example, PD-1 reinvigorates exhausted CD8+ T cells but can increase numbers and suppressive function of regulatory T cells. PD-1 bispecific antibodies that selectively target effector T cells may be more effective. Combination therapies that block PD-1 and deplete or attenuate Tregs also may enhance efficacy of PD-1 pathway blockade.
Third, what are the shared and unique molecular pathways triggered by coinhibitory receptors? Does co-expression of two or more receptors on T cells induce distinct signaling pathways compared to cells expressing one receptor? Transcriptional, proteomic and lipidomic approaches provide means to identify overlapping and distinct signaling nodes. Spatial technologies may localize receptor ligand interactions in specific tissues during cancer, autoimmunity and infections. Such studies could identify therapeutic synergy resulting from co-blockade on the same cell or different cells and inform synthetic biology strategies for targeting specific cellular interactions.
Fourth, what are the functions of coinhibitory receptors on non-T cells? The expression of these receptors on innate as well as adaptive immune cells points to a broader immunoregulatory role.
Fifth, the butyrophilin subfamily of the B7s has been perplexing but the recent finding that the B30.2 intracellular domain of the BTN3A1 butyrophilin binds intracellular ligands and transmits an “inside-out” signal to engage γδ cells reveals a new mechanism of immunoregulation. Do other intracellular ligands exist for butyrophilins and is this part of a broader form of regulation in other disease contexts?
The therapeutic potential of deeper understanding of coinhibitory pathways is underscored by FDA approval of CTLA-4, PD-1 and PD-L1 antibodies for cancer immunotherapy and recent development of PD-1 agonistic antibodies to treat autoimmune diseases. The answers to these questions will continue to illuminate our understanding of coinhibitory pathways in health and disease, paving the way for novel therapeutic strategies to effectively and safely treat a variety of immune-mediated disorders.
Acknowledgments:
We apologize to those whose papers were not cited due to space constraints. This work was supported by funding from NIH grants P01AI56299 and 5P50CA236749 (to A.H.S and G.J.F), P50CA101942 (to A.H.S., G.J.F., and K.P.B.), P01 AI108545 and 5P50CA127003 (to A.H.S) and the Barr Foundation (to K.P.B.). Figures were created using Biorender.com.
Footnotes
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 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.
Declarations of Interest:
A.H.S. has patents/pending royalties on the PD-1 pathway from Roche and Novartis. A.H.S. is on advisory boards for Elpiscience, Bicara, Monopteros, Fibrogen, Alixia, IOME, Corner Therapeutics, Bioentre, GlaxoSmithKline, Amgen, and Janssen. She also is on scientific advisory boards for the Massachusetts General Cancer Center, Program in Cellular and Molecular Medicine at Boston Children's Hospital, the Human Oncology and Pathogenesis Program at Memorial Sloan Kettering Cancer Center, the Johns Hopkins Bloomberg Kimmel Institute for Cancer Immunotherapy, and the Gladstone Institute and is an academic editor for the Journal of Experimental Medicine. A.H.S. has funding from Quark/Iome; AbbVie; and Taiwan Bio. GJF has patents/pending royalties on the PD-L1/PD-1 pathway from Roche, Merck MSD, Bristol-Myers-Squibb, Merck KGA, Boehringer-Ingelheim, AstraZeneca, Dako, Leica, Mayo Clinic, Eli Lilly, and Novartis. GJF has patents or patent applications on the use of PD-L1, PD-L2, PD-1, RGMb, HHLA2, KIR3DL3, and BTNL2 in cancer immunotherapy. GJF has served on advisory boards for iTeos, NextPoint, IgM, GV20, IOME, Bioentre, Santa Ana Bio, Simcere of America, and Geode. GJF has equity in Nextpoint, Triursus, Xios, iTeos, IgM, Trillium, Invaria, GV20, Bioentre, and Geode. The other authors declare no conflicts of interest.
References
- 1.Bretscher P, and Cohn M (1970). A theory of self-nonself discrimination. Science 169, 1042–1049. 10.1126/science.169.3950.1042. [DOI] [PubMed] [Google Scholar]
- 2.Cunningham AJ, and Lafferty KJ (1977). A simple conservative explanation of the H-2 restriction of interactions between lymphocytes. Scand J Immunol 6, 1–6. 10.1111/j.1365-3083.1977.tb00317.x. [DOI] [PubMed] [Google Scholar]
- 3.Lo B, Fritz JM, Su HC, Uzel G, Jordan MB, and Lenardo MJ (2016). CHAI and LATAIE: new genetic diseases of CTLA-4 checkpoint insufficiency. Blood 128, 1037–1042. 10.1182/blood-2016-04-712612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ogishi M, Yang R, Aytekin C, Langlais D, Bourgey M, Khan T, Ali FA, Rahman M, Delmonte OM, Chrabieh M, et al. (2021). Inherited PD-1 deficiency underlies tuberculosis and autoimmunity in a child. Nat Med 27, 1646–1654. 10.1038/s41591-021-01388-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, Lieb DJ, Chen JH, Frederick DT, Barzily-Rokni M, et al. (2018). Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 175, 998–1013.e1020. 10.1016/j.cell.2018.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, and Minato N (2002). Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 99, 12293–12297. 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, et al. (2006). PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354. 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
- 8.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, and Ahmed R (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687. 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
- 9.Esensten JH, Helou YA, Chopra G, Weiss A, and Bluestone JA (2016). CD28 Costimulation: From Mechanism to Therapy. Immunity 44, 973–988. 10.1016/j.immuni.2016.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Schildberg FA, Klein SR, Freeman GJ, and Sharpe AH (2016). Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. Immunity 44, 955–972. 10.1016/j.immuni.2016.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Borriello F, Sethna MP, Boyd SD, Schweitzer AN, Tivol EA, Jacoby D, Strom TB, Simpson EM, Freeman GJ, and Sharpe AH (1997). B7-1 and B7-2 have overlapping, critical roles in immunoglobulin class switching and germinal center formation. Immunity 6, 303–313. [DOI] [PubMed] [Google Scholar]
- 12.Edner NM, Carlesso G, Rush JS, and Walker LSK (2020). Targeting co-stimulatory molecules in autoimmune disease. Nat Rev Drug Discov 19, 860–883. 10.1038/s41573-020-0081-9. [DOI] [PubMed] [Google Scholar]
- 13.Panneton V, Chang J, Witalis M, Li J, and Suh WK (2019). Inducible T-cell co-stimulator: Signaling mechanisms in T follicular helper cells and beyond. Immunol Rev 291, 91–103. 10.1111/imr.12771. [DOI] [PubMed] [Google Scholar]
- 14.Molon B, Liboni C, and Viola A (2022). CD28 and chemokine receptors: Signalling amplifiers at the immunological synapse. Front Immunol 13, 938004. 10.3389/fimmu.2022.938004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bally AP, Austin JW, and Boss JM (2016). Genetic and Epigenetic Regulation of PD-1 Expression. J Immunol 196, 2431–2437. 10.4049/jimmunol.1502643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chamoto K, Yaguchi T, Tajima M, and Honjo T (2023). Insights from a 30-year journey: function, regulation and therapeutic modulation of PD1. Nat Rev Immunol 23, 682–695. 10.1038/s41577-023-00867-9. [DOI] [PubMed] [Google Scholar]
- 17.Sharpe AH, and Pauken KE (2018). The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18, 153–167. 10.1038/nri.2017.108. [DOI] [PubMed] [Google Scholar]
- 18.Finn PW, He H, Wang Y, Wang Z, Guan G, Listman J, and Perkins DL (1997). Synergistic induction of CTLA-4 expression by costimulation with TCR plus CD28 signals mediated by increased transcription and messenger ribonucleic acid stability. J Immunol 158, 4074–4081. [PubMed] [Google Scholar]
- 19.Gibson HM, Hedgcock CJ, Aufiero BM, Wilson AJ, Hafner MS, Tsokos GC, and Wong HK (2007). Induction of the CTLA-4 gene in human lymphocytes is dependent on NFAT binding the proximal promoter. J Immunol 179, 3831–3840. 10.4049/jimmunol.179.6.3831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Malquori L, Carsetti L, and Ruberti G (2008). The 3' UTR of the human CTLA4 mRNA can regulate mRNA stability and translational efficiency. Biochim Biophys Acta 1779, 60–65. 10.1016/j.bbagrm.2007.10.004. [DOI] [PubMed] [Google Scholar]
- 21.Skafi N, Fayyad-Kazan M, and Badran B (2020). Immunomodulatory role for MicroRNAs: Regulation of PD-1/PD-L1 and CTLA-4 immune checkpoints expression. Gene 754, 144888. 10.1016/j.gene.2020.144888. [DOI] [PubMed] [Google Scholar]
- 22.Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, and Sharpe AH (1995). Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3, 541–547. [DOI] [PubMed] [Google Scholar]
- 23.Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, and Mak TW (1995). Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270, 985–988. 10.1126/science.270.5238.985. [DOI] [PubMed] [Google Scholar]
- 24.Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, Bulashevska A, Petersen BS, Schaffer AA, Gruning BA, et al. (2014). Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20, 1410–1416. 10.1038/nm.3746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, Schickel JN, Tran DQ, Stoddard J, Zhang Y, et al. (2014). Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345, 1623–1627. 10.1126/science.1255904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, et al. (2015). AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349, 436–440. 10.1126/science.aaa1663. [DOI] [PubMed] [Google Scholar]
- 27.Serwas NK, Hoeger B, Ardy RC, Stulz SV, Sui Z, Memaran N, Meeths M, Krolo A, Yuce Petronczki O, Pfajfer L, et al. (2019). Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nat Commun 10, 3106. 10.1038/s41467-019-10812-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pawlak-Adamska E, Frydecka I, Bolanowski M, Tomkiewicz A, Jonkisz A, Karabon L, Partyka A, Nowak O, Szalinski M, and Daroszewski J (2017). CD28/CTLA-4/ICOS haplotypes confers susceptibility to Graves' disease and modulates clinical phenotype of disease. Endocrine 55, 186–199. 10.1007/s12020-016-1096-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mandelbrot DA, McAdam AJ, and Sharpe AH (1999). B7-1 or B7-2 is required to produce the lymphoproliferative phenotype in mice lacking cytotoxic T lymphocyte-associated antigen 4 (CTLA-4). J Exp Med 189, 435–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tai X, Van Laethem F, Sharpe AH, and Singer A (2007). Induction of autoimmune disease in CTLA-4-/- mice depends on a specific CD28 motif that is required for in vivo costimulation. Proc Natl Acad Sci U S A 104, 13756–13761. 10.1073/pnas.0706509104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, and Peach R (1994). Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1, 793–801. 10.1016/s1074-7613(94)80021-9. [DOI] [PubMed] [Google Scholar]
- 32.Greene JL, Leytze GM, Emswiler J, Peach R, Bajorath J, Cosand W, and Linsley PS (1996). Covalent dimerization of CD28/CTLA-4 and oligomerization of CD80/CD86 regulate T cell costimulatory interactions. J Biol Chem 271, 26762–26771. 10.1074/jbc.271.43.26762. [DOI] [PubMed] [Google Scholar]
- 33.Kennedy A, Waters E, Rowshanravan B, Hinze C, Williams C, Janman D, Fox TA, Booth C, Pesenacker AM, Halliday N, et al. (2022). Differences in CD80 and CD86 transendocytosis reveal CD86 as a key target for CTLA-4 immune regulation. Nat Immunol 23, 1365–1378. 10.1038/s41590-022-01289-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ovcinnikovs V, Ross EM, Petersone L, Edner NM, Heuts F, Ntavli E, Kogimtzis A, Kennedy A, Wang CJ, Bennett CL, et al. (2019). CTLA-4-mediated transendocytosis of costimulatory molecules primarily targets migratory dendritic cells. Sci Immunol 4. 10.1126/sciimmunol.aaw0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Marangoni F, Zhakyp A, Corsini M, Geels SN, Carrizosa E, Thelen M, Mani V, Prüßmann JN, Warner RD, Ozga AJ, et al. (2021). Expansion of tumor-associated Treg cells upon disruption of a CTLA-4-dependent feedback loop. Cell 184, 3998–4015.e3919. 10.1016/j.cell.2021.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tekguc M, Wing JB, Osaki M, Long J, and Sakaguchi S (2021). Treg-expressed CTLA-4 depletes CD80/CD86 by trogocytosis, releasing free PD-L1 on antigen-presenting cells. Proc Natl Acad Sci U S A 118. 10.1073/pnas.2023739118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Alegre ML, and Puccetti P (2003). Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 4, 1206–1212. 10.1038/ni1003. [DOI] [PubMed] [Google Scholar]
- 38.Chuang E, Fisher TS, Morgan RW, Robbins MD, Duerr JM, Vander Heiden MG, Gardner JP, Hambor JE, Neveu MJ, and Thompson CB (2000). The CD28 and CTLA-4 receptors associate with the serine/threonine phosphatase PP2A. Immunity 13, 313–322. 10.1016/s1074-7613(00)00031-5. [DOI] [PubMed] [Google Scholar]
- 39.Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, et al. (2004). Role of LAG-3 in regulatory T cells. Immunity 21, 503–513. 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
- 40.Joller N, Lozano E, Burkett PR, Patel B, Xiao S, Zhu C, Xia J, Tan TG, Sefik E, Yajnik V, et al. (2014). Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40, 569–581. 10.1016/j.immuni.2014.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pereira JA, Lanzar Z, Clark JT, Hart AP, Douglas BB, Shallberg L, O'Dea K, Christian DA, and Hunter CA (2023). PD-1 and CTLA-4 exert additive control of effector regulatory T cells at homeostasis. Front Immunol 14, 997376. 10.3389/fimmu.2023.997376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhang Q, Chikina M, Szymczak-Workman AL, Horne W, Kolls JK, Vignali KM, Normolle D, Bettini M, Workman CJ, and Vignali DAA (2017). LAG3 limits regulatory T cell proliferation and function in autoimmune diabetes. Sci Immunol 2. 10.1126/sciimmunol.aah4569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schmidt EM, Wang CJ, Ryan GA, Clough LE, Qureshi OS, Goodall M, Abbas AK, Sharpe AH, Sansom DM, and Walker LS (2009). Ctla-4 controls regulatory T cell peripheral homeostasis and is required for suppression of pancreatic islet autoimmunity. J Immunol 182, 274–282. 10.4049/jimmunol.182.1.274. [DOI] [PubMed] [Google Scholar]
- 44.Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, Nomura T, and Sakaguchi S (2008). CTLA-4 control over Foxp3+ regulatory T cell function. Science 322, 271–275. 10.1126/science.1160062. [DOI] [PubMed] [Google Scholar]
- 45.Klocke K, Sakaguchi S, Holmdahl R, and Wing K (2016). Induction of autoimmune disease by deletion of CTLA-4 in mice in adulthood. Proc Natl Acad Sci U S A 113, E2383–2392. 10.1073/pnas.1603892113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD, Arancibia-Carcamo CV, Sobel RA, Rudensky AY, Kuchroo VK, et al. (2015). Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med 212, 1603–1621. 10.1084/jem.20141030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wing JB, Ise W, Kurosaki T, and Sakaguchi S (2014). Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity 41, 1013–1025. 10.1016/j.immuni.2014.12.006. [DOI] [PubMed] [Google Scholar]
- 48.Sage PT, Paterson AM, Lovitch SB, and Sharpe AH (2014). The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 41, 1026–1039. 10.1016/j.immuni.2014.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li F, Zhao X, Zhang Y, Shao P, Ma X, Paradee WJ, Liu C, Wang J, and Xue HH (2021). T(FH) cells depend on Tcf1-intrinsic HDAC activity to suppress CTLA4 and guard B-cell help function. Proc Natl Acad Sci U S A 118. 10.1073/pnas.2014562118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, et al. (2013). Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med 210, 1695–1710. 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Arce Vargas F, Furness AJS, Litchfield K, Joshi K, Rosenthal R, Ghorani E, Solomon I, Lesko MH, Ruef N, Roddie C, et al. (2018). Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer Cell 33, 649–663 e644. 10.1016/j.ccell.2018.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, and Korman AJ (2013). Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res 1, 32–42. 10.1158/2326-6066.CIR-13-0013. [DOI] [PubMed] [Google Scholar]
- 53.Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, Wargo J, Allison JP, Ribas A, and Sharma P (2019). Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers. Clin Cancer Res 25, 1233–1238. 10.1158/1078-0432.CCR-18-0762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sato Y, Casson CN, Matsuda A, Kim JI, Shi JQ, Iwasaki S, Chen S, Modrell B, Chan C, Tavares D, et al. (2022). Fc-independent functions of anti-CTLA-4 antibodies contribute to anti-tumor efficacy. Cancer Immunol Immunother 71, 2421–2431. 10.1007/s00262-022-03170-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lax BM, Palmeri JR, Lutz EA, Sheen A, Stinson JA, Duhamel L, Santollani L, Kennedy A, Rothschilds AM, Spranger S, et al. (2023). Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc Natl Acad Sci U S A 120, e2300895120. 10.1073/pnas.2300895120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Knorr D, Leidner R, Jensen S, Meng R, Jones A, Ballesteros-Merino C, Bell RB, Baez M, Sprott D, Bifulco C, et al. (2023). FcyRIIB is a novel immune checkpoint in the tumor microenvironment limiting activity of Treg-targeting antibodies. bioRxiv 10.1101/2023.01.19.522856. [DOI] [PMC free article] [PubMed]
- 57.Zappasodi R, Serganova I, Cohen IJ, Maeda M, Shindo M, Senbabaoglu Y, Watson MJ, Leftin A, Maniyar R, Verma S, et al. (2021). CTLA-4 blockade drives loss of T(reg) stability in glycolysis-low tumours. Nature 591, 652–658. 10.1038/s41586-021-03326-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Khattri R, Auger JA, Griffin MD, Sharpe AH, and Bluestone JA (1999). Lymphoproliferative disorder in CTLA-4 knockout mice is characterized by CD28-regulated activation of Th2 responses. J Immunol 162, 5784–5791. [PubMed] [Google Scholar]
- 59.Yang Y, Li X, Ma Z, Wang C, Yang Q, Byrne-Steele M, Hong R, Min Q, Zhou G, Cheng Y, et al. (2021). CTLA-4 expression by B-1a B cells is essential for immune tolerance. Nature Communications 12, 525. 10.1038/s41467-020-20874-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Quandt D, Hoff H, Rudolph M, Fillatreau S, and Brunner-Weinzierl MC (2007). A new role of CTLA-4 on B cells in thymus-dependent immune responses in vivo. J Immunol 179, 7316–7324. 10.4049/jimmunol.179.11.7316. [DOI] [PubMed] [Google Scholar]
- 61.Pioli C, Gatta L, Ubaldi V, and Doria G (2000). Inhibition of IgG1 and IgE production by stimulation of the B cell CTLA-4 receptor. J Immunol 165, 5530–5536. 10.4049/jimmunol.165.10.5530. [DOI] [PubMed] [Google Scholar]
- 62.Haas KM (2023). Noncanonical B Cells: Characteristics of Uncharacteristic B Cells. J Immunol 211, 1257–1265. 10.4049/jimmunol.2200944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Do P, Beckwith KA, Cheney C, Tran M, Beaver L, Griffin BG, Mo X, Liu Y, Lapalombella R, Hertlein E, et al. (2019). Leukemic B Cell CTLA-4 Suppresses Costimulation of T Cells. J Immunol 202, 2806–2816. 10.4049/jimmunol.1801359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ahn E, Araki K, Hashimoto M, Li W, Riley JL, Cheung J, Sharpe AH, Freeman GJ, Irving BA, and Ahmed R (2018). Role of PD-1 during effector CD8 T cell differentiation. Proc Natl Acad Sci U S A 115, 4749–4754. 10.1073/pnas.1718217115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Youngblood B, Oestreich KJ, Ha SJ, Duraiswamy J, Akondy RS, West EE, Wei Z, Lu P, Austin JW, Riley JL, et al. (2011). Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8(+) T cells. Immunity 35, 400–412. 10.1016/j.immuni.2011.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Oestreich KJ, Yoon H, Ahmed R, and Boss JM (2008). NFATc1 regulates PD-1 expression upon T cell activation. J Immunol 181, 4832–4839. 10.4049/jimmunol.181.7.4832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Redd PS, Lu C, Klement JD, Ibrahim ML, Zhou G, Kumai T, Celis E, and Liu K (2018). H3K4me3 mediates the NF-kappaB p50 homodimer binding to the pdcd1 promoter to activate PD-1 transcription in T cells. Oncoimmunology 7, e1483302. 10.1080/2162402X.2018.1483302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, and Honjo T (2011). IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol 186, 2772–2779. 10.4049/jimmunol.1003208. [DOI] [PubMed] [Google Scholar]
- 69.Powell MD, Lu P, Neeld DK, Kania AK, George-Alexander LMM, Bally APR, Scharer CD, and Boss JM (2022). IL-6/STAT3 Signaling Axis Enhances and Prolongs Pdcd1 Expression in Murine CD8 T Cells. Immunohorizons 6, 872–882. 10.4049/immunohorizons.2100112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mathieu M, Cotta-Grand N, Daudelin JF, Thebault P, and Labrecque N (2013). Notch signaling regulates PD-1 expression during CD8(+) T-cell activation. Immunol Cell Biol 91, 82–88. 10.1038/icb.2012.53. [DOI] [PubMed] [Google Scholar]
- 71.Bally APR, Neeld DK, Lu P, Majumder P, Tang Y, Barwick BG, Wang Q, and Boss JM (2020). PD-1 Expression during Acute Infection Is Repressed through an LSD1-Blimp-1 Axis. J Immunol 204, 449–458. 10.4049/jimmunol.1900601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, Yates KB, Tsao HW, Godec J, LaFleur MW, Brown FD, et al. (2016). The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169. 10.1126/science.aae0491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen Z, Sen DR, Kurachi M, et al. (2016). Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165. 10.1126/science.aaf2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Agnellini P, Wolint P, Rehr M, Cahenzli J, Karrer U, and Oxenius A (2007). Impaired NFAT nuclear translocation results in split exhaustion of virus-specific CD8+ T cell functions during chronic viral infection. Proc Natl Acad Sci U S A 104, 4565–4570. 10.1073/pnas.0610335104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Staron MM, Gray SM, Marshall HD, Parish IA, Chen JH, Perry CJ, Cui G, Li MO, and Kaech SM (2014). The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8(+) T cells during chronic infection. Immunity 41, 802–814. 10.1016/j.immuni.2014.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Scott AC, Dundar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, Trivedi P, Menocal L, Appleby H, Camara S, et al. (2019). TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274. 10.1038/s41586-019-1324-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Alfei F, Kanev K, Hofmann M, Wu M, Ghoneim HE, Roelli P, Utzschneider DT, von Hoesslin M, Cullen JG, Fan Y, et al. (2019). TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269. 10.1038/s41586-019-1326-9. [DOI] [PubMed] [Google Scholar]
- 78.Seo H, Chen J, Gonzalez-Avalos E, Samaniego-Castruita D, Das A, Wang YH, Lopez-Moyado IF, Georges RO, Zhang W, Onodera A, et al. (2019). TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8(+) T cell exhaustion. Proc Natl Acad Sci U S A 116, 12410–12415. 10.1073/pnas.1905675116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yates KB, Tonnerre P, Martin GE, Gerdemann U, Al Abosy R, Comstock DE, Weiss SA, Wolski D, Tully DC, Chung RT, et al. (2021). Epigenetic scars of CD8(+) T cell exhaustion persist after cure of chronic infection in humans. Nat Immunol 22, 1020–1029. 10.1038/s41590-021-00979-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Abdel-Hakeem MS, Manne S, Beltra JC, Stelekati E, Chen Z, Nzingha K, Ali MA, Johnson JL, Giles JR, Mathew D, et al. (2021). Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat Immunol 22, 1008–1019. 10.1038/s41590-021-00975-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Soerens AG, Kunzli M, Quarnstrom CF, Scott MC, Swanson L, Locquiao JJ, Ghoneim HE, Zehn D, Youngblood B, Vezys V, and Masopust D (2023). Functional T cells are capable of supernumerary cell division and longevity. Nature 614, 762–766. 10.1038/s41586-022-05626-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sekine T, Perez-Potti A, Nguyen S, Gorin JB, Wu VH, Gostick E, Llewellyn-Lacey S, Hammer Q, Falck-Jones S, Vangeti S, et al. (2020). TOX is expressed by exhausted and polyfunctional human effector memory CD8(+) T cells. Sci Immunol 5. 10.1126/sciimmunol.aba7918. [DOI] [PubMed] [Google Scholar]
- 83.Okada M, Chikuma S, Kondo T, Hibino S, Machiyama H, Yokosuka T, Nakano M, and Yoshimura A (2017). Blockage of Core Fucosylation Reduces Cell-Surface Expression of PD-1 and Promotes Anti-tumor Immune Responses of T Cells. Cell Rep 20, 1017–1028. 10.1016/j.celrep.2017.07.027. [DOI] [PubMed] [Google Scholar]
- 84.Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, Liu H, Bai Y, Xue M, Hu R, et al. (2018). FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature 564, 130–135. 10.1038/s41586-018-0756-0. [DOI] [PubMed] [Google Scholar]
- 85.Wang X, He Q, Shen H, Xia A, Tian W, Yu W, and Sun B (2019). TOX promotes the exhaustion of antitumor CD8(+) T cells by preventing PD1 degradation in hepatocellular carcinoma. J Hepatol 71, 731–741. 10.1016/j.jhep.2019.05.015. [DOI] [PubMed] [Google Scholar]
- 86.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, et al. (2000). Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192, 1027–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, et al. (2001). PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2, 261–268. 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 88.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, and Vale RD (2017). T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433. 10.1126/science.aaf1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chan W, Cao YM, Zhao X, Schrom EC, Jia D, Song J, Sibener LV, Dong S, Fernandes RA, Bradfield CJ, et al. (2023). TCR ligand potency differentially impacts PD-1 inhibitory effects on diverse signaling pathways. J Exp Med 220. 10.1084/jem.20231242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Patsoukis N, Duke-Cohan JS, Chaudhri A, Aksoylar HI, Wang Q, Council A, Berg A, Freeman GJ, and Boussiotis VA (2020). Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD-1 dimerization and SHP-2 activation. Commun Biol 3, 128. 10.1038/s42003-020-0845-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, Qiu Y, Jussif JM, Carter LL, Wood CR, and Chaudhary D (2004). PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett 574, 37–41. 10.1016/j.febslet.2004.07.083. [DOI] [PubMed] [Google Scholar]
- 92.Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, Konieczny BT, Daugherty CZ, Koenig L, Yu K, et al. (2017). Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355, 1423–1427. 10.1126/science.aaf0683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Okazaki T, Maeda A, Nishimura H, Kurosaki T, and Honjo T (2001). PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A 98, 13866–13871. 10.1073/pnas.231486598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Rota G, Niogret C, Dang AT, Barros CR, Fonta NP, Alfei F, Morgado L, Zehn D, Birchmeier W, Vivier E, and Guarda G (2018). Shp-2 Is Dispensable for Establishing T Cell Exhaustion and for PD-1 Signaling In Vivo. Cell Rep 23, 39–49. 10.1016/j.celrep.2018.03.026. [DOI] [PubMed] [Google Scholar]
- 95.Christofides A, Katopodi XL, Cao C, Karagkouni D, Aliazis K, Yenyuwadee S, Aksoylar HI, Pal R, Mahmoud MAA, Strauss L, et al. (2023). SHP-2 and PD-1-SHP-2 signaling regulate myeloid cell differentiation and antitumor responses. Nat Immunol 24, 55–68. 10.1038/s41590-022-01385-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Boussiotis VA, and Patsoukis N (2022). Effects of PD-1 Signaling on Immunometabolic Reprogramming. Immunometabolism 4. 10.20900/immunometab20220007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Strauss L, Mahmoud MAA, Weaver JD, Tijaro-Ovalle NM, Christofides A, Wang Q, Pal R, Yuan M, Asara J, Patsoukis N, and Boussiotis VA (2020). Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 5. 10.1126/sciimmunol.aay1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wei F, Zhong S, Ma Z, Kong H, Medvec A, Ahmed R, Freeman GJ, Krogsgaard M, and Riley JL (2013). Strength of PD-1 signaling differentially affects T-cell effector functions. Proc Natl Acad Sci U S A 110, E2480–2489. 10.1073/pnas.1305394110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Pauken KE, Godec J, Odorizzi PM, Brown KE, Yates KB, Ngiow SF, Burke KP, Maleri S, Grande SM, Francisco LM, et al. (2020). The PD-1 Pathway Regulates Development and Function of Memory CD8+ T Cells following Respiratory Viral Infection. Cell Reports 31. 10.1016/j.celrep.2020.107827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Allie SR, Zhang W, Fuse S, and Usherwood EJ (2011). Programmed death 1 regulates development of central memory CD8 T cells after acute viral infection. J Immunol 186, 6280–6286. 10.4049/jimmunol.1003870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, and Honjo T (2005). Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A 102, 11823–11828. 10.1073/pnas.0505497102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Collier JL, Pauken KE, Lee CAA, Patterson DG, Markson SC, Conway TS, Fung ME, France JA, Mucciarone KN, Lian CG, et al. (2023). Single-cell profiling reveals unique features of diabetogenic T cells in anti-PD-1-induced type 1 diabetes mice. J Exp Med 220. 10.1084/jem.20221920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Odorizzi PM, Pauken KE, Paley MA, Sharpe A, and Wherry EJ (2015). Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J Exp Med 212, 1125–1137. 10.1084/jem.20142237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Im SJ, Konieczny BT, Hudson WH, Masopust D, and Ahmed R (2020). PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proc Natl Acad Sci U S A 117, 4292–4299. 10.1073/pnas.1917298117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, Yates KB, Lako A, Felt K, Naik GS, et al. (2019). Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 20, 326–336. 10.1038/s41590-019-0312-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, et al. (2006). Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med 12, 1198–1202. 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
- 107.Chang E, Pelosof L, Lemery S, Gong Y, Goldberg KB, Farrell AT, Keegan P, Veeraraghavan J, Wei G, Blumenthal GM, et al. (2021). Systematic Review of PD-1/PD-L1 Inhibitors in Oncology: From Personalized Medicine to Public Health. Oncologist 26, e1786–e1799. 10.1002/onco.13887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Nasti TH, Sharpe AH, et al. (2016). Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421. 10.1038/nature19330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, Casella V, Ngiow SF, Khan O, Huang YJ, et al. (2020). Developmental Relationships of Four Exhausted CD8(+) T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. Immunity 52, 825–841 e828. 10.1016/j.immuni.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, Carmona SJ, Scarpellino L, Gfeller D, Pradervand S, et al. (2019). Intratumoral Tcf1(+)PD-1(+)CD8(+) T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity 50, 195–211 e110. 10.1016/j.immuni.2018.12.021. [DOI] [PubMed] [Google Scholar]
- 111.Wang PH, Washburn RS, Mariuzza DL, Lin WW, Gill AL, Ahmed R, and Reiner SL (2023). Reciprocal transmission of activating and inhibitory signals and cell fate in regenerating T cells. Cell Rep 42, 113155. 10.1016/j.celrep.2023.113155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gill AL, Wang PH, Lee J, Hudson WH, Ando S, Araki K, Hu Y, Wieland A, Im S, Gavora A, et al. (2023). PD-1 blockade increases the self-renewal of stem-like CD8 T cells to compensate for their accelerated differentiation into effectors. Sci Immunol 8, eadg0539. 10.1126/sciimmunol.adg0539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Humblin E, Korpas I, Lu J, Filipescu D, van der Heide V, Goldstein S, Vaidya A, Soares-Schanoski A, Casati B, Selvan ME, et al. (2023). Sustained CD28 costimulation is required for self-renewal and differentiation of TCF-1(+) PD-1(+) CD8 T cells. Sci Immunol 8, eadg0878. 10.1126/sciimmunol.adg0878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Prokhnevska N, Cardenas MA, Valanparambil RM, Sobierajska E, Barwick BG, Jansen C, Reyes Moon A, Gregorova P, delBalzo L, Greenwald R, et al. (2023). CD8(+) T cell activation in cancer comprises an initial activation phase in lymph nodes followed by effector differentiation within the tumor. Immunity 56, 107–124 e105. 10.1016/j.immuni.2022.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wang Z, Wang S, Goplen NP, Li C, Cheon IS, Dai Q, Huang S, Shan J, Ma C, Ye Z, et al. (2019). PD-1(hi) CD8(+) resident memory T cells balance immunity and fibrotic sequelae. Sci Immunol 4. 10.1126/sciimmunol.aaw1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, Byrne A, Clarke K, Brown E, Teo ZL, et al. (2023). Intratumoral CD8(+) T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer. Cancer Cell 41, 585–601 e588. 10.1016/j.ccell.2023.01.004. [DOI] [PubMed] [Google Scholar]
- 117.Shwetank Abdelsamed, H.A., Frost EL, Schmitz HM, Mockus TE, Youngblood BA, and Lukacher AE (2017). Maintenance of PD-1 on brain-resident memory CD8 T cells is antigen independent. Immunol Cell Biol 95, 953–959. 10.1038/icb.2017.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, Tadros J, Ferretti AP, Liao S, Gurer C, et al. (2022). Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell 185, 2918–2935 e2929. 10.1016/j.cell.2022.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, Mog BJ, Hou W, Justesen S, Blosser R, et al. (2021). Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132. 10.1038/s41586-021-03752-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Luoma AM, Suo S, Williams HL, Sharova T, Sullivan K, Manos M, Bowling P, Hodi FS, Rahma O, Sullivan RJ, et al. (2020). Molecular Pathways of Colon Inflammation Induced by Cancer Immunotherapy. Cell 182, 655–671 e622. 10.1016/j.cell.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Weisberg SP, Carpenter DJ, Chait M, Dogra P, Gartrell-Corrado RD, Chen AX, Campbell S, Liu W, Saraf P, Snyder ME, et al. (2019). Tissue-Resident Memory T Cells Mediate Immune Homeostasis in the Human Pancreas through the PD-1/PD-L1 Pathway. Cell Rep 29, 3916–3932 e3915. 10.1016/j.celrep.2019.11.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Tan CL, Kuchroo JR, Sage PT, Liang D, Francisco LM, Buck J, Thaker YR, Zhang Q, McArdel SL, Juneja VR, et al. (2021). PD-1 restraint of regulatory T cell suppressive activity is critical for immune tolerance. J Exp Med 218. 10.1084/jem.20182232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Perry JA, Shallberg L, Clark JT, Gullicksrud JA, DeLong JH, Douglas BB, Hart AP, Lanzar Z, O'Dea K, Konradt C, et al. (2022). PD-L1-PD-1 interactions limit effector regulatory T cell populations at homeostasis and during infection. Nat Immunol 23, 743–756. 10.1038/s41590-022-01170-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, Sato E, Fukuoka S, Tada Y, Tanaka A, et al. (2019). PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A 116, 9999–10008. 10.1073/pnas.1822001116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kumagai S, Togashi Y, Kamada T, Sugiyama E, Nishinakamura H, Takeuchi Y, Vitaly K, Itahashi K, Maeda Y, Matsui S, et al. (2020). The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol 10.1038/s41590-020-0769-3. [DOI] [PubMed]
- 126.Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, Warmoes MO, de Cubas AA, MacIver NJ, Locasale JW, et al. (2016). Foxp3 and Toll-like receptor signaling balance T(reg) cell anabolic metabolism for suppression. Nat Immunol 17, 1459–1466. 10.1038/ni.3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Denize T, Jegede OA, Matar S, El Ahmar N, West DJ, Walton E, Bagheri AS, Savla V, Nabil Laimon Y, Gupta S, et al. (2023). PD-1 Expression on Intratumoral Regulatory T Cells is Associated with Lack of Benefit from Anti-PD-1 Therapy in Metastatic Clear Cell Renal Cell Carcinoma Patients. Clin Cancer Res 10.1158/1078-0432.CCR-23-2274. [DOI] [PMC free article] [PubMed]
- 128.Sage PT, Francisco LM, Carman CV, and Sharpe AH (2013). The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood. Nat Immunol 14, 152–161. 10.1038/ni.2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sage PT, Schildberg FA, Sobel RA, Kuchroo VK, Freeman GJ, and Sharpe AH (2018). Dendritic Cell PD-L1 Limits Autoimmunity and Follicular T Cell Differentiation and Function. J Immunol 200, 2592–2602. 10.4049/jimmunol.1701231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Good-Jacobson KL, Szumilas CG, Chen L, Sharpe AH, Tomayko MM, and Shlomchik MJ (2010). PD-1 regulates germinal center B cell survival and the formation and affinity of long-lived plasma cells. Nat Immunol 11, 535–542. 10.1038/ni.1877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Xiao X, Lao XM, Chen MM, Liu RX, Wei Y, Ouyang FZ, Chen DP, Zhao XY, Zhao Q, Li XF, et al. (2016). PD-1hi Identifies a Novel Regulatory B-cell Population in Human Hepatoma That Promotes Disease Progression. Cancer Discov 6, 546–559. 10.1158/2159-8290.CD-15-1408. [DOI] [PubMed] [Google Scholar]
- 132.Rao DA, Gurish MF, Marshall JL, Slowikowski K, Fonseka CY, Liu Y, Donlin LT, Henderson LA, Wei K, Mizoguchi F, et al. (2017). Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114. 10.1038/nature20810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Suzuki K, Tajima M, Tokumaru Y, Oshiro Y, Nagata S, Kamada H, Kihara M, Nakano K, Honjo T, and Ohta A (2023). Anti-PD-1 antibodies recognizing the membrane-proximal region are PD-1 agonists that can down-regulate inflammatory diseases. Sci Immunol 8, eadd4947. 10.1126/sciimmunol.add4947. [DOI] [PubMed] [Google Scholar]
- 134.Tuttle J, Drescher E, Simon-Campos JA, Emery P, Greenwald M, Kivitz A, Rha H, Yachi P, Kiley C, and Nirula A (2023). A Phase 2 Trial of Peresolimab for Adults with Rheumatoid Arthritis. N Engl J Med 388, 1853–1862. 10.1056/NEJMoa2209856. [DOI] [PubMed] [Google Scholar]
- 135.Nishimura H, Minato N, Nakano T, and Honjo T (1998). Immunological studies on PD-1 deficient mice: implication of PD-1 as a negative regulator for B cell responses. Int Immunol 10, 1563–1572. 10.1093/intimm/10.10.1563. [DOI] [PubMed] [Google Scholar]
- 136.Spurrier MA, Jennings-Gee JE, Daly CA, and Haas KM (2021). The PD-1 Regulatory Axis Inhibits T Cell-Independent B Cell Memory Generation and Reactivation. J Immunol 207, 1978–1989. 10.4049/jimmunol.2100336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.McKay JT, Egan RP, Yammani RD, Chen L, Shin T, Yagita H, and Haas KM (2015). PD-1 suppresses protective immunity to Streptococcus pneumoniae through a B cell-intrinsic mechanism. J Immunol 194, 2289–2299. 10.4049/jimmunol.1401673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Stojanovic A, Fiegler N, Brunner-Weinzierl M, and Cerwenka A (2014). CTLA-4 is expressed by activated mouse NK cells and inhibits NK Cell IFN-gamma production in response to mature dendritic cells. J Immunol 192, 4184–4191. 10.4049/jimmunol.1302091. [DOI] [PubMed] [Google Scholar]
- 139.Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault MC, Trevino TN, Azimi CS, Scheer AK, Randolph HE, Thompson TW, et al. (2018). Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest 128, 4654–4668. 10.1172/JCI99317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hasim MS, Marotel M, Hodgins JJ, Vulpis E, Makinson OJ, Asif S, Shih HY, Scheer AK, MacMillan O, Alonso FG, et al. (2022). When killers become thieves: Trogocytosed PD-1 inhibits NK cells in cancer. Sci Adv 8, eabj3286. 10.1126/sciadv.abj3286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Helou DG, Shafiei-Jahani P, Lo R, Howard E, Hurrell BP, Galle-Treger L, Painter JD, Lewis G, Soroosh P, Sharpe AH, and Akbari O (2020). PD-1 pathway regulates ILC2 metabolism and PD-1 agonist treatment ameliorates airway hyperreactivity. Nat Commun 11, 3998. 10.1038/s41467-020-17813-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jacquelot N, Seillet C, Wang M, Pizzolla A, Liao Y, Hediyeh-Zadeh S, Grisaru-Tal S, Louis C, Huang Q, Schreuder J, et al. (2021). Blockade of the co-inhibitory molecule PD-1 unleashes ILC2-dependent antitumor immunity in melanoma. Nat Immunol 22, 851–864. 10.1038/s41590-021-00943-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Oldenhove G, Boucquey E, Taquin A, Acolty V, Bonetti L, Ryffel B, Le Bert M, Englebert K, Boon L, and Moser M (2018). PD-1 Is Involved in the Dysregulation of Type 2 Innate Lymphoid Cells in a Murine Model of Obesity. Cell Rep 25, 2053–2060 e2054. 10.1016/j.celrep.2018.10.091. [DOI] [PubMed] [Google Scholar]
- 144.Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, et al. (2017). PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545, 495–499. 10.1038/nature22396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Klement JD, Redd PS, Lu C, Merting AD, Poschel DB, Yang D, Savage NM, Zhou G, Munn DH, Fallon PG, and Liu K (2023). Tumor PD-L1 engages myeloid PD-1 to suppress type I interferon to impair cytotoxic T lymphocyte recruitment. Cancer Cell 41, 620–636 e629. 10.1016/j.ccell.2023.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Yao S, Wang S, Zhu Y, Luo L, Zhu G, Flies S, Xu H, Ruff W, Broadwater M, Choi IH, et al. (2009). PD-1 on dendritic cells impedes innate immunity against bacterial infection. Blood 113, 5811–5818. 10.1182/blood-2009-02-203141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zhao J, Bang S, Furutani K, McGinnis A, Jiang C, Roberts A, Donnelly CR, He Q, James ML, Berger M, et al. (2023). PD-L1/PD-1 checkpoint pathway regulates hippocampal neuronal excitability and learning and memory behavior. Neuron 111, 2709–2726 e2709. 10.1016/j.neuron.2023.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wei SC, Sharma R, Anang NAS, Levine JH, Zhao Y, Mancuso JJ, Setty M, Sharma P, Wang J, Pe'er D, and Allison JP (2019). Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity 50, 1084–1098 e1010. 10.1016/j.immuni.2019.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. (2015). Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 373, 23–34. 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, Shabafrouz K, Ribi C, Cairoli A, Guex-Crosier Y, et al. (2019). Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol 16, 563–580. 10.1038/s41571-019-0218-0. [DOI] [PubMed] [Google Scholar]
- 151.Wei SC, Meijers WC, Axelrod ML, Anang NAS, Screever EM, Wescott EC, Johnson DB, Whitley E, Lehmann L, Courand PY, et al. (2020). A genetic mouse model recapitulates immune checkpoint inhibitor-associated myocarditis and supports a mechanism-based therapeutic intervention. Cancer Discov 10.1158/2159-8290.CD-20-0856. [DOI] [PMC free article] [PubMed]
- 152.Axelrod ML, Meijers WC, Screever EM, Qin J, Carroll MG, Sun X, Tannous E, Zhang Y, Sugiura A, Taylor BC, et al. (2022). T cells specific for alpha-myosin drive immunotherapy-related myocarditis. Nature 611, 818–826. 10.1038/s41586-022-05432-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Butte MJ, Keir ME, Phamduy TB, Sharpe AH, and Freeman GJ (2007). Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 27, 111–122. 10.1016/j.immuni.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Xiao Y, Yu S, Zhu B, Bedoret D, Bu X, Francisco LM, Hua P, Duke-Cohan JS, Umetsu DT, Sharpe AH, et al. (2014). RGMb is a novel binding partner for PD-L2 and its engagement with PD-L2 promotes respiratory tolerance. J Exp Med 211, 943–959. 10.1084/jem.20130790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Chaudhri A, Xiao Y, Klee AN, Wang X, Zhu B, and Freeman GJ (2018). PD-L1 Binds to B7-1 Only In Cis on the Same Cell Surface. Cancer Immunol Res 6, 921–929. 10.1158/2326-6066.Cir-17-0316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Sugiura D, Maruhashi T, Okazaki IM, Shimizu K, Maeda TK, Takemoto T, and Okazaki T (2019). Restriction of PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T cell responses. Science 364, 558–566. 10.1126/science.aav7062. [DOI] [PubMed] [Google Scholar]
- 157.Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, Xiao C, Bonorino C, Lu LF, Bui JD, and Hui E (2019). PD-L1:CD80 Cis-Heterodimer Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-4 Pathways. Immunity 51, 1059–1073 e1059. 10.1016/j.immuni.2019.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Garrett-Thomson SC, Massimi A, Fedorov EV, Bonanno JB, Scandiuzzi L, Hillerich B, Seidel RD 3rd, Love JD, Garforth SJ, Guha C, and Almo SC (2020). Mechanistic dissection of the PD-L1:B7-1 co-inhibitory immune complex. PLoS One 15, e0233578. 10.1371/journal.pone.0233578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zhao Y, Caron C, Chan YY, Lee CK, Xu X, Zhang J, Masubuchi T, Wu C, Bui JD, and Hui E (2023). cis-B7:CD28 interactions at invaginated synaptic membranes provide CD28 co-stimulation and promote CD8(+) T cell function and anti-tumor immunity. Immunity 56, 1187–1203 e1112. 10.1016/j.immuni.2023.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sugiura D, Okazaki IM, Maeda TK, Maruhashi T, Shimizu K, Arakaki R, Takemoto T, Ishimaru N, and Okazaki T (2022). PD-1 agonism by anti-CD80 inhibits T cell activation and alleviates autoimmunity. Nat Immunol 23, 399–410. 10.1038/s41590-021-01125-7. [DOI] [PubMed] [Google Scholar]
- 161.Paterson AM, Brown KE, Keir ME, Vanguri VK, Riella LV, Chandraker A, Sayegh MH, Blazar BR, Freeman GJ, and Sharpe AH (2011). The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol 187, 1097–1105. 10.4049/jimmunol.1003496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Yang J, Riella LV, Chock S, Liu T, Zhao X, Yuan X, Paterson AM, Watanabe T, Vanguri V, Yagita H, et al. (2011). The novel costimulatory programmed death ligand 1/B7.1 pathway is functional in inhibiting alloimmune responses in vivo. J Immunol 187, 1113–1119. 10.4049/jimmunol.1100056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Park JJ, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, Augustine MM, Yao S, Tsushima F, Narazaki H, Anand S, et al. (2010). B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 116, 1291–1298. 10.1182/blood-2010-01-265975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Daassi D, Mahoney KM, and Freeman GJ (2020). The importance of exosomal PDL1 in tumour immune evasion. Nat Rev Immunol 20, 209–215. 10.1038/s41577-019-0264-y. [DOI] [PubMed] [Google Scholar]
- 165.Mahoney KM, Shukla SA, Patsoukis N, Chaudhri A, Browne EP, Arazi A, Eisenhaure TM, Pendergraft WF 3rd, Hua P, Pham HC, et al. (2019). A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression. Cancer Immunol Immunother 68, 421–432. 10.1007/s00262-018-2282-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, Rodig S, Li J, Wu X, Butterfield LH, et al. (2017). Soluble PD-L1 as a Biomarker in Malignant Melanoma Treated with Checkpoint Blockade. Cancer Immunol Res 5, 480–492. 10.1158/2326-6066.CIR-16-0329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Frigola X, Inman BA, Lohse CM, Krco CJ, Cheville JC, Thompson RH, Leibovich B, Blute ML, Dong H, and Kwon ED (2011). Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res 17, 1915–1923. 10.1158/1078-0432.CCR-10-0250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Romero Y, Wise R, and Zolkiewska A (2020). Proteolytic processing of PD-L1 by ADAM proteases in breast cancer cells. Cancer Immunol Immunother 69, 43–55. 10.1007/s00262-019-02437-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, Montabana E, Lang UE, Fu Q, Fong L, and Blelloch R (2019). Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell 177, 414–427 e413. 10.1016/j.cell.2019.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, Yu Z, Yang J, Wang B, Sun H, et al. (2018). Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386. 10.1038/s41586-018-0392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Morrissey SM, and Yan J (2020). Exosomal PD-L1: Roles in Tumor Progression and Immunotherapy. Trends Cancer 6, 550–558. 10.1016/j.trecan.2020.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, Hayes JL, Lee K, Balaj L, Passaro C, et al. (2018). Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv 4, eaar2766. 10.1126/sciadv.aar2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Costantini A, Julie C, Dumenil C, Helias-Rodzewicz Z, Tisserand J, Dumoulin J, Giraud V, Labrune S, Chinet T, Emile JF, and Giroux Leprieur E (2018). Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. Oncoimmunology 7, e1452581. 10.1080/2162402X.2018.1452581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Gong B, Kiyotani K, Sakata S, Nagano S, Kumehara S, Baba S, Besse B, Yanagitani N, Friboulet L, Nishio M, et al. (2019). Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer. J Exp Med 216, 982–1000. 10.1084/jem.20180870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Sagawa R, Sakata S, Gong B, Seto Y, Takemoto A, Takagi S, Ninomiya H, Yanagitani N, Nakao M, Mun M, et al. (2022). Soluble PD-L1 works as a decoy in lung cancer immunotherapy via alternative polyadenylation. JCI Insight 7. 10.1172/jci.insight.153323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, and Whiteside TL (2018). Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin Cancer Res 24, 896–905. 10.1158/1078-0432.CCR-17-2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Fan Y, Che X, Qu J, Hou K, Wen T, Li Z, Li C, Wang S, Xu L, Liu Y, and Qu X (2019). Exosomal PD-L1 Retains Immunosuppressive Activity and is Associated with Gastric Cancer Prognosis. Ann Surg Oncol 26, 3745–3755. 10.1245/s10434-019-07431-7. [DOI] [PubMed] [Google Scholar]
- 178.Rossille D, Gressier M, Damotte D, Maucort-Boulch D, Pangault C, Semana G, Le Gouill S, Haioun C, Tarte K, Lamy T, et al. (2014). High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia 28, 2367–2375. 10.1038/leu.2014.137. [DOI] [PubMed] [Google Scholar]
- 179.Chmielewska I, Grenda A, Krawczyk P, Frak M, Kuznar Kaminska B, Mitura W, and Milanowski J (2023). The influence of plasma sPD-L1 concentration on the effectiveness of immunotherapy in advanced NSCLC patients. Cancer Immunol Immunother 10.1007/s00262-023-03552-x. [DOI] [PMC free article] [PubMed]
- 180.Liao G, Zhao Z, Qian Y, Ling X, Chen S, Li X, and Kong FS (2021). Prognostic Role of Soluble Programmed Death Ligand 1 in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front Oncol 11, 774131. 10.3389/fonc.2021.774131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Parra ER, Villalobos P, and Rodriguez-Canales J (2019). The Multiple Faces of Programmed Cell Death Ligand 1 Expression in Malignant and Nonmalignant Cells. Appl Immunohistochem Mol Morphol 27, 287–294. 10.1097/PAI.0000000000000602. [DOI] [PubMed] [Google Scholar]
- 182.Xiong W, Gao Y, Wei W, and Zhang J (2021). Extracellular and nuclear PD-L1 in modulating cancer immunotherapy. Trends Cancer 7, 837–846. 10.1016/j.trecan.2021.03.003. [DOI] [PubMed] [Google Scholar]
- 183.Ghebeh H, Lehe C, Barhoush E, Al-Romaih K, Tulbah A, Al-Alwan M, Hendrayani SF, Manogaran P, Alaiya A, Al-Tweigeri T, et al. (2010). Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: role of B7-H1 as an anti-apoptotic molecule. Breast Cancer Res 12, R48. 10.1186/bcr2605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Yu J, Qin B, Moyer AM, Nowsheen S, Tu X, Dong H, Boughey JC, Goetz MP, Weinshilboum R, Lou Z, and Wang L (2020). Regulation of sister chromatid cohesion by nuclear PD-L1. Cell Res 30, 590–601. 10.1038/s41422-020-0315-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Du W, Zhu J, Zeng Y, Liu T, Zhang Y, Cai T, Fu Y, Zhang W, Zhang R, Liu Z, and Huang JA (2021). KPNB1-mediated nuclear translocation of PD-L1 promotes non-small cell lung cancer cell proliferation via the Gas6/MerTK signaling pathway. Cell Death Differ 28, 1284–1300. 10.1038/s41418-020-00651-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Gao Y, Nihira NT, Bu X, Chu C, Zhang J, Kolodziejczyk A, Fan Y, Chan NT, Ma L, Liu J, et al. (2020). Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat Cell Biol 22, 1064–1075. 10.1038/s41556-020-0562-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Hou J, Zhao R, Xia W, Chang CW, You Y, Hsu JM, Nie L, Chen Y, Wang YC, Liu C, et al. (2020). PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol 22, 1264–1275. 10.1038/s41556-020-0575-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Samad TA, Rebbapragada A, Bell E, Zhang Y, Sidis Y, Jeong SJ, Campagna JA, Perusini S, Fabrizio DA, Schneyer AL, et al. (2005). DRAGON, a bone morphogenetic protein co-receptor. J Biol Chem 280, 14122–14129. 10.1074/jbc.M410034200. [DOI] [PubMed] [Google Scholar]
- 189.Conrad S, Stimpfle F, Montazeri S, Oldekamp J, Seid K, Alvarez-Bolado G, and Skutella T (2010). RGMb controls aggregation and migration of Neogenin-positive cells in vitro and in vivo. Mol Cell Neurosci 43, 222–231. 10.1016/j.mcn.2009.11.003. [DOI] [PubMed] [Google Scholar]
- 190.Yu S, Leung KM, Kim HY, Umetsu SE, Xiao Y, Albacker LA, Lee HJ, Umetsu DT, Freeman GJ, and DeKruyff RH (2019). Blockade of RGMb inhibits allergen-induced airways disease. J Allergy Clin Immunol 144, 94–108 e111. 10.1016/j.jaci.2018.12.1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Siebold C, Yamashita T, Monnier PP, Mueller BK, and Pasterkamp RJ (2017). RGMs: Structural Insights, Molecular Regulation, and Downstream Signaling. Trends Cell Biol 27, 365–378. 10.1016/j.tcb.2016.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Sekiya T, and Takaki S (2019). RGMB enhances the suppressive activity of the monomeric secreted form of CTLA-4. Sci Rep 9, 6984. 10.1038/s41598-019-43068-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Xia Y, Cortez-Retamozo V, Niederkofler V, Salie R, Chen S, Samad TA, Hong CC, Arber S, Vyas JM, Weissleder R, et al. (2011). Dragon (repulsive guidance molecule b) inhibits IL-6 expression in macrophages. J Immunol 186, 1369–1376. 10.4049/jimmunol.1002047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Nie X, Chen W, Zhu Y, Huang B, Yu W, Wu Z, Guo S, Zhu Y, Luo L, Wang S, and Chen L (2018). B7-DC (PD-L2) costimulation of CD4(+) T-helper 1 response via RGMb. Cell Mol Immunol 15, 888–897. 10.1038/cmi.2017.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Perez-Cruz M, Iliopoulou BP, Hsu K, Wu HH, Erkers T, Swaminathan K, Tang SW, Bader CS, Kambham N, Xie B, et al. (2022). Immunoregulatory effects of RGMb in gut inflammation. Front Immunol 13, 960329. 10.3389/fimmu.2022.960329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Shi Y, Zhong L, Li Y, Chen Y, Feng S, Wang M, Xia Y, and Tang S (2021). Repulsive Guidance Molecule b Deficiency Induces Gut Microbiota Dysbiosis and Increases the Susceptibility to Intestinal Inflammation in Mice. Front Microbiol 12, 648915. 10.3389/fmicb.2021.648915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Park JS, Gazzaniga FS, Wu M, Luthens AK, Gillis J, Zheng W, LaFleur MW, Johnson SB, Morad G, Park EM, et al. (2023). Targeting PD-L2-RGMb overcomes microbiome-related immunotherapy resistance. Nature 617, 377–385. 10.1038/s41586-023-06026-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Amatore F, Gorvel L, and Olive D (2020). Role of Inducible Co-Stimulator (ICOS) in cancer immunotherapy. Expert Opin Biol Ther 20, 141–150. 10.1080/14712598.2020.1693540. [DOI] [PubMed] [Google Scholar]
- 199.Getu AA, Tigabu A, Zhou M, Lu J, Fodstad O, and Tan M (2023). New frontiers in immune checkpoint B7-H3 (CD276) research and drug development. Mol Cancer 22, 43. 10.1186/s12943-023-01751-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Wang JY, and Wang WP (2020). B7-H4, a promising target for immunotherapy. Cell Immunol 347, 104008. 10.1016/j.cellimm.2019.104008. [DOI] [PubMed] [Google Scholar]
- 201.Zhao R, Chinai JM, Buhl S, Scandiuzzi L, Ray A, Jeon H, Ohaegbulam KC, Ghosh K, Zhao A, Scharff MD, and Zang X (2013). HHLA2 is a member of the B7 family and inhibits human CD4 and CD8 T-cell function. Proc Natl Acad Sci U S A 110, 9879–9884. 10.1073/pnas.1303524110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Zhu Y, Yao S, Iliopoulou BP, Han X, Augustine MM, Xu H, Phennicie RT, Flies SJ, Broadwater M, Ruff W, et al. (2013). B7-H5 costimulates human T cells via CD28H. Nat Commun 4, 2043. 10.1038/ncomms3043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Janakiram M, Chinai JM, Fineberg S, Fiser A, Montagna C, Medavarapu R, Castano E, Jeon H, Ohaegbulam KC, Zhao R, et al. (2015). Expression, Clinical Significance, and Receptor Identification of the Newest B7 Family Member HHLA2 Protein. Clin Cancer Res 21, 2359–2366. 10.1158/1078-0432.CCR-14-1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Mager DL, Hunter DG, Schertzer M, and Freeman JD (1999). Endogenous retroviruses provide the primary polyadenylation signal for two new human genes (HHLA2 and HHLA3). Genomics 59, 255–263. 10.1006/geno.1999.5877. [DOI] [PubMed] [Google Scholar]
- 205.Shigemura T, Perrot N, Huang Z, Bhatt RS, Sheshdeh AB, Ahmar NE, Ghandour F, Signoretti S, McDermott DF, Freeman GJ, and Mahoney KM (2023). Regulation of HHLA2 expression in kidney cancer and myeloid cells. BMC Cancer 23, 1039. 10.1186/s12885-023-11496-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Zhou QH, Li KW, Chen X, He HX, Peng SM, Peng SR, Wang Q, Li ZA, Tao YR, Cai WL, et al. (2020). HHLA2 and PD-L1 co-expression predicts poor prognosis in patients with clear cell renal cell carcinoma. J Immunother Cancer 8. 10.1136/jitc-2019-000157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Jing CY, Fu YP, Yi Y, Zhang MX, Zheng SS, Huang JL, Gan W, Xu X, Lin JJ, Zhang J, et al. (2019). HHLA2 in intrahepatic cholangiocarcinoma: an immune checkpoint with prognostic significance and wider expression compared with PD-L1. J Immunother Cancer 7, 77. 10.1186/s40425-019-0554-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Zhou Q, Li K, Lai Y, Yao K, Wang Q, Zhan X, Peng S, Cai W, Yao W, Zang X, et al. (2021). B7 score and T cell infiltration stratify immune status in prostate cancer. J Immunother Cancer 9. 10.1136/jitc-2021-002455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Bhatt RS, Berjis A, Konge JC, Mahoney KM, Klee AN, Freeman SS, Chen CH, Jegede OA, Catalano PJ, Pignon JC, et al. (2021). KIR3DL3 Is an Inhibitory Receptor for HHLA2 that Mediates an Alternative Immunoinhibitory Pathway to PD1. Cancer Immunol Res 9, 156–169. 10.1158/2326-6066.CIR-20-0315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Cheng H, Borczuk A, Janakiram M, Ren X, Lin J, Assal A, Halmos B, Perez-Soler R, and Zang X (2018). Wide Expression and Significance of Alternative Immune Checkpoint Molecules, B7x and HHLA2, in PD-L1-Negative Human Lung Cancers. Clin Cancer Res 24, 1954–1964. 10.1158/1078-0432.CCR-17-2924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Sun W, Li S, Tang G, Sun S, Luo Y, Bai R, Han L, Jiang X, Gao Y, Huang Z, et al. (2021). HHLA2 deficiency inhibits non-small cell lung cancer progression and THP-1 macrophage M2 polarization. Cancer Med 10, 5256–5269. 10.1002/cam4.4081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Guo H, Zhang C, Tang X, Zhang T, Liu Y, Yu H, Li Y, and Wang R (2022). HHLA2 Activates the JAK/STAT Signaling Pathway by Binding to TMIGD2 in Hepatocellular Carcinoma Cells. Inflammation 45, 1585–1599. 10.1007/s10753-022-01644-x. [DOI] [PubMed] [Google Scholar]
- 213.Ding L, Yu Q, Yang S, Yang WJ, Liu T, Xian JR, Tian TT, Li T, Chen W, Wang BL, et al. (2022). Comprehensive Analysis of HHLA2 as a Prognostic Biomarker and Its Association With Immune Infiltrates in Hepatocellular Carcinoma. Front Immunol 13, 831101. 10.3389/fimmu.2022.831101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Liao X, and Zhang D (2022). HHLA2 Immune Checkpoint Is a Novel Prognostic Predictor in Hepatocellular Carcinoma. Am J Clin Pathol 158, 62–69. 10.1093/ajcp/aqab221. [DOI] [PubMed] [Google Scholar]
- 215.Yang Z, Xu D, Ye X, Lin X, Zhang M, Su Y, Xie Q, and Ni W (2022). HHLA2 Used as a Potential Prognostic and Immunological Biomarker and Correlated with Tumor Microenvironment in Pan-Cancer. Biomed Res Int 2022, 3924400. 10.1155/2022/3924400. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 216.Qi Y, Deng G, Xu P, Zhang H, Yuan F, Geng R, Jiang H, Liu B, and Chen Q (2019). HHLA2 is a novel prognostic predictor and potential therapeutic target in malignant glioma. Oncol Rep 42, 2309–2322. 10.3892/or.2019.7343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Rieder SA, Wang J, White N, Qadri A, Menard C, Stephens G, Karnell JL, Rudd CE, and Kolbeck R (2021). B7-H7 (HHLA2) inhibits T-cell activation and proliferation in the presence of TCR and CD28 signaling. Cell Mol Immunol 18, 1503–1511. 10.1038/s41423-020-0361-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Wei Y, Ren X, Galbo PM Jr., Moerdler S, Wang H, Sica RA, Etemad-Gilbertson B, Shi L, Zhu L, Tang X, et al. (2021). KIR3DL3-HHLA2 is a human immunosuppressive pathway and a therapeutic target. Sci Immunol 6. 10.1126/sciimmunol.abf9792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Zhuang X, and Long EO (2019). CD28 Homolog Is a Strong Activator of Natural Killer Cells for Lysis of B7H7(+) Tumor Cells. Cancer Immunol Res 7, 939–951. 10.1158/2326-6066.CIR-18-0733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Mehta N, Maddineni S, Mathews II, Andres Parra Sperberg R, Huang PS, and Cochran JR (2019). Structure and Functional Binding Epitope of V-domain Ig Suppressor of T Cell Activation. Cell Rep 28, 2509–2516 e2505. 10.1016/j.celrep.2019.07.073. [DOI] [PubMed] [Google Scholar]
- 221.Yuan L, Tatineni J, Mahoney KM, and Freeman GJ (2021). VISTA: A Mediator of Quiescence and a Promising Target in Cancer Immunotherapy. Trends Immunol 42, 209–227. 10.1016/j.it.2020.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Flies DB, Wang S, Xu H, and Chen L (2011). Cutting edge: A monoclonal antibody specific for the programmed death-1 homolog prevents graft-versus-host disease in mouse models. J Immunol 187, 1537–1541. 10.4049/jimmunol.1100660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Wang J, Wu G, Manick B, Hernandez V, Renelt M, Erickson C, Guan J, Singh R, Rollins S, Solorz A, et al. (2019). VSIG-3 as a ligand of VISTA inhibits human T-cell function. Immunology 156, 74–85. 10.1111/imm.13001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Johnston RJ, Su LJ, Pinckney J, Critton D, Boyer E, Krishnakumar A, Corbett M, Rankin AL, Dibella R, Campbell L, et al. (2019). VISTA is an acidic pH-selective ligand for PSGL-1. Nature 574, 565–570. 10.1038/s41586-019-1674-5. [DOI] [PubMed] [Google Scholar]
- 225.Chen W, Qie C, Hu X, Wang L, Jiang J, Liu W, and Liu J (2022). A small molecule inhibitor of VSIG-8 prevents its binding to VISTA. Invest New Drugs 40, 690–699. 10.1007/s10637-022-01244-4. [DOI] [PubMed] [Google Scholar]
- 226.Smith FD, Pierce RH, Thisted T, and van der Horst EH (2023). Conditionally Active, pH-Sensitive Immunoregulatory Antibodies Targeting VISTA and CTLA-4 Lead an Emerging Class of Cancer Therapeutics. Antibodies (Basel) 12. 10.3390/antib12030055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Smith FD, Thisted T, Feng F, Kleschenko Y, Mukherjee A, Biesova Z, Takeuchi Y, Schreiber RD, van der Horst EH (2023). SNS-101, a clinical-stage pH-dependent and TME-selective anti-VISTA antibody with favorable pharmacokinetic and safety characteristics, promotes anti tumoral M1 macrophage polarization in PD-1 refractory settings. held in Milan, Italy, September 20, 2023.
- 228.Laszik Z, Jansen PJ, Cummings RD, Tedder TF, McEver RP, and Moore KL (1996). P-selectin glycoprotein ligand-1 is broadly expressed in cells of myeloid, lymphoid, and dendritic lineage and in some nonhematopoietic cells. Blood 88, 3010–3021. [PubMed] [Google Scholar]
- 229.Tinoco R, Carrette F, Barraza ML, Otero DC, Magana J, Bosenberg MW, Swain SL, and Bradley LM (2016). PSGL-1 Is an Immune Checkpoint Regulator that Promotes T Cell Exhaustion. Immunity 44, 1190–1203. 10.1016/j.immuni.2016.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.ElTanbouly MA, Croteau W, Noelle RJ, and Lines JL (2019). VISTA: a novel immunotherapy target for normalizing innate and adaptive immunity. Semin Immunol 42, 101308. 10.1016/j.smim.2019.101308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Wang L, Rubinstein R, Lines JL, Wasiuk A, Ahonen C, Guo Y, Lu LF, Gondek D, Wang Y, Fava RA, et al. (2011). VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med 208, 577–592. 10.1084/jem.20100619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, Zhao H, Chen J, Chen H, Efstathiou E, et al. (2017). VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med 23, 551–555. 10.1038/nm.4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Hong S, Yuan Q, Xia H, Zhu G, Feng Y, Wang Q, Zhang Z, He W, Lu J, Dong C, and Ni L (2019). Analysis of VISTA expression and function in renal cell carcinoma highlights VISTA as a potential target for immunotherapy. Protein Cell 10, 840–845. 10.1007/s13238-019-0642-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Guldner IH, Wang Q, Yang L, Golomb SM, Zhao Z, Lopez JA, Brunory A, Howe EN, Zhang Y, Palakurthi B, et al. (2020). CNS-Native Myeloid Cells Drive Immune Suppression in the Brain Metastatic Niche through Cxcl10. Cell 183, 1234–1248 e1225. 10.1016/j.cell.2020.09.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Liu J, Yuan Y, Chen W, Putra J, Suriawinata AA, Schenk AD, Miller HE, Guleria I, Barth RJ, Huang YH, and Wang L (2015). Immune-checkpoint proteins VISTA and PD-1 nonredundantly regulate murine T-cell responses. Proc Natl Acad Sci U S A 112, 6682–6687. 10.1073/pnas.1420370112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Le Mercier I, Chen W, Lines JL, Day M, Li J, Sergent P, Noelle RJ, and Wang L (2014). VISTA Regulates the Development of Protective Antitumor Immunity. Cancer Res 74, 1933–1944. 10.1158/0008-5472.CAN-13-1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Luk SJ, Schoppmeyer R, Ijsselsteijn ME, Somarakis A, Acem I, Remst DFG, Cox DT, van Bergen CAM, Briaire-de Bruijn I, Gronloh MLB, et al. (2023). VISTA Expression on Cancer-Associated Endothelium Selectively Prevents T-cell Extravasation. Cancer Immunol Res 11, 1480–1492. 10.1158/2326-6066.CIR-22-0759. [DOI] [PubMed] [Google Scholar]
- 238.ElTanbouly MA, Zhao Y, Nowak E, Li J, Schaafsma E, Le Mercier I, Ceeraz S, Lines JL, Peng C, Carriere C, et al. (2020). VISTA is a checkpoint regulator for naive T cell quiescence and peripheral tolerance. Science 367. 10.1126/science.aay0524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Han X, Vesely MD, Yang W, Sanmamed MF, Badri T, Alawa J, Lopez-Giraldez F, Gaule P, Lee SW, Zhang JP, et al. (2019). PD-1H (VISTA)-mediated suppression of autoimmunity in systemic and cutaneous lupus erythematosus. Sci Transl Med 11. 10.1126/scitranslmed.aax1159. [DOI] [PubMed] [Google Scholar]
- 240.Lines JL, Sempere LF, Broughton T, Wang L, and Noelle R (2014). VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res 2, 510–517. 10.1158/2326-6066.CIR-14-0072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Xu W, Dong J, Zheng Y, Zhou J, Yuan Y, Ta HM, Miller HE, Olson M, Rajasekaran K, Ernstoff MS, et al. (2019). Immune-Checkpoint Protein VISTA Regulates Antitumor Immunity by Controlling Myeloid Cell-Mediated Inflammation and Immunosuppression. Cancer Immunol Res 7, 1497–1510. 10.1158/2326-6066.CIR-18-0489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Li N, Xu W, Yuan Y, Ayithan N, Imai Y, Wu X, Miller H, Olson M, Feng Y, Huang YH, et al. (2017). Immune-checkpoint protein VISTA critically regulates the IL-23/IL-17 inflammatory axis. Sci Rep 7, 1485. 10.1038/s41598-017-01411-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Liu H, Li X, Hu L, Zhu M, He B, Luo L, and Chen L (2018). A crucial role of the PD-1H coinhibitory receptor in suppressing experimental asthma. Cell Mol Immunol 15, 838–845. 10.1038/cmi.2017.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Ceeraz S, Sergent PA, Plummer SF, Schned AR, Pechenick D, Burns CM, and Noelle RJ (2017). VISTA Deficiency Accelerates the Development of Fatal Murine Lupus Nephritis. Arthritis Rheumatol 69, 814–825. 10.1002/art.40020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Thisted T, Kleschenko Y, Takeuchi Y, Mukherjee A, Biesova Z, Cifuentes A, Pierce RH, Schreiber RD, van der Horst EH (2022). SNS-101, a monoclonal antibody that is highly selective for VISTA in the tumor microenvironment, demonstrates favorable pharmacokinetic and cytokine release characteristics and potentiates anti-PD-1 responsiveness held in New York, United States, 28 September 2022.
- 246.Compte E, Pontarotti P, Collette Y, Lopez M, and Olive D (2004). Frontline: Characterization of BT3 molecules belonging to the B7 family expressed on immune cells. Eur J Immunol 34, 2089–2099. 10.1002/eji.200425227. [DOI] [PubMed] [Google Scholar]
- 247.Payne KK, Mine JA, Biswas S, Chaurio RA, Perales-Puchalt A, Anadon CM, Costich TL, Harro CM, Walrath J, Ming Q, et al. (2020). BTN3A1 governs antitumor responses by coordinating alphabeta and gammadelta T cells. Science 369, 942–949. 10.1126/science.aay2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Harly C, Guillaume Y, Nedellec S, Peigne CM, Monkkonen H, Monkkonen J, Li J, Kuball J, Adams EJ, Netzer S, et al. (2012). Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human gammadelta T-cell subset. Blood 120, 2269–2279. 10.1182/blood-2012-05-430470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Chien YH, Meyer C, and Bonneville M (2014). gammadelta T cells: first line of defense and beyond. Annu Rev Immunol 32, 121–155. 10.1146/annurev-immunol-032713-120216. [DOI] [PubMed] [Google Scholar]
- 250.Gu S, Borowska MT, Boughter CT, and Adams EJ (2018). Butyrophilin3A proteins and Vgamma9Vdelta2 T cell activation. Semin Cell Dev Biol 84, 65–74. 10.1016/j.semcdb.2018.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Sandstrom A, Peigne CM, Leger A, Crooks JE, Konczak F, Gesnel MC, Breathnach R, Bonneville M, Scotet E, and Adams EJ (2014). The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vgamma9Vdelta2 T cells. Immunity 40, 490–500. 10.1016/j.immuni.2014.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Hsiao CH, Lin X, Barney RJ, Shippy RR, Li J, Vinogradova O, Wiemer DF, and Wiemer AJ (2014). Synthesis of a phosphoantigen prodrug that potently activates Vgamma9Vdelta2 T-lymphocytes. Chem Biol 21, 945–954. 10.1016/j.chembiol.2014.06.006. [DOI] [PubMed] [Google Scholar]
- 253.Gu S, Sachleben JR, Boughter CT, Nawrocka WI, Borowska MT, Tarrasch JT, Skiniotis G, Roux B, and Adams EJ (2017). Phosphoantigen-induced conformational change of butyrophilin 3A1 (BTN3A1) and its implication on Vgamma9Vdelta2 T cell activation. Proc Natl Acad Sci U S A 114, E7311–E7320. 10.1073/pnas.1707547114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Yuan L, Ma X, Yang Y, Qu Y, Li X, Zhu X, Ma W, Duan J, Xue J, Yang H, et al. (2023). Phosphoantigens glue butyrophilin 3A1 and 2A1 to activate Vgamma9Vdelta2 T cells. Nature 621, 840–848. 10.1038/s41586-023-06525-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Cano CE, Pasero C, De Gassart A, Kerneur C, Gabriac M, Fullana M, Granarolo E, Hoet R, Scotet E, Rafia C, et al. (2021). BTN2A1, an immune checkpoint targeting Vgamma9Vdelta2 T cell cytotoxicity against malignant cells. Cell Rep 36, 109359. 10.1016/j.celrep.2021.109359. [DOI] [PubMed] [Google Scholar]
- 256.Mamedov MR, Vedova S, Freimer JW, Sahu AD, Ramesh A, Arce MM, Meringa AD, Ota M, Chen PA, Hanspers K, et al. (2023). CRISPR screens decode cancer cell pathways that trigger gammadelta T cell detection. Nature 621, 188–195. 10.1038/s41586-023-06482-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.De Gassart A, Le KS, Brune P, Agaugue S, Sims J, Goubard A, Castellano R, Joalland N, Scotet E, Collette Y, et al. (2021). Development of ICT01, a first-in-class, anti-BTN3A antibody for activating Vgamma9Vdelta2 T cell-mediated antitumor immune response. Sci Transl Med 13, eabj0835. 10.1126/scitranslmed.abj0835. [DOI] [PubMed] [Google Scholar]
- 258.Arnett HA, Escobar SS, Gonzalez-Suarez E, Budelsky AL, Steffen LA, Boiani N, Zhang M, Siu G, Brewer AW, and Viney JL (2007). BTNL2, a butyrophilin/B7-like molecule, is a negative costimulatory molecule modulated in intestinal inflammation. J Immunol 178, 1523–1533. 10.4049/jimmunol.178.3.1523. [DOI] [PubMed] [Google Scholar]
- 259.Nguyen T, Liu XK, Zhang Y, and Dong C (2006). BTNL2, a butyrophilin-like molecule that functions to inhibit T cell activation. J Immunol 176, 7354–7360. 10.4049/jimmunol.176.12.7354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Rhodes DA, Reith W, and Trowsdale J (2016). Regulation of Immunity by Butyrophilins. Annu Rev Immunol 34, 151–172. 10.1146/annurev-immunol-041015-055435. [DOI] [PubMed] [Google Scholar]
- 261.Du Y, Peng Q, Cheng D, Pan T, Sun W, Wang H, Ma X, He R, Zhang H, Cui Z, et al. (2022). Cancer cell-expressed BTNL2 facilitates tumour immune escape via engagement with IL-17A-producing gammadelta T cells. Nat Commun 13, 231. 10.1038/s41467-021-27936-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Swanson RM, Gavin MA, Escobar SS, Rottman JB, Lipsky BP, Dube S, Li L, Bigler J, Wolfson M, Arnett HA, and Viney JL (2013). Butyrophilin-like 2 modulates B7 costimulation to induce Foxp3 expression and regulatory T cell development in mature T cells. J Immunol 190, 2027–2035. 10.4049/jimmunol.1201760. [DOI] [PubMed] [Google Scholar]
- 263.Tong X, Ma Y, Niu X, Yan Z, Liu S, Peng B, Peng S, and Fan H (2016). The BTNL2 G16071A gene polymorphism increases granulomatous disease susceptibility: A meta-analysis including FPRP test of 8710 participants. Medicine (Baltimore) 95, e4325. 10.1097/MD.0000000000004325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Valentonyte R, Hampe J, Huse K, Rosenstiel P, Albrecht M, Stenzel A, Nagy M, Gaede KI, Franke A, Haesler R, et al. (2005). Sarcoidosis is associated with a truncating splice site mutation in BTNL2. Nat Genet 37, 357–364. 10.1038/ng1519. [DOI] [PubMed] [Google Scholar]
- 265.Mitsunaga S, Hosomichi K, Okudaira Y, Nakaoka H, Kunii N, Suzuki Y, Kuwana M, Sato S, Kaneko Y, Homma Y, et al. (2013). Exome sequencing identifies novel rheumatoid arthritis-susceptible variants in the BTNL2. J Hum Genet 58, 210–215. 10.1038/jhg.2013.2. [DOI] [PubMed] [Google Scholar]
- 266.Lebrero-Fernandez C, Wenzel UA, Akeus P, Wang Y, Strid H, Simren M, Gustavsson B, Borjesson LG, Cardell SL, Ohman L, et al. (2016). Altered expression of Butyrophilin (BTN) and BTN-like (BTNL) genes in intestinal inflammation and colon cancer. Immun Inflamm Dis 4, 191–200. 10.1002/iid3.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Fitzgerald LM, Kumar A, Boyle EA, Zhang Y, McIntosh LM, Kolb S, Stott-Miller M, Smith T, Karyadi DM, Ostrander EA, et al. (2013). Germline missense variants in the BTNL2 gene are associated with prostate cancer susceptibility. Cancer Epidemiol Biomarkers Prev 22, 1520–1528. 10.1158/1055-9965.EPI-13-0345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Subramaniam KS, Spaulding E, Ivan E, Mutimura E, Kim RS, Liu X, Dong C, Feintuch CM, Zhang X, Anastos K, et al. (2015). The T-Cell Inhibitory Molecule Butyrophilin-Like 2 Is Up-regulated in Mild Plasmodium falciparum Infection and Is Protective During Experimental Cerebral Malaria. J Infect Dis 212, 1322–1331. 10.1093/infdis/jiv217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Panea C, Zhang R, VanValkenburgh J, Ni M, Adler C, Wei Y, Ochoa F, Schmahl J, Tang Y, Siao CJ, et al. (2021). Butyrophilin-like 2 regulates site-specific adaptations of intestinal gammadelta intraepithelial lymphocytes. Commun Biol 4, 913. 10.1038/s42003-021-02438-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Peng Q, Pan T, He R, Yi M, Feng L, Cui Z, Gao R, Wang H, Feng X, Li H, et al. (2023). BTNL2 promotes colitis-associated tumorigenesis in mice by regulating IL-22 production. EMBO Rep 24, e56034. 10.15252/embr.202256034. [DOI] [PMC free article] [PubMed] [Google Scholar]