Summary
Checkpoint blockade therapies that target inhibitory receptors on T cells have revolutionized clinical oncology. Antibodies targeting CTLA-4 or the PD-1/PD-L1 axis are now successfully used alone or in combination with chemotherapy for numerous tumor types. Despite the clinical success of checkpoint blockade therapies, tumors exploit multiple mechanisms to escape or subvert the anti-tumor T cell response. Within the tumor microenvironment, tumor-associated macrophages (TAM) can suppress T cell responses and facilitate tumor growth in various ways, ultimately debilitating clinical responses to T cell checkpoint inhibitors. There is therefore significant interest in identifying biologicals and drugs that target immunosuppressive TAM within the tumor microenvironment and can be combined with immune checkpoint inhibitors. Here we review approaches that are currently being evaluated to convert immunosuppressive TAM into immunostimulatory macrophages that promote T cell responses and tumor elimination.
Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment that impact anti-tumor immune responses and susceptibility to checkpoint blockade. TAMs are very heterogeneous and can be either immunosuppressive or immunostimulatory. Here, Molgora and Colonna review current strategies that aim to reprogram TAMs to enhance rather than inhibit immune responses.
Introduction
Macrophages provide a first line of defense during infections and other tissue insults through multiple mechanisms: a) phagocytosis of pathogens and damaged cells; b) production of bactericidal nitric oxide (NO) and reactive oxygen species (ROS); c) secretion of chemokines that recruit other immune cells; d) secretion of cytokines, such as IL-12, that activate CD8 T cells, TH1 cells and NK cells. Moreover, macrophages express Fc receptors for IgG that enable phagocytosis and killing of targets opsonized by IgG through antibody-dependent phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC) respectively 1,2. Conversely, macrophages also contribute to resolution of immune responses, tissue repair and restoration in several ways: a) secreting soluble factors that attenuate immune responses, such as IL-10, TGFβ, prostaglandin E2 (PGE2) and tryptophan catabolites generated by indoleamine 2,3-dioxygenase (IDO); b) releasing chemokines that attract Tregs; c) promoting angiogenesis through VGEF secretion; and d) producing metabolites generated by arginase I that promote wound healing 1,3. Although immune defense and tissue repair functions of macrophages were originally framed within the dual M1-M2 paradigm, it is now recognized that macrophage programs are complex and heterogeneous depending on context 1,4.
Beyond their contribution to host defense and tissue repair, macrophages are also one of the most abundant immune cell populations in human tumors and mouse tumor models 1,5,6. The origins, phenotypes and functions of tumor-associated macrophages (TAM) are heterogeneous, depending on the type of tumor and the microenvironment in which tumors develop 1. TAMs can derive from tissue-resident macrophages that seed tissues during fetal hematopoiesis and persist through self-renewal, as well as peripheral blood monocytes that infiltrate the tumor 7–14. Tumor-imprinted monocytes, known as monocyte-related myeloid-derived suppressor cells (M-MDSC), which differentiate into suppressive macrophages, have also been identified in patients and in tumor bearing mice 15. In general, the phenotypic and functional profile of TAMs parallel those observed during tissue repair, which promote suppression of anti-tumor immune responses, tumor growth, vascularization and metastatization 1 (Figure 1). In agreement with a predominant pro-tumorigenic impact, TAM infiltration is often associated with a poor prognosis and abbreviated survival for many different types of human cancer (e.g., breast, bladder, ovarian and gastric cancer and hematological malignancies) 16–20. Thus, therapeutic strategies have been devised to either limit macrophage recruitment into the tumor or deplete macrophages already present in it. Blockade of CCR2 can limit recruitment of monocytes into the tumor 6,21,22, whereas inhibitors of the receptor for CSF1 (CSF1R) and activation of the death receptor TRAIL-R impair maintenance and survival of TAMs within the tumor 23–25 (Figure 2). Clinical trials based on these strategies are ongoing, although their efficacy so far seems limited (see for review 1,5,26. Despite a predominant pro-tumorigenic impact, in certain contexts TAMs are capable of phagocytosing tumor cells, killing tumor cells through release of NO and ROS, secreting IL-12 and promoting anti-tumor CD8 T cell and Th1 responses, ultimately restricting tumor growth 26. Immunostimulatory and immunosuppressive TAMs can coexist within the same tumor. Thus, alternative therapeutic approaches have been designed that aim at reshaping the TAM landscape by converting the TAM profile from immunosuppressive into immunostimulatory. This goal can be achieved through various approaches: a) blockade of cell surface receptors on immunosuppressive TAMs, b) blockade of ligand-receptor pairs that inhibit phagocytosis, c) blockade of signaling and epigenetic mechanisms of TAM immunosuppression, and d) engagement of existing or artificially transduced activating receptors (Figure 2). In the following paragraphs we discuss prototypic target molecules for each of these strategies.
Figure 1. Pro-tumor and anti-tumor functions of TAMs.
Upper left. Immunosuppressive functions. TAMs can inhibit T cell, DC and NK cell activation through the immunosuppressive cytokines IL-10 and TGFβ; TAMs also suppress anti-tumor T cell functions through inhibitory receptor-ligand interactions, production of indoleamine 2,3-dioxygenase (IDO), release of arginase I catabolites and PGE2. Moreover, TAMs contribute to differentiation and recruitment of regulatory T cells (Tregs) via IL-10, TGFβ, CCL17 and CCL22.
Upper right. Immunostimulatory functions. TAMs can activate the anti-tumor immune response by recruiting and activating T cells, NK cells and DCs through the chemokines CXCL9 and CXCL10 and the pro-inflammatory cytokines IL-12, IL-15, IL-18. TAMs can eliminate Tregs via ADCC or ADCP during immunotherapy with monoclonal antibodies targeting Tregs.
Lower left. Direct pro-tumor functions. TAMs can promote resistance to therapy, angiogenesis, tumor cell survival and metastasization.
Lower right. Direct anti-tumor activities. TAMs can directly kill tumor cells via phagocytosis, antibody-dependent cellular phagocytosis (ADCP), antibody-dependent cellular cytotoxicity (ADCC), release of nitric oxide (NO) and reactive oxygen species (ROS).
Figure 2. Strategies to target tumor associated macrophages.
1) Inhibition of macrophage recruitment through blockade of CCL2-CCR2 axis; 2) Inhibition of macrophage differentiation and survival through blockade of CSF1-CSF1R axis and induction of TRAIL-driven apoptosis; 3) Macrophage reprogramming via blockade of inhibitory pathways or engagement of activation pathways; 4) Induction of macrophage-mediated phagocytosis through blockade of “don’t eat me” signals.
Cell surface receptors of immunosuppressive TAMs
Cell surface phenotypes, transcriptional profiles and functions of immunosuppressive TAMs are quite heterogeneous and change considerably depending on the tumor and the microenvironment. Notwithstanding, several cell surface molecules have been frequently associated with TAM immunosuppressive functions, including CD206 (MRC1), CD204 (MSR1), CX3CR1, MARCO, SIGLEC1, TREM2, CD9, CD63, PD-1 ligand (PD-L1), CD73, and MERTK (Figure 3). While some of these molecules are simply markers so far, others directly contribute to immunosuppression and have been targeted to reprogram TAMs into immune-stimulating cells (Figure 4). These latter include TREM2, MARCO, SIGLEC1, PD-L1 and CD73.
Figure 3. Macrophage distinct signatures.
Molecules associated with immunosuppressive (left), or immunostimulatory (right) signatures of TAMs.
Figure 4. Strategies to promote macrophage anti-tumor activities.
Left. Approaches to block immunosuppressive signals. Blocking mAbs targeting macrophage receptors SIGLEC-1, MARCO, TREM2, SIRP1a, SIGLEC-10, LILRB1, LILRB2, PD-1, PD-L1; PI3K inhibitors; HDAC inhibitors.
Right. Approaches to enhance activating signals. TLR agonists; IFNγ; FcR-mediated ADCC; agonistic anti-CD40 mAbs; CARs; DECTIN1 signaling.
TREM2.
TREM2 is a cell surface receptor of the Ig-superfamily expressed on many tissue macrophages that binds phospholipids, apoptotic cells and lipoproteins. TREM2 transmits intracellular signals through the adaptor molecule DAP12, which recruits the protein tyrosine kinase SYK. This signaling pathway activates multiple downstream mediators, including PI3K, PLCγ2, and MAPK, and cooperates with the CSF1R pathway 27–29. Metalloproteases regulate TREM2 surface expression by cleaving the ectodomain, which is released as soluble TREM2 30. TREM2 has been widely studied in Alzheimer’s disease because of its important role in sustaining the microglia response to brain accumulation of β-amyloid plaques 30. However, TREM2 is expressed in macrophages of various tissues in physiological and pathological conditions 30. In the adipose tissue, TREM2 is required to sustain a population of lipid-associated macrophages that prevent dysmetabolism caused by high-fat diet 31. In cirrhotic patients and a mouse model of non-alcoholic fatty liver disease, TREM2 is highly expressed in liver CD9+ macrophages, which resemble lipid-associated macrophages and protect hepatocytes from lipid accumulation 32–34. In atherosclerosis, TREM2 contributes to lipid catabolism in macrophages of atherosclerotic plaques 35. The expression of TREM2 has been recently reported in TAMs of cancer patients and mouse tumor models 36,37 and two studies showed that TREM2 sustains immunosuppressive TAMs 38,39. Using a novel technology that couples scRNAseq and intracellular protein measurements, one study identified a subset of TAMs in a model of MCA-induced sarcoma that coexpress the immunosuppressive marker arginase 1 (Arg1) and TREM2 38. This Arg1+Trem2+ subset was associated with tumor progression, whereas TREM2-deletion restricted tumor growth. In another study, TREM2-deletion and TREM2 blockade with a monoclonal antibody (mAb) reshaped the TAM landscape, facilitating T cell activation. The proportion of Mrc1+Cx3cr1+ TAMs declined, whereas the representation of Cd83+Cxcl9+ and Nos2+ TAMs increased. These changes in TAMs were associated with blunted tumor growth and an increased response to anti-PD-1 in models of sarcoma, colorectal cancer and breast cancer. Immunohistochemistry analysis of specimens from cancer patients revealed that TREM2 is specifically expressed by TAMs of many human tumor types (skin, liver, lung, breast, colon, stomach, bladder, kidney cancer, pancreatic cancer, lymphoma and glioma) 39. Moreover, high TREM2 expression in cohorts of patients with triple-negative breast carcinoma and colorectal cancer was associated with a negative prognosis. Together, these studies suggest that TREM2 blockade and, perhaps, TREM2 cleavage, can be promising approaches for TAM reprogramming. Whether TREM2 signaling promotes macrophage immunosuppression directly, by cooperating with CSF1R signaling, or through other mechanisms is currently unknown.
MARCO.
A member of the scavenger receptor family that is expressed by subsets of tissue macrophages, MARCO is upregulated upon bacterial infection and is expressed on TAMs in breast, colon cancer, endometrial cancer, melanoma and non-small cell lung cancer 40,41. TAMs expressing MARCO evinced a typical immunosuppressive signature in models of breast cancer, colon cancer and melanoma. Treatment with an anti-MARCO mAb was associated with improved tumor control and limited metastases in mouse models of melanoma and breast cancer 42. Combined blockade of MARCO and CTLA-4 was more efficient than single blockade of CTLA-4 in models of colon cancer and melanoma and was dependent on skewing of macrophage phenotypes 42. The mechanism by which anti-MARCO mAb impacts TAM function remains to be determined. However, MARCO+ TAMs were found close to tumor cell nests in non-small cell lung cancer patients, co-expressed other TAM immunosuppressive markers, such as PD-L1, CD163, and were associated with the expression of T cell inhibitory receptors and T cell inhibitory ligands such as PD-1, PD-L1, CTLA-4 and VISTA 40. Thus, combination of MARCO blockade with immunocheckpoint therapies might be a promising approach in clinical settings.
SIGLEC1.
SIGLEC1 (also known as CD169) is a phagocytic receptor of Ig-like lectin family that binds sialic acids and is expressed by normal macrophages and TAMs 43. In humans, SIGLEC1 expression correlated with poor disease prognosis in several cohorts and was one of the top genes with expression enriched in TAMs within breast cancer compared to macrophages from healthy breast tissue44. To address the impact of SIGLEC1+ macrophages, a recombinant mouse line has been developed in which the diphtheria toxin receptor is expressed under the control of the Siglec1 promoter, such that SIGLEC1+ macrophages can be eliminated in vivo by injection of diphtheria toxin. In a preclinical model of triple negative breast cancer, selective depletion of SIGLEC1+ macrophages was associated with improved tumor control and limited metastases, likely due to more robust CD8 T cell expansion45. In vitro, co-culture of breast cancer cell lines with macrophages heightened SIGLEC1 expression and induced PD-L1 expression through the JAK2 signaling pathway, bolstering their immunosuppressive function. While these studies demonstrate the immunosuppressive impact of SIGLEC1+ TAMs, it not yet clear that SIGLEC1 has a direct role the induction of immunosuppression.
Adenosine receptors.
Adenosine is mostly generated through the hydrolysis of adenosine triphosphate (ATP) released to the extracellular space by dying or stressed cells and immune cells 46. The cell membrane enzyme CD39 sequentially hydrolyzes ATP to adenosine diphosphate (ADP) and monophosphate (AMP); the ectonucleotidase CD73 then hydrolyzes AMP to adenosine, which signals through G-protein-coupled receptors (A1R, A2AR, A2BR, A3R). CD39, CD73 as well as adenosine receptors are widely expressed in both tumor cells and immune cells in the tumor microenvironment, including macrophages. Extracellular ATP promotes the inflammatory response in several conditions, whereas adenosine signaling has a broad anti-inflammatory effect 46. It was reported that adenosine could induce an “M2-like” differentiation of macrophages in the absence of other type 2 cytokines, such as IL-4 47. Moreover, A2B receptor signaling inhibited Interferon-γ (IFNγ) production in T cells, indirectly affecting macrophage polarization 48. Adenosine was also reported to suppress phagocytic activity, NO production and TNF expression of differentiated macrophages in vitro and in murine models of bacterial infections 49,50. In vitro inhibition of CD39 in macrophages promoted IL-1β production, possibly by preserving ATP and hindering adenosine accumulation 51. Along these lines, inhibition of CD73 with a siRNA emulsion given intranasally was associated with reduced glioblastoma growth and proportionally fewer CD206+ macrophages in a preclinical model 52. While the adenosine pathway has a broad immunosuppressive effect in the tumor microenvironment 53, specific and possibly inducible ablation of CD73, CD39 and adenosine receptor expression in macrophages will be essential to delineate the impact of adenosine on TAM function.
Ligand-receptor pairs that inhibit phagocytosis
Phagocytosis is triggered by “eat me” signals displayed on microorganisms and target cells that engage phagocytic receptors on macrophages. Conversely, healthy cells display “don’t eat me” signals that engage inhibitory receptors to prevent autoreactivity 54. Tumor cells often exploit “don’t eat me” signals to escape phagocytosis by macrophages. Thus, mAbs and other immunotherapeutic approaches have been developed to block “don’t eat me” signaling pathways and facilitate phagocytosis of tumor cells (Figure 4).
CD47 – SIRP1α pathway.
CD47 is a cell surface Ig-like molecule which is expressed in all cells. CD47 associates in cis with integrins, broadly contributing to migration, cell-cell fusion, activation, cytokine production, and phagocytosis. Moreover, CD47 acts as a “don’t eat me” signal by engaging in trans SIRP1α, a cell surface receptor that is constitutively expressed by macrophages, monocytes, neutrophils and some dendritic cells 55. The cytoplasmic domain of SIRP1α contains immunoreceptor tyrosine-based inhibitory motifs (ITIM). Upon binding CD47, SIRP1α ITIMs recruit the protein-tyrosine phosphatases SHP1 and SHP2, which transmit intracellular inhibitory signals 56. Among various targets, SIRP1α inhibits nonmuscle myosins that control cytoskeleton architecture, thereby blocking the formation of a phagocytic synapse between macrophages and tumor cells 56. In steady-state, CD47– SIRP1α interaction prevents phagocytosis of red blood cells by splenic macrophages 55. However, CD47 overexpression is a mechanism of immune evasion for human solid tumors (e.g. bladder and breast cancer) and hematological malignancies (e.g. myeloid leukemia and non-Hodgkin’s lymphoma) 57–60. Blockade of CD47 by mAbs in vitro restored macrophage phagocytosis of tumor cells 59,61,62. In vivo treatment of tumors with anti-CD47 mAbs in several humanized and syngeneic mouse models promoted phagocytosis of tumor cells along with immune activation and hence curbed tumor growth 61,63,64. MAbs and other molecules targeting the CD47-SIRP1α axis tested in clinical trials include mAbs blocking CD47 (Hu5F9-G4, SRF231, IBI188, AO-176), mAbs blocking SIRP1α (BI-765063), fusion proteins targeting CD47 (TTI-621, TTI-622, ALX148) (NCT03530683; NCT02663518), and bispecific mAbs targeting both CD47 and PD-1. Most of these treatments were well-tolerated and some (alone or in combination) showed a certain degree of efficacy in patients with ovarian cancer, fallopian tube cancer, acute myeloid leukemia, myelodysplastic syndrome (NCT02641002), B cell-lymphomas, cutaneous T cell lymphoma, late stage non-small cell lung cancer and gastric cancer (NCT02367196) 65–68. In addition to facilitating tumor cell phagocytosis, anti-CD47 mAb treatment promoted ADCP in a model of glioblastoma, as well as ADCC in models of ovarian and breast cancer 69–71. Accordingly, anti-CD47 mAbs with a functional Fc domain can be more efficacious than those with a mutated Fc lacking effector functions 68. Although current literature and clinical trials support the beneficial effects of targeting the CD47 – SIRP1α pathway, there are also some limitations. First, the broad expression of CD47 by different cell types, including red and white blood cells, can cause off-target sequestration of anti-CD47 mAbs and adverse effects like anemia, neutropenia and thrombocytopenia 66. In this regard, targeting SIRP1α instead of CD47 might be safer because of the restricted expression of SIRP1α on myeloid cells 72. Secondly, CD47 – SIRP1α blockade is not always sufficient to impact phagocytosis of tumor cells because of other “don’t eat me” signals coexpressed on tumor cells 54. Lastly, since it is ubiquitously expressed throughout the body, CD47 expression cannot be used to stratify patients and predict the response to anti-CD47 therapy 26. Recently, effective induction of phagocytosis of hematological tumors by CD47 blockade was shown to require concomitant adhesion of macrophages to hematopoietic tumor cells through SLAMF7 and integrin αMβ2 73. Thus, a better understanding of the complex mechanisms contributing to CD47-SIRP1α blockade might help identify patients who would benefit from this therapy.
MHC class I – LILRB1/2 pathway.
MHC class I (MHC-I) molecules can act as “don’t eat me” signals in cancer by interacting with two members of the human Leukocyte Ig-Like receptor (LILR) family, LILRB1 (also known as ILT2) and LILRB2 (also known as ILT4). LILRB1 and LILRB2 broadly recognize MHC-I molecules through two binding sites: one site interacts with the a3 domain of MHC-I heavy chain while another interacts with β2M 74,75. Because the MHC-I a3 domain and β2M are highly conserved across species, LILRB1 and LILB2 recognize both human classical (HLA-A, -B and -C) and non-classical (e.g. HLA-E, -G) MHC-I 76,77. LILRB1 and LILB2 contain cytoplasmic ITIMs that recruit SHP1 and SHP2, which mediate inhibition. LILRB1 is expressed by myeloid cells, B cells, subsets of CD8 T cells and NK cells; LILRB2 is only expressed on myeloid cells 78. In vitro studies demonstrated that deletion of MHC-I heavy chain or β2M in tumor cells or LILRB1 deletion in macrophages promotes tumor cell phagocytosis 79. In immunodeficient NOD-scid IL2Rgammanull mice, human MHC-I-deficient tumors were controlled more efficiently than MHC-I-expressing tumors. In C57BL/6 mice, macrophages more effectively controlled tumors deficient for both MHC-I and CD47 than did their CD47- and/or MHC-I- expressing counterparts 79. Thus, concomitant blockade of CD47 and MHC-I can be a promising approach to promote macrophage phagocytosis of tumor cells. In contrast to LILRB1 blockade, LILRB2 blockade did not enhance phagocytosis of tumor cells induced by anti-CD47 treatment in vitro 79. Perhaps LILRB2 blockade does not facilitate macrophage phagocytosis of tumor cells. However, anti-LILRB2 mAbs did enhance macrophage responses to LPS in vitro, inducing TNF upregulation and IL-10 downregulation 80. In a mouse model of lung cancer, anti-LILRB2 mAb treatment enhanced the response to immune checkpoint therapy and skewed TAMs toward an immunostimulatory phenotype 80. Thus, LILRB2 blockade may promote TAM reprogramming. The mouse counterparts of LILRs are known as paired Ig-like Receptors (PIRs) 81,82. However, PIRs include only one inhibitory receptor that recognizes MHC-I, PIRB, which is broadly expressed on hematopoietic cells. PIRB-deficiency was associated with reduced tumor growth in a lung cancer model. Myeloid cells isolated from tumor bearing mice had an “M1-like” signature in the absence of PIRB, characterized by increased expression of Nos2, Tnf, Il12 and Il1b and reduced expression of Arg1 and Il10. The discrepancy between the numbers of human and mouse genes encoding inhibitory LILRs and PIRs remains a challenge for translating mouse studies to humans; analysis of humanized mice may have to establish the impact of LILRB1 and LILRB2 in vivo 83.
CD24 – SIGLEC10 pathway.
CD24 is a highly glycosylated surface protein expressed by several solid tumors. CD24 is expressed on B cells, macrophages and dendritic cells and contains a cytoplasmic ITIM domain; it binds the inhibitory receptor SIGLEC10, a member of the Ig-like lectin family that binds sialic acids 84. The inhibitory effect of the CD24 – SIGLEC10 pathway has been documented in inflammation, adaptive immunity, autoimmune diseases and cancer 85–88. Recently, it was observed that CD24 is more abundantly expressed in most tumor types than other “don’t eat me” molecules (CD47, β2M) and is highly upregulated in ovarian cancer cells, triple negative, estrogen receptor- and progesterone receptor-positive breast cancer in comparison to the healthy tissue 89. Moreover, CD24 expression negatively correlated with disease prognosis and TAMs expressed high levels of SIGLEC10 89. CD24 deletion in tumor cell lines and mAb blockade of CD24 promoted phagocytosis of tumor cells in vitro; moreover, concomitant blockade of CD47 had a cooperative effect and enhanced phagocytosis. CD24-deficiency and CD24 targeting in tumor models in vivo facilitated control of tumor growth by TAMs 89. A clinical trial to test the safety and efficacy of CD24 targeting has been developed based on the administration of a fusion protein (CD24-Fc) in combination with anti-CTLA4 and anti-PD-1 in patients with metastatic melanoma, colon cancer and renal cell carcinoma (NCT04060407).
PD-1 – PD-L1 pathway.
PD-1 has been extensively characterized as a T cell inhibitory receptor induced by T cell activation that dampens effector functions 90. PD-1 ligand (PD-L1) expressed in tumor cells and immune cells in the tumor microenvironment has been shown to strongly inhibit T cells, thereby promoting tumor immune escape 90. Accordingly, PD-1–PD-L1 blockade has proved very efficacious in activating T cell responses in different cohorts of cancer patients 91,92. Interestingly, it was recently reported that PD-1 is not only expressed by T cells but also by other immune cells including human and murine TAMs, and hence may have T cell-independent functions 93. PD-1 expression in TAMs increased during tumor progression in murine models and cancer patients, and was negatively associated with macrophage phagocytic activity 93. Moreover, PD-L1 was also expressed by TAMs. PD-1–PD-L1 blockade in vivo led to improved macrophage-dependent tumor control and increased phagocytosis of tumor cells in the CT26 model of colorectal carcinoma and a model of osteosarcoma lung metastases 93,94. In murine models and patients with colorectal cancer, PD-1 expression in TAMs correlated with expression of markers of immune suppression (e.g. CD206) and weakened phagocytic potential 93. In vitro, anti-PD-L1 treatment promoted survival, proliferation and activation of both human and murine macrophages, corroborating that PD-1 – PD-L1 blockade can have a direct effect on macrophages 95. Further supporting this conclusion, anti-PD-1 treatment has been widely reported to affect macrophage function and polarization, mitigating their immunosuppressive phenotype 96. However, it should be noted that the concomitant restoration of T cell function and IFNγ production by PD-1 – PD-L1 blockade may have a considerable indirect effect on TAM phenotype, such that it is difficult to dissect macrophage-intrinsic from macrophage-extrinsic effects of anti-PD-1/PD-L1 blockade in many experimental settings.
Signaling and epigenetic mechanisms that promote immunosuppressive TAMs
Phosphoinositide-3-kinase gamma.
Phosphoinositide-3-kinase (PI3K) is a leukocyte-specific lipid kinase consisting of a regulatory and a catalytic subunit that phosphorylates phosphatidylinositol 4,5-bisphosphate to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3). PIP3 recruits PH domain-containing proteins such as AKT to the membrane, which activates signaling cascades involved in cell growth, survival, proliferation, motility and morphology. PI3K impacts macrophage differentiation and function 97. The gamma isoform of PI3K catalytic subunit (PI3Kγ), which is highly expressed in the tumor microenvironment, promotes a macrophage immunosuppressive phenotype 98. Genetic deletion or pharmacological inhibition of PI3Kγ in mouse tumor models broadly affected TAM number and phenotype: expression of MHCII and production of pro-inflammatory IL-12 increased, whereas expression of immunosuppressive markers (i.e. Arginase, CD206 and PD-L1) and secretion of anti-inflammatory IL-10 declined 99,100, as did VEGF production and consequent neoangiogenesis 100. TAM reprogramming was associated with enhanced T cell activation, T cell recruitment into the tumor, tumor control and response to anti-PD-1 in several models, including breast and lung cancer, as well as head and neck squamous cell carcinoma 99. Given the efficacy of PI3Kγ inhibitors in preclinical models, several strategies have been developed to inhibit PI3Kγ in the clinic. To date, ongoing clinical trials to target PI3Kγ have tested the PI3Kγ inhibitor IPI-549 alone or in combination with adenosine receptor antagonists, chemotherapy drugs (e.g. paclitaxel, doxorubicin), or anti-VEGF in triple negative breast cancer, ovarian and renal cancer patients. IPI-549 in combination with anti-PD-1 has been tested in non-small cell lung cancer, head and neck squamous cell carcinoma, triple negative breast cancer, bladder cancer and urothelial carcinoma (NCT03719326; NCT03961698; NCT02637531; NCT03980041). In a model of pancreatic cancer, it was observed that protein-tyrosine kinase BTK induced PI3Kγ activation and immunosuppression in TAMs. Blockade of either BTK or PI3Kγ was protective in the model and BTK blockade is currently under evaluation in a clinical trial in pancreatic adenocarcinoma patients (NCT02052492) 101.
Histone deacetylases (HDACs).
HDACs are enzymes that mediate epigenetic regulation of gene expression by removal of acetyl groups from an ε-N-acetyl lysine on histones 102. Abnormal HDAC expression patterns occur in several types of cancer and correlate with survival disadvantage in patients. HDAC blockade has been shown to be a promising strategy to help control aberrant gene expression in tumors and HDAC inhibitors have been tested in several clinical trials 103. Some studies have specifically addressed the impact of HDAC inhibitors on TAMs. TMP195 is a HDAC inhibitor that was shown to target TAMs and inhibit primary tumor growth and lung metastasization in a breast cancer model. The administration of TMP195 promoted monocyte recruitment and skewed macrophages towards a highly phagocytic phenotype 103. MP195 treatment tempered TAM production of CCL2, a chemoattractant for monocytes, whereas CCL1 production was elevated 104. TMP195 was also tested in preclinical models in combination with chemotherapy or anti-PD-1 and shown to enhance response to treatments 103. Treatment with HDAC inhibitors was also associated with poor recruitment of immunosuppressive macrophage subsets to the metastatic niche in preclinical models 105, suggesting that HDACs may have a beneficial effect in metastatic disease.
Prostaglandin E2 (PGE2).
PGE2 is a product of arachidonic acid metabolism synthetized by the cyclooxygenase (COX) enzymes. PGE2 is sensed by a family of G-coupled receptors (EP1–4) and was reported to promote tumor growth by directly impacting tumor cell proliferation, survival and migration. Moreover, PGE2 was shown to promote the establishment of an immunosuppressive microenvironment by inhibiting T and NK cell activation 106–108. TAMs express EP receptors 109 and there is growing evidence that PGE2 signaling in TAMs can contribute to skewing toward a pro-tumoral phenotype 106. In a model of mammary carcinoma, EP-2-deficient mice controlled tumors better than WT controls and this was associated with a reduction of immunosuppressive myeloid cells.
In vitro, it was observed that PGE2 can directly impact on bone marrow myeloid progenitors, driving the differentiation of suppressor cells 109. Given the protective effect of aspirin and other nonsteroidal anti-inflammatory drugs that target PGE2 in cancer, macrophages may contribute to this effect. Indeed, the treatment of tumor-bearing mice with a COX-2 inhibitor resulted in a lower tumor burden and this was associated with proportionally fewer suppressive myeloid cells 109. Recently, it was shown that PGE2 led to the enrichment of p50-NFκB in M-MDSCs within tumors in a sarcoma model; in turn, this engendered defective IFNγ-responsiveness and an immunosuppressive signature in the M-MDSCs 110. Furthermore, it was reported that PGE2 might also contribute to macrophage recruitment into the tumor in a mouse model of glioma; COX-2 inhibition was associated with lower CCL2 levels in the tumor and consequent ineffectual accumulation of myeloid cells 111. Since PGE2 acts on multiple cell types, it can have a broad effect in the tumor microenvironment; therefore, delineating the direct contribution of PGE2 sensing in TAMs in vivo requires further investigation.
Reprogramming immunosuppressive TAMs through activating receptors
Certain cell surface receptors expressed on immunosuppressive TAMs can deliver intracellular signals that override immunosuppression and skew TAMs towards immunostimulation (Figure 4). Most of these receptors are not unique to macrophages, but are also expressed in other immune and non-immune cells. One prototypic example is the TNF receptor family member CD40. Engagement of CD40 with mAbs has been extensively tested and shown to induce activation and cytokine production in DCs, as well as enhancing their ability to sustain T cell activation. In a pancreatic ductal adenocarcinoma model, a CD40 agonistic mAb stimulated TAMs to secrete high amounts of MMP13, which degraded fibrotic tissue and facilitated control of tumor growth 112,113. Among several mAbs targeting CD40 tested in clinical trials with promising outcomes, some were shown to have a direct effect on TAMs. Activation of toll-like receptors (TLRs) can also overcome TAM immunosuppression. Numerous TLR synthetic ligands have been tested in tumor models and clinical studies and some were shown to skew TAMs towards an anti-tumoral phenotype 114–120. In a melanoma model, local treatment with a TLR7/TLR8 agonist was associated with a modulation of macrophage phenotype that promoted tumor regression 121,122. Finally, the prototypic cytokine that drives M1-like macrophage polarization, IFNγ, was directly used to reprogram macrophages. In vivo administration IFNγ was tested in tumor models and IFNγ was injected intraperitoneally in patients with ovarian cancer, leading to favorable responses associated with increased immune activation 123. One important limitation of all described agonists of activating pathways is that they act on multiple cells in addition to TAMs, causing non-specific side-effects. Thus, it is likely that these therapeutic agents may be used as adjuvants in combination with other approaches rather than as stand-alone therapies.
One recently developed avenue to induce specific activation of TAMs is the transduction of chimeric antigen receptors (CARs). CARs consist of a single-chain variable fragment antibody specific for a tumor antigen, which is fused to a transmembrane domain that anchors the antibody to the cell membrane, followed by intracellular domains that transmit activation and costimulatory signals 124. Autologous T cells transduced with CARs (CAR-T) acquire tumor specificity as well as signals that drive full T cell activation. CAR-T cell therapy has been very effective in hematological malignancies, whereas success in solid tumors has been limited 125. Macrophages may be a valid alternative to T cells as recipient of CARs, because of their capacity to infiltrate tumors. Recent preclinical studies have addressed the potential anti-tumor impact of macrophages engineered to express a CAR targeting a tumor cell antigen (CAR-M) 126. The human macrophage cell line THP-1 was transduced with a first generation CAR targeting CD19 and was shown to acquire high phagocytic potential against CD19+ K562 tumor cells. Primary monocyte-derived macrophages were also engineered to express an anti-HER2 CAR, which promoted phagocytosis of an HER2-expressing tumor cell line 126. In vivo, treatment with CAR-Ms was protective in a HER2+ ovarian cancer model 126,127. Interestingly, it was observed that regardless of CAR expression, transduced macrophages displayed a pro-inflammatory phenotype that facilitated anti-tumor response, perhaps reflecting an adjuvant effect 126. Given the promising results using CAR-M, it will be important to test different tumor antigens.
Conclusions
As macrophage-targeting therapies are emerging as potential strategies to reprogram immunosuppressive TAMs, some important questions must be addressed: Which molecules should be chosen as targets? Which patients might benefit from TAM reprogramming? Should TAM based therapies be used alone, or in combination with either checkpoint blockade therapies or chemotherapies? Answering these questions requires a detailed knowledge of the TAM landscape in the tumor of interest. Single cell technologies developed in the last decade are providing a granular view of the macrophage phenotypic profiles in many human tumors and mouse models. One important result of these studies is demonstration of the great heterogeneity of TAMs. Multiple factors can imprint TAMs, including the tumor type, the tissue in which the tumor develops, and the presence of immune and stromal cells within the tumor. A detailed knowledge of TAM phenotype and function will help to stratify patients and choose the most appropriate TAM oriented therapies. Single cell technologies can further boost the discovery of novel targets for immunotherapy and the identification of TAM markers predictive of treatment outcome and prognosis. Various studies have suggested that TAMs may have different origins: some may originate from resident macrophages that seed the tissue during fetal hematopoiesis, whereas others may originate from bone marrow-derived blood monocytes 128. The recent development of fate map mice that can trace macrophages of bone marrow origin based on the expression of membrane-spanning 4-domains subfamily A member 3 (Ms4A3) will help delineate how the developmental origin of TAMs influences their response to tumor growth 129. Given the burgeoning availability of multi-omics data on the heterogeneous origins, phenotypes and functions TAM in human tumors, together with the data from clinical trials of TAM therapies in combination with checkpoint blockade therapies, artificial intelligence/machine learning analyses will be required to identify TAM pathways that can be successfully targeted in human cancer.
It is predictable that future studies will also delve into investigation of the complex and different mechanisms by which TAM control immune responses, which remain only partially understood. A recent study identified the tetraspanin MS4A4A as a M2-like pro-tumoral marker that is induced in macrophages by IL-4 or glucocorticoid hormones 130. In mouse metastasis models, MS4A4A-deficiency in macrophages impaired signaling of the surface receptor DECTIN1, which promotes the ability of macrophages to activate NK cells. Thus, MS4A4A-deficiency ultimately undermined NK cell-mediated resistance to metastases 130. In another study, APOE emerged as a biomarker of prognosis and response to therapy in melanoma patients 131. Of the three allelic APOE variants (APOE2, APOE3 and APOE4), APOE4 was associated with attenuated disease progression and an improved response to anti-PD1. The beneficial effect of APOE4 on tumor control was recapitulated in a mouse model expressing the same human variant and was associated with fewer MDSCs in the tumor. Remarkably, APOE is a TREM2 ligand, and both APOE and TREM2 variants are genetically linked to Alzheimer’s disease 30. Thus, the protective role of APOE4 in tumors may be linked to that of TREM2 in TAMs 131. We envision that the complexity of mechanisms through which TAMs control anti-tumor immune responses will reflect the diversity of TAMs infiltrating the tumors.
Acknowledgments
MM is a recipient of the Cancer Research Institute-Lloyd J. Old Memorial Fellowship in Tumor Immunology. MC is supported by the National Institutes of Health (RF1AG05148501 and R21AG059176). We thank Susan Gilfillan for helpful discussion.
Footnotes
Declaration of interests
M. Colonna receives research support from Pfizer and NGM Biotechnology, is a scientific advisory board member of NGM Biotechnology and Vigil, is a consultant for Cell Signaling Technologies and has a patent to TREM2 pending.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Bibliography
- 1.Mantovani A, Marchesi F, Malesci A, Laghi L, and Allavena P. (2017). Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14, 399–416. 10.1038/nrclinonc.2016.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Weiskopf K, and Weissman IL (2015). Macrophages are critical effectors of antibody therapies for cancer. MAbs 7, 303–310. 10.1080/19420862.2015.1011450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.O’Neill LA, and Pearce EJ (2016). Immunometabolism governs dendritic cell and macrophage function. J Exp Med 213, 15–23. 10.1084/jem.20151570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al. (2014). Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20. 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cassetta L, and Pollard JW (2018). Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov 17, 887–904. 10.1038/nrd.2018.169. [DOI] [PubMed] [Google Scholar]
- 6.Bottazzi B, Polentarutti N, Acero R, Balsari A, Boraschi D, Ghezzi P, Salmona M, and Mantovani A. (1983). Regulation of the macrophage content of neoplasms by chemoattractants. Science 220, 210–212. 10.1126/science.6828888. [DOI] [PubMed] [Google Scholar]
- 7.Muller S, Kohanbash G, Liu SJ, Alvarado B, Carrera D, Bhaduri A, Watchmaker PB, Yagnik G, Di Lullo E, Malatesta M, et al. (2017). Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol 18, 234. 10.1186/s13059-017-1362-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, Pamer EG, and Li MO (2014). The cellular and molecular origin of tumor-associated macrophages. Science 344, 921–925. 10.1126/science.1252510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA, and Pollard JW (2011). CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222–225. 10.1038/nature10138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Loyher PL, Hamon P, Laviron M, Meghraoui-Kheddar A, Goncalves E, Deng Z, Torstensson S, Bercovici N, Baudesson de Chanville C, Combadiere B, et al. (2018). Macrophages of distinct origins contribute to tumor development in the lung. J Exp Med 215, 2536–2553. 10.1084/jem.20180534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhu Y, Herndon JM, Sojka DK, Kim KW, Knolhoff BL, Zuo C, Cullinan DR, Luo J, Bearden AR, Lavine KJ, et al. (2017). Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 47, 597. 10.1016/j.immuni.2017.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kwak T, Wang F, Deng H, Condamine T, Kumar V, Perego M, Kossenkov A, Montaner LJ, Xu X, Xu W, et al. (2020). Distinct Populations of Immune-Suppressive Macrophages Differentiate from Monocytic Myeloid-Derived Suppressor Cells in Cancer. Cell Rep 33, 108571. 10.1016/j.celrep.2020.108571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sharma A, Seow JJW, Dutertre CA, Pai R, Bleriot C, Mishra A, Wong RMM, Singh GSN, Sudhagar S, Khalilnezhad S, et al. (2020). Onco-fetal Reprogramming of Endothelial Cells Drives Immunosuppressive Macrophages in Hepatocellular Carcinoma. Cell 183, 377–394 e321. 10.1016/j.cell.2020.08.040. [DOI] [PubMed] [Google Scholar]
- 14.Haldar M, and Murphy KM (2014). Origin, development, and homeostasis of tissue-resident macrophages. Immunol Rev 262, 25–35. 10.1111/imr.12215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, et al. (2016). Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 7, 12150. 10.1038/ncomms12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhao X, Qu J, Sun Y, Wang J, Liu X, Wang F, Zhang H, Wang W, Ma X, Gao X, and Zhang S. (2017). Prognostic significance of tumor-associated macrophages in breast cancer: a meta-analysis of the literature. Oncotarget 8, 30576–30586. 10.18632/oncotarget.15736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang J, Chang L, Zhang X, Zhou Z, and Gao Y. (2021). Meta-Analysis of the Prognostic and Clinical Value of Tumor-Associated Macrophages in Hepatocellular Carcinoma. J Invest Surg 34, 297–306. 10.1080/08941939.2019.1631411. [DOI] [PubMed] [Google Scholar]
- 18.Yuan X, Zhang J, Li D, Mao Y, Mo F, Du W, and Ma X. (2017). Prognostic significance of tumor-associated macrophages in ovarian cancer: A meta-analysis. Gynecol Oncol 147, 181–187. 10.1016/j.ygyno.2017.07.007. [DOI] [PubMed] [Google Scholar]
- 19.Yin S, Huang J, Li Z, Zhang J, Luo J, Lu C, Xu H, and Xu H. (2017). The Prognostic and Clinicopathological Significance of Tumor-Associated Macrophages in Patients with Gastric Cancer: A Meta-Analysis. PLoS One 12, e0170042. 10.1371/journal.pone.0170042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Yang Z, Zhang M, Peng R, Liu J, Wang F, Li Y, Zhao Q, and Liu J. (2020). The prognostic and clinicopathological value of tumor-associated macrophages in patients with colorectal cancer: a systematic review and meta-analysis. Int J Colorectal Dis 35, 1651–1661. 10.1007/s00384-020-03686-9. [DOI] [PubMed] [Google Scholar]
- 21.Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z, Junt T, and Bentires-Alj M. (2014). Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature 515, 130–133. 10.1038/nature13862. [DOI] [PubMed] [Google Scholar]
- 22.Loberg RD, Ying C, Craig M, Day LL, Sargent E, Neeley C, Wojno K, Snyder LA, Yan L, and Pienta KJ (2007). Targeting CCL2 with systemic delivery of neutralizing antibodies induces prostate cancer tumor regression in vivo. Cancer Res 67, 9417–9424. 10.1158/0008-5472.CAN-07-1286. [DOI] [PubMed] [Google Scholar]
- 23.DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA, et al. (2011). Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov 1, 54–67. 10.1158/2159-8274.CD-10-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V, et al. (2013). CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 19, 1264–1272. 10.1038/nm.3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Banerjee P, Zhang R, Ivan C, Galletti G, Clise-Dwyer K, Barbaglio F, Scarfo L, Aracil M, Klein C, Wierda W, et al. (2019). Trabectedin Reveals a Strategy of Immunomodulation in Chronic Lymphocytic Leukemia. Cancer Immunol Res 7, 2036–2051. 10.1158/2326-6066.CIR-19-0152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Majety M, Runza V, Lehmann C, Hoves S, and Ries CH (2018). A drug development perspective on targeting tumor-associated myeloid cells. FEBS J 285, 763–776. 10.1111/febs.14277. [DOI] [PubMed] [Google Scholar]
- 27.Ulland TK, Song WM, Huang SC, Ulrich JD, Sergushichev A, Beatty WL, Loboda AA, Zhou Y, Cairns NJ, Kambal A, et al. (2017). TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s Disease. Cell 170, 649–663 e613. 10.1016/j.cell.2017.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Peng Q, Malhotra S, Torchia JA, Kerr WG, Coggeshall KM, and Humphrey MB (2010). TREM2- and DAP12-dependent activation of PI3K requires DAP10 and is inhibited by SHIP1. Sci Signal 3, ra38. 10.1126/scisignal.2000500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Otero K, Shinohara M, Zhao H, Cella M, Gilfillan S, Colucci A, Faccio R, Ross FP, Teitelbaum SL, Takayanagi H, and Colonna M. (2012). TREM2 and beta-catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. J Immunol 188, 2612–2621. 10.4049/jimmunol.1102836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ulland TK, and Colonna M. (2018). TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol 14, 667–675. 10.1038/s41582-018-0072-1. [DOI] [PubMed] [Google Scholar]
- 31.Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, Lundgren P, Bleriot C, Liu Z, Deczkowska A, et al. (2019). Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell 178, 686–698 e614. 10.1016/j.cell.2019.05.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Daemen S, Gainullina A, Kalugotla G, He L, Chan MM, Beals JW, Liss KH, Klein S, Feldstein AE, Finck BN, et al. (2021). Dynamic Shifts in the Composition of Resident and Recruited Macrophages Influence Tissue Remodeling in NASH. Cell Rep 34, 108626. 10.1016/j.celrep.2020.108626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ramachandran P, Dobie R, Wilson-Kanamori JR, Dora EF, Henderson BEP, Luu NT, Portman JR, Matchett KP, Brice M, Marwick JA, et al. (2019). Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518. 10.1038/s41586-019-1631-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hou J, Zhang J, Cui P, Zhou Y, Liu C, Wu X, Ji Y, Wang S, Cheng B, Ye H, et al. (2021). TREM2 sustains macrophage-hepatocyte metabolic coordination in nonalcoholic fatty liver disease and sepsis. J Clin Invest 131. 10.1172/JCI135197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cochain C, Vafadarnejad E, Arampatzi P, Pelisek J, Winkels H, Ley K, Wolf D, Saliba AE, and Zernecke A. (2018). Single-Cell RNA-Seq Reveals the Transcriptional Landscape and Heterogeneity of Aortic Macrophages in Murine Atherosclerosis. Circ Res 122, 1661–1674. 10.1161/CIRCRESAHA.117.312509. [DOI] [PubMed] [Google Scholar]
- 36.Song Q, Hawkins GA, Wudel L, Chou PC, Forbes E, Pullikuth AK, Liu L, Jin G, Craddock L, Topaloglu U, et al. (2019). Dissecting intratumoral myeloid cell plasticity by single cell RNA-seq. Cancer Med 8, 3072–3085. 10.1002/cam4.2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C, Remark R, Sweeney R, Becker CD, Levine JH, et al. (2017). Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 169, 750–765 e717. 10.1016/j.cell.2017.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Katzenelenbogen Y, Sheban F, Yalin A, Yofe I, Svetlichnyy D, Jaitin DA, Bornstein C, Moshe A, Keren-Shaul H, Cohen M, et al. (2020). Coupled scRNA-Seq and Intracellular Protein Activity Reveal an Immunosuppressive Role of TREM2 in Cancer. Cell 182, 872–885 e819. 10.1016/j.cell.2020.06.032. [DOI] [PubMed] [Google Scholar]
- 39.Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, Brioschi S, Bugatti M, Omodei AS, Ricci B, et al. (2020). TREM2 Modulation Remodels the Tumor Myeloid Landscape Enhancing Anti-PD-1 Immunotherapy. Cell 182, 886–900 e817. 10.1016/j.cell.2020.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.La Fleur L, Boura VF, Alexeyenko A, Berglund A, Ponten V, Mattsson JSM, Djureinovic D, Persson J, Brunnstrom H, Isaksson J, et al. (2018). Expression of scavenger receptor MARCO defines a targetable tumor-associated macrophage subset in non-small cell lung cancer. Int J Cancer 143, 1741–1752. 10.1002/ijc.31545. [DOI] [PubMed] [Google Scholar]
- 41.Kraal G, van der Laan LJ, Elomaa O, and Tryggvason K. (2000). The macrophage receptor MARCO. Microbes Infect 2, 313–316. 10.1016/s1286-4579(00)00296-3. [DOI] [PubMed] [Google Scholar]
- 42.Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J, Dahan R, Harris RA, Rantalainen M, Klevebring D, et al. (2016). Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep 15, 2000–2011. 10.1016/j.celrep.2016.04.084. [DOI] [PubMed] [Google Scholar]
- 43.O’Neill AS, van den Berg TK, and Mullen GE (2013). Sialoadhesin - a macrophage-restricted marker of immunoregulation and inflammation. Immunology 138, 198–207. 10.1111/imm.12042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cassetta L, Fragkogianni S, Sims AH, Swierczak A, Forrester LM, Zhang H, Soong DYH, Cotechini T, Anur P, Lin EY, et al. (2019). Human Tumor-Associated Macrophage and Monocyte Transcriptional Landscapes Reveal Cancer-Specific Reprogramming, Biomarkers, and Therapeutic Targets. Cancer Cell 35, 588–602 e510. 10.1016/j.ccell.2019.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Jing W, Guo X, Wang G, Bi Y, Han L, Zhu Q, Qiu C, Tanaka M, and Zhao Y. (2020). Breast cancer cells promote CD169(+) macrophage-associated immunosuppression through JAK2-mediated PD-L1 upregulation on macrophages. Int Immunopharmacol 78, 106012. 10.1016/j.intimp.2019.106012. [DOI] [PubMed] [Google Scholar]
- 46.Strakhova R, Cadassou O, Cros-Perrial E, and Jordheim LP (2020). Regulation of tumor infiltrated innate immune cells by adenosine. Purinergic Signal 16, 289–295. 10.1007/s11302-020-09701-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ferrante CJ, Pinhal-Enfield G, Elson G, Cronstein BN, Hasko G, Outram S, and Leibovich SJ (2013). The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Ralpha) signaling. Inflammation 36, 921–931. 10.1007/s10753-013-9621-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xaus J, Mirabet M, Lloberas J, Soler C, Lluis C, Franco R, and Celada A. (1999). IFNgamma up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J Immunol 162, 3607–3614. [PubMed] [Google Scholar]
- 49.Hasko G, Szabo C, Nemeth ZH, Kvetan V, Pastores SM, and Vizi ES (1996). Adenosine receptor agonists differentially regulate IL-10, TNF-alpha, and nitric oxide production in RAW 264.7 macrophages and in endotoxemic mice. J Immunol 157, 4634–4640. [PubMed] [Google Scholar]
- 50.Belikoff BG, Hatfield S, Georgiev P, Ohta A, Lukashev D, Buras JA, Remick DG, and Sitkovsky M. (2011). A2B adenosine receptor blockade enhances macrophage-mediated bacterial phagocytosis and improves polymicrobial sepsis survival in mice. J Immunol 186, 2444–2453. 10.4049/jimmunol.1001567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Perrot I, Michaud HA, Giraudon-Paoli M, Augier S, Docquier A, Gros L, Courtois R, Dejou C, Jecko D, Becquart O, et al. (2019). Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. Cell Rep 27, 2411–2425 e2419. 10.1016/j.celrep.2019.04.091. [DOI] [PubMed] [Google Scholar]
- 52.Azambuja JH, Schuh RS, Michels LR, Iser IC, Beckenkamp LR, Roliano GG, Lenz GS, Scholl JN, Sevigny J, Wink MR, et al. (2020). Blockade of CD73 delays glioblastoma growth by modulating the immune environment. Cancer Immunol Immunother 69, 1801–1812. 10.1007/s00262-020-02569-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Antonioli L, Pacher P, Vizi ES, and Hasko G. (2013). CD39 and CD73 in immunity and inflammation. Trends Mol Med 19, 355–367. 10.1016/j.molmed.2013.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hochreiter-Hufford A, and Ravichandran KS (2013). Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb Perspect Biol 5, a008748. 10.1101/cshperspect.a008748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, and Lindberg FP (2000). Role of CD47 as a marker of self on red blood cells. Science 288, 2051–2054. 10.1126/science.288.5473.2051. [DOI] [PubMed] [Google Scholar]
- 56.Barclay AN, and Van den Berg TK (2014). The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: structure, function, and therapeutic target. Annu Rev Immunol 32, 25–50. 10.1146/annurev-immunol-032713-120142. [DOI] [PubMed] [Google Scholar]
- 57.Manna PP, and Frazier WA (2004). CD47 mediates killing of breast tumor cells via Gi-dependent inhibition of protein kinase A. Cancer Res 64, 1026–1036. 10.1158/0008-5472.can-03-1708. [DOI] [PubMed] [Google Scholar]
- 58.Kim MJ, Lee JC, Lee JJ, Kim S, Lee SG, Park SW, Sung MW, and Heo DS (2008). Association of CD47 with natural killer cell-mediated cytotoxicity of head-and-neck squamous cell carcinoma lines. Tumour Biol 29, 28–34. 10.1159/000132568. [DOI] [PubMed] [Google Scholar]
- 59.Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, Traver D, van Rooijen N, and Weissman IL (2009). CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285. 10.1016/j.cell.2009.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chan KS, Espinosa I, Chao M, Wong D, Ailles L, Diehn M, Gill H, Presti J Jr., Chang HY, van de Rijn M, et al. (2009). Identification, molecular characterization, clinical prognosis, and therapeutic targeting of human bladder tumor-initiating cells. Proc Natl Acad Sci U S A 106, 14016–14021. 10.1073/pnas.0906549106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, Jan M, Cha AC, Chan CK, Tan BT, et al. (2010). Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142, 699–713. 10.1016/j.cell.2010.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr., van Rooijen N, and Weissman IL (2009). CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 138, 286–299. 10.1016/j.cell.2009.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Xiao Z, Chung H, Banan B, Manning PT, Ott KC, Lin S, Capoccia BJ, Subramanian V, Hiebsch RR, Upadhya GA, et al. (2015). Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma. Cancer Lett 360, 302–309. 10.1016/j.canlet.2015.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, et al. (2012). The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A 109, 6662–6667. 10.1073/pnas.1121623109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Swoboda DM, and Sallman DA (2020). The promise of macrophage directed checkpoint inhibitors in myeloid malignancies. Best Pract Res Clin Haematol 33, 101221. 10.1016/j.beha.2020.101221. [DOI] [PubMed] [Google Scholar]
- 66.Sikic BI, Lakhani N, Patnaik A, Shah SA, Chandana SR, Rasco D, Colevas AD, O’Rourke T, Narayanan S, Papadopoulos K, et al. (2019). First-in-Human, First-in-Class Phase I Trial of the Anti-CD47 Antibody Hu5F9-G4 in Patients With Advanced Cancers. J Clin Oncol 37, 946–953. 10.1200/JCO.18.02018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lopez-Yrigoyen M, Cassetta L, and Pollard JW (2020). Macrophage targeting in cancer. Ann N Y Acad Sci. 10.1111/nyas.14377. [DOI] [PubMed] [Google Scholar]
- 68.Huang Y, Ma Y, Gao P, and Yao Z. (2017). Targeting CD47: the achievements and concerns of current studies on cancer immunotherapy. J Thorac Dis 9, E168–E174. 10.21037/jtd.2017.02.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, Finetti P, Van Egmond M, Matozaki T, Kraal G, et al. (2011). CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A 108, 18342–18347. 10.1073/pnas.1106550108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Liu R, Wei H, Gao P, Yu H, Wang K, Fu Z, Ju B, Zhao M, Dong S, Li Z, et al. (2017). CD47 promotes ovarian cancer progression by inhibiting macrophage phagocytosis. Oncotarget 8, 39021–39032. 10.18632/oncotarget.16547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Cheung A, Opzoomer J, Ilieva KM, Gazinska P, Hoffmann RM, Mirza H, Marlow R, Francesch-Domenech E, Fittall M, Dominguez Rodriguez D, et al. (2018). Anti-Folate Receptor Alpha-Directed Antibody Therapies Restrict the Growth of Triple-negative Breast Cancer. Clin Cancer Res 24, 5098–5111. 10.1158/1078-0432.CCR-18-0652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Yanagita T, Murata Y, Tanaka D, Motegi SI, Arai E, Daniwijaya EW, Hazama D, Washio K, Saito Y, Kotani T, et al. (2017). Anti-SIRPalpha antibodies as a potential new tool for cancer immunotherapy. JCI Insight 2, e89140. 10.1172/jci.insight.89140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chen J, Zhong MC, Guo H, Davidson D, Mishel S, Lu Y, Rhee I, Perez-Quintero LA, Zhang S, Cruz-Munoz ME, et al. (2017). SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 544, 493–497. 10.1038/nature22076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kuroki K, Matsubara H, Kanda R, Miyashita N, Shiroishi M, Fukunaga Y, Kamishikiryo J, Fukunaga A, Fukuhara H, Hirose K, et al. (2019). Structural and Functional Basis for LILRB Immune Checkpoint Receptor Recognition of HLA-G Isoforms. J Immunol 203, 3386–3394. 10.4049/jimmunol.1900562. [DOI] [PubMed] [Google Scholar]
- 75.Willcox BE, Thomas LM, and Bjorkman PJ (2003). Crystal structure of HLA-A2 bound to LIR-1, a host and viral major histocompatibility complex receptor. Nat Immunol 4, 913–919. 10.1038/ni961. [DOI] [PubMed] [Google Scholar]
- 76.Colonna M, Navarro F, Bellon T, Llano M, Garcia P, Samaridis J, Angman L, Cella M, and Lopez-Botet M. (1997). A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells. J Exp Med 186, 1809–1818. 10.1084/jem.186.11.1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Allan DS, McMichael AJ, and Braud VM (2000). The ILT family of leukocyte receptors. Immunobiology 202, 34–41. 10.1016/S0171-2985(00)80050-9. [DOI] [PubMed] [Google Scholar]
- 78.Cella M, Nakajima H, Facchetti F, Hoffmann T, and Colonna M. (2000). ILT receptors at the interface between lymphoid and myeloid cells. Curr Top Microbiol Immunol 251, 161–166. 10.1007/978-3-642-57276-0_20. [DOI] [PubMed] [Google Scholar]
- 79.Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, George BM, Markovic M, Ring NG, Tsai JM, et al. (2018). Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol 19, 76–84. 10.1038/s41590-017-0004-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen HM, van der Touw W, Wang YS, Kang K, Mai S, Zhang J, Alsina-Beauchamp D, Duty JA, Mungamuri SK, Zhang B, et al. (2018). Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest 128, 5647–5662. 10.1172/JCI97570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Takai T. (2005). Paired immunoglobulin-like receptors and their MHC class I recognition. Immunology 115, 433–440. 10.1111/j.1365-2567.2005.02177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kubagawa H, Burrows PD, and Cooper MD (1997). A novel pair of immunoglobulin-like receptors expressed by B cells and myeloid cells. Proc Natl Acad Sci U S A 94, 5261–5266. 10.1073/pnas.94.10.5261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ma G, Pan PY, Eisenstein S, Divino CM, Lowell CA, Takai T, and Chen SH (2011). Paired immunoglobin-like receptor-B regulates the suppressive function and fate of myeloid-derived suppressor cells. Immunity 34, 385–395. 10.1016/j.immuni.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Crocker PR, Paulson JC, and Varki A. (2007). Siglecs and their roles in the immune system. Nat Rev Immunol 7, 255–266. 10.1038/nri2056. [DOI] [PubMed] [Google Scholar]
- 85.Chen W, Han C, Xie B, Hu X, Yu Q, Shi L, Wang Q, Li D, Wang J, Zheng P, et al. (2013). Induction of Siglec-G by RNA viruses inhibits the innate immune response by promoting RIG-I degradation. Cell 152, 467–478. 10.1016/j.cell.2013.01.011. [DOI] [PubMed] [Google Scholar]
- 86.Chen GY, Chen X, King S, Cavassani KA, Cheng J, Zheng X, Cao H, Yu H, Qu J, Fang D, et al. (2011). Amelioration of sepsis by inhibiting sialidase-mediated disruption of the CD24-SiglecG interaction. Nat Biotechnol 29, 428–435. 10.1038/nbt.1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Chen GY, Tang J, Zheng P, and Liu Y. (2009). CD24 and Siglec-10 selectively repress tissue damage-induced immune responses. Science 323, 1722–1725. 10.1126/science.1168988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Toubai T, Hou G, Mathewson N, Liu C, Wang Y, Oravecz-Wilson K, Cummings E, Rossi C, Evers R, Sun Y, et al. (2014). Siglec-G-CD24 axis controls the severity of graft-versus-host disease in mice. Blood 123, 3512–3523. 10.1182/blood-2013-12-545335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, Krishnan V, Hatakeyama J, Dorigo O, Barkal LJ, and Weissman IL (2019). CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 572, 392–396. 10.1038/s41586-019-1456-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Baumeister SH, Freeman GJ, Dranoff G, and Sharpe AH (2016). Coinhibitory Pathways in Immunotherapy for Cancer. Annu Rev Immunol 34, 539–573. 10.1146/annurev-immunol-032414-112049. [DOI] [PubMed] [Google Scholar]
- 91.Zou W, Wolchok JD, and Chen L. (2016). PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 8, 328rv324. 10.1126/scitranslmed.aad7118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Sosman JA, Atkins MB, Leming PD, et al. (2019). Five-Year Survival and Correlates Among Patients With Advanced Melanoma, Renal Cell Carcinoma, or Non-Small Cell Lung Cancer Treated With Nivolumab. JAMA Oncol. 10.1001/jamaoncol.2019.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, et al. (2017). PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545, 495–499. 10.1038/nature22396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Dhupkar P, Gordon N, Stewart J, and Kleinerman ES (2018). Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases. Cancer Med 7, 2654–2664. 10.1002/cam4.1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Hartley GP, Chow L, Ammons DT, Wheat WH, and Dow SW (2018). Programmed Cell Death Ligand 1 (PD-L1) Signaling Regulates Macrophage Proliferation and Activation. Cancer Immunol Res 6, 1260–1273. 10.1158/2326-6066.CIR-17-0537. [DOI] [PubMed] [Google Scholar]
- 96.Gubin MM, Esaulova E, Ward JP, Malkova ON, Runci D, Wong P, Noguchi T, Arthur CD, Meng W, Alspach E, et al. (2018). High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell 175, 1014–1030 e1019. 10.1016/j.cell.2018.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, and Tsatsanis C. (2017). Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J Immunol 198, 1006–1014. 10.4049/jimmunol.1601515. [DOI] [PubMed] [Google Scholar]
- 98.Schmid MC, Avraamides CJ, Dippold HC, Franco I, Foubert P, Ellies LG, Acevedo LM, Manglicmot JR, Song X, Wrasidlo W, et al. (2011). Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3kgamma, a single convergent point promoting tumor inflammation and progression. Cancer Cell 19, 715–727. 10.1016/j.ccr.2011.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, Woo G, Nguyen AV, Figueiredo CC, Foubert P, et al. (2016). PI3Kgamma is a molecular switch that controls immune suppression. Nature 539, 437–442. 10.1038/nature19834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Joshi S, Singh AR, Zulcic M, and Durden DL (2014). A macrophage-dominant PI3K isoform controls hypoxia-induced HIF1alpha and HIF2alpha stability and tumor growth, angiogenesis, and metastasis. Mol Cancer Res 12, 1520–1531. 10.1158/1541-7786.MCR-13-0682. [DOI] [PubMed] [Google Scholar]
- 101.Gunderson AJ, Kaneda MM, Tsujikawa T, Nguyen AV, Affara NI, Ruffell B, Gorjestani S, Liudahl SM, Truitt M, Olson P, et al. (2016). Bruton Tyrosine Kinase-Dependent Immune Cell Cross-talk Drives Pancreas Cancer. Cancer Discov 6, 270–285. 10.1158/2159-8290.CD-15-0827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hojfeldt JW, Agger K, and Helin K. (2013). Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov 12, 917–930. 10.1038/nrd4154. [DOI] [PubMed] [Google Scholar]
- 103.Guerriero JL, Sotayo A, Ponichtera HE, Castrillon JA, Pourzia AL, Schad S, Johnson SF, Carrasco RD, Lazo S, Bronson RT, et al. (2017). Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature 543, 428–432. 10.1038/nature21409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lobera M, Madauss KP, Pohlhaus DT, Wright QG, Trocha M, Schmidt DR, Baloglu E, Trump RP, Head MS, Hofmann GA, et al. (2013). Selective class IIa histone deacetylase inhibition via a nonchelating zinc-binding group. Nat Chem Biol 9, 319–325. 10.1038/nchembio.1223. [DOI] [PubMed] [Google Scholar]
- 105.Lu Z, Zou J, Li S, Topper MJ, Tao Y, Zhang H, Jiao X, Xie W, Kong X, Vaz M, et al. (2020). Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature 579, 284–290. 10.1038/s41586-020-2054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wang D, and Dubois RN (2010). Eicosanoids and cancer. Nat Rev Cancer 10, 181–193. 10.1038/nrc2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zelenay S, van der Veen AG, Bottcher JP, Snelgrove KJ, Rogers N, Acton SE, Chakravarty P, Girotti MR, Marais R, Quezada SA, et al. (2015). Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 162, 1257–1270. 10.1016/j.cell.2015.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bonavita E, Bromley CP, Jonsson G, Pelly VS, Sahoo S, Walwyn-Brown K, Mensurado S, Moeini A, Flanagan E, Bell CR, et al. (2020). Antagonistic Inflammatory Phenotypes Dictate Tumor Fate and Response to Immune Checkpoint Blockade. Immunity 53, 1215–1229 e1218. 10.1016/j.immuni.2020.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Sinha P, Clements VK, Fulton AM, and Ostrand-Rosenberg S. (2007). Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res 67, 4507–4513. 10.1158/0008-5472.CAN-06-4174. [DOI] [PubMed] [Google Scholar]
- 110.Porta C, Consonni FM, Morlacchi S, Sangaletti S, Bleve A, Totaro MG, Larghi P, Rimoldi M, Tripodo C, Strauss L, et al. (2020). Tumor-Derived Prostaglandin E2 Promotes p50 NF-kappaB-Dependent Differentiation of Monocytic MDSCs. Cancer Res 80, 2874–2888. 10.1158/0008-5472.CAN-19-2843. [DOI] [PubMed] [Google Scholar]
- 111.Fujita M, Kohanbash G, Fellows-Mayle W, Hamilton RL, Komohara Y, Decker SA, Ohlfest JR, and Okada H. (2011). COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res 71, 2664–2674. 10.1158/0008-5472.CAN-10-3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, and Beatty GL (2016). IFNgamma and CCL2 Cooperate to Redirect Tumor-Infiltrating Monocytes to Degrade Fibrosis and Enhance Chemotherapy Efficacy in Pancreatic Carcinoma. Cancer Discov 6, 400–413. 10.1158/2159-8290.CD-15-1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL, et al. (2011). CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331, 1612–1616. 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Monk BJ, Brady MF, Aghajanian C, Lankes HA, Rizack T, Leach J, Fowler JM, Higgins R, Hanjani P, Morgan M, et al. (2017). A phase 2, randomized, double-blind, placebo- controlled study of chemo-immunotherapy combination using motolimod with pegylated liposomal doxorubicin in recurrent or persistent ovarian cancer: a Gynecologic Oncology Group partners study. Ann Oncol 28, 996–1004. 10.1093/annonc/mdx049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Chow LQM, Morishima C, Eaton KD, Baik CS, Goulart BH, Anderson LN, Manjarrez KL, Dietsch GN, Bryan JK, Hershberg RM, et al. (2017). Phase Ib Trial of the Toll-like Receptor 8 Agonist, Motolimod (VTX-2337), Combined with Cetuximab in Patients with Recurrent or Metastatic SCCHN. Clin Cancer Res 23, 2442–2450. 10.1158/1078-0432.CCR-16-1934. [DOI] [PubMed] [Google Scholar]
- 116.Ribas A, Medina T, Kummar S, Amin A, Kalbasi A, Drabick JJ, Barve M, Daniels GA, Wong DJ, Schmidt EV, et al. (2018). SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase Ib, Multicenter Study. Cancer Discov 8, 1250–1257. 10.1158/2159-8290.CD-18-0280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kyi C, Roudko V, Sabado R, Saenger Y, Loging W, Mandeli J, Thin TH, Lehrer D, Donovan M, Posner M, et al. (2018). Therapeutic Immune Modulation against Solid Cancers with Intratumoral Poly-ICLC: A Pilot Trial. Clin Cancer Res 24, 4937–4948. 10.1158/1078-0432.CCR-17-1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kell SA, Kachura MA, Renn A, Traquina P, Coffman RL, and Campbell JD (2019). Preclinical development of the TLR9 agonist DV281 as an inhaled aerosolized immunotherapeutic for lung cancer: Pharmacological profile in mice, non-human primates, and human primary cells. Int Immunopharmacol 66, 296–308. 10.1016/j.intimp.2018.11.019. [DOI] [PubMed] [Google Scholar]
- 119.Adams S, Kozhaya L, Martiniuk F, Meng TC, Chiriboga L, Liebes L, Hochman T, Shuman N, Axelrod D, Speyer J, et al. (2012). Topical TLR7 agonist imiquimod can induce immune-mediated rejection of skin metastases in patients with breast cancer. Clin Cancer Res 18, 6748–6757. 10.1158/1078-0432.CCR-12-1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Camargo JA, Passos GR, Ferrari KL, Billis A, Saad MJA, and Reis LO (2018). Intravesical Immunomodulatory Imiquimod Enhances Bacillus Calmette-Guerin Downregulation of Nonmuscle-invasive Bladder Cancer. Clin Genitourin Cancer 16, e587–e593. 10.1016/j.clgc.2017.10.019. [DOI] [PubMed] [Google Scholar]
- 121.Rodell CB, Arlauckas SP, Cuccarese MF, Garris CS, Li R, Ahmed MS, Kohler RH, Pittet MJ, and Weissleder R. (2018). TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng 2, 578–588. 10.1038/s41551-018-0236-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Singh M, Khong H, Dai Z, Huang XF, Wargo JA, Cooper ZA, Vasilakos JP, Hwu P, and Overwijk WW (2014). Effective innate and adaptive antimelanoma immunity through localized TLR7/8 activation. J Immunol 193, 4722–4731. 10.4049/jimmunol.1401160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Pujade-Lauraine E, Guastalla JP, Colombo N, Devillier P, Francois E, Fumoleau P, Monnier A, Nooy M, Mignot L, Bugat R, et al. (1996). Intraperitoneal recombinant interferon gamma in ovarian cancer patients with residual disease at second-look laparotomy. J Clin Oncol 14, 343–350. 10.1200/JCO.1996.14.2.343. [DOI] [PubMed] [Google Scholar]
- 124.Larson RC, and Maus MV (2021). Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat Rev Cancer 21, 145–161. 10.1038/s41568-020-00323-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kingwell K. (2017). CAR T therapies drive into new terrain. Nat Rev Drug Discov 16, 301–304. 10.1038/nrd.2017.84. [DOI] [PubMed] [Google Scholar]
- 126.Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, Schmierer M, Gabrusiewicz K, Anderson NR, Petty NE, et al. (2020). Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol 38, 947–953. 10.1038/s41587-020-0462-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zhang W, Liu L, Su H, Liu Q, Shen J, Dai H, Zheng W, Lu Y, Zhang W, Bei Y, and Shen P. (2019). Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix. Br J Cancer 121, 837–845. 10.1038/s41416-019-0578-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Franklin RA, and Li MO (2016). Ontogeny of Tumor-associated Macrophages and Its Implication in Cancer Regulation. Trends Cancer 2, 20–34. 10.1016/j.trecan.2015.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Liu Z, Gu Y, Chakarov S, Bleriot C, Kwok I, Chen X, Shin A, Huang W, Dress RJ, Dutertre CA, et al. (2019). Fate Mapping via Ms4a3-Expression History Traces Monocyte-Derived Cells. Cell 178, 1509–1525 e1519. 10.1016/j.cell.2019.08.009. [DOI] [PubMed] [Google Scholar]
- 130.Mattiola I, Tomay F, De Pizzol M, Silva-Gomes R, Savino B, Gulic T, Doni A, Lonardi S, Astrid Boutet M, Nerviani A, et al. (2019). The macrophage tetraspan MS4A4A enhances dectin-1-dependent NK cell-mediated resistance to metastasis. Nat Immunol 20, 1012–1022. 10.1038/s41590-019-0417-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ostendorf BN, Bilanovic J, Adaku N, Tafreshian KN, Tavora B, Vaughan RD, and Tavazoie SF (2020). Common germline variants of the human APOE gene modulate melanoma progression and survival. Nat Med 26, 1048–1053. 10.1038/s41591-020-0879-3. [DOI] [PMC free article] [PubMed] [Google Scholar]