Open Access

Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review)

  • Authors:
    • Anna Kazakova
    • Tatiana Sudarskikh
    • Oleg Kovalev
    • Julia Kzhyshkowska
    • Irina Larionova
  • View Affiliations

  • Published online on: January 16, 2023     https://doi.org/10.3892/ijo.2023.5480
  • Article Number: 32
  • Copyright: © Kazakova et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor‑associated macrophages (TAMs) are crucial cells of the tumor microenvironment (TME), which belong to the innate immune system and regulate primary tumor growth, immunosuppression, angiogenesis, extracellular matrix remodeling and metastasis. The review discusses current knowledge of essential cell‑cell interactions of TAMs within the TME of solid tumors. It summarizes the mechanisms of stromal cell (including cancer‑associated fibroblasts and endothelial cells)‑mediated monocyte recruitment and regulation of differentiation, as well as pro‑tumor and antitumor polarization of TAMs. Additionally, it focuses on the perivascular TAM subpopulations that regulate angiogenesis and lymphangiogenesis. It describes the possible mechanisms of reciprocal interactions of TAMs with other immune cells responsible for immunosuppression. Finally, it highlights the perspectives for novel therapeutic approaches to use combined cellular targets that include TAMs and other stromal and immune cells in the TME. The collected data demonstrated the importance of understanding cell‑cell interactions in the TME to prevent distant metastasis and reduce the risk of tumor recurrence.

1. Introduction

Tumors exist in constant interaction with components of the tumor microenvironment (TME), which includes stromal cells [such as cancer-associated fibroblasts (CAFs), endothelial cells (ECs) and adipocytes], and immune cells [such as tumor-associated macrophages (TAMs), cancer-associated neutrophils, dendritic cells (DCs), natural killer (NK) cells, T lymphocytes and B lymphocytes], as well as extracellular matrix (ECM) (1,2). Cells in the TME are involved in reciprocal interaction with tumor cells, which eventually leads to enhanced proliferation, metastasis and chemoresistance (1,3,4).

TAMs are key innate immune cells involved in regulation of primary tumor growth, antitumor adaptive immune response, tumor angiogenesis, ECM remodeling and metastasis (5,6). TAMs originate either from circulating monocytes derived from bone marrow hematopoietic stem cells (recruited) or yolk sac progenitors (resident) (7). These cells are attracted into the primary tumor and activated by various signals in the TME (6).

TAMs are classified as M1 (classically activated macrophages) and M2 (alternatively activated macrophages) (5). M1 TAMs secrete pro-inflammatory cytokines IL-12, TNFα, C-X-C motif chemokine ligand (CXCL)-10 and IFN-γ, and produce high levels of nitric oxide synthase (NOS), while M2 TAMs secrete anti-inflammatory cytokines IL-10, IL-4 and IL-13, and express scavenger receptors, including CD163, CD206, CD204, macrophage receptor with collagenous structure (MARCO) and stabilin-1 (5,6). TAMs are plastic cells that can change their polarization from M1 to M2 and vice versa in response to various stimuli from the TME, including cytokines, growth factors and matrix remodeling proteins (8,9). Cells in the TME are genetically more stable compared with tumor cells, which makes them useful as therapeutic targets (10,11).

In the present review, data on the main mechanisms of the TAM interaction with crucial components of the TME, including CAFs, blood and lymphatic ECs (LECs), and diverse immune cell infiltrates, were collected. It also discusses possible therapeutic approaches that combine targeting of TAMs and other stromal or immune cells.

2. CAFs

CAFs are heterogenous stromal cells originating from different precursor cells, including resident tissue fibroblasts (via activation), bone marrow mesenchymal stem cells (via differentiation), hematopoietic stem cells (via differentiation), epithelial cells via epithelial-mesenchymal transition (EMT) and ECs via endothelial-mesenchymal transition (EndoMT) (12,13). Tumor cells activate EndoMT through production of cytokines and growth factors, among which TGF-α and TGF-β are the most studied (14,15). In some tumors, such as melanoma, pancreatic cancer and lung cancer, EndoMT provides 30-40% of CAFs (16,17).

In the TME, CAFs possess functions critical for supporting cancer cell proliferation, angiogenesis, lymphangiogenesis, invasion and metastasis. CAFs promote tumor progression via various mechanisms, including cytokine and chemokine secretion, as well as ECM remodeling that occurs due to basement membrane degradation and interstitial stroma disintegration (18-26).

CAFs induce monocyte recruitment

Numerous studies have demonstrated that CAFs contribute to monocyte recruitment (Fig. 1A) (27-29). In a Transwell migration assay, conditioned medium (CM) of colon cancer-derived CAFs was more efficient in attracting monocytes compared with CM of macrophages, differentiated from peripheral blood mononuclear cells, or human HCT116 and DLD-1 colon cancer cells (27). There are two key factors promoting monocyte recruitment: Macrophage colony-stimulating factor [M-CSF or colony stimulating factor 1 (CSF1)] and monocyte chemoattractant protein 1 [MCP-1; or C-C motif chemokine ligand (CCL2)] (27,30,31). Co-culture of CAFs with monocyte-derived macrophages (MDMs) revealed high expression of CCL2 in MDMs, indicating CAFs to be one of the major inducers of CCL2 production in macrophages (27). M-CSF is mainly produced by CAFs but not by tumor cells or macrophages (27). Inhibition of M-CSF/M-CSF receptor (M-CSFR) signaling results in decreased CCL2, CD163 and M-CSFR expression (27). The role of M-CSF in myeloid cell survival, differentiation and function has been demonstrated in other studies (32,33). IL-8 produced by CAFs derived from human colorectal cancer (CRC) tissue has been reported to recruit monocytes, while IL-6 promoted their adhesion to the tumor cells (28). CM from CAFs induced vascular cell adhesion molecule 1 (VCAM-1) expression in CRC cells, which is associated with enriched infiltration of TAMs in the TME (28). A Transwell assay revealed that CAF-derived C-X-C motif chemokine receptor (CXCR)2 and IL-8 facilitated monocyte migration in a dose-dependent manner (28).

Figure 1

Functional crosstalk between TAMs and stromal components. (A) TAMs mutually interact with CAFs in the tumor microenvironment. CAFs produce various soluble factors that act as chemoattractants, activation and polarization agents for monocytes/macrophages. (B) TAMs are crucial regulators of tumor neovascularization and metastasis. In the TME, TAMs are polarized to the M2 state. Vascular ECs and LECs can recruit monocytes and macrophages and induce M2 polarization by producing various soluble (Ang2, VEGF-A, VEGF-C, IL-1β, IL-6, M-CSF and IL-33) and membranes (CX3CL1, GAL8 and stabilin-1) factors. M2 TAMs secrete proteins (LCN2, MMP9 and MMP2) and growth factors (VEGF-C and VEGF-A) that are essential for vascular and LEC proliferation and differentiation. M2 TAMs also facilitate degradation of the basement membrane of blood and lymphatic vessels and can destabilize EC junctions. Perivascular TAMs (PDPN+, LYVE1+ and TEM) are specific TAM subpopulations inducing angiogenesis. Ang2, angiopoetin 2; BM, basement membrane; CAFs, cancer-associated fibroblasts; CCL, C-C motif chemokine ligand; CX3CL1, C-X3-C motif chemokine ligand 1; CXCL, C-X-C motif chemokine ligand; EC, endothelial cells; GAL8, galectin-8; GAS6, growth arrest specific 6; HSP, heat-shock protein; LCN2, lipocalin 2; LEC, lymphatic EC; LYVE1, lymphatic vessel endothelial hyaluronan receptor 1; M-CSF, macrophage colony-stimulating factor; PDPN, podoplanin; ROS, reactive oxygen species; RS-1, receptor stabilin-1; SEMA7A, semaforin 7A; ST2, suppression of tumorigenicity 2; TAMs, tumor-associated macrophages; TEM, Tie2-expressing macrophages; Tie2, TEK receptor tyrosine kinase; TNFR, TNF receptor.

THP-1 human monocytes co-cultured with human oral squamous cell carcinoma (OSCC)-derived CAFs acquired the M2 phenotype, which manifested in elevated expression levels of M2 markers IL-10 and TGFβ and increased adhesion, compared with control propidium monoazide-treated macrophages (34). Triple co-culture of THP-1 monocytes, CAFs and OSCC cells revealed a more pronounced effect on monocyte differentiation and further macrophage polarization (34). Furthermore, tumor cells from colon, breast and oral cancer stimulate CAFs to produce higher levels of IL-6, CXCL5 and granulocyte M-CSF (GM-CSF/CSF-2) (34). CAFs isolated from human lung squamous cell carcinoma expressed monocyte- and neutrophil-attracting chemokines, including CCL2, CCL7, CXCL1, CXCL5 and CXCL8 (35). CAFs in co-culture with macrophages exhibited higher expression levels of M-CSF, IL-6, IL-8, hepatocyte growth factor and CCL2 (36). In this in vitro system, macrophages were differentiated from peripheral blood monocytes of healthy donors, and CAFs were obtained by mesenchymal stem cell co-cultivation with esophageal squamous cell carcinoma cell lines (36). CAFs obtained from human breast cancer (BC) tissue induced the recruitment of monocytes via CCL2 and stromal cell-derived factor-1 (SDF-1), and promoted M2 polarization (37).

In human hepatocellular carcinoma (HCC), CAFs express endosialin, a transmembrane glycoprotein that is also expressed in tumor cells (38). Endosialin can interact with CD68, thereby recruiting macrophages into the TME (38). Endosialin-positive CAFs can promote M2 polarization of macrophages through growth arrest specific 6 factor secretion (Fig. 1A) (38). In the TME, CAFs are the main producers of CXCL12 and CXCL16, which are monocyte chemoattractants that induce M2 polarization (29). In TAMs differentiated from the THP-1 cell line, CXCL12 stimulated the secretion of plasminogen activator inhibitor-1, inducing tumor cell proliferation, angiogenesis, metastasis, EMT, cell apoptosis inhibition and drug resistance (29). Immunohistochemistry (IHC) analysis of primary triple-negative BC (TNBC) revealed that both tumor cells and CAFs expressed CXCL16 (39). CM of CAFs isolated from human tumors affected the migration of human peripheral blood monocytes and, to a lesser extent, M2 macrophages (39). Monocyte migration towards CXCL16 was more pronounced compared with that towards CXCL12 (39). In vitro, CAFs isolated from human prostate carcinoma tissue induced the recruitment of macrophages, obtained from healthy donor monocytes, towards tumor cells and macrophage M2 polarization via SDF-1 (40). Other factors, including CCL2 and CXCL14, mainly affect myeloid cell recruitment to the tumor site; however, to the best of our knowledge, their role in macrophage polarization has not yet been studied (40).

CAFs facilitate M2 macrophage polarization

CAFs serve pivotal roles not only in monocyte recruitment and differentiation but also in M2 polarization in tumors (28,39). Cross-talk between CAFs and TAMs mediated by different factors has been described in a number of studies and in various tumor types, including esophageal and lung cancer (35,36,41). In vitro, CAFs derived from human CRC tissue induced monocyte differentiation and M2 TAM polarization via the upregulation of M2 markers CD206 and CD163 (28). CAFs promoted M2 recruitment in a CRC xenograft mouse model by increasing VCAM-1 expression in tumor cells (28). Flow cytometry analysis of tumor tissue demonstrated that the expression levels of VCAM-1 in CRC cells were associated with higher infiltration of CD11b+CD68+ cells, specifically CD163+CD206+ TAMs (28). A positive association between CD163/CD206 and VCAM-1 expression in colon adenocarcinoma was observed (28). TAMs induced by CAFs promoted the migration of SW480 CRC cells and suppressed NK cell functions in vitro (28). Increased tumor cell invasion has been demonstrated in collagen gel co-cultures with TAMs and CAFs or in triple co-culture with colon cancer cells (HCT116 and DLD-1) (27). Human monocyte-derived TAMs weakly induced invasion, whereas human colon cancer-derived CAFs exhibited a more pronounced effect (27). CM derived from co-culture of CAFs and TAMs had the most prominent effect on tumor cell migration compared with CM from mono- and triple co-cultures (27).

CM obtained from CAFs isolated from human pancreatic ductal adenocarcinoma (PDAC) tissue induced M2 polarization of human peripheral blood monocytes in a co-culture system, as well as in monoculture (42). Stimulation of human peripheral blood monocytes by pancreatic CAF CM induced CD206 expression and increased reactive oxygen species production. M-CSF produced by CAFs is responsible for generating oxidative stress resulting in M2 polarization (Fig. 1A) (42). In vitro, CAF-stimulated TAMs enhanced pancreatic tumor cell proliferation, invasion and migration (42). Another factor involved in macrophage polarization is heat-shock protein 90 α (HSP90α) (43). CAFs isolated from murine PDAC tissue promoted M2 polarization in mouse RAW264.7 macrophages by secreting HSP90α, which induced a feedforward loop of HSP90α secretion in macrophages (43). In CRC, EndoMT-derived CAFs also exhibit high HSP90α expression (17).

In a transgenic mouse model of BC, genetic ablation of chitinase 3 like 1 (CHI3L1) in CAFs attenuated tumor growth, macrophage recruitment and M2 polarization (44). In vivo, injection of CHI3L1 facilitated macrophage infiltration and increased angiogenesis (44). Several studies have reported that overexpression of IL-33 in CAFs induced repolarization of M1 macrophages into M2 macrophages (45,46). Genome-wide expression profiling demonstrated upregulation of M2-related genes [arginase 1 (Arg1), C-C motif chemokine receptor (CCR)3, Cd163, Cdh1, F13a1, Hmox1, Il1r2, mannose receptor C-type 1 (MRC1), Pdcdl1g2, Serpinb2 and Stab1] in IL-33-stimulated macrophages compared with non-stimulated macrophages (46). In a study in humans, as well as in a mouse model, it has been demonstrated that IL-33-stimulated TAMs actively promoted cancer metastasis (45,46). The possible mechanism is associated with the IL-33-NF-κB-MMP9-laminin axis (45,46).

CAFs express fibroblast-specific protease fibroblast-activated protein (FAP), which is involved in metastasis and modification of collagen matrices, as well as inflammation (47). In vitro and in vivo, class A scavenger receptor (SR-A)-expressing macrophages exhibited higher adhesion to FAP-cleaved collagen (47). In a BC mouse model, CAFs increased peritoneal macrophage adhesion to FAP-cleaved type I collagen recognized by macrophages (47). Macrophages with a lack of SR-A on their surface exhibited decreased attachment compared with SR-A-expressing ones (47).

Macrophages induce transdifferentiation of fibroblasts

Macrophages are able to affect 'mesenchymal-mesenchymal transition' (MMT) in fibroblasts, thereby activating their pro-tumor phenotype (48). MMT is the process during which fibroblasts acquire the properties of mesenchymal cells after undergoing transdifferentiation into myofibroblasts (48). In a human prostate cancer model, M2 macrophages activated healthy fibroblasts through MMT and converted them into CAFs, which are traditionally referred to as myofibroblasts (40,49). M1 macrophages are also able to activate fibroblasts but to a lesser extent than M2 macrophages (40). In a TNBC model, macrophages actively expressed CD163 and the immunosuppressive factors S100A9 and collagen VI, activating CAFs (39). Myeloid cells induced the functional differentiation of fibroblasts into α-smooth muscle actin (αSMA)+ CAFs. In turn, CAFs facilitated the migration of monocytes via CXCL16 secretion (39).

In conclusion, the evidence indicates that CAFs serve a major role in recruiting and pro-tumor polarization of monocytes/macrophages, while TAMs can induce transdifferentiation of CAFs.

3. Blood vessel ECs

The vascular system is a highly branched network lined by ECs that supply tissues with oxygen and nutrients (50). Lack of oxygen is a primary stimulus for angiogenesis (50). During this process, ECs require nutrients and energy to maintain motility and biosynthesis of biomolecules, which are essential for cell duplication (50). Tumor cells produce several factors that induce angiogenesis, including VEGF, the main angiogenesis activator, fibroblast growth factor (FGF), which stimulates EC proliferation and induces neovascularization, angiopoetin 2 (Ang2), responsible for remodeling and stabilization of novel capillary tubes, IL-8, which induces EC proliferation, and MMP2, which is a key factor for basement membrane degradation and ECM remodeling (51). Tumor-associated vessels are structurally abnormal; they are dilated, convoluted and exceptionally permeant due to transcellular holes and the lack of the basement membrane (51,52). Additionally, tumor vessel walls may consist of both endothelial and tumor cells (51-53). Tumor cells recruit ECs from adjacent tissues to invade into the tumor stroma and form neovessels (54). Tumor ECs (TECs) exhibit higher expression levels of proangiogenic factors, such as VEGFR-1, VEGFR-2, VEGFR-3, VEGF-D, angiopoietin receptor TEK receptor tyrosine kinase (Tie2) and angiopoietin 1, compared with normal ECs (53). TECs also express adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1), VCAM-1 and E-selectin, which are necessary for interaction with stromal and tumor cells (53). TECs are highly proliferative and self-sustaining (53).

TAMs serve an emerging role in the stimulation of neoangiogenesis (55). IHC analysis of PDAC mice tissue revealed a positive association between M2 subpopulations and microvessel density (MVD) (56). An association between CD31 and CD206, as well as CD31 and CD163, was identified (56). In a lung adenocarcinoma model, M2 macrophages exhibited high microRNA-942 expression, which was associated with lung cancer metastasis (56). M2 macrophage-derived exosomes consistently increased the migration of both tumor cells and HUVECs, and increased the formation of blood vessels in vitro and in vivo (56).

Macrophages can contribute to tumor angiogenesis by secreting basic FGF, macrophage migration inhibitory factor, platelet-activating factor, prostaglandin E2, osteopontin, adrenomedullin, VEGF-A, epidermal growth factor, placental growth factor, TGF-β, TNFα, IL-1β, IL-8, CCL2, CXCL8 and CXCL12 (57-59). A specific subpopulation of TAMs, which highly expresses immune checkpoint molecule B7 homolog 3 protein, also known as CD276, promoted angiogenesis and induced an immunosuppressive TME in a mouse model of TNBC (60).

TAMs can induce vascular mimicry, a formation of new blood vessels associated with an acquisition of vascular cell features or functions by tumor cells of a non-vascular origin (61). This phenomenon has been described in BC, ovarian cancer, prostate cancer, lung cancer and glioma (61,62). Macrophage-induced vascular mimicry is an IL-6- and cyclooxygenase-2 (COX-2)-dependent process (63). Vascular mimicry density is positively associated with M2 polarization (63). This is explained by the presence of hypoxia in the TME, which promotes macrophage infiltration and vessel formation (63).

ECs recruit monocytes/macrophages

Monocytes and TAMs can be recruited to the perivascular sites by ECs themselves (63). ECs express Ang2, stabilin-1 and C-X3-C motif chemokine ligand 1 (CX3CL1), facilitating the recruitment of macrophages (63). In vitro, Tie2+ monocytes differentiated from THP-1 cells actively migrated towards EC-derived Ang2 (Fig. 1B) (64). High Ang2 expression is associated with increased MVD and poor prognosis in several cancer types, including high grade serous ovarian, lung and gastric cancer (63,65,66). Ang2 expression is upregulated in response to anti-VEGF therapy, resulting in treatment failure due to increased TAM infiltration (64). Therefore, anti-VEGF and anti-Ang2 combined therapy can improve the effectiveness of anti-angiogenic therapy (67).

ECs are able to recruit monocytes and TAMs through direct interaction via adhesion molecules (68). Stabilin-1 (Clever-1) is an adhesion molecule that is expressed by ECs and involved in monocyte/macrophage and regulatory T cell (Treg) recruitment (68,69). Macrophages interact with stabilin-1 via the scavenger receptor stabilin-1 (Fig. 1B) (70). Stabilin-1 blockade in mice decreased monocyte binding to tumor vessels by 70%, whereas no inhibition of lymphocyte binding was observed (69).

In a mouse model of ovarian cancer, apoptosis signal-regulating kinase 1, expressed by the vascular endothelium, was responsible for EC activation and TAM recruitment, without affecting lymphocyte recruitment (71). CX3CL1 expression in ECs is involved in C-X3-C motif chemokine receptor 1 (CX3CR1)-dependent recruitment of NK cells, CD8+ T cells and CX3CR1+ non-classical monocytes (63). CX3CL1 exists both in a soluble form, acting as a monocyte chemoattractant, and in a membrane form, promoting CX3CR1 monocyte adhesion (Fig. 1B) (63).

Perivascular macrophages

In perivascular sites, macrophages are present in two forms: Migratory TAMs and perivascular TAMs (PvTAMs) (72). Migratory TAMs are responsible for tumor cell migration, thereby promoting cancer metastasis (73). An in vitro migration assay demonstrated that rat mammary adenocarcinoma tumor cells migrating in streams with TAMs, obtained from murine bone marrow, moved at higher speeds, from greater distances towards HUVECs coated onto Sephadex beads compared with tumor cells migrating without TAMs (73). PvTAMs are a specific subpopulation of TAMs residing close to blood vasculature (<15-20 µm), which serves an important role in proangiogenic niche formation and tumor metastasis (74,75). PvTAMs are associated with increased tumor angiogenesis, distant metastasis, poor prognosis and tumor recurrence after chemotherapy (75). PvTAMs are found in structures defined as the TME of metastasis (TMEM), and are responsible for VEGF-A production, resulting in transient vascular leakiness, as well as tumor cell extravasation and distant metastasis (76). The TMEM consists of PvTAMs, invasive tumor cells and ECs (76). In macrophage-depleted mice, the CD68+ TAMs amount returned to normal levels in 4 days, and TAMs were rarely found in tight proximity to CD31+ ECs (72). TAMs accumulated in perivascular sites and developed PvTAM properties after 7 days (72). It has been demonstrated that PvTAMs are originated from CCR2+ monocytes and are recruited to the perivascular space by perivascular CAFs (77). Perivascular CAFs secrete CXCL12, IL-33 and SDF-1α in response to cancer cell activation (63). High infiltration of macrophages in vascular sites can be responsible for pro-metastatic niche formation (78). In a BC model, perivascular macrophages were activated by the cancer cell-derived ECM protein tenascin (TNC) (78). PvTAMs contributed to vascular niche formation via nitric oxide and TNF secretion, activating ECs (78). TNC deficiency decreased metastasis formation in vivo (78). In BC, PvTAMs are originated mainly from bone marrow-derived interstitial macrophages, but not from resident macrophages, and have a mixed M1/M2 phenotype (78). Immunofluorescence analysis of metastatic lung tissue demonstrated that VEGFR1+F4/80+ interstitial macrophages were found in close proximity to CD31+ ECs (78). This suggested their pivotal role in induction of the perivascular niche (78). PvTAMs in mouse mesentery mainly express the M2 marker CD206 (79). Co-culture of M1 and M2 TAMs with ECs demonstrated that M2 TAMs exhibited greater binding to ECs compared with M1 TAMs (79). Immunofluorescence analysis of human glioblastoma multiforme (GBM) tissues revealed that CD68+CD206+ macrophages were localized in proximity to CD31+ ECs (80). Similarly, in an orthotopic genetic murine GBM model, F4/80+CD206+ macrophages were localized near ECs (80). GBM-derived ECs induced M2 polarization of macrophages via M-CSF and IL-6 expression (80). IL-33 secreted by pericytes and CAFs promotes M2 polarization and induced MMP9 expression in M2 TAMs facilitating metastasis development via the IL-33-ST2-NF-κB-MMP9-laminin pathway in a mouse model of pancreatic cancer (Fig. 1B) (46). PvTAMs can serve an immunosuppressive role in perivascular sites (75). They exhibit high secretion of IL-10, which leads to cytotoxic T cells and Treg suppression in perivascular sites (75).

Anticancer therapy causes vascular damage and hypoxia, leading to increased recruitment of myeloid cells, proliferation of ECs and migration of pericytes, which are involved in reconstruction of the vessels in order to restore oxygen levels and provide nutrients for tissue repair (81). The main role in this process is assigned to the Ang-Tie2 pathway (63,75). Tie2 is a receptor that is broadly expressed on vascular ECs and on a subpopulation of macrophages [Tie2-expressing macrophages (TEMs)] (75). Ang2 is a proangiogenic factor and chemokine for TEMs, which also serves a role in regulating the recruitment and/or activation of perivascular macrophages in the TMEM (75). TEMs are a critical subpopulation involved in tumor angiogenesis and are able to adhere to ECs (81). TEMs are essential for vascular anastomosis and formation of new vessels (75,81). Ang2 acts as an inducer of M2 polarization and Tie2+ monocyte recruitment via the upregulation of M2 markers (IL-10 and MRC1) and angiogenesis-related genes (cathepsin B and thymidine phosphorylase) (63).

In a mouse model of fibrosarcoma, tumor relapse after chemotherapy was associated with active vessel reconstruction within 14 days (81). FACS analysis revealed that the proportion of CD11b+Tie2+ cells was increased compared with that in untreated mice (81). Gene expression profiling of TEMs isolated from mouse mammary tumors revealed increased expression of pro-tumoral genes, including MMP9, VEGFA, CXCL12, toll like receptor 4, neuropilin 1 (NRP1) and platelet derived growth factor (PDGF) subunit B (75). Immunofluorescence analysis of the TMEM obtained from tumor-bearing mice demonstrated that a major fraction of PvTAMs was Tie2+/VEGFA+ macrophages (82). VEGFA expression in TEMs was elevated compared with that in the adjacent ECs and surrounding tumor tissue (82). TMEM-associated macrophages exhibited expression of the TEM markers MRC1, CD11b and F4/80 (82). Anti-VEGFA treatment decreased the amount of circulating tumor cells and vascular permeability (82). In a mammary mouse model, Tie2+ PvTAMs increased vessel permeability via VEGF-A-dependent destabilization of EC junctions (Fig. 1B) (82).

Another specific subpopulation of PvTAMs exhibit lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) expression, which is considered to be a marker for lymphatic endothelium (77). In a mouse model of BC, LYVE1 was expressed by pro-angiogenic TAMs located near the blood vasculature (77). LYVE1+ PvTAMs formed nest structures in perivascular niches in a CCR5-dependent manner (77). CCR5 depletion led to decreased tumor growth in mice (77). Impaired nest formation resulted in increased chemosensitivity in tumor-bearing mice treated with fluorouracil (77). LYVE1+ PvTAMs are also responsible for αSMA+ CAFs expansion within the perivascular niche (Fig. 1B) (74). PvTAMs are a major source of PDGFC in the perivascular niche, which is the ligand for PDGF receptor α expressed on αSMA+ CAFs (74). PDGF blockade led to decreased migration of αSMA+ CAFs (74). This observation revealed reciprocal interactions between PvTAMs and perivascular CAFs (PvCAFs), since PvCAFs are responsible for monocyte/macrophage recruitment into perivascular sites (77).

4. LECs

Several studies have demonstrated the important role of TAMs in tumor lymphangiogenesis (83-87). TAMs are positively associated with lymph node metastasis and lymphatic vessel density (LVD) in numerous cancer types, including lung cancer, tongue OSCC, pancreatic cancer, gastric cancer, cervical cancer and ovarian cancer (88-95).

VEGF-C is a known lymphangiogenic factor that is most essential for LEC proliferation and lymphatic vessel sprouting (96). TAM expression of VEGF-C is stimulated by different stimuli, such as TNFα/TNF receptor 1 (TNFR1) signaling (97-99), IL-1β secretion by tumor cells (98), and via VEGFR activation by endothelial cell-derived VEGF-C and VEGF-D (Fig. 1B) (99,100).

TAMs induce lymphangiogenesis

In a murine BC model, sphingolipid sphingosine-1-phosphate receptor 1 (S1PR1) overexpression in TAMs promoted pulmonary metastasis and tumor lymphangiogenesis in vivo (101). The mechanism included the upregulation of inflammasome-related gene NLR family pyrin domain containing 3 in S1PR1+ TAMs followed by IL-1β production (101). Lung-derived LECs stimulated by IL-1β exhibit increased VEGF-C expression and a high proliferation rate (101). M2 TAMs activated by tumor cell-derived IL-1 promote lymphangiogenesis and lymph node metastasis through the secretion of VEGF-A and VEGF-C (102). S1P protein derived from dying breast tumor cells stimulated the expression of lipocalin 2 (LCN2) in TAMs in mouse mammary tumor virus-polyoma middle tumor-antigen mice (103). LCN2 promoted LEC differentiation and tube formation via activation of VEGF-C expression (103).

TNFα stimulates morphological changes and migration of LECs in a VEGFR-3-dependent manner in vitro (97). Interaction of tumor cell-derived TNFα with macrophage TNFR1 triggers VEGF-C secretion by CD206+ TAMs, amplifying lymphangiogenesis and lymphatic metastasis in a murine lung cancer model (97). TNFα and IL-1β secretion by tumor cells stimulated VEGF-C expression in macrophages in a co-culture of a melanoma cell line and murine peritoneal macrophages (98). Macrophages differentiated from monocytes of healthy donors promoted BC cell adhesion to LECs in vitro in an IL-1β-dependent manner, suggesting a possible mechanism of lymphatic metastasis (104). It has been suggested that lymph node metastasis associated transcript 1-induced CCL2 expression in bladder cancer cells could promote TAM recruitment, which contributed to lymphangiogenesis and lymphatic metastasis via VEGF-C expression (105).

CM from co-culture of cervical cancer cell lines and macrophages differentiated from THP-1 monocytes stimulated tube formation of human LECs (93). IL-1β, IL-8, VEGF-C and VEGF-A secretion was elevated in CM (93). LECs isolated after a tube formation assay exhibited increased expression levels of VEGFR-3 and podoplanin (PDPN) (93). In vitro, CM from TAMs isolated from human epithelial ovarian cancer (EOC) ascites induced LEC proliferation, migration and tube formation (95).

The interaction between TAMs differentiated from human peripheral monocytes and LECs isolated from metastatic lymph nodes of human gastric cancer has been examined in co-culture (106). Expression levels of MMP9 and adhesion molecules (CD44, neural cell adhesion molecule 1, ICAM-1, VCAM-1, E-selectin and osteopontin) were upregulated in capillary-forming LECs (106). TAMs in contact co-culture with LECs exhibited increased expression levels of various cytokines, including IL-1β, IL-6, IL-8, IL-10, IL-12 and IL-18 (106). CXCL2 expression was upregulated, whereas CXCL6 and CXCR2 expression was downregulated in macrophages in contact with LECs (106). In human papillary thyroid carcinoma, both M2 TAMs and cancer cells secrete MMP2 responsible for lymphatic vessel basement membrane degradation that could further lead to lymphatic invasion (107).

Lymphatic macrophage subpopulations

In a murine BC 4T1 model, a specific subpopulation of TAMs was localized near lymphatic vessels (83). TAMs isolated from 4T1 tumors exhibited high expression levels of PDPN, a heavily O-glycosylated small mucin-type transmembrane glycoprotein, and were referred to as PDPN-expressing macrophages (PoEMs) (83). PoEMs interact with LECs via PDPN-galectin 8 interaction and promote tumor lympangiogenesis (83). PDPN knockout in mice led to reduced tumor lymphatic growth and lymphatic metastasis (83). In murine mammary tumors with high SEMA7 expression, an elevated amount of PDPN+ TAMs was observed (84). In vitro, treatment of murine macrophages with recombinant SEMA7A increased PDPN expression as well as macrophage motility and adherence to the human dermal LEC monolayer and promoted lymphangiogenesis (84). The adhesion of macrophages to the LEC is mediated by the upregulation of β1 integrin and the interaction of PDPN with its receptor CLEC-2 (Fig. 1B) (84).

IHC staining of human cervical squamous cell carcinoma tissue demonstrated that, under hypoxic conditions, lymphatic vessels were encapsulated by CD163+ TAMs, and the LVD was higher compared with that under normoxic conditions (108). These structures were referred to as lymphatic vessels encapsulated by TAMs (LVEM) (108). Hypoxic TAMs exhibited high IL-10 expression (108). A popliteal lymphatic metastasis mouse model revealed that IL-10 promoted lymphangiogenesis and LVEM formation (108). TAM-derived IL-10 activated LEC production of CCL1, which resulted in further TAM recruitment and sustained LVEM formation and lymphatic metastasis (108).

VEGF-C and VEGF-D-expressing TAMs form small clusters close to lymphatic microvessels and tumor surfaces of human cervical squamous carcinoma (99). All VEGF-C and VEGF-D-expressing TAMs also express VEGFR-3, but in a granular intracellular pattern, and not on the cell surface membrane (99). In human mammary carcinoma, VEGF-C-expressing TAMs have been reported as morphologically different from other macrophages (109). They were generally larger, oval or irregular in shape and had abundant cytoplasm, and were located mainly in peritumoral regions (109). Macrophage numbers and VEGF-C expression were positively associated with LVD and LEC proliferation (109).

TEMs that interact with ECs can also be enriched in lymphovascular regions (85). TEMs express β4 integrin, which interacts with LECs that express laminin-5, a β4 integrin ligand (85). In human BC tissues, TEMs are mainly found in the tumor center and tumor invasive edges and account for 95% of the whole population of CD14+ cells (86). Confocal microscopy analysis of human BC tissue revealed that TEMs expressed the lymphatic markers EGFR-2, VEGFR-3, LYVE1, PDPN and prospero homeobox 1 protein (PROX-1) (86). TEMs exhibited lymphangiogenic activity (86). Injection of human breast tumor-derived TEMs into BC xenograft mice resulted in increased tumor size and vascularization (86). Injection of TEMs into mouse cornea resulted in 800-fold higher LVD compared with that of cornea injected with buffer only (87). Additionally, TEMs are associated with lymph node metastasis in patients with BC (86).

Myeloid cells can contribute to lymphangiogenesis by differentiating into myeloid-LEC progenitors (M-LECP) (110-112). In human BC, M-LECP have been identified as cells co-expressing CD68 and LEC markers (LYVE1, PDPN, VEGFR-3 and PROX-1) (113). Human M-LECPs are usually identified by additional expression of a marker of progenitor cells, such as CD133 (111). The density of M-LECP is associated with LVD and lymphatic metastasis in human BC (113).

LYVE1+ macrophages have been reported to be closely localized to the developing embryonic lymphatic vasculature in mice (114). In some cases, LYVE1+ macrophages were integrated into jugular lymph sacs and dermal lymphatic vessels (114). Reverse transcription-quantitative PCR analysis revealed that LYVE1+ macrophages expressed LYVE1, MRC1, Cd163, Stab1, NRP1, Plxnd1 and Sema6d, which revealed overlap with the gene signature of embryonic and tumor-derived TEMs, suggesting a role of LYVE1+ macrophages in tumor lymphangiogenesis (114).

5. CD4+ T cells

CD4+ lymphocytes include several subsets: T helper 1 (Th1), T helper 2 (Th2), Tregs and follicular helper T cells (115). Tumor-infiltrating CD4+ cells express increased levels of forkhead box P3 (Foxp3; specific for Treg), IL-17 (specific for Th17), and Th2-associated cytokines IL-4 and IL-13 (115). Cytokines secreted by Th1 lead to the classical activation of M1 macrophages, and cytokines secreted by Th2 contribute to the polarization of macrophages towards M2 (116,117). The cytokine profile of Th1 and M1 macrophages is associated with the stimulation of antitumor immunity, in contrast to the cytokine profile of Th2 and M2 macrophages, which are pro-tumoral (117).

Th1

Th1 cytokines affect the polarization of macrophages, and the latter, in turn, can change the differentiation of Th1 and Th2 (117). The predominance of Th1 in the tumor inhibits the maturation of tumor-infiltrating CD11b+F4/80+ macrophages isolated from mouse liver cancer tissue (117). It has also been suggested that CD11b+F4/80+ macrophages, rather than Tregs, serve a major role in Th1 suppression (117). Fragile site-associated tumor suppressor (Fats)−/− bone marrow-derived macrophages (BMDMs) isolated from fat-deficient mice stimulated CD4+ T lymphocyte differentiation to the Th1 phenotype in vitro (118). In vivo, a similar effect was observed when Fats−/− BMDMs were transferred to mice injected subcutaneously with melanoma cells (118). CD4+ Th1 cytokines activate CD8+ T cells, which display antitumor functions in vivo (118). In addition, CD4+ Th1 produce IFN-γ, which promotes additional polarization of macrophages towards the M1 phenotype (Fig. 2) (118). Receptor-interacting serine/threonine protein kinase 1 (RIP1) regulated macrophage differentiation into a pro-tumor phenotype, and RIP1 inhibition in TAMs altered the macrophage phenotype towards M1 in a mouse model of PDAC (119). This induced Th differentiation toward a mixed Th1/Th17 phenotype via a TNFα-dependent mechanism (119). The antitumor effect of gemcitabine in combination with anti-programmed cell death protein 1 (PD-1) antibodies was mediated by activation of Th1 and M1 macrophages in a mouse model of liver metastasis of pancreatic cancer (120).

Th2

Th2, similarly to M2 macrophages, are pro-tumor cells, and associated with tumor progression and metastasis (121,122). Th2 cytokines are involved both in repolarization of macrophages towards M2, and in the suppression of Th1 and activation of tumor growth signaling pathways (121). In a mouse model of spontaneous pancreatic islet carcinogenesis, tumor macrophages with altered polarization after irradiation of the pancreatic region synthesized an increased level of inducible NOS (iNOS; specific for M1 macrophages), which activated the cytokine profile of Th1 and inhibited Th2 cytokines (121). M2 pro-tumor macrophage activity was enhanced by CD4+ cells expressing higher levels of IL-4, IL-13 and IL-10 (specific for Th2) compared with IFNγ (specific for Th1) or IL-17 (specific for Th17) expression in a mouse BC model (Fig. 2) (123). In another study using a mouse model of BC, it was demonstrated that depletion of TAMs in the tumor resulted in a decrease in the number of CD4+ T cells that express Th2 cytokines (122).

Th17

Th17 are a subset of CD4+ T cells that predominantly synthesize IL-17 (115,123). Th17 serve a role in inflammatory and autoimmune diseases, including cancer development (124,125). Th17 in cancer are considered as pro-inflammatory cells with an antitumor function; however, there are conflicting results depending on the type and stage of cancer (125-129). A high level of Th17 is associated with improved survival in patients with ovarian cancer (126). Th17 is positively associated with IFN-γ+CD8+ and IFN-γ+CD4+ T cells but negatively associated with Tregs (126). In vitro, TAMs isolated from ovarian tumors induce Th17 predominantly through IL-1β synthesis (Fig. 2). Despite the fact that TAMs serve an important role in the activation of Th17 in the TME, the increase in the amount of Th17 is limited by Tregs (126). A high Treg/Th17 ratio in patients with EOC is associated with increased tumor burden, shorter survival time and cancer metastasis (127). The imbalance in the Treg/Th17 ratio is mediated by exosomes released from human monocyte-derived TAMs (127). Exosomes contain microRNA, which suppresses the synthesis of pro-inflammatory cytokines (IL-6 and TNFα) and the anti-inflammatory cytokine IL-4 by T cells, and promotes the activation of anti-inflammatory cytokines (TGFβ and IL-10) (127). In a mouse intestinal adenoma model, production of IL-17 promoted tumor growth (128). In vivo, blockade of IL-1β synthesized by intestinal macrophages reduced the differentiation of Th17 in tumors and reduced the growth of polyps in the intestine (128). Furthermore, in patients with gastric cancer, an increase in Th17 has been observed at an early stage and their decrease has been observed in advanced stages of cancer (129).

Tregs

Tregs belong to the population of CD4+ T lymphocytes, serving a crucial role in adaptive immunity by suppressing immune cells to attack tumor cells (130). In normal non-tumor tissues, Tregs are important regulators of autoimmune reactions (130). Tregs can be recruited to pathological sites, e.g., inflammation and tumor sites (131). This process occurs due to a gradient of C-C motif and C-X-C motif chemokines, which are expressed by abnormal cells (131). Tregs induced by the TME differ from the host ones by exhibiting more pronounced immunosuppressive activity (132). Usually, Tregs are defined as Foxp3+ cells; however, due to inherent heterogeneity they include both immunosuppressive and non-suppressive cells (133). This fact complicates the study of Tregs in various types of cancer and the use of Tregs in anticancer therapy (133).

An increase in the number of TAMs along with Tregs is observed during carcinogenesis and associated with unfavorable prognosis in patients with colorectal (134), lung (135) and ovarian (136) cancer. In the tumor stroma of lung cancer, MARCO-expressing macrophages have an immunosuppressive phenotype, increasing the proliferation of Tregs, which is associated with a negative prognosis (135). In liver cancer in mice, intratumor triggering receptor expressed on myeloid cells 1 (TREM-1)+ macrophages suppress the activity of CD8+ T cells by activating Tregs (137). The inhibition of TREM-1+ macrophages does not eliminate the depletion of CD8+ T cells, indicating that Tregs cause CD8+ T cell dysfunction (137).

Direct interaction of TAMs with Tregs has been demonstrated in numerous studies (137-141). TAMs synthesize a large amount of chemoattractants that facilitate the recruitment of Tregs in tumors (Fig. 2) (138). The key chemokines include CCL17 and CCL22 (138). Therapeutic use of immunomodulator imiquimod reduced the production of CCL22 of CD11b+ TAMs in a melanoma model, resulting in a decrease in the population of Foxp3+ Tregs and an increase in the antitumor response (139). In the hypoxic tumor environment, TREM-1+ macrophages activated the synthesis of the CCL20 ligand, which has an affinity to the CCR6 receptors on Tregs, stimulating Treg recruitment, immunosuppression and tumor growth in a mouse orthotopic liver tumor model (137). Macrophages isolated from malignant pleural effusions of patients with lung cancer exhibit increased CCL22 expression (140). CCL22 induces the recruitment of Tregs, which, through positive feedback, affect macrophages via the production of IL-8 (140). IL-8 increases the production of TGF-β by macrophages, which suppresses the immune system and induces carcinogenesis (140). TAMs from lung (3LL-R) tumor-bearing mice were treated with agonists to liver X receptor (a transcription factor from the nuclear receptor family), which inhibited the secretion of CCL22 and CCL17 from TAMs (141). The macrophages were then stimulated in vitro with GM-CSF or IL-4, and a decrease in the synthesis of CCL17 and CCL22 by the M2 macrophages was observed (141). This was associated with a decrease in the number of Tregs in a mouse model of syngeneic Lewis lung carcinoma (LLC) (141).

In a mouse CRC model, it was found that the homing and migration of Tregs into the tumor depended on the interaction of CCR6 receptor with ligand CCL20 produced by both tumor cells and macrophages (142). Macrophages isolated from the tumor tissue of patients with CRC express high levels of CXCL11, which has an affinity to CXCR3 receptor expressed on Foxp3+IL-17+ Tregs (143). The depletion of macrophages by injection of diphtheria toxin into CD11b diphtheria toxin receptor (CD11b-DTR) mice decreased CCL20 mRNA and protein expression in the CRC tissue, indicating that TAMs are required for release of CCL20 from tumor cells and recruitment of Tregs (Fig. 2) (142). In mice injected with melanoma or colon adenocarcinoma cells, Tregs inhibited the activity of tumor-suppressing CD8+ T cells, which in turn led to an increase in the M2 macrophage phenotype (144).

M2 macrophages derived from human peripheral blood monocytes induce T cell differentiation towards Tregs via IL-10 secretion (136). In patients with EOC, an increased number of TAMs and Tregs, and elevated levels of IL-10 were found in tissue samples, and these were associated with decreased survival time (136). In a mouse model of ovarian cancer, TAMs secreted exosomes containing microRNAs, which affected the differentiation of CD4+ T cells and increased the number of immunosuppressive Tregs via the STAT3 mechanism (127). In vitro, recombinant human IL-23 secreted predominantly by TAMs enhanced the immunosuppressive activity of Tregs, as indicated by increased expression levels of PD-1 and CD69, as well as the secretion of IL-10 and TGFβ (Fig. 2) (145).

Co-cultivation of CD4+CD25Foxp3 T cells with M2 macrophages stimulated by the supernatant of a laryngeal carcinoma cell line resulted in the change of phenotype of T cells towards CD4+CD25+Foxp3+ Tregs (146). In turn, the cultivation of Tregs with tumor cells and human peripheral blood-derived monocytes stimulated the expression of M2-specific markers in monocytes (146). Tregs can express an increased level of ligand-activated aryl hydrocarbon receptor transcription factor (AHR) (147). AHR-expressing Tregs exhibit increased secretion of immunosuppressive factors [IL-10, VEGF and cytotoxic T-lymphocyte associated protein 4 (CTLA-4)] (147). Co-cultivation of Tregs pretreated with kynurenine, which activates AHR, resulted in the predominance of the M2 macrophage phenotype (147).

6. CD8+ T lymphocytes

CD8+ T lymphocytes are one of the key elements of adaptive immunity in infectious and oncological diseases (148,149). CD8+ cells are activated by interacting with major histocompatibility complex-1 molecules and various costimulatory signals and cytokines (148). Cytotoxic CD8+ T cells are able to recognize and directly participate in the destruction of transformed pathogenic carrier cells, in particular tumor cells (149). An increased amount of CD8+ T lymphocytes is associated with improved patient survival (150).

During tumor development, M2-polarized macrophages can affect CD8+ T lymphocytes by suppressing their cytotoxic activity, while M1 TAMs are associated with a higher density of CD8+ T cells (151). The antitumor immune response is mediated by T cell-dependent reprogramming of TAMs towards the M1 phenotype in tumor tissue, which has been demonstrated in a mouse model of LLC (152). Direct contact of CD8+ cells with stromal CD11c+CD206+ TAMs decreased T cell motility followed by their exhaustion in human lung squamous-cell carcinoma (150). In a mouse model of lung adenocarcinoma, the depletion of tissue-resident macrophages by injection of diphtheria toxin into CD169-DTR mice contributed to the active accumulation of CD8+ T cells in tumor tissues, reducing tumor invasiveness and growth (153). In mouse model of HCC, a CCR2 antagonist inhibited tumor infiltration by macrophages, which was associated with an increase in the number and cytotoxicity of CD8+ T cells and a decrease in tumor size (154).

TAMs regulate the recruitment of CD8+ T cells to the tumor site

CD8+ T lymphocytes are recruited to the tumor by both tumor cells themselves and macrophages (155-158). The chemokines CCL5, CXCL9 and CXCL10 are critical for attracting CD8+ T cells, which has been demonstrated in various cancer types: Breast, colon, ovarian, lung and uterus cancer (155-158). The chemokine receptors CCR5 (specific to CCL5) and CXCR3 (specific to CXCL9) have been found on the surface of T cells (155,156). TAM-derived CCL2 does not directly affect the activity of CD8+ T cells; however, its overexpression reduces the synthesis of CXCL9, one of the key CD8+ T cell chemoattracting cytokines (159). The effect of CCL5 is lower than that of CXCL9 since CXCR3 is predominantly expressed on stem PD-1low CD8+ T cells, while CCR5 is increased on exhausted PD-1high CD8+ T cells (155). The expression of CCL5 and CXCL9 is associated with the expression of CD8A, a cytotoxic T cell glycoprotein involved in intercellular interaction (156). In the tumor tissue of patients with lung cancer, M1 macrophages are the main source of CXCL9 (151). In a mouse model of HCC, high expression of the Sonic hedgehog protein activated the 'hedgehog' pathway associated with an increase in the number of M2 macrophages and a decrease in the expression of the chemoattracting cytokines CXCL9 and CXCL10, followed by the inhibition of CD8+ T cell migration (158).

CCL5 is predominantly expressed by tumor cells and stimulates macrophages to synthesize CXCL9 (155). Tumor-infiltrating lymphocytes express IFNγ, which in turn stimulates CXCL9 synthesis in macrophages (Fig. 2) (156). In a mouse model of HCC, CXCL10 together with CXCL9 affected the cytotoxic index of CD8+ T cells (158). The decrease in the expression of these cytokines was associated with an increase in tumor infiltration by CD163+ TAMs (158).

TAMs induce apoptosis in CD8+ cells

In a mouse liver metastasis model, hepatic monocyte-derived CD11b+F4/80+ macrophages could induce immunosuppression of CD8+ T lymphocytes via Fas-Fas ligand (FasL) interaction (160). Binding of FasL, expressed by macrophages, to Fas on the surface of CD8+ T cells leads to the activation of the apoptotic pathways in CD8+ T cells (160). In mouse colon carcinoma, fibrosarcoma, methylcholanthrene-induced sarcoma and lymphoma models, F4/80+ macrophages isolated from tumor tissue had higher levels of iNOS and arginase I inhibited T cell activity and activated their apoptosis (161).

TAMs suppress the cytotoxicity of CD8+ T cells

M-CSF affects the proliferation and differentiation of macrophages via M-CSFR receptor (162). The M-CSF/M-CSFR axis is able to suppress the cytotoxicity of CD8+ T cells (163). The use of anti-bodies blocking M-CSFR reduced the infiltration of TAMs and increased CD8+ T cell activation in a mouse colon carcinoma model (163). A decrease in M-CSF expression increases the expression of IFN-γ by T cells (164). The IFNγ-IFNγ receptor 1 axis between CD8+ T cells and TAMs reduces macrophage polarization into the M2 phenotype (Fig. 2) (144).

In subcutaneous mouse breast and colon tumor models, monoacylglycerol lipase deficiency resulted in lipid accumulation in TAMs and their polarization into M2 phenotype, resulting in reduced CD8+ T cell activity (165). The decreased expression of COX-2 increased the number of TAMs, which was associated with an increase in CD8+ T cell infiltration and improved survival of mice in a mammary carcinoma model (166). Blockage of COX-2 reduced the expression of macrophage pro-tumor markers (arginase-1, IL-10 and iNOS), which reversed the suppression of tumor-infiltrating lymphocytes (166).

P65 is one of the proteins of the NF-κB family, and its activation leads to increased expression of T-cell inhibitory molecule B7x (also referred to as B7-H4 or B7S1) on the TAM surface and a decrease in IL-10 in TAMs, suppressing the activation of tumor-infiltrating CD8+ T lymphocytes during lung tumorigenesis (167). However, inactivation of macrophage PI(3) kinase, which is an inhibitor of NF-κB, promoted CD8+ T cell activation in tumors in mouse models of implanted human papillomavirus (HPV)+ and HPV head and neck squamous cell, lung and breast carcinoma (168).

Antigen presentation to CD8+ T lymphocytes

Macrophages and DCs are antigen-presenting cells (169). The ability of macrophages to present antigens to T lymphocytes can lead to exhaustion of T cells; however, this property can be used in therapy to stimulate T cell cytotoxicity (170-172). Interferon regulatory factor 8, as a transcription factor, controls DC-like properties in TAMs and induces antigen acquisition and presentation by macrophages (170). Furthermore, macrophages presenting tumor cell antigens depleted CD8+ T cells in a mouse BC model (170). Macrophages express signal regulatory protein α interacting with the CD47 'do not eat me' signal on tumor cells, which leads to the termination of phagocytosis of tumor cells by TAMs (Fig. 2) (171). The use of anti-CD47 antibodies promotes efficient phagocytosis of cancer cells, antigen presentation to CD8+ T cells and stimulation of CD8+ T cell activity by TAMs in vivo and in vitro (171). DCs interacting with macrophages also regulate the cytotoxic response in CD8+ T cells (172). In BC, macrophages produce IL-10, which suppresses the production of IL-12 by DCs (172). IL-12 is required for the cytotoxic response of CD8+ T cells but its blockade inhibits the T cell response (172).

Interaction between macrophages and CD8+ T cells via programmed death-ligand 1 (PD-L1)/PD-1

Inside the tumor, TAMs and DCs are the main sources of PD-L1 (173,174), while CD8+ T cells express the PD-1 receptor (150,159). The PD-L1/PD-1 interaction axis inhibits the cytotoxic activity of T lymphocytes (173), which reduces the effectiveness of the use of immune checkpoint blockade agents (155). An increased amount of progranulin, which is expressed in a number of tumors, could lead to the upregulation of PD-L1 in macrophages, followed by their polarization towards the M2 phenotype and suppression of the activity of CD8+ T cells in a mouse BC model (173).

Blockade of PD-1 enhances CD8+ T cell activity via increased production of IFN-γ and TNFα (Fig. 2) (174,175). One of the key markers of the anti-PD-1-induced antitumor response is CXCL9, which is a chemoattracting and activating cytokine for CD8+ T cells (175). In tumor samples from patients with recurrent glioblastoma who received neoadjuvant anti-PD-1 therapy, the possibility of enhancing the interaction between the CD8+ receptor CXCR3 and CXCL9/10 was confirmed using a bioinformatic interactome analysis based on single-cell data (176).

Several studies have demonstrated that anti-PD-1 therapy combined with macrophage inhibitors is more efficient in terms of increasing the immune response and suppression of TAM activity compared with mono-immunotherapy (150,159,163). In mouse models of glioblastoma and ovarian cancer, anti-PD-1 therapy in combination with the blockade of LIF (anti-IL-6 class cytokine that affects cell proliferation) increased survival and promoted tumor regression more effectively than mono-therapy (159). The use of a M-CSF inhibitor in combination with anti-PD-1 therapy increased the migration and infiltration of CD8+ T cells in a mouse BC model (150). Another macrophage inhibitor, lenvatinib, in combination with an anti-PD-1 agent, increased the percentage of CD8+ T cells, followed by a decrease in tumor growth and vascular invasion in a mouse colon carcinoma model (163).

7. B cells

B cells are important components of the immune defense against cancer. Opinions on the role of B cells in tumor carcinogenesis vary based on collected evidence on both pro-tumor and antitumor activity of B cells (177-181). For example, pro-tumor activity of B cells can be mediated by the recruitment of TAMs via immune complexes and Fcγ receptor interactions, whereas the beneficial effect of B cells is related to the activation of antitumor immunity through antigen presentation or direct cytotoxicity towards tumor cells via secretion of granzyme B and TNF-related apoptosis-inducing ligand (179,181). The complexity of studying B cells in carcinogenesis is associated with difficulties in isolating and purifying B cells (180). Furthermore, there are several functional phenotypes depending on the TME (177).

Evidence indicates that B cells and macrophages can have a common bipotential progenitor in bone marrow (182). In the early stages of development, pre-B cells co-expressing B-cell and myeloid markers can differentiate into macrophages, due to the receptors of B cells not being rearranged, which indicates the plasticity of B cells (182).

When studying the cross-talk of Bruton's tyrosine kinase (BTK)-expressing immune cells in a mouse model of pancreatic carcinoma and in vitro, the important role of the interaction between B cells and macrophages was noted (179). Activation of macrophage Fc receptors initiates BTK signaling via the PI3K-dependent signaling pathway and activates macrophage repolarization towards the M2 phenotype and promotes tumor development (179). Tumor B cells enhance the change in macrophage phenotype in favor of type 2 by influencing macrophage Fc receptors (179).

The antitumor or pro-tumor function of B cells can be mediated by antibody synthesis (179). B-cell-expressed IgG binds to macrophages and induces macrophage-mediated phagocytosis of tumor cells through antibody-dependent cellular cytotoxicity in breast, pancreatic and gastric cancer but not in clear cell renal cell cancer, squamous cell carcinoma and non-small cell lung cancer (179). IgG secretion by B cells is associated with poor prognosis (183).

Despite these findings, to the best of our knowledge, the direct mechanisms of the interaction of TAMs and B cells in the TME remain unknown (Fig. 3).

8. NK cells

NK cells are part of the innate immune system (184). Together with macrophages, they are among the first cells recruited to the site of infection (185). NK cells have cytotoxic activity against infected or transformed cells that helps to kill tumor cells even when the latter do not express major histocompatibility complex class 1 or its expression is reduced (184,186).

In turn, the TME can reduce the cytotoxic activity of NK cells against tumor cells and inhibit the expression of NK receptors responsible for the recognition and killing of tumor cells (187,188). Macrophages are able to change the expression of activating and inhibiting NK cell receptors (187,189-192). M2 TAMs suppress the cytotoxic activity of NK cells (193). Co-culture of NK cells isolated from the murine spleen and peritoneal or bone marrow M2 macrophages revealed suppression of the expression of CD27 activation marker, while co-culture with M1-like macrophages under similar conditions did not suppress the cytotoxicity of NK cells (187).

In patients with stage II and III esophageal cancer, high tumor infiltration by TAMs and low infiltration by NK cells are associated with poor prognosis (194). Macrophages/monocytes isolated from autologous nontumor and tumor tissues of patients with gastric cancer cultured together with allogeneic peripheral blood NK cells reduced the expression of IFNγ and TNFα in NK cells co-cultured with tumor-associated monocytes/macrophages (190). In addition, a decrease in the expression of cell proliferation marker Ki-67 by NK cells was observed, which together indicated a decrease in the functional activity of NK cells (190). MARCO-expressing TAMs exhibited immunosuppressive properties against NK cells in a mouse melanoma model (195).

One of the mechanisms by which TAMs interact with NK cells is the secretion of TGFβ1 by macrophages/monocytes, which, in a gastric cancer ex vivo model, induced functional impairment of NK cells (190). In vitro, TGFβ1 affected the expression of activation receptors, natural cytotoxicity triggering receptor 3 (NKp30) and killer cell lectin like receptor K1 (NKG2D), in NK cells isolated from human peripheral blood mononuclear cells (191). A decrease in the expression of NKp30 and NKG2D suppresses the cytotoxic activity of NK cells against a melanoma cell line (191). In peripheral blood NK cells from patients with prostate cancer, TGFβ is able to increase the surface expression of CD56 and CD9 receptors, which decrease the cytotoxic activity of NK cells, and reduce the expression of NK cell activation receptor NKG2D in vitro (192). Similarly, in a mouse lymphoma model, M2-like macrophages suppressed the activity of NK cells by secreting TGF-β, which decreased the expression of degranulation factor CD107a by NK cells (187).

Activation of the natural cytotoxicity receptors 46 and DNAX accessory molecule-1 receptors on NK cells results in the lysis of human peripheral blood monocyte-derived M2 macrophages (196,197). Resting NK cells do not exhibit cytolytic activity against macrophages; however, activated NK cells lyse M0 and M2 macrophages (Fig. 3) (197). In co-culture, monocytes obtained from peripheral blood and synovial fluid of patients with inflammatory arthritis could activate NK cells via increased expression of the proliferation marker CD69 and elevated secretion of IFN-γ by NK cells (198). Additionally, NK cells increased the secretion of the pro-inflammatory cytokine TNFα by monocytes (198). IFN-γ secreted by NK cells altered macrophage polarization to an M1 phenotype in a mouse model of sarcoma (199).

In addition to antitumor activity, NK cells can be involved in pro-tumor polarization of TAMs (192). Co-cultivation of human MDMs with NK cells isolated from the blood of patients with prostate cancer revealed a decrease in the expression of M1-like factors (IL-12, TNFα, IFNγ and IL-1β) and an increase in the expression of M2-like factors (CD206, Arg1, IL-10, TGFβ and CXCL8) (Fig. 3) (192).

9. Neutrophils

Neutrophils are immune cells that serve an important role in the development of inflammation (200). Similar to macrophages, neutrophils are classified into N1 and N2: Antitumor and pro-tumor, respectively (201). Activated neutrophils are able to secrete IL-8 and TNFα, which recruit macrophages into the inflammation site (202). The interaction of macrophages with neutrophils likely occurs through the macrophage mannose receptor highly expressed by macrophages and through neutrophil-secreted myeloperoxidase (Fig. 3) (202). Neutrophil-derived CCL2 and CCL17 affected the migration of macrophages into the tumor in a mouse model of HCC (203). In another study, granulocyte colony-stimulating factor receptor-depleted mice had neutropenia, which was associated with increased tumor progression, compared with wild-type mice in a 3-methylcholanthrene-induced sarcoma model (200). An increase in neutropenia was associated with an increase in the expression of the M2 macrophage genes and was associated with poor prognosis (200). Tumor monocytes derived from patients with non-small cell lung cancer are also able to affect the migration of neutrophils via CXCL3, a ligand for the neutrophil receptor CXCR2 (204).

The co-cultivation of tumor cells simultaneously with macrophages and neutrophils resulted in increased proliferation, invasion and colony formation of tumor cells compared with those of tumor cells cultured with a monoculture of macrophages or neutrophils (205). The data were confirmed in three intrahepatic cholangiocarcinoma cell lines: SG231, RBE and HuCCT1 (205).

10. Combined therapeutic approaches based on TAMs and other cells of the TME

Anti-CAF and anti-TAM combined therapy can be a promising option in antitumor immunotherapy (206-208). In murine models of lung cancer, melanoma and colon cancer, immune modifying poly(lactic-co-glycolic acid) nanoparticles (referred to as ONP-302) were used to target CAFs and TAMs (209). The authors of the previous study suggested that ONP-302 was able to alter the polarization of TAMs and MDSCs from pro-tumor to antitumor state. The same mechanism was described for CAFs (209).

Combined anti-VEGFA/Ang2 and anti-CD40 therapy led to vascular pruning and normalization of blood vessels, myeloid cell activation, including M1-like TAM skewing in colon cancer, melanoma and BC models in vivo (210). Tie2 expression in TAMs and ECs could be another possible target for combined therapy (211). The Tie2-targeting anti-tumor hydrophobic peptide T4 was modified to generate self-assembly nanoparticles (P-T4) (211). T4 interacted with Tie2 and inhibited signal transduction, cell migration and angiogenesis (211). P-T4 exerted an inhibitory effect on the viability of Tie2-positive RAW264.7 cells and ECs in vitro, and reduced vessel density and distant metastasis formation in vivo (211). SAR131675 is a dose-dependent selective inhibitor of VEGFR-3 tyrosine kinase activity (212). In a murine BC model, SAR131675 inhibited primary human lymphatic cells in vitro and reduced the migration of cancer cells into lymph nodes and lungs in tumor-bearing mice (212). IHC staining of tumor tissues isolated from murine mammary tumors revealed reduced macrophage infiltration, suggesting that reduction of tumor growth might be associated with decreased TAM infiltration (212).

The use of immunotherapy against both macrophages and Tregs can help improve the prognosis of patients with cancer, since there is a positive association between TAMs and Tregs (146). Mono-immunotherapy against TAMs or Tregs was not efficient in a colon cancer in vivo model (213). Deletion of the CSF1 gene in colon adenocarcinoma cells (MC38 line) led to a decrease in the population of M2 macrophages in vivo but an increase in the number of Foxp3+ Tregs followed by slight tumor growth in mice (213). In turn, depletion of Foxp3+ Tregs in tumor-bearing mice upon administration of diphtheria toxin resulted in an increase in colony-stimulating factor 1 receptor (CSF1R)+ TAMs and slight tumor growth (213). In a mouse model of head and neck cancer, radiotherapy in combination with anti-CD25 depletion of Tregs resulted in reduced tumor growth and an increase in the M1/M2 ratio (214). Notably, the use of radiotherapy alone led to an increase in the proportion of M2 macrophages (214).

In a mouse pancreatic cancer model, a CSF1R blockade and PD-1/CTLA-4 antagonist combination therapy reduced tumor progression more effectively than single therapy (215). In a mouse melanoma model, the use of the CSF1R inhibitor in combination with the transfer of naive CD8+ T cells activated with a synthetic peptide vaccine resulted in a reduction in tumor growth (216). The use of regorafenib (an inhibitor of protein kinases, including VEGFR, CSF1R and others) in combination with anti-PD1 therapy inhibited tumor development due to a decrease in M2 macrophages and Tregs, and an increase in CD8+ T cells and M1 macrophages inside the tumor in a mouse colon cancer model (217). In mice with gastrointestinal stromal tumors, anti-CD40 therapy activated TAMs against tumor cells and recruited more monocytes from the bone marrow (218). Additionally, the efficacy of treatment with the combination of anti-CD40 with imatinib (affects the KIT mutation characteristic of this type of cancer) was completely dependent on the presence of macrophages in the tumor and partially on the presence of CD8+ T cells (218).

Various interleukins are widely used in cancer immunotherapy (195,219,220). Pro-inflammatory cytokines such as IL-12, IL-15 and IL-18 are involved in the regulation of activity, proliferation and cytotoxicity of NK cells and other cells of myeloid origin (221). In a mouse model of melanoma, combination therapy with a DNA vaccine based on endoglin and IL-12 gene therapy was applied (220). Combination therapy resulted in the re-polarization of TAMs from M2 phenotype to M1 phenotype, which was associated with an increase in the number of NK cells by >3 times compared with the control group (220). Treatment of mouse macrophages with polyinosinic:polycytidylic acid (stimulator of type I IFN) revealed an increase in the synthesis of ligands for NKG2D NK cell receptor, which increased NK cell cytotoxic activity against lymphoma tumor cells in vitro (222). In a mouse model of melanoma treated with AIP therapy that included a combination of an antitumor antibody ('A'), an extended half-life IL-2 ('I'), and an anti-PD-1 ('P'), the therapeutic effect was dependent on the presence of NK cells and macrophages in the tumor, which, under the influence of therapy, were reprogrammed into an antitumor phenotype (223).

During the development of methods of therapy for pancreatic adenocarcinoma, it was revealed that the simultaneous inhibition of CCR2+ macrophages and CXCR2+ neutrophils enhanced antitumor immunity (224).

11. Conclusions

The present review demonstrates the broad network of TAM interactions with stromal and immune components of the TME via cell-to-cell and extracellular mechanisms. The collected data indicate that CAFs are an essential source in the TME for monocyte recruitment and M2 TAM polarization. Despite the involvement of TAMs in ECM remodeling, they have a minor impact on CAF activity, only inducing MMT. After polarization, TAMs are able to interact with naive fibroblasts and promote their differentiation into functional CAFs, creating a self-sustaining loop.

TAMs are also involved in tumor angiogenesis and lympangiogenesis via reciprocal interaction with blood ECs and LECs, respectively. ECs are able to recruit TAMs to the tumor site, where the latter are differentiated toward perivascular and lymphatic vessel-associated macrophage subpopulations promoting proliferation and migration of ECs, as well as growth of novel vessels.

The crosstalk between TAMs and other immune cells can be crucial for the balance between the pro-tumor and anti-tumor immune state and regulation of immunosuppression.

Finally, combined targeting of TAMs and other cells of the TME can be decisive to achieve advanced efficacy of anticancer treatment. Most studies have focused on targeting stromal or immune cells alone (225-228). However, it is critical to not exclude cell-cell crosstalk within the TME and future studies should aim to identify possible targets in these interactions in order to increase the effectiveness of antitumor therapy. Combined therapeutic approaches should be considered to prevent distant metastasis and reduce the risk of tumor recurrence.

Availability of data and materials

Not applicable.

Authors' contributions

The research project was designed, organized and reviewed by IL, and reviewed and critiqued by JK. AK, TS, OK and IL performed the literature research, wrote the first draft of manuscript and prepared the figures. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Ms. Militsa Rakina (Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia) for the professional English editing.

Funding

The present study was supported by Tomsk State University Development Programme (Priority-2030; grant no. HY 2.1.1.22 MJI).

Abbreviations:

BMDM

bone marrow-derived macrophage

CAF

cancer-associated fibroblast

CM

conditioned medium

DC

dendritic cell

EC

endothelial cell

ECM

extracellular matrix

EMT

epithelial-mesenchymal transition

EndoMT

endothelial-mesenchymal transition

IHC

immunohistochemistry

LEC

lymphatic endothelial cell

LVD

lymphatic vessel density

MDM

monocyte-derived macrophage

MMT

mesenchymal-mesenchymal transition

MVD

microvessel density

NK cell

natural killer cell

PoEM

podoplanin-expressing macrophage

PvTAM

perivascular tumor-associated macrophage

TAM

tumor-associated macrophage

TEC

tumor endothelial cell

TEM

Tie2-expressing macrophage

TME

tumor microenvironment

TMEM

tumor microenvironment of metastasis

References

1 

Yuan Y, Jiang YC, Sun CK and Chen QM: Role of the tumor microenvironment in tumor progression and the clinical applications (Review). Oncol Rep. 35:2499–2515. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Whiteside TL: The tumor microenvironment and its role in promoting tumor growth. Oncogene. 27:5904–5912. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Werb Z and Lu P: The role of stroma in tumor development. Cancer J. 21:250–253. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, Kolahian S, Javaheri T and Zare P: Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 18:592020. View Article : Google Scholar : PubMed/NCBI

5 

Larionova I, Tuguzbaeva G, Ponomaryova A, Stakheyeva M, Cherdyntseva N, Pavlov V, Choinzonov E and Kzhyshkowska J: Tumor-associated macrophages in human breast, colorectal, lung, ovarian and prostate cancers. Front Oncol. 10:5665112020. View Article : Google Scholar : PubMed/NCBI

6 

Lin Y, Xu J and Lan H: Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J Hematol Oncol. 12:762019. View Article : Google Scholar : PubMed/NCBI

7 

Pan Y, Yu Y, Wang X and Zhang T: Tumor-associated macrophages in tumor immunity. Front Immunol. 11:5830842020. View Article : Google Scholar : PubMed/NCBI

8 

Larionova I, Kazakova E, Patysheva M and Kzhyshkowska J: Transcriptional, epigenetic and metabolic programming of tumor-associated macrophages. Cancers (Basel). 12:14112020. View Article : Google Scholar : PubMed/NCBI

9 

Gao J, Liang Y and Wang L: Shaping polarization of tumor-associated macrophages in cancer immunotherapy. Front Immunol. 13:8887132022. View Article : Google Scholar : PubMed/NCBI

10 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Marusyk A, Almendro V and Polyak K: Intra-tumour heterogeneity: A looking glass for cancer? Nat Rev Cancer. 12:323–334. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Gordillo CH, Sa ndova l P, Muñoz-Her nández P, Pascual-Antón L, López-Cabrera M and Jiménez-Heffernan JA: Mesothelial-to-mesenchymal transition contributes to the generation of carcinoma-associated fibroblasts in locally advanced primary colorectal carcinomas. Cancers (Basel). 12:4992020. View Article : Google Scholar : PubMed/NCBI

13 

Shiga K, Hara M, Nagasaki T, Sato T, Takahashi H and Takeyama H: Cancer-associated fibroblasts: Their characteristics and their roles in tumor growth. Cancers (Basel). 7:2443–2458. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Adjuto-Saccone M, Soubeyran P, Garcia J, Audebert S, Camoin L, Rubis M, Roques J, Binétruy B, Iovanna JL and Tournaire R: TNF-α induces endothelial-mesenchymal transition promoting stromal development of pancreatic adenocarcinoma. Cell Death Dis. 12:6492021. View Article : Google Scholar

15 

Ciszewski WM, Sobierajska K, Wawro ME, Klopocka W, Chefczyńska N, Muzyczuk A, Siekacz K, Wujkowska A and Niewiarowska J: The ILK-MMP9-MRTF axis is crucial for EndMT differentiation of endothelial cells in a tumor microenvironment. Biochim Biophys Acta Mol Cell Res. 1864:2283–2296. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Fan CS, Chen LL, Hsu TA, Chen CC, Chua KV, Li CP and Huang TS: Endothelial-mesenchymal transition harnesses HSP90α-secreting M2-macrophages to exacerbate pancreatic ductal adenocarcinoma. J Hematol Oncol. 12:1382019. View Article : Google Scholar

17 

Fan CS, Chen WS, Chen LL, Chen CC, Hsu YT, Chua KV, Wang HD and Huang TS: Osteopontin-integrin engagement induces HIF-1α-TCF12-mediated endothelial-mesenchymal transition to exacerbate colorectal cancer. Oncotarget. 9:4998–5015. 2017. View Article : Google Scholar

18 

Chen C, Li WJ, Weng JJ, Chen ZJ, Wen YY, Deng T, Le HB, Zhang YK and Zhang BJ: Cancer-associated fibroblasts, matrix metalloproteinase-9 and lymphatic vessel density are associated with progression from adenocarcinoma in situ to invasive adenocarcinoma of the lung. Oncol Lett. 20:1302020. View Article : Google Scholar : PubMed/NCBI

19 

Xie J, Qi X, Wang Y, Yin X, Xu W, Han S, Cai Y and Han W: Cancer-associated fibroblasts secrete hypoxia-induced serglycin to promote head and neck squamous cell carcinoma tumor cell growth in vitro and in vivo by activating the Wnt/β-catenin pathway. Cell Oncol (Dordr). 44:661–671. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Fullár A, Dudás J, Oláh L, Hollósi P, Papp Z, Sobel G, Karászi K, Paku S, Baghy K and Kovalszky I: Remodeling of extracellular matrix by normal and tumor-associated fibroblasts promotes cervical cancer progression. BMC Cancer. 15:2562015. View Article : Google Scholar : PubMed/NCBI

21 

Erdogan B and Webb DJ: Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans. 45:229–236. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Ren J, Smid M, Iaria J, Salvatori DCF, van Dam H, Zhu HJ, Martens JWM and Ten Dijke P: Cancer-associated fibroblast-derived Gremlin 1 promotes breast cancer progression. Breast Cancer Res. 21:1092019. View Article : Google Scholar : PubMed/NCBI

23 

Takahashi M, Kobayashi H, Mizutani Y, Hara A, Iida T, Miyai Y, Asai N and Enomoto A: Roles of the mesenchymal stromal/stem cell marker meflin/Islr in cancer fibrosis. Front Cell Dev Biol. 9:7499242021. View Article : Google Scholar : PubMed/NCBI

24 

Attieh Y, Clark AG, Grass C, Richon S, Pocard M, Mariani P, Elkhatib N, Betz T, Gurchenkov B and Vignjevic DM: Cancer-associated fibroblasts lead tumor invasion through integrin-β3-dependent fibronectin assembly. J Cell Biol. 216:3509–3520. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Zhang J, Fu B, Li M and Mi S: Secretome of activated fibroblasts induced by exosomes for the discovery of biomarkers in non-small cell lung cancer. Small. 17:e20047502021. View Article : Google Scholar

26 

Glentis A, Oertle P, Mariani P, Chikina A, El Marjou F, Attieh Y, Zaccarini F, Lae M, Loew D, Dingli F, et al: Cancer-associated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane. Nat Commun. 8:9242017. View Article : Google Scholar : PubMed/NCBI

27 

Stadler M, Pudelko K, Biermeier A, Walterskirchen N, Gaigneaux A, Weindorfer C, Harrer N, Klett H, Hengstschläger M, Schüler J, et al: Stromal fibroblasts shape the myeloid phenotype in normal colon and colorectal cancer and induce CD163 and CCL2 expression in macrophages. Cancer Lett. 520:184–200. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Zhang R, Qi F, Zhao F, Li G, Shao S, Zhang X, Yuan L and Feng Y: Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 10:2732019. View Article : Google Scholar : PubMed/NCBI

29 

Chen S, Morine Y, Tokuda K, Yamada S, Saito Y, Nishi M, Ikemoto T and Shimada M: Cancer-associated fibroblast-induced M2-polarized macrophages promote hepatocellular carcinoma progression via the plasminogen activator inhibitor-1 pathway. Int J Oncol. 59:592021. View Article : Google Scholar :

30 

Olingy CE, Dinh HQ and Hedrick CC: Monocyte heterogeneity and functions in cancer. J Leukoc Biol. 106:309–322. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Hao Q, Vadgama JV and Wang P: CCL2/CCR2 signaling in cancer pathogenesis. Cell Commun Signal. 18:822020. View Article : Google Scholar : PubMed/NCBI

32 

Gomez-Roca CA, Italiano A, Le Tourneau C, Cassier PA, Toulmonde M, D'Angelo SP, Campone M, Weber KL, Loirat D, Cannarile MA, et al: Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages. Ann Oncol. 30:1381–1392. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH and Rüttinger D: Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 5:532017. View Article : Google Scholar : PubMed/NCBI

34 

Cho H, Seo Y, Loke KM, Kim SW, Oh SM, Kim JH, Soh J, Kim HS, Lee H, Kim J, et al: Cancer-stimulated CAFs enhance monocyte differentiation and protumoral TAM Activation via IL6 and GM-CSF secretion. Clin Cancer Res. 24:5407–5421. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, Afshar R, Georgiev P, Sze MA, Song XS, et al: Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res. 8:436–450. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, Nishio M, Shigeoka M, Kakeji Y and Yokozaki H: Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 99:777–792. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Gok Yavuz B, Gunaydin G, Gedik ME, Kosemehmetoglu K, Karakoc D, Ozgur F and Guc D: Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1+ TAMs. Sci Rep. 9:31722019. View Article : Google Scholar

38 

Yang F, Wei Y, Han D, Li Y, Shi S, Jiao D, Wu J, Zhang Q, Shi C, Yang L, et al: Interaction with CD68 and regulation of GAS6 expression by endosialin in fibroblasts drives recruitment and polarization of macrophages in hepatocellular carcinoma. Cancer Res. 80:3892–3905. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Allaoui R, Bergenfelz C, Mohlin S, Hagerling C, Salari K, Werb Z, Anderson RL, Ethier SP, Jirström K, Påhlman S, et al: Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat Commun. 7:130502016. View Article : Google Scholar : PubMed/NCBI

40 

Comito G, Giannoni E, Segura CP, Barcellos-de-Souza P, Raspollini MR, Baroni G, Lanciotti M, Serni S and Chiarugi P: Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 33:2423–2431. 2014. View Article : Google Scholar

41 

Hegab AE, Ozaki M, Kameyama N, Gao J, Kagawa S, Yasuda H, Soejima K, Yin Y, Guzy RD, Nakamura Y, et al: Effect of FGF/FGFR pathway blocking on lung adenocarcinoma and its cancer-associated fibroblasts. J Pathol. 249:193–205. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Zhang A, Qian Y, Ye Z, Chen H, Xie H, Zhou L, Shen Y and Zheng S: Cancer-associated fibroblasts promote M2 polarization of macrophages in pancreatic ductal adenocarcinoma. Cancer Med. 6:463–470. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Chua KV, Fan CS, Chen CC, Chen LL, Hsieh SC and Huang TS: Octyl gallate induces pancreatic ductal adenocarcinoma cell apoptosis and suppresses endothelial-mesenchymal transition-promoted M2-macrophages, HSP90α secretion, and tumor growth. Cells. 9:912019. View Article : Google Scholar

44 

Cohen N, Shani O, Raz Y, Sharon Y, Hoffman D, Abramovitz L and Erez N: Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene. 36:4457–4468. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Zhou Q, Wu X, Wang X, Yu Z, Pan T, Li Z, Chang X, Jin Z, Li J, Zhu Z, et al: The reciprocal interaction between tumor cells and activated fibroblasts mediated by TNF-α/IL-33/ST2L signaling promotes gastric cancer metastasis. Oncogene. 39:1414–1428. 2020. View Article : Google Scholar

46 

Andersson P, Yang Y, Hosaka K, Zhang Y, Fischer C, Braun H, Liu S, Yu G, Liu S, Beyaert R, et al: Molecular mechanisms of IL-33-mediated stromal interactions in cancer metastasis. JCI Insight. 3:e1223752018. View Article : Google Scholar : PubMed/NCBI

47 

Mazur A, Holthoff E, Vadali S, Kelly T and Post SR: Cleavage of type I collagen by fibroblast activation protein-α enhances class A scavenger receptor mediated macrophage adhesion. PLoS One. 11:e01502872016. View Article : Google Scholar

48 

Cat B, Stuhlmann D, Steinbrenner H, Alili L, Holtkötter O, Sies H and Brenneisen P: Enhancement of tumor invasion depends on transdifferentiation of skin fibroblasts mediated by reactive oxygen species. J Cell Sci. 119:2727–2738. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, Dang Y, Chu Y, Fan J and He R: FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 76:4124–4135. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Potente M and Carmeliet P: The Link between angiogenesis and endothelial metabolism. Annu Rev Physiol. 79:43–66. 2017. View Article : Google Scholar

51 

Papetti M and Herman IM: Mechanisms of normal and tumor-derived angiogenesis. Am J Physiol Cell Physiol. 282:C947–C970. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Hida K and Maishi N: Abnormalities of tumor endothelial cells and cancer progression. Oral Sci Int. 15:1–6. 2018. View Article : Google Scholar

53 

Hida K, Maishi N, Takeda R and Hida Y: The roles of tumor endothelial cells in cancer metastasis. Metastasis Brisbane (AU): Sergi CM: Exon Publications; 2022, View Article : Google Scholar

54 

Wang WY, Lin D, Jarman EH, Polacheck WJ and Baker BM: Functional angiogenesis requires microenvironmental cues balancing endothelial cell migration and proliferation. Lab Chip. 20:1153–1166. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Yang D, Guo P, He T and Powell CA: Role of endothelial cells in tumor microenvironment. Clin Transl Med. 11:e4502021. View Article : Google Scholar : PubMed/NCBI

56 

Wei K, Ma Z, Yang F, Zhao X, Jiang W, Pan C, Li Z, Pan X, He Z, Xu J, et al: M2 macrophage-derived exosomes promote lung adenocarcinoma progression by delivering miR-942. Cancer Lett. 526:205–216. 2022. View Article : Google Scholar

57 

Baradaran A, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Shadbad MA, Khosravi N, Derakhshani A, Alemohammad H, Afrashteh Nour M, Safarpour H, et al: The cross-talk between tumor-associated macrophages and tumor endothelium: Recent advances in macrophage-based cancer immunotherapy. Biomed Pharmacother. 146:1125882022. View Article : Google Scholar : PubMed/NCBI

58 

Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, Movahedi K, Houbracken I, Schouppe E, Elkrim Y, et al: Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 74:24–30. 2014. View Article : Google Scholar

59 

Hughes R, Qian BZ, Rowan C, Muthana M, Keklikoglou I, Olson OC, Tazzyman S, Danson S, Addison C, Clemons M, et al: Perivascular M2 macrophages stimulate tumor relapse after chemotherapy. Cancer Res. 75:3479–3491. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Cheng N, Bei Y, Song Y, Zhang W, Xu L, Zhang W, Yang N, Bai X, Shu Y and Shen P: B7-H3 augments the pro-angiogenic function of tumor-associated macrophages and acts as a novel adjuvant target for triple-negative breast cancer therapy. Biochem Pharmacol. 183:1142982021. View Article : Google Scholar

61 

Treps L, Faure S and Clere N: Vasculogenic mimicry, a complex and devious process favoring tumorigenesis-Interest in making it a therapeutic target. Pharmacol Ther. 223:1078052021. View Article : Google Scholar

62 

Zhang L, Xu Y, Sun J, Chen W, Zhao L, Ma C, Wang Q, Sun J, Huang B, Zhang Y, et al: M2-like tumor-associated macrophages drive vasculogenic mimicry through amplification of IL-6 expression in glioma cells. Oncotarget. 8:819–832. 2017. View Article : Google Scholar :

63 

Delprat V and Michiels C: A bi-directional dialog between vascular cells and monocytes/macrophages regulates tumor progression. Cancer Metastasis Rev. 40:477–500. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Cortes-Santiago N, Hossain MB, Gabrusiewicz K, Fan X, Gumin J, Marini FC, Alonso MM, Lang F, Yung WK, Fueyo J, et al: Soluble Tie2 overrides the heightened invasion induced by anti-angiogenesis therapies in gliomas. Oncotarget. 7:16146–16157. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Sopo M, Sallinen H, Hämäläinen K, Kivelä A, Ylä-Herttuala S, Kosma VM, Keski-Nisula L and Anttila M: High expression of Tie-2 predicts poor prognosis in primary high grade serous ovarian cancer. PLoS One. 15:e02414842020. View Article : Google Scholar : PubMed/NCBI

66 

Dong Z, Chen J, Yang X, Zheng W, Wang L, Fang M, Wu M, Yao M and Yao D: Ang-2 promotes lung cancer metastasis by increasing epithelial-mesenchymal transition. Oncotarget. 9:12705–12717. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, Taus A, Albanell J, Sablin MP, Alt M, et al: First-in-human phase I study of single-agent vanucizumab, a first-in-class bispecific anti-angiopoietin-2/anti-VEGF-A antibody, in adult patients with advanced solid tumors. Clin Cancer Res. 24:1536–1545. 2018. View Article : Google Scholar

68 

Karikoski M, Irjala H, Maksimow M, Miiluniemi M, Granfors K, Hernesniemi S, Elima K, Moldenhauer G, Schledzewski K, Kzhyshkowska J, et al: Clever-1/stabilin-1 regulates lymphocyte migration within lymphatics and leukocyte entrance to sites of inflammation. Eur J Immunol. 39:3477–3487. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Karikoski M, Marttila-Ichihara F, Elima K, Rantakari P, Hollmén M, Kelkka T, Gerke H, Huovinen V, Irjala H, Holmdahl R, et al: Clever-1/stabilin-1 controls cancer growth and metastasis. Clin Cancer Res. 20:6452–6464. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Riabov V, Yin S, Song B, Avdic A, Schledzewski K, Ovsiy I, Gratchev A, Llopis Verdiell M, Sticht C, Schmuttermaier C, et al: Stabilin-1 is expressed in human breast cancer and supports tumor growth in mammary adenocarcinoma mouse model. Oncotarget. 7:31097–31110. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Yin M, Zhou HJ, Zhang J, Lin C, Li H, Li X, Li Y, Zhang H, Breckenridge DG, Ji W and Min W: ASK1-dependent endothelial cell activation is critical in ovarian cancer growth and metastasis. JCI Insight. 2:e918282017. View Article : Google Scholar : PubMed/NCBI

72 

Arwert EN, Harney AS, Entenberg D, Wang Y, Sahai E, Pollard JW and Condeelis JS: A unidirectional transition from migratory to perivascular macrophage is required for tumor cell intravasation. Cell Rep. 23:1239–1248. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Leung E, Xue A, Wang Y, Rougerie P, Sharma VP, Eddy R, Cox D and Condeelis J: Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene. 36:2680–2692. 2017. View Article : Google Scholar :

74 

Opzoomer JW, Anstee JE, Dean I, Hill EJ, Bouybayoune I, Caron J, Muliaditan T, Gordon P, Sosnowska D, Nuamah R, et al: Macrophages orchestrate the expansion of a proangiogenic perivascular niche during cancer progression. Sci Adv. 7:eabg95182021. View Article : Google Scholar : PubMed/NCBI

75 

Lewis CE, Harney AS and Pollard JW: The multifaceted role of perivascular macrophages in tumors. Cancer Cell. 30:3652016. View Article : Google Scholar : PubMed/NCBI

76 

Sparano JA, Gray R, Oktay MH, Entenberg D, Rohan T, Xue X, Donovan M, Peterson M, Shuber A, Hamilton DA, et al: A metastasis biomarker (MetaSite Breast™ Score) is associated with distant recurrence in hormone receptor-positive, HER2-negative early-stage breast cancer. NPJ Breast Cancer. 3:422017. View Article : Google Scholar

77 

Anstee JE, Opzoomer JW, Dean I, Muller HP, Bahri M, Liakath-Ali K, Liu Z, Choy D, Caron J, Sosnowska D, et al: Perivascular macrophages collaborate to facilitate chemotherapy resistance in cancer. bioRxiv. 2022.2002.2003.478952. 2022.

78 

Hongu T, Pein M, Insua-Rodríguez J, Gutjahr E, Mattavelli G, Meier J, Decker K, Descot A, Bozza M, Harbottle R, et al: Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs. Nat Cancer. 3:486–504. 2022. View Article : Google Scholar : PubMed/NCBI

79 

He H, Mack JJ, Güç E, Warren CM, Squadrito ML, Kilarski WW, Baer C, Freshman RD, McDonald AI, Ziyad S, et al: Perivascular macrophages limit permeability. Arterioscler Thromb Vasc Biol. 36:2203–2212. 2016. View Article : Google Scholar : PubMed/NCBI

80 

Wang Q, He Z, Huang M, Liu T, Wang Y, Xu H, Duan H, Ma P, Zhang L, Zamvil SS, et al: Vascular niche IL-6 induces alternative macrophage activation in glioblastoma through HIF-2α. Nat Commun. 9:5592018. View Article : Google Scholar

81 

Chen L, Li J, Wang F, Dai C, Wu F, Liu X, Li T, Glauben R, Zhang Y, Nie G, et al: Tie2 expression on macrophages is required for blood vessel reconstruction and tumor relapse after chemotherapy. Cancer Res. 76:6828–6838. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian BZ, Oktay MH, Pollard JW, Jones JG and Condeelis JS: Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov. 5:932–943. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Bieniasz-Krzywiec P, Martín-Pérez R, Ehling M, García-Caballero M, Pinioti S, Pretto S, Kroes R, Aldeni C, Di Matteo M, Prenen H, et al: Podoplanin-expressing macrophages promote lymphangiogenesis and lymphoinvasion in breast cancer. Cell Metab. 30:917–936.e10. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Elder AM, Tamburini BAJ, Crump LS, Black SA, Wessells VM, Schedin PJ, Borges VF and Lyons TR: Semaphorin 7A promotes macrophage-mediated lymphatic remodeling during postpartum mammary gland involution and in breast cancer. Cancer Res. 78:6473–6485. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Evans R, Flores-Borja F, Nassiri S, Miranda E, Lawler K, Grigoriadis A, Monypenny J, Gillet C, Owen J, Gordon P, et al: Integrin-mediated macrophage adhesion promotes lymphovascular dissemination in breast cancer. Cell Rep. 27:1967–1978.e4. 2019. View Article : Google Scholar : PubMed/NCBI

86 

Bron S, Henry L, Faes-Van't Hull E, Turrini R, Vanhecke D, Guex N, Ifticene-Treboux A, Marina Iancu E, Semilietof A, Rufer N, et al: TIE-2-expressing monocytes are lymphangiogenic and associate specifically with lymphatics of human breast cancer. Oncoimmunology. 5:e10738822015. View Article : Google Scholar

87 

Moussai D, Mitsui H, Pettersen JS, Pierson KC, Shah KR, Suárez-Fariñas M, Cardinale IR, Bluth MJ, Krueger JG and Carucci JA: The human cutaneous squamous cell carcinoma microenvironment is characterized by increased lymphatic density and enhanced expression of macrophage-derived VEGF-C. J Invest Dermatol. 131:229–236. 2011. View Article : Google Scholar

88 

Zhang BC, Gao J, Wang J, Rao ZG, Wang BC and Gao JF: Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma. Med Oncol. 28:1447–1452. 2011. View Article : Google Scholar

89 

Zhang B, Yao G, Zhang Y and Gao J, Yang B, Rao Z and Gao J: M2-polarized tumor-associated macrophages are associated with poor prognoses resulting from accelerated lymphangiogenesis in lung adenocarcinoma. Clinics (Sao Paulo). 66:1879–1886. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Yamagata Y, Tomioka H, Sakamoto K, Sato K, Harada H, Ikeda T and Kayamori K: CD163-positive macrophages within the tumor stroma are associated with lymphangiogenesis and lymph node metastasis in oral squamous cell carcinoma. J Oral Maxillofac Surg. 75:2144–2153. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Kurahara H, Takao S, Maemura K, Mataki Y, Kuwahata T, Maeda K, Sakoda M, Iino S, Ishigami S, Ueno S, et al: M2-polarized tumor-associated macrophage infiltration of regional lymph nodes is associated with nodal lymphangiogenesis and occult nodal involvement in pN0 pancreatic cancer. Pancreas. 42:155–159. 2013. View Article : Google Scholar

92 

Wu H, Xu JB, He YL, Peng JJ, Zhang XH, Chen CQ, Li W and Cai SR: Tumor-associated macrophages promote angiogenesis and lymphangiogenesis of gastric cancer. J Surg Oncol. 106:462–468. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Ding H, Cai J, Mao M, Fang Y, Huang Z, Jia J, Li T, Xu L, Wang J, Zhou J, et al: Tumor-associated macrophages induce lymphangiogenesis in cervical cancer via interaction with tumor cells. APMIS. 122:1059–1069. 2014.PubMed/NCBI

94 

Utrera-Barillas D, Castro-Manrreza M, Castellanos E, Gutiérrez-Rodríguez M, Arciniega-Ruíz de Esparza O, García-Cebada J, Velazquez JR, Flores-Reséndiz D, Hernández-Hernández D and Benítez-Bribiesca L: The role of macrophages and mast cells in lymphangiogenesis and angiogenesis in cervical carcinogenesis. Exp Mol Pathol. 89:190–196. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Zhang W, Tian J and Hao Q: HMGB1 combining with tumor-associated macrophages enhanced lymphangiogenesis in human epithelial ovarian cancer. Tumour Biol. 35:2175–2186. 2014. View Article : Google Scholar

96 

Sweat RS, Sloas DC and Murfee WL: VEGF-C induces lymphangiogenesis and angiogenesis in the rat mesentery culture model. Microcirculation. 21:532–540. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Ji H, Cao R, Yang Y, Zhang Y, Iwamoto H, Lim S, Nakamura M, Andersson P, Wang J, Sun Y, et al: TNFR1 mediates TNF-α-induced tumour lymphangiogenesis and metastasis by modulating VEGF-C-VEGFR3 signalling. Nat Commun. 5:49442014. View Article : Google Scholar

98 

Peppicelli S, Bianchini F and Calorini L: Inflammatory cytokines induce vascular endothelial growth factor-C expression in melanoma-associated macrophages and stimulate melanoma lymph node metastasis. Oncol Lett. 8:1133–1138. 2014. View Article : Google Scholar : PubMed/NCBI

99 

Schoppmann SF, Birner P, Stöckl J, Kalt R, Ullrich R, Caucig C, Kriehuber E, Nagy K, Alitalo K and Kerjaschki D: Tumor-associated macrophages express lymphatic endothelial growth factors and are related to peritumoral lymphangiogenesis. Am J Pathol. 161:947–956. 2002. View Article : Google Scholar : PubMed/NCBI

100 

Mazzone M and Bergers G: Regulation of blood and lymphatic vessels by immune cells in tumors and metastasis. Annu Rev Physiol. 81:535–560. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Weichand B, Popp R, Dziumbla S, Mora J, Strack E, Elwakeel E, Frank AC, Scholich K, Pierre S, Syed SN, et al: S1PR1 on tumor-associated macrophages promotes lymphangiogenesis and metastasis via NLRP3/IL-1β. J Exp Med. 214:2695–2713. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Watari K, Shibata T, Kawahara A, Sata K, Nabeshima H, Shinoda A, Abe H, Azuma K, Murakami Y, Izumi H, et al: Tumor-derived interleukin-1 promotes lymphangiogenesis and lymph node metastasis through M2-type macrophages. PLoS One. 9:e995682014. View Article : Google Scholar : PubMed/NCBI

103 

Jung M, Ören B, Mora J, Mertens C, Dziumbla S, Popp R, Weigert A, Grossmann N, Fleming I and Brüne B: Lipocalin 2 from macrophages stimulated by tumor cell-derived sphingosine 1-phosphate promotes lymphangiogenesis and tumor metastasis. Sci Signal. 9:ra642016. View Article : Google Scholar : PubMed/NCBI

104 

Storr SJ, Safuan S, Ahmad N, El-Refaee M, Jackson AM and Martin SG: Macrophage-derived interleukin-1beta promotes human breast cancer cell migration and lymphatic adhesion in vitro. Cancer Immunol Immunother. 66:1287–1294. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Chen C, He W, Huang J, Wang B, Li H, Cai Q, Su F, Bi J, Liu H, Zhang B, et al: LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment. Nat Commun. 9:38262018. View Article : Google Scholar : PubMed/NCBI

106 

Tauchi Y, Tanaka H, Kumamoto K, Tokumoto M, Sakimura C, Sakurai K, Kimura K, Toyokawa T, Amano R, Kubo N, et al: Tumor-associated macrophages induce capillary morphogenesis of lymphatic endothelial cells derived from human gastric cancer. Cancer Sci. 107:1101–1109. 2016. View Article : Google Scholar : PubMed/NCBI

107 

Kabasawa T, Ohe R, Aung NY, Urano Y, Kitaoka T, Tamazawa N, Utsunomiya A and Yamakawa M: Potential role of M2 TAMs around lymphatic vessels during lymphatic invasion in papillary thyroid carcinoma. Sci Rep. 11:11502021. View Article : Google Scholar : PubMed/NCBI

108 

Chen XJ, Wei WF, Wang ZC, Wang N, Guo CH, Zhou CF, Liang LJ, Wu S, Liang L and Wang W: A novel lymphatic pattern promotes metastasis of cervical cancer in a hypoxic tumour-associated macrophage-dependent manner. Angiogenesis. 24:549–565. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Ding M, Fu X, Tan H, Wang R, Chen Z and Ding S: The effect of vascular endothelial growth factor C expression in tumor-associated macrophages on lymphangiogenesis and lymphatic metastasis in breast cancer. Mol Med Rep. 6:1023–1029. 2012. View Article : Google Scholar : PubMed/NCBI

110 

Ran S and Wilber A: Novel role of immature myeloid cells in formation of new lymphatic vessels associated with inflammation and tumors. J Leukoc Biol. 102:253–263. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Ran S and Volk-Draper L: Lymphatic endothelial cell progenitors in the tumor microenvironment. Adv Exp Med Biol. 1234:87–105. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Ran S and Montgomery KE: Macrophage-mediated lymphangiogenesis: The emerging role of macrophages as lymphatic endothelial progenitors. Cancers (Basel). 4:618–657. 2012. View Article : Google Scholar : PubMed/NCBI

113 

Volk-Draper L, Patel R, Bhattarai N, Yang J, Wilber A, DeNardo D and Ran S: Myeloid-derived lymphatic endothelial cell progenitors significantly contribute to lymphatic metastasis in clinical breast cancer. Am J Pathol. 189:2269–2292. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Gordon EJ, Rao S, Pollard JW, Nutt SL, Lang RA and Harvey NL: Macrophages define dermal lymphatic vessel calibre during development by regulating lymphatic endothelial cell proliferation. Development. 137:3899–3910. 2010. View Article : Google Scholar : PubMed/NCBI

115 

Dannenmann SR, Thielicke J, Stöckli M, Matter C, von Boehmer L, Cecconi V, Hermanns T, Hefermehl L, Schraml P, Moch H, et al: Tumor-associated macrophages subvert T-cell function and correlate with reduced survival in clear cell renal cell carcinoma. Oncoimmunology. 2:e235622013. View Article : Google Scholar : PubMed/NCBI

116 

Gordon S and Martinez FO: Alternative activation of macrophages: Mechanism and functions. Immunity. 32:593–604. 2010. View Article : Google Scholar : PubMed/NCBI

117 

He Z and Zhang S: Tumor-associated macrophages and their functional transformation in the hypoxic tumor microenvironment. Front Immunol. 12:7413052021. View Article : Google Scholar : PubMed/NCBI

118 

Zhang L, Zhang K, Zhang J, Zhu J, Xi Q, Wang H, Zhang Z, Cheng Y, Yang G, Liu H, et al: Loss of fragile site-associated tumor suppressor promotes antitumor immunity via macrophage polarization. Nat Commun. 12:43002021. View Article : Google Scholar : PubMed/NCBI

119 

Wang W, Marinis JM, Beal AM, Savadkar S, Wu Y, Khan M, Taunk PS, Wu N, Su W, Wu J, et al: RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell. 34:757–774.e7. 2018. View Article : Google Scholar : PubMed/NCBI

120 

Ho TTB, Nasti A, Seki A, Komura T, Inui H, Kozaka T, Kitamura Y, Shiba K, Yamashita T, Yamashita T, et al: Combination of gemcitabine and anti-PD-1 antibody enhances the anticancer effect of M1 macrophages and the Th1 response in a murine model of pancreatic cancer liver metastasis. J Immunother Cancer. 8:e0013672020. View Article : Google Scholar : PubMed/NCBI

121 

Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, et al: Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 24:589–602. 2013. View Article : Google Scholar : PubMed/NCBI

122 

Shiao SL, Ruffell B, DeNardo DG, Faddegon BA, Park CC and Coussens LM: TH2-polarized CD4(+) T cells and macrophages limit efficacy of radiotherapy. Cancer Immunol Res. 3:518–525. 2015. View Article : Google Scholar : PubMed/NCBI

123 

DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N and Coussens LM: CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 16:91–102. 2009. View Article : Google Scholar : PubMed/NCBI

124 

Yasuda K, Takeuchi Y and Hirota K: The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol. 41:283–297. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Chang SH: T helper 17 (Th17) cells and interleukin-17 (IL-17) in cancer. Arch Pharm Res. 42:549–559. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, Huang E, Finlayson E, Simeone D, Welling TH, et al: Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 114:1141–1149. 2009. View Article : Google Scholar : PubMed/NCBI

127 

Zhou J, Li X, Wu X, Zhang T, Zhu Q and Wang X, Wang H, Wang K, Lin Y and Wang X: Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. 6:1578–1592. 2018. View Article : Google Scholar : PubMed/NCBI

128 

Yang JY, Jie Z, Mathews A, Zhou X, Li Y, Gu M, Xie X, Ko CJ, Cheng X, Qi Y, et al: Intestinal epithelial tbk1 prevents differentiation of T-helper 17 cells and tumorigenesis in mice. Gastroenterology. 159:1793–1806. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Maruyama T, Kono K, Mizukami Y, Kawaguchi Y, Mimura K, Watanabe M, Izawa S and Fujii H: Distribution of Th17 cells and FoxP3(+) regulatory T cells in tumor-infiltrating lymphocytes, tumor-draining lymph nodes and peripheral blood lymphocytes in patients with gastric cancer. Cancer Sci. 101:1947–1954. 2010. View Article : Google Scholar : PubMed/NCBI

130 

Knochelmann HM, Dwyer CJ, Bailey SR, Amaya SM, Elston DM, Mazza-McCrann JM and Paulos CM: When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell Mol Immunol. 15:458–469. 2018. View Article : Google Scholar : PubMed/NCBI

131 

Ohue Y and Nishikawa H: Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 110:2080–2089. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Whiteside TL: What are regulatory T cells (Treg) regulating in cancer and why? Semin Cancer Biol. 22:327–334. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Tanaka A and Sakaguchi S: Regulatory T cells in cancer immunotherapy. Cell Res. 27:109–118. 2017. View Article : Google Scholar :

134 

Waniczek D, Lorenc Z, Śnietura M, Wesecki M, Kopec A and Muc-Wierzgoń M: Tumor-associated macrophages and regulatory T cells infiltration and the clinical outcome in colorectal cancer. Arch Immunol Ther Exp (Warsz). 65:445–454. 2017. View Article : Google Scholar : PubMed/NCBI

135 

La Fleur L, Botling J, He F, Pelicano C, Zhou C, He C, Palano G, Mezheyeuski A, Micke P, Ravetch JV, et al: Targeting MARCO and IL37R on immunosuppressive macrophages in lung cancer blocks regulatory T cells and supports cytotoxic lymphocyte function. Cancer Res. 81:956–967. 2021. View Article : Google Scholar

136 

Zhu Q, Wu X, Wu Y and Wang X: Interaction between Treg cells and tumor-associated macrophages in the tumor microenvironment of epithelial ovarian cancer. Oncol Rep. 36:3472–3478. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Wu Q, Zhou W, Yin S, Zhou Y, Chen T, Qian J, Su R, Hong L, Lu H, Zhang F, et al: Blocking triggering receptor expressed on myeloid cells-1-positive tumor-associated macrophages induced by hypoxia reverses immunosuppression and anti-programmed cell death ligand 1 resistance in liver cancer. Hepatology. 70:198–214. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Fujimura T, Kambayashi Y, Fujisawa Y, Hidaka T and Aiba S: Tumor-associated macrophages: Therapeutic targets for skin cancer. Front Oncol. 8:32018. View Article : Google Scholar : PubMed/NCBI

139 

Furudate S, Fujimura T, Kambayashi Y, Kakizaki A, Hidaka T and Aiba S: Immunomodulatory effect of imiquimod through CCL22 produced by tumor-associated macrophages in B16F10 melanomas. Anticancer Res. 37:3461–3471. 2017.PubMed/NCBI

140 

Wang D, Yang L, Yue D, Cao L, Li L, Wang D, Ping Y, Shen Z, Zheng Y, Wang L and Zhang Y: Macrophage-derived CCL22 promotes an immunosuppressive tumor microenvironment via IL-8 in malignant pleural effusion. Cancer Lett. 452:244–253. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Carbó JM, León TE, Font-Díaz J, De la Rosa JV, Castrillo A, Picard FR, Staudenraus D, Huber M, Cedó L, Escolà-Gil JC, et al: Pharmacologic activation of LXR alters the expression profile of tumor-associated macrophages and the abundance of regulatory T cells in the tumor microenvironment. Cancer Res. 81:968–985. 2021. View Article : Google Scholar

142 

Liu J, Zhang N, Li Q, Zhang W, Ke F, Leng Q and Wang H, Chen J and Wang H: Tumor-associated macrophages recruit CCR6+ regulatory T cells and promote the development of colorectal cancer via enhancing CCL20 production in mice. PLoS One. 6:e194952011. View Article : Google Scholar : PubMed/NCBI

143 

Yang S, Wang B, Guan C, Wu B, Cai C, Wang M, Zhang B, Liu T and Yang P: Foxp3+IL-17+ T cells promote development of cancer-initiating cells in colorectal cancer. J Leukoc Biol. 89:85–91. 2011. View Article : Google Scholar

144 

Liu C, Chikina M, Deshpande R, Menk AV, Wang T, Tabib T, Brunazzi EA, Vignali KM, Sun M, Stolz DB, et al: Treg cells promote the SREBP1-dependent metabolic fitness of tumor-promoting macrophages via repression of CD8+ T cell-derived interferon-γ. Immunity. 51:381–397.e6. 2019. View Article : Google Scholar

145 

Fu Q, Xu L, Wang Y, Jiang Q, Liu Z, Zhang J, Zhou Q, Zeng H, Tong S, Wang T, et al: Tumor-associated macrophage-derived Interleukin-23 interlinks kidney cancer glutamine addiction with immune evasion. Eur Urol. 75:752–763. 2019. View Article : Google Scholar

146 

Sun W, Wei FQ, Li WJ, Wei JW, Zhong H, Wen YH, Lei WB, Chen L, Li H, Lin HQ, et al: A positive-feedback loop between tumour infiltrating activated Treg cells and type 2-skewed macrophages is essential for progression of laryngeal squamous cell carcinoma. Br J Cancer. 117:1631–1643. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Campesato LF, Budhu S, Tchaicha J, Weng CH, Gigoux M, Cohen IJ, Redmond D, Mangarin L, Pourpe S, Liu C, et al: Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine. Nat Commun. 11:40112020. View Article : Google Scholar : PubMed/NCBI

148 

Raskov H, Orhan A, Christensen JP and Gögenur I: Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer. 124:359–367. 2021. View Article : Google Scholar

149 

Reiser J and Banerjee A: Effector, memory, and dysfunctional CD8(+) T cell fates in the antitumor immune response. J Immunol Res. 2016:89412602016. View Article : Google Scholar : PubMed/NCBI

150 

Peranzoni E, Lemoine J, Vimeux L, Feuillet V, Barrin S, Kantari-Mimoun C, Bercovici N, Guérin M, Biton J, Ouakrim H, et al: Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci USA. 115:E4041–E4050. 2018. View Article : Google Scholar : PubMed/NCBI

151 

Garrido-Martin EM, Mellows TWP, Clarke J, Ganesan AP, Wood O, Cazaly A, Seumois G, Chee SJ, Alzetani A, King EV, et al: M1hot tumor-associated macrophages boost tissue-resident memory T cells infiltration and survival in human lung cancer. J Immunother Cancer. 8:e0007782020. View Article : Google Scholar

152 

Dai X, Lu L, Deng S, Meng J, Wan C, Huang J, Sun Y, Hu Y, Wu B, Wu G, et al: USP7 targeting modulates anti-tumor immune response by reprogramming tumor-associated macrophages in lung cancer. Theranostics. 10:9332–9347. 2020. View Article : Google Scholar : PubMed/NCBI

153 

Casanova-Acebes M, Dalla E, Leader AM, LeBerichel J, Nikolic J, Morales BM, Brown M, Chang C, Troncoso L, Chen ST, et al: Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature. 595:578–584. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Yao W, Ba Q, Li X, Li H, Zhang S, Yuan Y, Wang F, Duan X, Li J, Zhang W and Wang H: A natural CCR2 antagonist relieves tumor-associated macrophage-mediated immunosuppression to produce a therapeutic effect for liver cancer. EBioMedicine. 22:58–67. 2017. View Article : Google Scholar : PubMed/NCBI

155 

Dolina JS, Van Braeckel-Budimir N, Thomas GD and Salek-Ardakani S: CD8+ T cell exhaustion in cancer. Front Immunol. 12:7152342021. View Article : Google Scholar

156 

Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PA, Lanitis E, Duraiswamy J, Tanyi JL, Benencia F, et al: Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell. 35:885–900.e10. 2019. View Article : Google Scholar : PubMed/NCBI

157 

House IG, Savas P, Lai J, Chen AXY, Oliver AJ, Teo ZL, Todd KL, Henderson MA, Giuffrida L, Petley EV, et al: Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin Cancer Res. 26:487–504. 2020. View Article : Google Scholar

158 

Petty AJ, Li A, Wang X, Dai R, Heyman B, Hsu D, Huang X and Yang Y: Hedgehog signaling promotes tumor-associated macrophage polarization to suppress intratumoral CD8+ T cell recruitment. J Clin Invest. 129:5151–5162. 2019. View Article : Google Scholar : PubMed/NCBI

159 

Pascual-García M, Bonfill-Teixidor E, Planas-Rigol E, Rubio-Perez C, Iurlaro R, Arias A, Cuartas I, Sala-Hojman A, Escudero L, Martínez-Ricarte F, et al: LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8+ T cell tumor-infiltration impairing anti-PD1 therapy. Nat Commun. 10:24162019. View Article : Google Scholar

160 

Yu J, Green MD, Li S, Sun Y, Journey SN, Choi JE, Rizvi SM, Qin A, Waninger JJ, Lang X, et al: Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat Med. 27:152–164. 2021. View Article : Google Scholar : PubMed/NCBI

161 

Kusmartsev S and Gabrilovich DI: STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 174:4880–4891. 2005. View Article : Google Scholar : PubMed/NCBI

162 

Stanley ER and Chitu V: CSF-1 receptor signaling in myeloid cells. Cold Spring Harb Perspect Biol. 6:a0218572014. View Article : Google Scholar : PubMed/NCBI

163 

Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, Ito J, Tachino S, Hori Y, Matsuki M, et al: Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 14:e02125132019. View Article : Google Scholar : PubMed/NCBI

164 

Mehta AK, Cheney EM, Hartl CA, Pantelidou C, Oliwa M, Castrillon JA, Lin JR, Hurst KE, de Oliveira Taveira M, Johnson NT, et al: Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. Nat Cancer. 2:66–82. 2021. View Article : Google Scholar : PubMed/NCBI

165 

Xiang W, Shi R, Kang X, Zhang X, Chen P, Zhang L, Hou A, Wang R, Zhao Y, Zhao K, et al: Monoacylglycerol lipase regulates cannabinoid receptor 2-dependent macrophage activation and cancer progression. Nat Commun. 9:25742018. View Article : Google Scholar : PubMed/NCBI

166 

Chen EP, Markosyan N, Connolly E, Lawson JA, Li X, Grant GR, Grosser T, FitzGerald GA and Smyth EM: Myeloid cell COX-2 deletion reduces mammary tumor growth through enhanced cytotoxic T-lymphocyte function. Carcinogenesis. 35:1788–1797. 2014. View Article : Google Scholar : PubMed/NCBI

167 

Li L, Han L, Sun F, Zhou J, Ohaegbulam KC, Tang X, Zang X, Steinbrecher KA, Qu Z and Xiao G: NF-κB RelA renders tumor-associated macrophages resistant to and capable of directly suppressing CD8+ T cells for tumor promotion. Oncoimmunology. 7:e14352502018. View Article : Google Scholar

168 

Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, Woo G, Nguyen AV, Figueiredo CC, Foubert P, et al: PI3Kγ is a molecular switch that controls immune suppression. Nature. 539:437–442. 2016. View Article : Google Scholar : PubMed/NCBI

169 

McIntire RH and Hunt JS: Antigen presenting cells and HLA-G-a review. Placenta. 26(Suppl A): S104–S109. 2005. View Article : Google Scholar

170 

Nixon BG, Kuo F, Liu M, Capistrano K, Do M, Franklin RA, Wu X, Kansler ER, Srivastava RM, Purohit TA, et al: IRF8 governs tumor-associated macrophage control of T cell exhaustion. bioRxiv. 2020.2003.2012.989731. 2020.

171 

Tseng D, Volkmer JP, Willingham SB, Contreras-Trujillo H, Fathman JW, Fernhoff NB, Seita J, Inlay MA, Weiskopf K, Miyanishi M and Weissman IL: Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc Natl Acad Sci USA. 110:11103–11108. 2013. View Article : Google Scholar : PubMed/NCBI

172 

Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho CM, Pryer N, Daniel D, Hwang ES, Rugo HS and Coussens LM: Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 26:623–637. 2014. View Article : Google Scholar : PubMed/NCBI

173 

Fang W, Zhou T, Shi H, Yao M, Zhang D, Qian H, Zeng Q, Wang Y, Jin F, Chai C and Chen T: Progranulin induces immune escape in breast cancer via up-regulating PD-L1 expression on tumor-associated macrophages (TAMs) and promoting CD8+ T cell exclusion. J Exp Clin Cancer Res. 40:42021. View Article : Google Scholar

174 

Lin H, Wei S, Hurt EM, Green MD, Zhao L, Vatan L, Szeliga W, Herbst R, Harms PW, Fecher LA, et al: Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest. 128:805–815. 2018. View Article : Google Scholar : PubMed/NCBI

175 

Chow MT, Ozga AJ, Servis RL, Frederick DT, Lo JA, Fisher DE, Freeman GJ, Boland GM and Luster AD: Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity. 50:1498–1512.e5. 2019. View Article : Google Scholar : PubMed/NCBI

176 

Lee AH, Sun L, Mochizuki AY, Reynoso JG, Orpilla J, Chow F, Kienzler JC, Everson RG, Nathanson DA, Bensinger SJ, 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. 12:69382021. View Article : Google Scholar : PubMed/NCBI

177 

Tsou P, Katayama H, Ostrin EJ and Hanash SM: The emerging role of B cells in tumor immunity. Cancer Res. 76:5597–5601. 2016. View Article : Google Scholar : PubMed/NCBI

178 

Nelson BH: CD20+ B cells: The other tumor-infiltrating lymphocytes. J Immunol. 185:4977–4982. 2010. View Article : Google Scholar : PubMed/NCBI

179 

Gunderson AJ, Kaneda MM, Tsujikawa T, Nguyen AV, Affara NI, Ruffell B, Gorjestani S, Liudahl SM, Truitt M, Olson P, et al: Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov. 6:270–285. 2016. View Article : Google Scholar

180 

Fremd C, Schuetz F, Sohn C, Beckhove P and Domschke C: B cell-regulated immune responses in tumor models and cancer patients. Oncoimmunology. 2:e254432013. View Article : Google Scholar : PubMed/NCBI

181 

Wang SS, Liu W, Ly D, Xu H, Qu L and Zhang L: Tumor-infiltrating B cells: Their role and application in anti-tumor immunity in lung cancer. Cell Mol Immunol. 16:6–18. 2019. View Article : Google Scholar :

182 

Audzevich T, Bashford-Rogers R, Mabbott NA, Frampton D, Freeman TC, Potocnik A, Kellam P and Gilroy DW: Pre/pro-B cells generate macrophage populations during homeostasis and inflammation. Proc Natl Acad Sci USA. 114:E3954–E3963. 2017. View Article : Google Scholar : PubMed/NCBI

183 

Fridman WH, Petitprez F, Meylan M, Chen TW, Sun CM, Roumenina LT and Sautès-Fridman C: B cells and cancer: To B or not to B? J Exp Med. 218:e202008512021. View Article : Google Scholar : PubMed/NCBI

184 

Gonzalez H, Hagerling C and Werb Z: Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 32:1267–1284. 2018. View Article : Google Scholar : PubMed/NCBI

185 

Michel T, Hentges F and Zimmer J: Consequences of the cross-talk between monocytes/macrophages and natural killer cells. Front Immunol. 3:4032013. View Article : Google Scholar

186 

Sivori S, Pende D, Quatrini L, Pietra G, Della Chiesa M, Vacca P, Tumino N, Moretta F, Mingari MC, Locatelli F and Moretta L: NK cells and ILCs in tumor immunotherapy. Mol Aspects Med. 80:1008702021. View Article : Google Scholar

187 

Krneta T, Gillgrass A, Poznanski S, Chew M, Lee AJ, Kolb M and Ashkar AA: M2-polarized and tumor-associated macrophages alter NK cell phenotype and function in a contact-dependent manner. J Leukoc Biol. 101:285–295. 2017. View Article : Google Scholar

188 

Platonova S, Cherfils-Vicini J, Damotte D, Crozet L, Vieillard V, Validire P, André P, Dieu-Nosjean MC, Alifano M, Régnard JF, et al: Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 71:5412–5422. 2011. View Article : Google Scholar : PubMed/NCBI

189 

Kloss M, Decker P, Baltz KM, Baessler T, Jung G, Rammensee HG, Steinle A, Krusch M and Salih HR: Interaction of monocytes with NK cells upon Toll-like receptor-induced expression of the NKG2D ligand MICA. J Immunol. 181:6711–6719. 2008. View Article : Google Scholar : PubMed/NCBI

190 

Peng LS, Zhang JY, Teng YS, Zhao YL, Wang TT, Mao FY, Lv YP, Cheng P, Li WH, Chen N, et al: Tumor-associated monocytes/macrophages impair NK-cell function via TGFβ1 in human gastric cancer. Cancer Immunol Res. 5:248–256. 2017. View Article : Google Scholar : PubMed/NCBI

191 

Castriconi R, Cantoni C, Della Chiesa M, Vitale M, Marcenaro E, Conte R, Biassoni R, Bottino C, Moretta L and Moretta A: Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: Consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci USA. 100:4120–4125. 2003. View Article : Google Scholar : PubMed/NCBI

192 

Gallazzi M, Baci D, Mortara L, Bosi A, Buono G, Naselli A, Guarneri A, Dehò F, Capogrosso P, Albini A, et al: Prostate cancer peripheral blood NK cells show enhanced CD9, CD49a, CXCR4, CXCL8, MMP-9 production and secrete monocyte-recruiting and polarizing factors. Front Immunol. 11:5861262021. View Article : Google Scholar : PubMed/NCBI

193 

Malekghasemi S, Majidi J, Baghbanzadeh A, Abdolalizadeh J, Baradaran B and Aghebati-Maleki L: Tumor-associated macrophages: Protumoral macrophages in inflammatory tumor microenvironment. Adv Pharm Bull. 10:556–565. 2020. View Article : Google Scholar : PubMed/NCBI

194 

Xu B, Chen L, Li J, Zheng X, Shi L, Wu C and Jiang J: Prognostic value of tumor infiltrating NK cells and macrophages in stage II+III esophageal cancer patients. Oncotarget. 7:74904–74916. 2016. View Article : Google Scholar : PubMed/NCBI

195 

Eisinger S, Sarhan D, Boura VF, Ibarlucea-Benitez I, Tyystjärvi S, Oliynyk G, Arsenian-Henriksson M, Lane D, Wikström SL, Kiessling R, et al: Targeting a scavenger receptor on tumor-associated macrophages activates tumor cell killing by natural killer cells. Proc Natl Acad Sci USA. 117:32005–32016. 2020. View Article : Google Scholar : PubMed/NCBI

196 

Bellora F, Castriconi R, Dondero A, Pessino A, Nencioni A, Liggieri G, Moretta L, Mantovani A, Moretta A and Bottino C: TLR activation of tumor-associated macrophages from ovarian cancer patients triggers cytolytic activity of NK cells. Eur J Immunol. 44:1814–1822. 2014. View Article : Google Scholar : PubMed/NCBI

197 

Bellora F, Castriconi R, Dondero A, Reggiardo G, Moretta L, Mantovani A, Moretta A and Bottino C: The interaction of human natural killer cells with either unpolarized or polarized macrophages results in different functional outcomes. Proc Natl Acad Sci USA. 107:21659–21664. 2010. View Article : Google Scholar : PubMed/NCBI

198 

Dalbeth N, Gundle R, Davies RJO, Lee YCG, McMichael AJ and Callan MFC: CD56bright NK cells are enriched at inflammatory sites and can engage with monocytes in a reciprocal program of activation. J Immunol. 173:6418–6426. 2004. View Article : Google Scholar : PubMed/NCBI

199 

O'Sullivan T, Saddawi-Konefka R, Vermi W, Koebel CM, Arthur C, White JM, Uppaluri R, Andrews DM, Ngiow SF, Teng MW, et al: Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J Exp Med. 209:1869–1882. 2012. View Article : Google Scholar : PubMed/NCBI

200 

Ponzetta A, Carriero R, Carnevale S, Barbagallo M, Molgora M, Perucchini C, Magrini E, Gianni F, Kunderfranco P, Polentarutti N, et al: Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell. 178:346–360.e24. 2019. View Article : Google Scholar : PubMed/NCBI

201 

Wu L and Zhang XH: Tumor-associated neutrophils and macrophages-heterogenous but not chaotic. Front Immunol. 11:5539672020. View Article : Google Scholar : PubMed/NCBI

202 

Kim J and Bae JS: Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm. 2016:60581472016. View Article : Google Scholar : PubMed/NCBI

203 

Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z and Zhou J: Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. 150:1646–1658.e17. 2016. View Article : Google Scholar : PubMed/NCBI

204 

Zilionis R, Engblom C, Pfirschke C, Savova V, Zemmour D, Saatcioglu HD, Krishnan I, Maroni G, Meyerovitz CV, Kerwin CM, et al: Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 50:1317–1334.e10. 2019. View Article : Google Scholar : PubMed/NCBI

205 

Zhou Z, Wang P, Sun R, Li J, Hu Z, Xin H, Luo C, Zhou J, Fan J and Zhou S: Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J Immunother Cancer. 9:e0019462021. View Article : Google Scholar : PubMed/NCBI

206 

Gunaydin G: CAFs interacting with TAMs in tumor microenvironment to enhance tumorigenesis and immune evasion. Front Oncol. 11:6683492021. View Article : Google Scholar : PubMed/NCBI

207 

Sweeny L, Liu Z, Lancaster W, Hart J, Hartman YE and Rosenthal EL: Inhibition of fibroblasts reduced head and neck cancer growth by targeting fibroblast growth factor receptor. Laryngoscope. 122:1539–1544. 2012. View Article : Google Scholar : PubMed/NCBI

208 

Vu HN, Miller WJ, O'Connor SA, He M, Schafer PH, Payvandi F, Muller GW, Stirling DI and Libutti SK: CC-5079: A small molecule with MKP1, antiangiogenic, and antitumor activity. J Surg Res. 164:116–125. 2010. View Article : Google Scholar

209 

Donthireddy L, Vonteddu P, Murthy T, Kwak T, Eraslan RN, Podojil JR, Elhofy A, Boyne MT II, Puisis JJ, Veglia F, et al: ONP-302 nanoparticles inhibit tumor growth by altering tumor-associated macrophages and cancer-associated fibroblasts. J Cancer. 13:1933–1944. 2022. View Article : Google Scholar : PubMed/NCBI

210 

Kashyap AS, Schmittnaegel M, Rigamonti N, Pais-Ferreira D, Mueller P, Buchi M, Ooi CH, Kreuzaler M, Hirschmann P, Guichard A, et al: Optimized antiangiogenic reprogramming of the tumor microenvironment potentiates CD40 immunotherapy. Proc Natl Acad Sci USA. 117:541–551. 2020. View Article : Google Scholar : PubMed/NCBI

211 

Zhang L, Qi Y, Min H, Ni C, Wang F, Wang B, Qin H, Zhang Y, Liu G, Qin Y, et al: Cooperatively responsive peptide nanotherapeutic that regulates angiopoietin receptor Tie2 activity in tumor microenvironment to prevent breast tumor relapse after chemotherapy. ACS Nano. 13:5091–5102. 2019. View Article : Google Scholar : PubMed/NCBI

212 

Alam A, Blanc I, Gueguen-Dorbes G, Duclos O, Bonnin J, Barron P, Laplace MC, Morin G, Gaujarengues F, Dol F, et al: SAR131675, a potent and selective VEGFR-3-TK inhibitor with antilymphangiogenic, antitumoral, and antimetastatic activities. Mol Cancer Ther. 11:1637–1649. 2012. View Article : Google Scholar : PubMed/NCBI

213 

Gyori D, Lim EL, Grant FM, Spensberger D, Roychoudhuri R, Shuttleworth SJ, Okkenhaug K, Stephens LR and Hawkins PT: Compensation between CSF1R+ macrophages and Foxp3+ Treg cells drives resistance to tumor immunotherapy. JCI Insight. 3:e1206312018. View Article : Google Scholar : PubMed/NCBI

214 

Oweida AJ, Darragh L, Phan A, Binder D, Bhatia S, Mueller A, Court BV, Milner D, Raben D, Woessner R, et al: STAT3 modulation of regulatory T cells in response to radiation therapy in head and neck cancer. J Natl Cancer Inst. 111:1339–1349. 2019. View Article : Google Scholar : PubMed/NCBI

215 

Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, Wang-Gillam A, Goedegebuure SP, Linehan DC and DeNardo DG: CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 74:5057–5069. 2014. View Article : Google Scholar : PubMed/NCBI

216 

Sluijter M, van der Sluis TC, van der Velden PA, Versluis M, West BL, van der Burg SH and van Hall T: Inhibition of CSF-1R supports T-cell mediated melanoma therapy. PLoS One. 9:e1042302014. View Article : Google Scholar : PubMed/NCBI

217 

Doleschel D, Hoff S, Koletnik S, Rix A, Zopf D, Kiessling F and Lederle W: Regorafenib enhances anti-PD1 immunotherapy efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J Exp Clin Cancer Res. 40:2882021. View Article : Google Scholar : PubMed/NCBI

218 

Zhang JQ, Zeng S, Vitiello GA, Seifert AM, Medina BD, Beckman MJ, Loo JK, Santamaria-Barria J, Maltbaek JH, Param NJ, et al: Macrophages and CD8+ T cells mediate the antitumor efficacy of combined CD40 ligation and imatinib therapy in gastrointestinal stromal tumors. Cancer Immunol Res. 6:434–447. 2018. View Article : Google Scholar : PubMed/NCBI

219 

Zhang M, Wen B, Anton OM, Yao Z, Dubois S, Ju W, Sato N, DiLillo DJ, Bamford RN, Ravetch JV and Waldmann TA: IL-15 enhanced antibody-dependent cellular cytotoxicity mediated by NK cells and macrophages. Proc Natl Acad Sci USA. 115:E10915–E10924. 2018. View Article : Google Scholar : PubMed/NCBI

220 

Jarosz-Biej M, Kamińska N, Matuszczak S, Cichoń T, Pamuła-Piłat J, Czapla J, Smolarczyk R, Skwarzyńska D, Kulik K and Szala S: M1-like macrophages change tumor blood vessels and microenvironment in murine melanoma. PLoS One. 13:e01910122018. View Article : Google Scholar : PubMed/NCBI

221 

Gaggero S, Witt K, Carlsten M and Mitra S: Cytokines orchestrating the natural killer-myeloid cell crosstalk in the tumor microenvironment: implications for natural killer cell-based cancer immunotherapy. Front Immunol. 11:6212252021. View Article : Google Scholar : PubMed/NCBI

222 

Zhou Z, Zhang C, Zhang J and Tian Z: Macrophages help NK cells to attack tumor cells by stimulatory NKG2D ligand but protect themselves from NK killing by inhibitory ligand Qa-1. PLoS One. 7:e369282012. View Article : Google Scholar : PubMed/NCBI

223 

Wang C, Cui A, Bukenya M, Aung A, Pradhan D, Whittaker CA, Agarwal Y, Thomas A, Liang S, Amlashi P, et al: Reprogramming NK cells and macrophages via combined antibody and cytokine therapy primes tumors for elimination by checkpoint blockade. Cell Rep. 37:1100212021. View Article : Google Scholar : PubMed/NCBI

224 

Nywening TM, Belt BA, Cullinan DR, Panni RZ, Han BJ, Sanford DE, Jacobs RC, Ye J, Patel AA, Gillanders WE, et al: Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut. 67:1112–1123. 2018. View Article : Google Scholar

225 

Li X, Yao W, Yuan Y, Chen P, Li B, Li J, Chu R, Song H, Xie D, Jiang X and Wang H: Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 66:157–167. 2017. View Article : Google Scholar

226 

Veillette A and Tang Z: Signaling regulatory protein (SIRP) α-CD47 blockade joins the ranks of immune checkpoint inhibition. J Clin Oncol. 37:1012–1014. 2019. View Article : Google Scholar : PubMed/NCBI

227 

Wesolowski R, Sharma N, Reebel L, Rodal MB, Peck A, West BL, Marimuthu A, Severson P, Karlin DA, Dowlati A, et al: Phase Ib study of the combination of pexidartinib (PLX3397), a CSF-1R inhibitor, and paclitaxel in patients with advanced solid tumors. Ther Adv Med Oncol. 11:17588359198542382019. View Article : Google Scholar : PubMed/NCBI

228 

Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S and Zhou H: Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther. 6:2182021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2023
Volume 62 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kazakova A, Sudarskikh T, Kovalev O, Kzhyshkowska J and Larionova I: Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review). Int J Oncol 62: 32, 2023
APA
Kazakova, A., Sudarskikh, T., Kovalev, O., Kzhyshkowska, J., & Larionova, I. (2023). Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review). International Journal of Oncology, 62, 32. https://doi.org/10.3892/ijo.2023.5480
MLA
Kazakova, A., Sudarskikh, T., Kovalev, O., Kzhyshkowska, J., Larionova, I."Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review)". International Journal of Oncology 62.2 (2023): 32.
Chicago
Kazakova, A., Sudarskikh, T., Kovalev, O., Kzhyshkowska, J., Larionova, I."Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review)". International Journal of Oncology 62, no. 2 (2023): 32. https://doi.org/10.3892/ijo.2023.5480