Next Article in Journal
Excess Heme Promotes the Migration and Infiltration of Macrophages in Endometrial Hyperplasia Complicated with Abnormal Uterine Bleeding
Next Article in Special Issue
Proinflammatory and Cancer-Promoting Pathobiont Fusobacterium nucleatum Directly Targets Colorectal Cancer Stem Cells
Previous Article in Journal
Neuroprotective Effects of Pharmacological Hypothermia on Hyperglycolysis and Gluconeogenesis in Rats after Ischemic Stroke
Previous Article in Special Issue
Targeting of the Peritumoral Adipose Tissue Microenvironment as an Innovative Antitumor Therapeutic Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells

1
Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China
2
Department of Surgery, Nanjing Lishui People’s Hospital, Nanjing 211200, China
3
Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 10 April 2022 / Revised: 15 June 2022 / Accepted: 16 June 2022 / Published: 19 June 2022

Abstract

:
Cancer stem cells (CSCs) are a subset of highly tumorigenic cells in tumors. They have enhanced self-renewal properties, are usually chemo-radioresistant, and can promote tumor recurrence and metastasis. They can recruit macrophages into the tumor microenvironment and differentiate them into tumor-associated macrophages (TAMs). TAMs maintain CSC stemness and construct niches that are favorable for CSC survival. However, how CSCs and TAMs interact is not completely understood. An understanding on these mechanisms can provide additional targeting strategies for eliminating CSCs. In this review, we comprehensively summarize the reported mechanisms of crosstalk between CSCs and TAMs and update the related signaling pathways involved in tumor progression. In addition, we discuss potential therapies targeting CSC–TAM interaction, including targeting macrophage recruitment and polarization by CSCs and inhibiting the TAM-induced promotion of CSC stemness. This review also provides the perspective on the major challenge for developing potential therapeutic strategies to overcome CSC-TAM crosstalk.

1. Introduction

Cancer stem cells (CSCs), also called tumor initiation cells, are specialized tumor cells that promote tumor initiation and progression [1]. The existence of CSCs was first confirmed in 1994 when Dick et al. grouped acute myeloid leukemia (AML) cells according to the expression of cell surface markers; when AML with CD34+CD38 surface markers were transplanted into severe combined immunodeficiency (SCID) mice, numerous progenitor cell colonies were formed. However, non-CD34+CD38 AML cells had limited proliferative capacity. Therefore, the CD34+CD38 population was considered to be CSCs [2]. Since then, CSCs have been reported in other cancers: breast cancer (CSC markers: CD44, CD24, and ALDH-1) [3,4], colon cancer (CSC markers: CD24, CD44, CD133, and LGR5) [5,6,7], and melanoma (CSC markers: CD34 and ABCB5) [8,9]. CSCs are characterized by their self-renewal and differentiation potential that contribute to chemo-radioresistant and tumor initiation, which primarily account for the failure of current anticancer therapies in CSC-containing cancers [4]. In addition, CSC stemness is maintained mainly through the CSC niche, which is conducive to CSC survival, thereby making it difficult for drugs or other therapeutic methods to completely kill them, which leads to tumor recurrence [10]. Therefore, the eradication of CSCs is considered a promising method for improving cancer survival rates and even for curing cancer.
The CSC niche is a part of the tumor microenvironment (TME) and is similar to the normal adult stem cell niche that regulates the biological activity of stem cells in the form of cell–cell contacts and secreted factors [11,12]. The TME comprises fibroblasts, immune cells, endothelial cells, perivascular cells, extracellular matrix (ECM) components, and cytokine networks, among which tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME [11]. TAMs can be recruited by CSCs and participate in TME formation; hence, they are beneficial to CSC survival [13]. Some studies have reported using CSCs derived from human or murine cholangiocarcinoma, hepatocellular carcinoma (HCC), or glioblastoma (GBM) cells; under in vitro spheroid culture, many factors regulate monocytes/macrophages in the supernatant, including CC-motif chemokine ligand (CCL) 2, CCL5, colony-stimulating factor (CSF)-1, interleukin (IL)-13, transforming growth factor (TGF)-β, and periosteum proteins (POSTN). These cytokines recruit circulating monocytes as well as surrounding tissue macrophage precursors into the TME and polarize them into TAMs [14,15,16]. After recruiting macrophages into the TME, CSCs can avoid phagocytosis by macrophages through protective mechanisms [17]. In clinical studies, CD47 expression has been reported to be upregulated in CSCs of HCC [18], pancreatic ductal adenocarcinoma [19], and lung cancer [20]. CD47 interacts with signal regulatory protein α (SIRP-α), which is expressed on TAMs, to prevent CSCs from being phagocytosed. Macrophages recruited into the TME can be divided into classically activated M1 macrophages and alternatively activated M2 macrophages [21]. M1 macrophages are induced and activated by interferon (IFN)-γ and lipopolysaccharides. Activated M1 macrophages typically express CD40 and CD86 and secrete higher levels of proinflammatory factors than M2 macrophages, such as IL-1α, tumor necrosis factor (TNF), and IL-12, which mainly exert an antitumor immune response and directly kill tumor cells in the early stage of a tumor [22]. Therefore, injecting M1 macrophages into the tumor tissue may be a potential therapeutic strategy for cancer [23], but the specific mechanism of action remains unclear. Activated M2 macrophages, however, play a role opposite to that of M1 macrophages—promoting ECM remodeling and tumor growth and metastasis [24]. M2 macrophages support CSC signatures, and consequently, they are also called M2 TAMs [25]. They are induced by IL-4 and IL-13, often with CD163 and CD206 as markers [26]. M2 macrophages can secrete soluble mediators, such as platelet-derived growth factor (PDGF), vascular epidermal growth factor (VEGF), and cytokines, such as IL-6, IL-10, and TNF-α [15,27,28]. These soluble mediators and cytokines can maintain the stemness characteristics of tumor cells by regulating the nuclear factor-κB (NF-κB), signal transducer and activator of transcription (STAT)3, AKT, and Hippo pathways [14,27,29,30,31,32]. These findings highlight the importance of molecular crosstalk between CSCs and macrophages in tumor progression; targeting these molecular mechanisms may provide more effective immunotherapies.
The CSC niche provides favorable conditions for drug resistance, tumor recurrence, and metastasis. TAMs, as the main immune cells in the TME, also play a crucial role in cancer malignancy. In this review, we summarize the potential mechanisms of interaction between CSCs and TAMs and discuss the current molecular targeting studies thereof. An overview of these emerging insights can provide new directions for antitumor strategies.

2. Secretory Molecules of CSCs Induce Macrophage Recruitment and Tumor-Promoting Characteristics of the TME

The functions of CSCs, such as immune escape, therapy resistance, metastasis, and maintenance of stemness, are largely dependent on the secretion of soluble factors (chemokines, interleukins, growth factors, or secreted proteins) and extracellular vesicles [33,34,35]. Specifically, CSCs signal by secreting soluble factors to recruit peripheral macrophages to the TME to become part of the tumor niche and by inducing macrophage transformation into TAMs [36]. A comprehensive understanding of the underlying mechanisms of macrophage recruitment and polarization by CSCs is beneficial for determining how to interrupt the mutual support pathway between CSCs and TAMs. Figure 1 presents the molecular interaction mechanisms between TAMs and CSCs.

2.1. CSCs Recruit Macrophages into the TME

Many cytokines secreted by tumor cells can recruit macrophages to the TME, but some of them are exclusively produced by CSCs: CCL2, CCL3, CCL5, CXC motif chemokine ligand 12 (CXCL12), olfactomedin-like 3 (OLFML3), stromal-cell-derived factor-1b (Sdf1b), IL-6, IL-33, and CSF-1 [37,38,39,40,41,42,43,44,45] (Figure 1). CSCs contribute to the infiltration of macrophages or microglia to promote their own metastatic spread [38,39,43,46]. Here, we summarize the cytokines and soluble protein molecules secreted for macrophage recruitment by CSCs in different tumor types (Table 1).
Table 1. CSCs secrete cytokines and other soluble protein molecules to recruit macrophages.
Table 1. CSCs secrete cytokines and other soluble protein molecules to recruit macrophages.
Chemokines Produced by CSCsChemokine ReceptorType of CancerAssociated Signaling Pathway/Mechanism of Chemokine SecretionReference
CCL2 1CCR2Glioblastoma IFNγ[47]
Breast cancerβ-Catenin[48]
Bladder cancerNot shown[39]
Hepatocellular carcinomaHippo [43]
CCL3CCR1/CCR5Bladder cancerNot shown[49,50]
LeukemiaNot shown[51]
CCL5CCR5, CCR1, CCR2Glioblastoma PI3K 2/AKT[52,53]
Optic gliomaNot shown[54]
Hepatocellular carcinomaNot shown[55]
CCL8CCR2Bladder cancerNot shown[56]
Cutaneous squamous cell carcinomaNot shown[57]
CCL9CCR1Non-small cell lung cancerNot shown[58]
CXCL5 3CXCR2Hepatocellular carcinomaSox9[59]
CXCL12CXCR4LeukemiaNot shown[42]
Colon cancerNF-κB 4[60]
CXCL14Not shownLung cancerNot shown[61]
IL-10IL-10ROvarian cancerWNT[62]
IL-33ST2Squamous cell carcinoma NRF2[44]
CSF1 5CSF1RHepatocellular carcinomaHippo [43]
Non-small cell lung cancerIRF5[63]
Glioblastoma STAT3 6[64]
TGF-β 7Not shownGlioblastoma STAT3[64]
MIC-1 8Not shownGlioblastoma Not shown[64]
Pancreatic cancerNot shown[65]
LOX 9Not shownPancreatic cancerNot shown[66]
VEGF 10VEGFRColorectal cancerSix1/MAPK 11 & PI3K/AKT/mTOR [67,68]
OLFML3 12Not shownGlioblastomaCLOCK-BMAL1[40]
Sdf1 13Cxcr4bBrain tumorAKT1[41]
POSTN 14Integrinαvβ₃GlioblastomaNot shown[69]
CX3CL1CX3CR1Testicular germ cell tumorsNot shown[70]
HGF 15Not shownGlioblastomaHGF-MET axis[71]
MIFCXCR4GlioblastomaNot shown[72]
FGL2Not shownGlioblastomaNot shown[73]
1 C-C-motif chemokine ligand 2; 2 Phosphoinositide 3-kinase; 3 CXC motif chemokine ligand 5; 4 Nuclear factor-kappa B; 5 Colony-stimulating factor-1; 6 Signal transducer and activator of transcription (STAT)3; 7 Transforming growth factor β; 8 Macrophage inhibitory cytokine-1; 9 Lipoxygenase; 10 Vascular epidermal growth factor; 11 Mitogen-activated protein kinase; 12 Olfactomedin like 3; 13 Stromal-cell-derived factor-1b; 14 Periostin; 15 Hepatocyte growth factor.

2.1.1. CSCs Recruit Macrophages through the CC Chemokine Family and the CXC Chemokine Subfamily

The CC chemokine family and the CXC chemokine subfamily are involved in macrophage recruitment in tumors (or CSCs) [74]. CCL2, a CC chemokine, is secreted by CSCs of different tumor types to promote macrophage infiltration. CCL2 overexpression is associated with poor patient prognosis in various tumor types [75] (Table 1). In studies on GBM [38], prostate cancer [76], and bladder cancer [39], CCL2 expression was significantly increased in CSCs compared with non-CSCs, and the macrophage count in the TME was also increased. Another study showed that the blockade of CCL2 binding to its receptor CCR2 on macrophages or depletion of CCL2 decreased monocyte recruitment and prolonged tumor survival in mice [43]. Thus, CCL2 may play a vital role in macrophage recruitment. Studies have attempted to uncover why CCL2 is upregulated in CSCs. In a study of liver CSCs, CCL2 expression was associated with Yes-associated protein, which acts as a transcriptional coactivator and can upregulate CCL2 expression [43]. Furthermore, the upregulation of CCL2 is due to the increase of the stemness-related transcription factor TWIST, and the increased CCL2 secretion promotes the infiltration of M2 macrophages in lung cancer [77]. GBM stem cells (GSCs) have stable epigenetic changes that initiate a myeloid-dependent transcriptional program that expresses myeloid-specific master transcription factors such as interferon regulatory factor (IRF)1 and IRF8 [47]. Together, these findings reveal that CSCs may induce macrophage recruitment by secreting CCL2; however, the underlying mechanism remains unclear.
CCL3, also known as macrophage inflammatory protein-1α, is a ligand for the receptors CCR1 and CCR5 (Figure 1) and promotes macrophage recruitment [78]. CCL3 is secreted in CD14+ bladder CSCs rather than CD14 cancer cells, and macrophage infiltration is also increased in the TME [49]. However, another study determined that CCL3 recruits macrophages to the TME to exert anticancer activities [79]. These results indicate that CSCs can secrete CCL3 to recruit macrophages with antitumor properties in the TME.
Other chemokine members also participate in macrophage recruitment. In a study on GBM, CCL5 expression was significantly higher in GSCs compared with low-grade gliomas and noncancerous tissues, whereas GSCs did not express CCR5 (Figure 1). GSCs may secrete CCL5, which binds to microglial CCR5 and causes microglial infiltration [52,53]. Microglia are known as the macrophages of the nervous system, and their biological properties are similar to those of peripheral macrophages [80]. Notably, CCL5 is produced exclusively by CSCs, highlighting their unique role in macrophage recruitment. Furthermore, the ability to recruit microglia through CCL5 is enhanced when PTEN or FGFR1 is mutated [53]. By contrast, a study reported that CCL8 secreted by CSCs can recruit M1 macrophages and increase the number of CD16 and CD80 macrophages in the TME [57]. However, the study did not explain why CSCs recruited M1 macrophages, which are thought to have antitumor properties. In fact, tumor stromal mesenchymal stem cells (MSCs) in the TME are also involved in macrophage recruitment, and MSCs highly express various chemokines, especially CCL2, which increase the recruitment of monocytes/macrophages to the TME [81]. In conclusion, accumulating evidence indicates that the CC family of chemokines secreted by the CSCs of different cancer types are involved in macrophage recruitment.
Chemokines in the CXC family secreted by CSCs are also involved in macrophage recruitment. CXCL5 expression is higher in CD44+ CSCs compared with non-CSCs [82]. CXCL5 acts as a key downstream mediator of Sox9, which is a biomarker of liver CSCs. Sox9 can bind to the promoter of CXCL5 to induce CXCL5 secretion, and a high CXCL5 level enhances macrophage recruitment in HCC [59]. Furthermore, the CXCL12 level has a significant positive correlation with macrophage infiltration in tumors [60]. CD133+ GCSC can also secrete CXCL12, which binds to CXCR4 (CXCL12 receptor) on macrophages to recruit macrophages to the TME [83]. Similar results were also observed in breast cancer [84]. These data suggest that both the CC and CXC chemokine families secreted by CSCs play a crucial role in macrophage recruitment. Targeting these chemokines may inhibit macrophage recruitment by CSCs, thereby hindering CSC niche formation.

2.1.2. CSCs Recruit Macrophages by Secreting IL-33

CSCs recruit and induce myeloid cells to differentiate into TAMs by releasing IL-33 [85] (Figure 1). IL-33 combined with ST2 on TAMs stimulates TAMs to secrete TGF-β. TGF-β can regulate CSCs through a negative feedback mechanism, activate NRF2-mediated antioxidant responses, and release IL-33, thus promoting macrophage recruitment [44]. This suggests that macrophage recruitment by CSCs is an interactive process. Interruption of the IL-33–TGF-β signaling loop provides a potential therapeutic option for preventing CSC niche formation.

2.1.3. CSCs Recruit Macrophages through Other Types of Secreted Proteins

In addition to chemokines and ILs, CSCs can also secrete various other soluble cellular proteins to promote macrophage recruitment into the TME, such as CSF-1 [86] (Figure 1). Yamashina et al. reported that CSCs stimulated by interferon regulatory factor 5 (IRF5) could activate the IRF5/CSF-1 pathway in non-small-cell lung cancer (NSCLC), thereby increasing CSF-1 secretion and causing an increase in the number of M2 macrophages with CSF-1 receptors [63]. However, GSCs can produce CSF-1 and cytokines, such as TGF-β1 and macrophage inhibitory cytokine (MIC)-1, to recruit surrounding microglia [64]. These results suggest that CSCs promote macrophage recruitment by secreting these cytokines, but what drives CSCs to secrete these soluble cytokines is unclear. Targeting these cytokines may effectively block macrophage recruitment into the TME and prevent macrophages from participating in CSC niche formation.
CSCs can also promote macrophage recruitment by secreting other soluble proteins, including lipoxygenase (LOX), VEGF-A, OLFML3, IRF, Sdf1, and POSTN (Figure 1). In a mouse model of pancreatic cancer, Janakiram et al. demonstrated that LOX can also promote macrophage recruitment [66]. In a model of natural killer T cell loss, stimulation of 5-LOX expression in CSCs resulted in a synchronous increase in the number of M2-type macrophages, suggesting that LOX may be involved in macrophage recruitment [66]. In GBM, binding of the circadian regulator CLOCK and its heterodimeric partner BMAL1 upregulates the transcription of OLFML3, which acts as a chemokine and simultaneously recruits immunosuppressive microglia to the TME to enhance CSC stemness [40]. In the early stage of brain tumors, GSCs, marked by Sox2, were found to promote macrophage/microglia infiltration through the Sdf1b–CXCr4b signaling pathway, and in vivo confocal imaging revealed that macrophages/microglia and CSCs have highly dynamic interactions and do not lead to the phagocytosis of CSCs [41]. In GBM, CSCs secrete more POSTN than non-CSCs to recruit macrophages, and neutralization of POSTN can significantly reduce TAM density [87]. POSTN binds to integrin αvβ₃ receptors on macrophages to increase their recruitment, which can be inhibited by blocking this signaling with the RGD peptide [69]. These results reveal that CSCs promote macrophage recruitment by secreting several soluble proteins. Targeting these pathways can be a new strategy for cancer treatment.

2.2. CSCs Promote Macrophage Polarization in the TME

When macrophages infiltrate the TME, CSC-secreted cytokines can induce the polarization of macrophages into M2 TAMs. These cytokines include CC chemokines, the IL family, prostaglandin E2 (PGE2), WNT1-inducible signaling pathway protein 1 (WISP1), and exosomes, some of which are also involved in macrophage recruitment (Figure 1). Table 2 summarizes the CSC-secreted cytokines and soluble protein molecules that promote macrophage polarization in different tumor types.
Table 2. CSCs secreted molecules to induce the polarization of macrophages.
Table 2. CSCs secreted molecules to induce the polarization of macrophages.
Chemokines Produced by CSCsChemokine ReceptorType of CancerAssociated Signaling Pathway/Mechanism of Cytokine SecretionReference
CCL2CCR2Breast cancerβ-catenin/CCL2 axis[48]
IL-4IL-4RBreast cancerERK1/2 1[88]
IL-6IL-6RGlioblastomaTLR4[89]
Triple negative breast cancerHLF/MCT-1[90,91]
IL-8IL-8ROvarian cancerSTAT3[92]
IL-10IL-10ROvarian cancerPPARγ 2/NF-κB[93]
IL-13IL-13RCholangiocarcinomaNot shown[94]
IL-34IL-34RCholangiocarcinomaNot shown[94]
IL-33ST-2Squamous cell carcinoma NRF2[44]
CSF1CSF1RGlioblastoma Not shown[64]
Non-small cell lung cancer Oct4[95]
Breast cancerNot shown[96]
CSF2CSFR2Triple negative breast cancerNF-κB[97]
Pancreatic cancerNot shown[98]
WISP1Integrinα6β1Glioblastoma WNT/β-catenin[99]
TGF-βTGFBRGlioblastoma Not shown[64]
Pancreatic cancerNot shown[98]
Hepatocellular carcinomaTLR4[100]
MIC-1Not shownGlioblastoma Not shown[64]
PGE2 3EP2Ovarian cancerCOX-2 4/PGE2 [101]
GlioblastomaARS2 5/MAGL[102]
OsteoactivinNot shownCholangiocarcinomaNot shown[94]
ExosomesNot shownPancreatic cancerNot shown[103,104]
GlioblastomaNot shown[105]
Esophageal squamous cell carcinoma Not shown[106]
1 Extracellular signal-regulated protein kinases 1 and 2; 2 Peroxisome proliferator-activated receptor-γ; 3 Prostaglandin E2; 4 Cyclo-oxygenase 2; 5 Arsenite-resistance protein 2.

2.2.1. CSCs Induce Macrophage Polarization by Secreting CC Chemokines

Some chemokines secreted by CSCs, including CCL2 and CCL5, can also promote macrophage polarization while recruiting macrophages. For example, breast CSCs (BCSCs) activate the β-catenin pathway to regulate CCL2 expression and promote macrophage polarization to the M2 type [48] (Table 2). In addition, CSCs promote macrophage polarization through the β-catenin/CCL2 axis. However, macrophages secrete CCL2 to maintain the characteristics of BCSCs [48]. Zhuang et al. reported that CCL5 can transform M0 macrophages (inactivated macrophages) into M2 TAMs when cocultured with hepatoma cells; the result is the expression of high levels of IL-10, IL-12, and TNF-α [55]. However, the mechanism by which activated TAMs secrete cytokines via the CCL5–CCR5 pathway remains unclear, thus necessitating further research.

2.2.2. CSCs Promote Macrophage Polarization by Secreting ILs

To date, most studies support the idea that CSCs induce macrophage polarization in the TME by secreting ILs, including IL-6, IL-8, IL-10, IL-13, and IL-34 (Table 2). Weng et al. reported that MCT-1-overexpressing triple-negative breast cancer (TNBC) cells secrete IL-6, which activates monocytes/macrophages to an M2 TAM-like phenotype via the JAK2/STAT3 pathway and that this pathway can be inhibited after neutralization with IL-6 antibodies [90,91]. In liver cancer, M2 polarization of macrophages is also regulated via the IL-6/STAT3 pathway, the inhibition of which causes macrophages to be polarized to M1 [107]. In addition, CSCs may secrete IL-8 to promote macrophage polarization. IL-8 levels are increased when ovarian CSCs (OCSCs) are cocultured with macrophages [92]. OCSC can promote M2 polarization of macrophages via the IL-8/STAT3 signaling pathway. In addition, IL-10 is highly expressed in OCSCs and induces macrophage polarization toward M2 TAMs by activating the PPARγ/NF-κB pathway [93]. These findings suggest that CSCs can induce macrophage polarization in the TME by secreting ILs, but the specific underlying pathways remain unclear.

2.2.3. CSCs Promote Macrophage Polarization through Other Cytokines and Soluble Protein Molecules

The cytokine CSF secreted by CSCs, which promotes macrophage recruitment, can also cause macrophage polarization. In pancreatic cancer, TGF-β1 and CSF-2 secreted by pancreatic CSCs (Pa-CSCs) were reported to be positively correlated with the levels of M2 TAM-related molecules. In addition, the use of TGF-β1 or CSF-2 inhibitors repressed the polarization of M2 TAMs, indicating that Pa-CSCs can promote macrophage polarization to M2 TAMs through TGF-β1 and CSF-2 [98]. High CSF-2 levels can be secreted by TNBC cells in an NF-κB-dependent manner to promote macrophage polarization into M2 macrophages [97]. CSF-1 can also promote macrophage polarization in a glioma model [64]. M1 macrophages promote lung cancer to express octamer 4 (Oct4) to upregulate CSF-1 gene expression, and CSF-1 stimulates the activation of M2 macrophages [95]. Notably, M1 macrophages induce CSF-1 gene expression in CSCs, thereby stimulating M2 TAM activation and extending our understanding of TAMs.
CSCs can also secrete prostaglandin F2 (PGF2), WISP1, and other soluble protein molecules to promote the activation of macrophages in the TME. In GSCs, PGF2, produced via the ARS2–MAGL signaling pathway, can induce the transformation of macrophages to M2 TAMs [102]. In addition, OCSCs secrete PGE2 via the COX-2/PGE2 pathway, which activates Janus kinase (JAK) signaling in M2 macrophages and significantly increases the secretion of IL-10 and CD206+ on M2 macrophages [101]. CSCs may activate the related pathway of M2 macrophage polarization by secreting PGF2. Furthermore, GSCs induce the secretion of WISP1 via the WNT pathway, supporting M2 polarization of macrophages in a paracrine manner. In addition, inhibition of WNT/β-catenin-WISP1 signaling can significantly reduce macrophage polarization [99]. To summarize, CSCs can transform macrophages into TAMs by secreting soluble proteins.
The direct contact of some proteins on CSCs and TAMs can also promote macrophage polarization. CSCs can bind phosphatidylserine to apoptotic cell membranes by binding to receptor tyrosine kinase family protein molecules (Tyro3, Axl, and MerTK) on TAMs using bridging ligand Gas6 and protein S, and post-efferocytosis, macrophages are polarized to an M2-like phenotype [108]. This finding suggests that CSCs are involved in macrophage transformation into TAMs by secreting these proteins in different tumor types. More studies on macrophage transformation into TAMs are warranted to reveal the immunosuppressive mechanisms in malignant tumors, and these pathways will provide reliable ideas for future tumor therapy.

2.2.4. CSCs Polarize Macrophages by Secreting Exosomes

CSCs can also induce macrophage transformation into TAMs by producing exosomes in the TME [103,104]. GSC-derived exosomes can enter monocytes to reconstitute the actin cytoskeleton, thereby inducing their transformation into M2 TAMs [105]. The same result was also observed in esophageal squamous cell carcinoma [106]. In conclusion, CSCs may stimulate macrophage polarization by secreting exosomes and releasing miRNAs; this may provide a direction for the development of new biomarkers.

3. TAMs Enhance the Stemness Characteristics of Tumor Cells

The supporting role of TAMs in CSCs is crucial. After polarization, TAMs actively participate in CSC niche formation and play a crucial role in maintaining the self-renewal capacity of CSCs and tumor initiation. Notably, cytokines produced by TAMs can enable “differentiated” cancer cells to regain the characteristics of CSCs and maintain CSC stemness in tumors. This process of regaining the characteristics of CSCs transpires mainly through epithelial–mesenchymal transition (EMT) [109]. Table 3 summarizes the TAMs that support CSC stemness by secreting cytokines, soluble proteins, and exosomes in different tumor types.
Table 3. TAMs secreted molecules to enhance the properties of CSCs.
Table 3. TAMs secreted molecules to enhance the properties of CSCs.
TAM TypeFactor of TAM ProductionType of CancerAssociated Signaling Pathway/Mechanism of TAMs on CSCsReference
M2CCL2Breast cancerβ-catenin [48]
CCL5Prostate cancerβ-catenin/STAT3[110]
GlioblastomaNot shown[52]
CCL7Ovarian cancerCCR3/MM9 1[111]
CCL8GlioblastomaCCR1/CCR5/ERK1/2[112]
Gastric cancerJAK1/STAT3[113]
CCL17Hepatocellular carcinomaWNT/β-catenin[114]
CCL22Hepatocellular carcinomaNot shown[115]
CXCL1Breast cancerNot shown[116]
CXCL7GlioblastomaNot shown[73]
CXCL8Breast cancerCXCR2[117]
IL-1βGlioblastomap38 MAPK[118]
IL-6GlioblastomaNot shown[89]
Ovarian cancerWNT[62]
Hepatocellular carcinomaSTAT3[119]
Breast cancerSTAT3[120]
IL-8Ovarian cancerJAK2/STAT3 [92]
IL-10Ovarian cancerWNT [62]
Non-small cell lung cancerJAK1/STAT1/NF-κB/Notch1[121]
IL-18Gastric cancerNot shown[122]
IL-35Breast cancerNot shown[123]
TGF-βHepatocellular carcinomaEMT[124]
Pancreatic cancerTGF-β1/smad2/3 axis[125]
Squamous cell carcinomaNRF2[44]
Lung cancerNot shown[126]
Breast cancerEMT[127]
TNF-αClear cell renal cell carcinomaNF-κB[128]
ChordomaPI3K/AKT[129]
EGF 1Breast cancerEGFR/STAT3/Sox2[130]
GPNMB 2Breast cancerPI3K/AKT/mTOR & β-catenin/MAPKs/AMPK/Src[131]
PTN 3LymphomaPTPRZ1/β-catenin[132]
GlioblastomaPTN-PTPRZ1[133]
IGF 4Thyroid cancerPI3K/AKT/mTOR[134]
PDGF-BB 5Breast cancerNot shown[135]
hCAP-18Pancreatic ductal adenocarcinomaNot shown[136]
S100A9 6Hepatocellular carcinomaNF-κB[137]
HMGB1 7Non-small cell lung cancerNF-κB[138]
MFG-E8 8Colorectal cancerSTAT3/Sonic Hedgehog[139]
extracellular vesiclesPancreatic cancerKLF3[140]
M1IL-6Oral squamous cell carcinomaJAK/STAT3[141]
IL-12GlioblastomaNot shown[142]
TNF-αLung cancerNot shown[143]
Oral cancerNot shown[144]
1 Epidermal growth factor; 2 Glycoprotein nonmetastatic B; 3 Pleiotrophin; 4 Inslin-like growth factor; 5 Platelet-derived growth factor-BB; 6 Matrix metalloproteinase 9; 7 High mobility group box 1; 8 Milk-fat globule-epidermal growth factor-VIII.

3.1. CC and CXC Chemokines Are Secreted by TAMs to Maintain Tumor Cell Stemness Characteristics

Notably, the current study shows that TAMs maintain the characteristics of CSCs by secreting CC chemokines, such as CCL2, CCL5, CCL7, CCL8, and CCL17 (Table 3) (Figure 1). As described in Section 2.2.1, β-catenin secretion by BCSCs directly regulates CCL2 expression, promoting macrophage recruitment and TAM polarization. TAMs also express CCL2 and promote the maintenance of BCSC properties [48]. Similarly, CCL5, a chemokine that induces the recruitment and activation of macrophages, has also been found to be secreted by TAMs to promote CSC properties. In GBM, TAMs secrete CCL5, which enhances GSC stemness and tumor invasion via the CCL5–CCR5 pathway [52]. CCL5 activates β-catenin/STAT3 signaling to promote the self-renewal and tumor metastasis of prostate CSCs (PCSCs), and its knockdown significantly inhibits the metastasis of prostate cancer and the self-renewal capacity of PCSCs [110]. TAMs secrete CCL7, which binds to CCR3 to activate MMP9 expression in ovarian cancer cells, significantly increasing their invasiveness [111]. In addition, TAMs secrete large amounts of CCL8 in the TME, binding to receptors CCR1 and CCR5 on glioma cells, which activates ERK1/2 and can induce the aggressive and stemness characteristics of GBM [112]. However, TAM-derived CCL-17, CCL-22, TGF-β, IL-10, arginase 1, and galectin-3 can significantly reduce tumor growth and metastatic sites, but the specific mechanism remains unclear [145]. In summary, TAMs can secrete CC chemokines to support the growth of CSCs and targeting these chemokines may effectively block the positive feedback loop-like association between CSCs and TAMs.
TAMs also secrete CXC chemokines (CXCL1 and CXCL7) that support CSC-like characteristics (Table 3). M2 TAMs secrete CXCL1 in breast cancer and promote tumor metastasis. The use of CXCL1 promoter inhibitors reduced the migration of breast cancer cells and the number of BCSCs, suggesting that TAMs may support BCSC survival by secreting CXCL1 [116]. In addition, CXCL7 secretion was significantly positively correlated with higher numbers of TAMs and the enhanced stemness of GBM. FGL2 produced by GBM induces macrophage recruitment and promotes macrophages to exhibit TAM-like properties. Activated TAMs secrete CXCL7 via the CD16/SYK/PI3K/HIF1α pathway, thereby enhancing the stem-like characteristics of GBM cells [73]. In conclusion, TAMs secrete CC and CXC chemokines to maintain CSC stemness. Therefore, blocking the secretion of these chemokines can block CSC–TAM interactions, thus effectively inhibiting tumor growth and metastasis.

3.2. TAMs Support the Stemness of Tumor Cells by Secreting ILs

TAMs can also promote the self-renewal and metastasis of CSCs by secreting ILs. IL-6, a common IL factor, plays a vital role in promoting tumorigenesis. Activation of the TLR4 signaling pathway on microglia promotes the secretion of IL-6, and IL-6 can support GSC in regulating tumor growth [89]. In human HCC samples, the same results were observed. TAMs activated STAT3 to produce IL-6 and induced an increase in the number of CD44+ hepatoma cells [119]. Another study reported that M2 TAMs could also secrete IL-6 through the WNT ligand WNT5B, inducing the enrichment of CSCs in ovarian cancer [62]. Moreover, CSC-derived WNT ligands can drive the activation of M2 macrophages. Thus, a positive feedback loop is formed between CSCs and M2 TAMs through WNTs. In addition, M1 macrophages can secrete IL-6. In oral squamous cell carcinoma cells, M1 TAMs can promote EMT and induce the CSC phenotype by upregulating the expression of MME and MMP14. However, M1 TAMs secrete IL-6, increasing CSC stemness by activating the JAK/STAT3 pathway [141]. These findings indicate that TAMs secrete IL-6 through the TLR4 signaling pathway and STAT3 pathway and that IL-6 promotes the chemoresistance and invasiveness of CSCs by activating the JAK/STAT3 pathway and WNT pathway in CSCs, forming a CSC–TAM–CSC positive feedback loop.
TAMs can also secrete IL-10, IL-8, IL-12, and IL-35, which can promote the stemness characteristics of tumor cells (Figure 1). In NSCLC, IL-10 is derived from TAMs via the JAK1/STAT1/NF-κB/Notch1 signaling pathway and induces CSC-like properties in NSCLC cells. Blocking IL-10/JAK1 signaling significantly inhibits TAM-mediated CSC-related genes, such as Sox2, Oct4, and Nanog [121]. This pathway may provide a new potential therapeutic target for the treatment of NSCLC. In addition, IL-8 secreted by TAMs in ovarian cancer via the STAT3 pathway can also increase the stemness characteristics of OCSC [146]. Notably, in GBM, GSCs express higher amounts of active spin cleavage-like products (ALPs) than non-GSCs. ALPs promote secretion of IL-12 by M1 TAMs, which can enhance the activity of GSCs and drive the formation of the GSC niche [142]. In addition, the expression of RON signaling in TAMs induces the secretion of IL-35, which supports the self-renewal and metastasis of CSCs; however, the specific mechanism remains under investigation [123]. Together, these findings reveal that TAMs secrete ILs to promote the expression of CSC genes in various cancer models. In addition, M1 macrophages can secrete IL-6 and IL-12, inducing the expression of CSC genes. Studies continue to find more IL factors secreted by TAMs, and they play a crucial role in the maintenance of CSC stemness, but further research is required to clarify the precise role of TAMs in the maintenance of CSC stemness.

3.3. Other Cytokines and Soluble Protein Molecules Secreted by TAMs to Promote the Stemness of Tumor Cells

TAMs can also directly support tumor stemness by secreting cytokines, such as TGF-β1, FGF, and PGE2 (Figure 1). TAMs secrete more TGF-β1 than macrophages of other phenotypes, and TGF-β1 activates EMT and acquires CSC-like properties [124]. Furthermore, FGF expression in macrophages significantly increases the stemness of GBM cells. FGF1, the most abundant FGF in the FGF family, was upregulated in TAMs cocultured with GBM and enhanced the stemness characteristics of GMB [147]. In addition, in a breast cancer model, TAMs were noted to establish a paracrine EGFR/STAT3/Sox2 signaling pathway and increase Sox2, Oct4, Nanog, AbcG2, and Sca-1 gene expression [130]. TAMs can also secrete another cytokine, PGE2. Overexpression of IL-33 by colon cancer cells induces macrophage recruitment and stimulates them to produce PGE2, thereby supporting the stemness of colon cancer cells [148]. These findings show that TAMs are crucial in supporting CSCs and targeting the cytokines that facilitate this process can block tumor cells from acquiring or maintaining stemness characteristics.
TAMs can also support CSCs by secreting soluble protein molecules, such as GPNMB, TNF-α, pleiotrophin (PTN), growth factor-BB, immunomodulatory cationic antimicrobial peptide 18/LL-37 (hCAP-18/LL-37), and S100A9. GPNMB is a glycoprotein that is highly expressed in macrophages and microglia and promotes inflammation. However, in mouse tumor models, M2 TAMs preferentially express soluble GPNMB and bind to the CD44 receptor on tumor cells to trigger CSC proliferation [131]. In addition, in clear-cell renal cell carcinoma (ccRCC), TAMs secrete TNF-α, which may upregulate CD44 expression through NF-κB signaling and enhance CSC stemness [128]. PTN is also a soluble protein molecule secreted by TAMs, which is significantly positively correlated with the expression of PTPRZ1 (PTN receptor) in lymphoma. PTN induces the generation of lymphoma stem cells via the β-catenin pathway [132]. In addition, GSCs can upregulate the expression of PTPRZ1 and activate PTPRZ1 signaling by binding to PTN secreted by M2 TAMs to maintain GSC stemness [133]. Together, these results indicate that PTN secreted by TAMs plays a crucial role in the maintenance of CSC stemness. A recent study revealed that TAMs can also secrete IGF-1 and IGF-2 to promote PI3K/AKT/mTOR signaling and increase the stemness characteristics of thyroid cancer [134]. In pancreatic ductal adenocarcinoma, TAMs secrete hCAP-18/LL-37, creating a paracrine niche beneficial to CSCs [136]. S100A9, a secreted protein associated with the TME, is significantly upregulated in TAMs [149]. It activates the NF-κB signaling pathway in a Ca2+-dependent manner to enhance the stem cell-like properties of hepatoma cells. Simultaneously, treatment of tumor cells with S100A9 significantly increased CCL2 expression, recruited and polarized more macrophages, and formed a CSC–TAM positive feedback loop [137]. These findings suggest that TAMs produce abundant protein molecules in the TME, and these abundant proteins promote the generation and maintenance of CSCs by activating CSC-related pathways.

3.4. TAMs Promote Tumor Cell Stemness through Surface Protein Molecules

TAMs can also directly bind to CSCs to support CSC stemness by expressing corresponding protein molecules on the cell surface, including CD90, EphA4, and SIRP1α. In breast cancer, EMT programmatically upregulates the expression of CD90 and EphA4. CD90 directly binds to receptors on TAMs to mediate physical effects and activate the NF-κB pathway in CSCs and induce the production of cytokines to maintain stemness [21] (Figure 1). In addition, CD47 expression was observed to be elevated in CD133+ lung cancer cells. CD47 also binds to SIRP1α on macrophages, thus inhibiting phagocytosis [20] and supporting immune escape. In conclusion, the surface protein molecules expressed on the surface of TAM-CSC, after sufficient contact, play a significant role in immunosuppression; these surface molecules can be used as immune checkpoints to improve the efficacy of immunotherapy.

3.5. TAMs Promote Tumor Cell Stemness by Secreting Extracellular Vesicles

TAM-derived extracellular vesicles can also promote tumor invasion. The expression of microRNA-21-5p is upregulated in M2 macrophage-derived extracellular vesicles and combines with KLF3 to induce Pa-CSC differentiation [140]. Accordingly, nanocarriers inspired by M2 macrophage microvesicles can penetrate tumor tissue to improve the accessibility of CSCs [150]. Therefore, the use of extracellular vesicles can produce an effective tumor treatment effect.

4. Therapeutic Strategies Targeting CSCs and TAMs

Macrophages constitute the bulk of immune cells. They have tumor-promoting and immunosuppressive roles in cancer [151,152]. For example, the number of TAMs in human tumors is positively correlated with tumor grade and negatively correlated with patient survival in cancers such as GBM, lymphoma, breast, and pancreatic cancers [153,154,155,156,157]. However, when TAMs lose immunosuppressive signals or undergo repolarization, survival is increased in mice and in patients with different types of cancer [158]. In recent tumor treatment studies, macrophage therapy has received great attention due to the contribution of macrophages to CSC growth, metastasis, and promotion of the CSC niche. Several common immunotherapeutic strategies include depleting monocytes to prevent TAM production and recruitment, inducing macrophage polarization toward the M1 phenotype and promoting phagocytosis, targeting TAMs supporting the CSC niche, and suppressing CSC occurrence and metastasis [159]. These strategies are now being tested to enhance antitumor immunity, in conjunction with either conventional chemo-radiotherapy or T cell–mediated immunotherapy [159]. We discuss some potential pathways that can translate into therapeutic strategies that interfere with the interaction between TAMs and CSCs, inhibiting TAM-mediated immunosuppression. Figure 2 presents several key targets that regulate CSC and TAM properties during tumor progression. In addition, we also list the relevant agents targeting TAM in ongoing clinical trials (Table 4).

4.1. Targeting Tumor-Associated Macrophage Recruitment and Survival

TAMs mainly originate from monocyte precursor cells in the circulation system. One strategy to deplete TAMs is to block the replenishment of TAMs through monocyte recruitment or by targeting M2 macrophage markers to specifically clear TAMs from the TME (Figure 2).

4.1.1. CSF1–CSF1R Inhibitors

Blocking the CSF1–CSF1R signaling pathway can inhibit macrophage recruitment to the CSC niche (Figure 2). The CSF1–CSF1R axis can promote TAM differentiation, survival, and recruitment [160]. Blockade of this signaling pathway can greatly reduce the number of TAMs or repolarize M2 macrophages to M1 macrophages in the TME [161,162,163]. Studies have reported that blockade of the CSF1–CSF1R axis improved the effects of immunotherapy [164,165]. Emactuzumab, a monoclonal antibody to CSF1, blocks the activation of this signaling pathway by inhibiting CSF1–CSF1R dimerization. Emactuzumab treatment can reduce the number of F4/80+ TAMs in CSC niches and increase the ratio of CD8+/CD4+ T cells, thereby improving antitumor immune function. Emactuzumab combined with paclitaxel can significantly reduce the number of CSF1R+ TAMs and effectively inhibit the development of advanced solid tumor CSCs [166]. Pexidartinib, an inhibitor of tyrosine kinases on CSF1R, can inhibit the activation of TAMs to block their tumor-promoting effects, thereby reducing the CSC count in the TME. Pexidartinib has been approved by the FDA as an alternative to surgery for tenosynovial giant cell tumor [167,168,169]. The use of monoclonal antibodies or small-molecule inhibitors that target the CSF1–CSF1R signaling pathway can successfully reduce the number of TAMs in the CSC niche and increase the number of infiltrating T cells, thereby enhancing antitumor immunity (Figure 2) [160].
In addition, the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced vaccine (GVAX), a cell-based tumor vaccine transduced by the GM-CSF gene, can be used to block the CSF2–CSF2R signaling pathway, which inhibits TAM activation [170]. Following GVAX inoculation, sustained local release of GM-CSF significantly increased levels of M1 macrophages, dendritic cell populations, and CD8+ T cells, exerting antitumor immunity effects [170]. In addition, the IL-2 vaccine can also induce specific antitumor immunity in mice, and the combination of GVAX and IL-2 vaccine was noted to have better antitumor effects than the single treatment [171]. These results suggest that GVAX can induce strong tumor immunity and can be used in tumor immunotherapy.

4.1.2. CCL2–CCR2 Inhibitors

The migration and recruitment of monocytes from bone marrow to the CSC niche occurs mainly via the CCL2–CCR2 signaling pathway. Therefore, targeting the CCL2–CCR2 axis effectively inhibits the number of invasive TAMs in the CSC niche [172,173] and synergistically improves the efficacy of chemo-radiotherapy and immunotherapy (Figure 2) [174]. TAM activity can also be inhibited by monoclonal antibodies or small-molecule inhibitors targeting CCL2 or CCR2. Carlumab and PF04136309 play a key role in targeting the CCL2–CCR2 signaling pathway. Carlumab is a monoclonal antibody with a high binding ability to CCL2, which can reduce the number of CD68+ TAMs in the CSC niche, increase the phagocytic ability of macrophages, and effectively control the occurrence and metastasis of CSCs [175,176]. PF04136309, a CCR2 antagonist, inhibits the metastatic infiltration of CCR2+ macrophages and reduces the proportion of TAMs in the CSC niche [177]. In addition, in esophageal cancer cells, aspirin can reduce the recruitment of M2 macrophages, the expression of CSC marker genes CD90 and Nanog, and the ability of CSCs to form spheroids by inhibiting CCL2 expression on TAMs [178].
The combined application of siRNA and drugs to block the CCL2/CCR2–STAT3 axis can inhibit the activity of TAMs in the CSC niche, thus enhancing the phagocytic activity of macrophages and inhibiting tumor growth and metastasis [179]. In addition, siRNA complexed with TAT cell-penetrating peptide (Ca-TAT) can silence genes more effectively than traditional antibody neutralization [75].
Low-dose photodynamic therapy can significantly inhibit CCL2 secretion in MSCs, possibly limiting the infiltration of precancerous macrophages [180]. However, whether it can inhibit CSC secretion of CCL2 remains to be further investigated.

4.1.3. CXCL12–CXCR4 Inhibitors

The CXCL12–CXCR4 signaling pathway is associated with macrophage recruitment, and its blockade in combination with traditional therapy has shown antitumor efficacy (Figure 2) [181]. Some small-molecule antagonists against the CXCL12–CXCR4 axis can be used for antitumor therapy, such as small peptide CXCR4 antagonists, nonpeptide CXCR4 antagonists, anti-CXCR4 antibodies, modified CXCL12 agonists, and antagonists [182,183]. Plerixafor is a CXCR4 antagonist that inhibits CXCL12–CXCR4-mediated chemotaxis and reduces macrophage recruitment in CSC niches. CXCR4 is highly expressed in most CSCs, and the combination of nonpeptide CXCR4 antagonists with etoposide and cisplatin reduces CSC proliferation and inhibits tumor growth [184]. In addition, highly diffused CD133+ CXCR4+ cells (metastasis promoter cells, MICs) can trigger distant metastasis and resist conventional chemotherapy. A new CXCR4 inhibitory peptide can prevent the expression of CD73, CD38, and IL-10 by MICs, thus inhibiting macrophage transformation into TAMs and rescuing the antitumor cytotoxic effects of T cells [185].
However, these target molecules for TAM-targeted therapy may exist in cell populations other than TAMs. For example, CCR2 and CXCR4 are also expressed on lymphocytes [186,187,188]. Simultaneous targeting of these molecules may lead to altered functions of these immune cells as well as various other complications; therefore, future studies should attempt to identify more specific targets.

4.2. Targeting TAM Activation

Another strategy is to reprogram macrophages to repolarize toward the M1 antitumor phenotype from the M2 tumor-promoting phenotype and enhance macrophage phagocytosis to produce antitumor effects (Figure 2).

4.2.1. TAMR Inhibitors

TAMR, a collective term for Tyro3, Axl, and MerTK, is a group of tyrosine kinase receptors that are ubiquitously expressed on CSCs and various immune cells. Axl and MERTK are overexpressed in CSCs, which can promote M2 polarization of macrophages [189,190,191]. The interaction of TAMR with ligands Gas6 and protein S can stimulate M2 polarization, promote the production of immunosuppressive cytokines, prevent the activation of immune T cells, limit the inflammatory response, and favor CSC survival [192]. Therefore, blocking the TAMR signaling pathway may effectively inhibit the activities of TAMs and CSCs (Figure 2). Researchers have discovered that UNC2025, a small-molecule inhibitor of MERTK, can inhibit macrophage transformation into TAMs, increase the phagocytosis of M1 macrophages, and reduce the CSC count [193].

4.2.2. Anti-MARCO Antibody

MARCO, also known as SCARA2, is a macrophage receptor with a collagenous structure that drives the transformation of macrophages into TAMs. Macrophages expressing MARCO represent a subset of macrophages with a protumor and anti-inflammatory immunosuppressive phenotype [194,195]. Therefore, targeting MARCO may effectively inhibit TAM activation (Figure 2). In breast and colon cancer and melanoma models, the use of anti-MARCO antibodies can reprogram TAMs toward the M1 phenotype, reduce the proportion of TAMs in the CSC niche, increase tumor immunogenicity, and improve the efficacy of antitumor immunotherapy [194,196].

4.2.3. Toll-Like Receptors and Stimulator of Interferon Gene Agonists

Toll-like receptors (TLRs) on macrophages can trigger TAM repolarization. TLRs can bind to some endogenous molecules, participate in antivirus and antitumor regulation by activating NF-κB, and produce various immune-stimulating cytokines, including type I IFN, which increase the expression of MHC-I, promote the repolarization of TAMs to the M1 phenotype, and enhance the specific recognition of CSCs by the immune system [197,198]. Encouraging results have been obtained within the TLR3, TLR7, TLR8, and TLR9 agonists in mouse tumor models. Among these, the TLR7 agonists imiquimod and 852A have shown potential antitumor ability and durable therapeutic effects in preclinical trials [199]. Another key factor regulating innate immunity is the stimulator of interferon genes (STING), which also leads to the production of immune-stimulating cytokines [200]. Activation of TLRs and STING can induce good antitumor effects in the body [201,202,203]. On the basis of these results, TLR and STING agonists are being used in CSC treatment trials in combination with other chemotherapy and immunotherapies (Figure 2) [202,203].

4.2.4. Others

PI3Kγ signaling can promote the immunosuppressive activity of TAMs. In melanoma, lung, and pancreatic cancer cell models, the blockade of PI3Kγ expression in TAMs activates NF-κB, which promotes the immunostimulatory transcriptional program, rescues CD8+ T cell cytotoxicity, and promotes the regression of CSCs and prolongs survival in mouse models in combination with immune checkpoint inhibitor therapy [151,158].
TAMs can express histone deacetylases (HDACs), and HDAC inhibitors can perform epigenetic reprogramming of macrophages, repolarizing them toward the M1 phenotype, and induce T cell toxicity [204]. Inhibition of HDAC2 expression in TAMs through siRNA knockdown or by using pharmacological inhibitors (ISAHA and VPA) can induce TAM repolarization, activate the antitumor phenotype of macrophages, help T-lymphocyte responses, and increase the sensitivity of CSCs to chemotherapy and immune checkpoint inhibition [204]. Furthermore, the histone methyltransferase EZH2 utilizes a methylation modification to inhibit miR-454-3p and promotes the m6A modification of PTEN to polarize macrophages toward the M2 phenotype. Of note, combined treatment with EZH2 inhibitors and PD-1 inhibitors restored the cytotoxicity of macrophages and blocked the progression of CSCs [205,206,207,208,209,210,211,212].
IFN-γ is a potent antiviral bioactive substance, and celecoxib is a nonsteroidal anti-inflammatory drug. In a study, their combination decreased the M2/M1 macrophage ratio in the CSC niche reduced the expression of MMP-2, MMP-9, and VEGF of TAMs; and inhibited microangiogenesis in the CSC niche [213]. Thus, IFN-γ and celecoxib may inhibit CSC development by regulating the ratio of M2/M1 macrophages in the TME.
Folic acid oral solution (OOS) has anti-inflammatory, antioxidant, and antitumor effects and can inhibit M2 macrophages in vitro and in vivo without side effects. In addition, OOS can target plastic CSCs as well as the TME that supports CSCs [214]. Therefore, OOS, which can counteract the hypoxic state and inhibit the activity of TAMs and CSCs in the TME, can be a candidate for adjuvant therapy in malignant tumors.
ZnO/NPs loaded with the chemotherapeutic drug doxorubicin (Dox) are multifunctional and multitargeted nanodrug carriers. ZnO(Dox)/NPs can promote Dox-induced polarization of macrophages to an M1 phenotype in vivo and increase the sensitivity of CSCs to doxorubicin treatment. ZnO(Dox)/NPs can also effectively downregulate CD44, a key surface marker of CSCs, reduce CSC stemness, and inhibit CSC development [215].

4.3. Targeting CSC–TAM Interaction

The molecules that mediate CSC–TAM interaction are important for maintaining CSC stemness. Accordingly, specifically interfering with this interaction and disrupting the plasticity of CSCs and TAMs in the TME may lead to more effective antitumor responses (Figure 2) [19,216].

4.3.1. Blockade of the SIRP-α–CD47 Pathway

SIRP-α can inhibit the phagocytosis of CSCs by TAMs and promote tumor immune evasion (Figure 2) [217]. Therefore, blocking the CD47–SIRP-α pathway can interfere with CSC–TAM interaction [218,219]. Several studies are currently underway to test the efficacy of monoclonal antibodies and small-molecule inhibitors targeting SIRP-α or CD47.
CD47 deletion may reduce the stemness properties of glioma CSCs. Anti-CD47 antibody therapy increased the phagocytic activity of macrophages to glioma stem cells, decreased cancer stemness, and resulted in a significant inhibition of tumor growth [219]. Subsequently, the combination of CD47 blockers and the antimicrotubule drug cabazitaxel promoted programmed cell clearance (PRCR) in TNBC cells as well as the activation of NF-κB and generation of immune-stimulating cell factors, which promoted macrophage polarization to the M1 phenotype and inhibited the activity and self-renewal of CSCs [220]. Accumulating evidence suggests that targeting CD47 may be a safe, effective, and promising treatment strategy for CSCs. Similarly, anti-SIRP-α antibodies can increase macrophage-mediated phagocytosis and T-cell-mediated cytotoxicity to CSCs and reduce the resistance of CSCs to antiangiogenic therapy [221,222]. Notably, blocking the SIRP-α-CD47 signaling pathway in combination with chemoradiotherapy can further improve the antitumor therapeutic effect [19]. Moreover, targeting E3-ligase-mediated CD47 degradation to boost anticancer immunity also has been discussed [223]. The above findings suggest that blocking the SIRP-α-CD47 signaling pathway with monoclonal antibodies or small-molecule inhibitors can effectively block CSC–TAM interaction, thereby inhibiting the progression of CSCs and reducing the tumor burden in vivo.

4.3.2. Targeting IL-Mediated Signaling Pathways

Cytokines such as IL-6 and IL-10 are essential for maintaining CSC stemness and promoting TAM activation, and their inhibitors have shown potent antitumor activity [224]. IL-6 secreted by TAMs can promote the proliferation and development of CSCs, and IL-6 levels correlate with the specific phenotype of CSCs. CD44+ HCC cell lines exhibit CSC activity in vitro; however, tocilizumab, a monoclonal antibody to the IL-6 receptor, can inhibit IL-6 function and downregulate TAM-mediated CD44+ cell activity, reducing the stemness of CD44+ cells [119]. TAMs can activate the Sonic Hedgehog (SHH) and STAT3 signaling pathways in CSCs by releasing milk fat globule-EGF factor 8 (MFG-E8), which plays a vital role in regulating the activity of CSCs and enhances the resistance to antitumor drugs [225,226]. IL-6 can increase the tumorigenic activity of CSCs by enhancing MFG-E8-induced activity, and the combined blockade of IL-6 and MFG-E8 can significantly reduce the number of primary tumor-derived CSCs, whereas blockade of IL-6 alone or MFG-E8 has only a partial antitumor effect. Furthermore, the inhibition of the MFG-E8-activated SHH and STAT3 pathways significantly increased cisplatin (CDDP)-induced apoptosis of CSCs [139]. In addition, blocking the IL-6/STAT3 signaling pathway has also shown some antitumor activity, with STAT3 involved in the recruitment and polarization of TAMs [64,227]. Anti-IL-6 or anti-IL-6R antibodies or small-molecule inhibitors targeting STAT3 can inhibit the activity of CSCs and M2 polarization of macrophages, thus inhibiting the progression of CSCs [39,91]. STAT3 knockdown by siRNA prevented tumor cell proliferation and blocked CSC-mediated immunosuppression [228,229].
A study reported an antitumor drug candidate, CBP501, which inhibits the lipopolysaccharide-induced secretion of TNF-α, IL-6, and IL-10 from TAMs. Coculture of CBP501 with lipopolysaccharide-treated TAMs inhibited the formation of tumor spheroid cells. In addition, CBP501 can also inhibit the expression of CSC marker ABCG2 by inhibiting the interaction between VCAM-1+ cancer cells and VLA-4+ macrophages [143].
IL-8 and CXCR1/2 are a pair of homologous receptors. IL-8 enhances the tumor-promoting activity of TAMs and promotes the progression of CSCs by helping the interaction between CSC and TAM. The IL-8/CXCR1/2 signaling pathway is partially mediated by the EGFR/HER2 signaling pathway. Therefore, the simultaneous application of CXCR1/2 inhibitors and HER2 inhibitors in cancer therapy can inhibit the tumor-promoting activity of TAMs, inhibit the proliferation and migration of CSCs, and improve the overall survival rate of patients with cancer [230,231].
IL-33 can upregulate the expression of MMP-9 in TAMs. MMP-9 can modify activated receptors on NK cells, CD8+ T cells, CD4+ T cells, as well as inhibit the expression of antigen-presenting molecule MHC-I on the surface of CSCs, which interferes with the interdependence between CSCs and TAMs [232]. These findings suggest that IL-33-induced TAMs can significantly reduce the toxic effects of immune cells on CSCs through MMP-9 blockade [233,234]. Therefore, the researchers reasoned that the antitumor effect of immune cells could be enhanced by inhibiting the proteolytic activity of MMPs, namely by targeting the IL-33–TAMs–MMP-9 axis-mediated immune tolerance of CSCs; this represents a new therapeutic modality against CSCs.

4.3.3. Others

Targeting promoting-cancer pathways, such as WNT, Notch, and Hedgehog, can help disrupt the interdependence between CSCs and TAMs [235]. During CSC–TAM interaction, activated WNT signaling initiated by TAMs can promote the chemoresistance and invasive phenotypes of CSCs [62], suggesting that targeting the WNT pathway may inhibit the activity of CSCs and TAMs.
The EGFR/STAT3/Sox2 paracrine signaling pathway, which mediates CSC–TAM interaction, can promote the expression of the CSC marker gene Sox2, thus enhancing CSC stemness, and increase the ability of drug excretion and chemotherapy resistance in vivo [130]. Therefore, knockdown of Sox2 in CSCs by siRNA can inhibit the TAM-mediated stemness of CSCs, thereby inhibiting tumor progression [130]. In addition, targeting EGFR and STAT3 by small-molecule inhibitors AG1478 and CDDO-Im, respectively, can also effectively block this signaling pathway [130]. Other pathway inhibitors, such as Vismodegib, an SHH pathway inhibitor, can be used to treat metastatic or advanced localized basal cell carcinoma [236]. In addition, imatinib is a tyrosine kinase inhibitor that can inhibit STAT6 phosphorylation and nuclear translocation, thereby promoting macrophage-induced phagocytosis and reducing CSC growth and migration [237]. Notably, these signaling pathways also act on normal tissue cells. Therefore, to minimize adverse effects, researchers should find therapeutic pathways that specifically target CSCs or TAMs, which is undoubtedly a major challenge.
One study demonstrated that inhibition of CSC-specific POSTN and its related pathways or the GBM derivative CCL5 disrupted CSC–TAM interaction in mouse models of GBM and prostate cancer [69,110]. Maraviroc is a CCR5 antagonist that significantly inhibits the proliferation, colony formation, and migration of CCR5+ CSCs [52].
Chemokine ligand 8 (CXCL8) is a key chemokine secreted by TAMs that can mediate CSC–TAM interaction. CXCL8 significantly increased the migration, invasion, and EMT events of breast cancer cells, as well as the self-renewal of BCSCs [117]. Although danirixin is a selective antagonist of CXC chemokine receptor 2 (CXCR2), in vivo analysis confirmed that it can also inhibit the activity of CXCL8, thus inhibiting the self-renewal and metastasis properties of BCSCs [238,239]. These results suggest that danirixin treatment significantly abrogates TAMs/CXCL8-mediated tumor-promoting efficacy.
Metformin is a traditional drug commonly used to treat type 2 diabetes. It can significantly reduce the proportion of ALDH+CD133+ CSCs in patients with ovarian cancer and can decrease HCC stemness [240,241]. In addition, metformin may also increase macrophage phagocytosis by reprogramming the functional activity of macrophages [242], thereby affecting the interaction of TAMs with CSCs, reducing CSC levels, and inhibiting tumor growth.
Survivin, an antiapoptotic protein, is present in human adipose tissue-derived stem cells (HASCs) and is a diagnostic marker of tumor recurrence. One study reported that HASCs isolated from obese patients had significantly elevated survivin expression; at the same time, the antiapoptotic capacity of HASCs and the activity of soluble proinflammatory cytokines (such as IL-1β) secreted by M1 macrophages were also improved [243]. Therefore, targeting survivin may present a new approach to cancer treatment.
Despite the attractive therapeutic strategies targeting CSCs, they lack efficient and specific CSC targets. Furthermore, due to their high plasticity, CSCs may become reestablished after the treatment-induced loss of stemness. Similarly, some TAM-targeting strategies, such as antagonizing CSF1R, also have only a weak antitumor effect, which may be due to some altered pathways in the TME leading to unknown therapy resistance mechanisms of TAMs.

5. Summary and Perspective

CSCs represent a small portion of the cancer cells in a tumor, but they have very powerful self-renewal properties, are often resistant to chemotherapy and radiation therapy, and promote tumor recurrence and metastasis. This article reviewed the molecular mechanism of how CSC–TAM interaction maintains tumor progression. This bidirectional effect is manifested at several levels, including the role of CSCs in promoting the recruitment and polarization of TAMs and the role of TAMs in promoting CSC stemness and CSC niche formation. We also described potential therapeutic strategies, including various monoclonal antibodies, small-molecule inhibitors, agonists, and antagonists, that involve targeting (1) tumor-associated macrophage recruitment and survival, (2) tumor-associated macrophage activation, and (3) cytokines and chemokines involved in CSC–TAM interaction. However, most treatments are still in preclinical studies. A major challenge is the high plasticity of CSCs and the lack of efficient and specific therapeutic targets. In addition, macrophages play various important roles in the immune system, and excessive depletion of these cells can seriously affect immune function. To address these issues, new therapeutic strategies are required to target markers of TAMs and CSCs efficiently and specifically to precisely target these tumor immunosuppressive cell populations. Furthermore, current knowledge of TAMs and CSCs relies heavily on tumor engraftment analysis in syngeneic and xenograft models; these models do not exhibit the exact same TME as the primary tumor, nor do they fully mimic the interaction between human CSCs and TAMs. Therefore, developing a mouse model with a human-like immune system that can support the growth of human primary tumors will elucidate the relationship between TAMs and CSCs.
In conclusion, the interaction between TAMs and CSCs during tumor progression constitutes a positive feedback loop and exerts structural maintenance and protective effects on the CSC niche. Although many therapeutic strategies have been proposed to target TAMs and CSCs, the results remain unsatisfactory. Further elucidation of the mechanisms and roles of CSC–TAM interaction in the progression of CSCs and using TAM-specific and CSC-specific targeted immunotherapy to treat CSCs are warranted to develop more effective therapies with minimal adverse effects.

Author Contributions

The draft was written and edited by S.L., G.Y. and P.Y.; N.C. and X.C. reviewed and edited the manuscript; W.-H.Y. and X.Y. wrote the manuscript and supervised the entire work. All authors agree to be responsible for the publication. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the following: Guangzhou key medical discipline construction project fund; the National Natural Science Foundation of China (81872138 and 82172789); China Medical University YingTsai Young Scholar Award (CMU108-YTY-04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Due to the limited space, we apologize for any publications that may be omitted from the references.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhou, H.M.; Zhang, J.G.; Zhang, X.; Li, Q. Targeting cancer stem cells for reversing therapy resistance: Mechanism, signaling, and prospective agents. Signal Transduct. Target. Ther. 2021, 6, 62. [Google Scholar] [CrossRef] [PubMed]
  2. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
  3. Abdoli Shadbad, M.; Hosseinkhani, N.; Asadzadeh, Z.; Derakhshani, A.; Karim Ahangar, N.; Hemmat, N.; Lotfinejad, P.; Brunetti, O.; Silvestris, N.; Baradaran, B. A Systematic Review to Clarify the Prognostic Values of CD44 and CD44(+)CD24(-) Phenotype in Triple-Negative Breast Cancer Patients: Lessons Learned and the Road Ahead. Front. Oncol. 2021, 11, 689839. [Google Scholar] [CrossRef] [PubMed]
  4. Zeng, X.; Liu, C.; Yao, J.; Wan, H.; Wan, G.; Li, Y.; Chen, N. Breast cancer stem cells, heterogeneity, targeting therapies and therapeutic implications. Pharmacol. Res. 2021, 163, 105320. [Google Scholar] [CrossRef]
  5. Kemper, K.; Prasetyanti, P.R.; De Lau, W.; Rodermond, H.; Clevers, H.; Medema, J.P. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells 2012, 30, 2378–2386. [Google Scholar] [CrossRef]
  6. Wang, Z.; Tang, Y.; Xie, L.; Huang, A.; Xue, C.; Gu, Z.; Wang, K.; Zong, S. The Prognostic and Clinical Value of CD44 in Colorectal Cancer: A Meta-Analysis. Front. Oncol. 2019, 9, 309. [Google Scholar] [CrossRef]
  7. Huang, J.L.; Oshi, M.; Endo, I.; Takabe, K. Clinical relevance of stem cell surface markers CD133, CD24, and CD44 in colorectal cancer. Am. J. Cancer Res. 2021, 11, 5141–5154. [Google Scholar]
  8. Chong, C.; Müller, M.; Pak, H.; Harnett, D.; Huber, F.; Grun, D.; Leleu, M.; Auger, A.; Arnaud, M.; Stevenson, B.J.; et al. Integrated proteogenomic deep sequencing and analytics accurately identify non-canonical peptides in tumor immunopeptidomes. Nat. Commun. 2020, 11, 1293. [Google Scholar] [CrossRef] [Green Version]
  9. Rambow, F.; Marine, J.C.; Goding, C.R. Melanoma plasticity and phenotypic diversity: Therapeutic barriers and opportunities. Genes Dev. 2019, 33, 1295–1318. [Google Scholar] [CrossRef] [Green Version]
  10. Prieto-Vila, M.; Takahashi, R.U.; Usuba, W.; Kohama, I.; Ochiya, T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int. J. Mol. Sci. 2017, 18, 2574. [Google Scholar] [CrossRef] [Green Version]
  11. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chen, B.; Ye, P.; Chen, Y.; Liu, T.; Cha, J.H.; Yan, X.; Yang, W.H. Involvement of the Estrogen and Progesterone Axis in Cancer Stemness: Elucidating Molecular Mechanisms and Clinical Significance. Front. Oncol. 2020, 10, 1657. [Google Scholar] [CrossRef] [PubMed]
  13. Al Haq, A.T.; Tseng, H.Y.; Chen, L.M.; Wang, C.C.; Hsu, H.L. Targeting prooxidant MnSOD effect inhibits triple-negative breast cancer (TNBC) progression and M2 macrophage functions under the oncogenic stress. Cell Death Dis. 2022, 13, 49. [Google Scholar] [CrossRef] [PubMed]
  14. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  15. Song, W.; Mazzieri, R.; Yang, T.; Gobe, G.C. Translational Significance for Tumor Metastasis of Tumor-Associated Macrophages and Epithelial-Mesenchymal Transition. Front. Immunol. 2017, 8, 1106. [Google Scholar] [CrossRef] [Green Version]
  16. Zeng, J.; Liu, Z.; Sun, S.; Xie, J.; Cao, L.; Lv, P.; Nie, S.; Zhang, B.; Xie, B.; Peng, S.; et al. Tumor-associated macrophages recruited by periostin in intrahepatic cholangiocarcinoma stem cells. Oncol. Lett. 2018, 15, 8681–8686. [Google Scholar] [CrossRef]
  17. Zhang, H.; Lu, H.; Xiang, L.; Bullen, J.W.; Zhang, C.; Samanta, D.; Gilkes, D.M.; He, J.; Semenza, G.L. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, E6215–E6223. [Google Scholar] [CrossRef] [Green Version]
  18. Castelli, G.; Pelosi, E.; Testa, U. Liver Cancer: Molecular Characterization, Clonal Evolution and Cancer Stem Cells. Cancers 2017, 9, 127. [Google Scholar] [CrossRef] [Green Version]
  19. Cioffi, M.; Trabulo, S.; Hidalgo, M.; Costello, E.; Greenhalf, W.; Erkan, M.; Kleeff, J.; Sainz, B., Jr.; Heeschen, C. Inhibition of CD47 Effectively Targets Pancreatic Cancer Stem Cells via Dual Mechanisms. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 2325–2337. [Google Scholar] [CrossRef] [Green Version]
  20. Liu, L.; Zhang, L.; Yang, L.; Li, H.; Li, R.; Yu, J.; Yang, L.; Wei, F.; Yan, C.; Sun, Q.; et al. Anti-CD47 Antibody as a Targeted Therapeutic Agent for Human Lung Cancer and Cancer Stem Cells. Front. Immunol. 2017, 8, 404. [Google Scholar] [CrossRef] [Green Version]
  21. Lu, H.; Clauser, K.R.; Tam, W.L.; Fröse, J.; Ye, X.; Eaton, E.N.; Reinhardt, F.; Donnenberg, V.S.; Bhargava, R.; Carr, S.A.; et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat. Cell Biol. 2014, 16, 1105–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front. Immunol. 2019, 10, 1084. [Google Scholar] [CrossRef] [PubMed]
  23. Guerra, A.D.; Yeung, O.W.H.; Qi, X.; Kao, W.J.; Man, K. The Anti-Tumor Effects of M1 Macrophage-Loaded Poly (ethylene glycol) and Gelatin-Based Hydrogels on Hepatocellular Carcinoma. Theranostics 2017, 7, 3732–3744. [Google Scholar] [CrossRef] [PubMed]
  24. Nusblat, L.M.; Carroll, M.J.; Roth, C.M. Crosstalk between M2 macrophages and glioma stem cells. Cell. Oncol. 2017, 40, 471–482. [Google Scholar] [CrossRef] [PubMed]
  25. Shao, X.J.; Xiang, S.F.; Chen, Y.Q.; Zhang, N.; Cao, J.; Zhu, H.; Yang, B.; Zhou, Q.; Ying, M.D.; He, Q.J. Inhibition of M2-like macrophages by all-trans retinoic acid prevents cancer initiation and stemness in osteosarcoma cells. Acta Pharmacol. Sin. 2019, 40, 1343–1350. [Google Scholar] [CrossRef]
  26. Ito, A.; Kagawa, S.; Sakamoto, S.; Kuwada, K.; Kajioka, H.; Yoshimoto, M.; Kikuchi, S.; Kuroda, S.; Yoshida, R.; Tazawa, H.; et al. Extracellular vesicles shed from gastric cancer mediate protumor macrophage differentiation. BMC Cancer 2021, 21, 102. [Google Scholar] [CrossRef]
  27. Pedraza-Brindis, E.J.; Sánchez-Reyes, K.; Hernández-Flores, G.; Bravo-Cuellar, A.; Jave-Suárez, L.F.; Aguilar-Lemarroy, A.; Gómez-Lomelí, P.; López-López, B.A.; Ortiz-Lazareno, P.C. Culture supernatants of cervical cancer cells induce an M2 phenotypic profile in THP-1 macrophages. Cell. Immunol. 2016, 310, 42–52. [Google Scholar] [CrossRef]
  28. Sánchez-Reyes, K.; Pedraza-Brindis, E.J.; Hernández-Flores, G.; Bravo-Cuellar, A.; López-López, B.A.; Rosas-González, V.C.; Ortiz-Lazareno, P.C. The supernatant of cervical carcinoma cells lines induces a decrease in phosphorylation of STAT-1 and NF-κB transcription factors associated with changes in profiles of cytokines and growth factors in macrophages derived from U937 cells. Innate Immun. 2019, 25, 344–355. [Google Scholar] [CrossRef]
  29. Wang, D.; Wang, X.; Si, M.; Yang, J.; Sun, S.; Wu, H.; Cui, S.; Qu, X.; Yu, X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020, 474, 36–52. [Google Scholar] [CrossRef]
  30. Gok Yavuz, B.; Gunaydin, G.; Gedik, M.E.; Kosemehmetoglu, K.; Karakoc, D.; Ozgur, F.; Guc, D. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci. Rep. 2019, 9, 3172. [Google Scholar] [CrossRef]
  31. Cavalcante, R.S.; Ishikawa, U.; Silva, E.S.; Silva-Júnior, A.A.; Araújo, A.A.; Cruz, L.J.; Chan, A.B.; de Araújo Júnior, R.F. STAT3/NF-κB signalling disruption in M2 tumour-associated macrophages is a major target of PLGA nanocarriers/PD-L1 antibody immunomodulatory therapy in breast cancer. Br. J. Pharmacol. 2021, 178, 2284–2304. [Google Scholar] [CrossRef] [PubMed]
  32. Ouzounova, M.; Lee, E.; Piranlioglu, R.; El Andaloussi, A.; Kolhe, R.; Demirci, M.F.; Marasco, D.; Asm, I.; Chadli, A.; Hassan, K.A.; et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat. Commun. 2017, 8, 14979. [Google Scholar] [CrossRef] [PubMed]
  33. Clara, J.A.; Monge, C.; Yang, Y.; Takebe, N. Targeting signalling pathways and the immune microenvironment of cancer stem cells—A clinical update. Nat. Rev. Clin. Oncol. 2020, 17, 204–232. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, J.; Zheng, Y.; Zhao, M. Exosome-Based Cancer Therapy: Implication for Targeting Cancer Stem Cells. Front. Pharmacol. 2016, 7, 533. [Google Scholar] [CrossRef] [Green Version]
  35. Lee, H.H.; Wang, Y.N.; Yang, W.H.; Xia, W.; Wei, Y.; Chan, L.C.; Wang, Y.H.; Jiang, Z.; Xu, S.; Yao, J.; et al. Human ribonuclease 1 serves as a secretory ligand of ephrin A4 receptor and induces breast tumor initiation. Nat. Commun. 2021, 12, 2788. [Google Scholar] [CrossRef]
  36. Cheng, H.; Wang, Z.; Fu, L.; Xu, T. Macrophage Polarization in the Development and Progression of Ovarian Cancers: An Overview. Front. Oncol. 2019, 9, 421. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, P.; Hsu, W.H.; Han, J.; Xia, Y.; DePinho, R.A. Cancer Stemness Meets Immunity: From Mechanism to Therapy. Cell Rep. 2021, 34, 108597. [Google Scholar] [CrossRef]
  38. Wei, J.; Chen, P.; Gupta, P.; Ott, M.; Zamler, D.; Kassab, C.; Bhat, K.P.; Curran, M.A.; de Groot, J.F.; Heimberger, A.B. Immune biology of glioma-associated macrophages and microglia: Functional and therapeutic implications. Neuro-Oncol. 2020, 22, 180–194. [Google Scholar] [CrossRef]
  39. Kobatake, K.; Ikeda, K.I.; Nakata, Y.; Yamasaki, N.; Ueda, T.; Kanai, A.; Sentani, K.; Sera, Y.; Hayashi, T.; Koizumi, M.; et al. Kdm6a Deficiency Activates Inflammatory Pathways, Promotes M2 Macrophage Polarization, and Causes Bladder Cancer in Cooperation with p53 Dysfunction. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2065–2079. [Google Scholar] [CrossRef]
  40. Chen, P.; Hsu, W.H.; Chang, A.; Tan, Z.; Lan, Z.; Zhou, A.; Spring, D.J.; Lang, F.F.; Wang, Y.A.; DePinho, R.A. Circadian Regulator CLOCK Recruits Immune-Suppressive Microglia into the GBM Tumor Microenvironment. Cancer Discov. 2020, 10, 371–381. [Google Scholar] [CrossRef] [Green Version]
  41. Chia, K.; Mazzolini, J.; Mione, M.; Sieger, D. Tumor initiating cells induce Cxcr4-mediated infiltration of pro-tumoral macrophages into the brain. eLife 2018, 7, e31918. [Google Scholar] [CrossRef] [PubMed]
  42. Roversi, F.M.; Bueno, M.L.P.; Pericole, F.V.; Saad, S.T.O. Hematopoietic Cell Kinase (HCK) Is a Player of the Crosstalk between Hematopoietic Cells and Bone Marrow Niche through CXCL12/CXCR4 Axis. Front. Cell Dev. Biol. 2021, 9, 634044. [Google Scholar] [CrossRef] [PubMed]
  43. Guo, X.; Zhao, Y.; Yan, H.; Yang, Y.; Shen, S.; Dai, X.; Ji, X.; Ji, F.; Gong, X.G.; Li, L.; et al. Single tumor-initiating cells evade immune clearance by recruiting type II macrophages. Genes Dev. 2017, 31, 247–259. [Google Scholar] [CrossRef] [Green Version]
  44. Taniguchi, S.; Elhance, A.; Van Duzer, A.; Kumar, S.; Leitenberger, J.J.; Oshimori, N. Tumor-initiating cells establish an IL-33-TGF-β niche signaling loop to promote cancer progression. Science 2020, 369, eaay1813. [Google Scholar] [CrossRef]
  45. Pathria, P.; Louis, T.L.; Varner, J.A. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol. 2019, 40, 310–327. [Google Scholar] [CrossRef]
  46. Qian, B.Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef] [Green Version]
  47. Gangoso, E.; Southgate, B.; Bradley, L.; Rus, S.; Galvez-Cancino, F.; McGivern, N.; Güç, E.; Kapourani, C.A.; Byron, A.; Ferguson, K.M.; et al. Glioblastomas acquire myeloid-affiliated transcriptional programs via epigenetic immunoediting to elicit immune evasion. Cell 2021, 184, 2454–2470.e2426. [Google Scholar] [CrossRef]
  48. Zhang, F.; Li, P.; Liu, S.; Yang, M.; Zeng, S.; Deng, J.; Chen, D.; Yi, Y.; Liu, H. β-Catenin-CCL2 feedback loop mediates crosstalk between cancer cells and macrophages that regulates breast cancer stem cells. Oncogene 2021, 40, 5854–5865. [Google Scholar] [CrossRef]
  49. Cheah, M.T.; Chen, J.Y.; Sahoo, D.; Contreras-Trujillo, H.; Volkmer, A.K.; Scheeren, F.A.; Volkmer, J.P.; Weissman, I.L. CD14-expressing cancer cells establish the inflammatory and proliferative tumor microenvironment in bladder cancer. Proc. Natl. Acad. Sci. USA 2015, 112, 4725–4730. [Google Scholar] [CrossRef] [Green Version]
  50. Korbecki, J.; Grochans, S.; Gutowska, I.; Barczak, K.; Baranowska-Bosiacka, I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int. J. Mol. Sci. 2020, 21, 7619. [Google Scholar] [CrossRef]
  51. Baba, T.; Mukaida, N. Role of macrophage inflammatory protein (MIP)-1α/CCL3 in leukemogenesis. Mol. Cell. Oncol. 2014, 1, e29899. [Google Scholar] [CrossRef] [PubMed]
  52. Novak, M.; Koprivnikar Krajnc, M.; Hrastar, B.; Breznik, B.; Majc, B.; Mlinar, M.; Rotter, A.; Porčnik, A.; Mlakar, J.; Stare, K.; et al. CCR5-Mediated Signaling Is Involved in Invasion of Glioblastoma Cells in Its Microenvironment. Int. J. Mol. Sci. 2020, 21, 4199. [Google Scholar] [CrossRef] [PubMed]
  53. Kranjc, M.K.; Novak, M.; Pestell, R.G.; Lah, T.T. Cytokine CCL5 and receptor CCR5 axis in glioblastoma multiforme. Radiol. Oncol. 2019, 53, 397–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Guo, X.; Pan, Y.; Gutmann, D.H. Genetic and genomic alterations differentially dictate low-grade glioma growth through cancer stem cell-specific chemokine recruitment of T cells and microglia. Neuro-Oncol. 2019, 21, 1250–1262. [Google Scholar] [CrossRef]
  55. Zhuang, Y.; Zhao, X.; Yuan, B.; Zeng, Z.; Chen, Y. Blocking the CCL5-CCR5 Axis Using Maraviroc Promotes M1 Polarization of Macrophages Cocultured with Irradiated Hepatoma Cells. J. Hepatocell. Carcinoma 2021, 8, 599–611. [Google Scholar] [CrossRef]
  56. Li, Y.; Chen, X.; Li, D.; Yang, Z.; Bai, Y.; Hu, S.; Liu, Z.; Gu, J.; Zhang, X. Identification of prognostic and therapeutic value of CC chemokines in Urothelial bladder cancer: Evidence from comprehensive bioinformatic analysis. BMC Urol. 2021, 21, 173. [Google Scholar] [CrossRef] [PubMed]
  57. Ji, J.; Wang, P.; Zhou, Q.; Zhu, L.; Zhang, H.; Zhang, Y.; Zheng, Z.; Bhatta, A.K.; Zhang, G.; Wang, X. CCL8 enhances sensitivity of cutaneous squamous cell carcinoma to photodynamic therapy by recruiting M1 macrophages. Photodiagn. Photodyn. Ther. 2019, 26, 235–243. [Google Scholar] [CrossRef]
  58. Kortlever, R.M.; Sodir, N.M.; Wilson, C.H.; Burkhart, D.L.; Pellegrinet, L.; Brown Swigart, L.; Littlewood, T.D.; Evan, G.I. Myc Cooperates with Ras by Programming Inflammation and Immune Suppression. Cell 2017, 171, 1301–1315.e1314. [Google Scholar] [CrossRef]
  59. Ren, Z.; Chen, Y.; Shi, L.; Shao, F.; Sun, Y.; Ge, J.; Zhang, J.; Zang, Y. Sox9/CXCL5 axis facilitates tumour cell growth and invasion in hepatocellular carcinoma. FEBS J. 2022. [Google Scholar] [CrossRef]
  60. Jiang, X.; Cao, G.; Gao, G.; Wang, W.; Zhao, J.; Gao, C. Triptolide decreases tumor-associated macrophages infiltration and M2 polarization to remodel colon cancer immune microenvironment via inhibiting tumor-derived CXCL12. J. Cell. Physiol. 2021, 236, 193–204. [Google Scholar] [CrossRef]
  61. Takiguchi, S.; Korenaga, N.; Inoue, K.; Sugi, E.; Kataoka, Y.; Matsusue, K.; Futagami, K.; Li, Y.J.; Kukita, T.; Teramoto, N.; et al. Involvement of CXCL14 in osteolytic bone metastasis from lung cancer. Int. J. Oncol. 2014, 44, 1316–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Raghavan, S.; Mehta, P.; Xie, Y.; Lei, Y.L.; Mehta, G. Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J. Immunother. Cancer 2019, 7, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yamashina, T.; Baghdadi, M.; Yoneda, A.; Kinoshita, I.; Suzu, S.; Dosaka-Akita, H.; Jinushi, M. Cancer stem-like cells derived from chemoresistant tumors have a unique capacity to prime tumorigenic myeloid cells. Cancer Res. 2014, 74, 2698–2709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wu, A.; Wei, J.; Kong, L.Y.; Wang, Y.; Priebe, W.; Qiao, W.; Sawaya, R.; Heimberger, A.B. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro-Oncol. 2010, 12, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  65. Jelski, W.; Mroczko, B. Biochemical diagnostics of pancreatic cancer—Present and future. Clin. Chim. Acta Int. J. Clin. Chem. 2019, 498, 47–51. [Google Scholar] [CrossRef] [PubMed]
  66. Janakiram, N.B.; Mohammed, A.; Bryant, T.; Ritchie, R.; Stratton, N.; Jackson, L.; Lightfoot, S.; Benbrook, D.M.; Asch, A.S.; Lang, M.L.; et al. Loss of natural killer T cells promotes pancreatic cancer in LSL-Kras(G12D/+) mice. Immunology 2017, 152, 36–51. [Google Scholar] [CrossRef] [Green Version]
  67. Tan, X.; Zhang, Z.; Yao, H.; Shen, L. Tim-4 promotes the growth of colorectal cancer by activating angiogenesis and recruiting tumor-associated macrophages via the PI3K/AKT/mTOR signaling pathway. Cancer Lett. 2018, 436, 119–128. [Google Scholar] [CrossRef]
  68. Xu, H.; Zhang, Y.; Peña, M.M.; Pirisi, L.; Creek, K.E. Six1 promotes colorectal cancer growth and metastasis by stimulating angiogenesis and recruiting tumor-associated macrophages. Carcinogenesis 2017, 38, 281–292. [Google Scholar] [CrossRef] [Green Version]
  69. Zhou, W.; Ke, S.Q.; Huang, Z.; Flavahan, W.; Fang, X.; Paul, J.; Wu, L.; Sloan, A.E.; McLendon, R.E.; Li, X.; et al. Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat. Cell Biol. 2015, 17, 170–182. [Google Scholar] [CrossRef] [Green Version]
  70. Batool, A.; Wang, Y.Q.; Hao, X.X.; Chen, S.R.; Liu, Y.X. A miR-125b/CSF1-CX3CL1/tumor-associated macrophage recruitment axis controls testicular germ cell tumor growth. Cell Death Dis. 2018, 9, 962. [Google Scholar] [CrossRef]
  71. Sa, J.K.; Kim, S.H.; Lee, J.K.; Cho, H.J.; Shin, Y.J.; Shin, H.; Koo, H.; Kim, D.; Lee, M.; Kang, W.; et al. Identification of genomic and molecular traits that present therapeutic vulnerability to HGF-targeted therapy in glioblastoma. Neuro-Oncol. 2019, 21, 222–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Otvos, B.; Silver, D.J.; Mulkearns-Hubert, E.E.; Alvarado, A.G.; Turaga, S.M.; Sorensen, M.D.; Rayman, P.; Flavahan, W.A.; Hale, J.S.; Stoltz, K.; et al. Cancer Stem Cell-Secreted Macrophage Migration Inhibitory Factor Stimulates Myeloid Derived Suppressor Cell Function and Facilitates Glioblastoma Immune Evasion. Stem Cells 2016, 34, 2026–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yan, J.; Zhao, Q.; Wang, J.; Tian, X.; Wang, J.; Xia, X.; Ott, M.; Rao, G.; Heimberger, A.B.; Li, S. FGL2-wired macrophages secrete CXCL7 to regulate the stem-like functionality of glioma cells. Cancer Lett. 2021, 506, 83–94. [Google Scholar] [CrossRef] [PubMed]
  74. Bauer, D.; Mazzio, E.; Soliman, K.F.A. Whole Transcriptomic Analysis of Apigenin on TNFα Immuno-activated MDA-MB-231 Breast Cancer Cells. Cancer Genom. Proteom. 2019, 16, 421–431. [Google Scholar] [CrossRef] [Green Version]
  75. Fang, W.B.; Yao, M.; Brummer, G.; Acevedo, D.; Alhakamy, N.; Berkland, C.; Cheng, N. Targeted gene silencing of CCL2 inhibits triple negative breast cancer progression by blocking cancer stem cell renewal and M2 macrophage recruitment. Oncotarget 2016, 7, 49349–49367. [Google Scholar] [CrossRef]
  76. Zhang, J.; Patel, L.; Pienta, K.J. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev. 2010, 21, 41–48. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, J.; Wang, X.; Wang, Y.; Li, S.; Wang, X. Krüppel like factor 6 splice variant 1 (KLF6-SV1) overexpression recruits macrophages to participate in lung cancer metastasis by up-regulating TWIST1. Cancer Biol. Ther. 2019, 20, 680–691. [Google Scholar] [CrossRef]
  78. Aldinucci, D.; Lorenzon, D.; Cattaruzza, L.; Pinto, A.; Gloghini, A.; Carbone, A.; Colombatti, A. Expression of CCR5 receptors on Reed-Sternberg cells and Hodgkin lymphoma cell lines: Involvement of CCL5/Rantes in tumor cell growth and microenvironmental interactions. Int. J. Cancer 2008, 122, 769–776. [Google Scholar] [CrossRef]
  79. Kodama, T.; Koma, Y.I.; Arai, N.; Kido, A.; Urakawa, N.; Nishio, M.; Shigeoka, M.; Yokozaki, H. CCL3-CCR5 axis contributes to progression of esophageal squamous cell carcinoma by promoting cell migration and invasion via Akt and ERK pathways. Lab. Investig. J. Tech. Methods Pathol. 2020, 100, 1140–1157. [Google Scholar] [CrossRef]
  80. Gutmann, D.H.; Kettenmann, H. Microglia/Brain Macrophages as Central Drivers of Brain Tumor Pathobiology. Neuron 2019, 104, 442–449. [Google Scholar] [CrossRef]
  81. Shi, H.; Sun, Y.; Ruan, H.; Ji, C.; Zhang, J.; Wu, P.; Li, L.; Huang, C.; Jia, Y.; Zhang, X.; et al. 3,3’-Diindolylmethane Promotes Gastric Cancer Progression via β-TrCP-Mediated NF-κB Activation in Gastric Cancer-Derived MSCs. Front. Oncol. 2021, 11, 603533. [Google Scholar] [CrossRef] [PubMed]
  82. Özcan, Y.; Çağlar, F.; Celik, S.; Demir, A.B.; Erçetin, A.P.; Altun, Z.; Aktas, S. The role of cancer stem cells in immunotherapy for bladder cancer: An in vitro study. Urol. Oncol. 2020, 38, 476–487. [Google Scholar] [CrossRef] [PubMed]
  83. Pavon, L.F.; Sibov, T.T.; de Souza, A.V.; da Cruz, E.F.; Malheiros, S.M.F.; Cabral, F.R.; de Souza, J.G.; Boufleur, P.; de Oliveira, D.M.; de Toledo, S.R.C.; et al. Tropism of mesenchymal stem cell toward CD133(+) stem cell of glioblastoma in vitro and promote tumor proliferation in vivo. Stem Cell Res. Ther. 2018, 9, 310. [Google Scholar] [CrossRef] [PubMed]
  84. Wani, N.; Nasser, M.W.; Ahirwar, D.K.; Zhao, H.; Miao, Z.; Shilo, K.; Ganju, R.K. C-X-C motif chemokine 12/C-X-C chemokine receptor type 7 signaling regulates breast cancer growth and metastasis by modulating the tumor microenvironment. Breast Cancer Res. BCR 2014, 16, R54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. De Boeck, A.; Ahn, B.Y.; D’Mello, C.; Lun, X.; Menon, S.V.; Alshehri, M.M.; Szulzewsky, F.; Shen, Y.; Khan, L.; Dang, N.H.; et al. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat. Commun. 2020, 11, 4997. [Google Scholar] [CrossRef]
  86. Liu, M.; Sun, X.; Shi, S. MORC2 Enhances Tumor Growth by Promoting Angiogenesis and Tumor-Associated Macrophage Recruitment via Wnt/β-Catenin in Lung Cancer. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 51, 1679–1694. [Google Scholar] [CrossRef]
  87. Liu, Y.; Li, X.; Zhang, Y.; Wang, H.; Rong, X.; Peng, J.; He, L.; Peng, Y. An miR-340-5p-macrophage feedback loop modulates the progression and tumor microenvironment of glioblastoma multiforme. Oncogene 2019, 38, 7399–7415. [Google Scholar] [CrossRef]
  88. Hu, J.; Jo, M.; Eastman, B.M.; Gilder, A.S.; Bui, J.D.; Gonias, S.L. uPAR induces expression of transforming growth factor β and interleukin-4 in cancer cells to promote tumor-permissive conditioning of macrophages. Am. J. Pathol. 2014, 184, 3384–3393. [Google Scholar] [CrossRef] [Green Version]
  89. Dzaye, O.; Hu, F.; Derkow, K.; Haage, V.; Euskirchen, P.; Harms, C.; Lehnardt, S.; Synowitz, M.; Wolf, S.A.; Kettenmann, H. Glioma Stem Cells but Not Bulk Glioma Cells Upregulate IL-6 Secretion in Microglia/Brain Macrophages via Toll-like Receptor 4 Signaling. J. Neuropathol. Exp. Neurol. 2016, 75, 429–440. [Google Scholar] [CrossRef] [Green Version]
  90. Li, H.; Yang, P.; Wang, J.; Zhang, J.; Ma, Q.; Jiang, Y.; Wu, Y.; Han, T.; Xiang, D. HLF regulates ferroptosis, development and chemoresistance of triple-negative breast cancer by activating tumor cell-macrophage crosstalk. J. Hematol. Oncol. 2022, 15, 2. [Google Scholar] [CrossRef]
  91. Weng, Y.S.; Tseng, H.Y.; Chen, Y.A.; Shen, P.C.; Al Haq, A.T.; Chen, L.M.; Tung, Y.C.; Hsu, H.L. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol. Cancer 2019, 18, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Ning, Y.; Cui, Y.; Li, X.; Cao, X.; Chen, A.; Xu, C.; Cao, J.; Luo, X. Co-culture of ovarian cancer stem-like cells with macrophages induced SKOV3 cells stemness via IL-8/STAT3 signaling. Biomed. Pharmacother. 2018, 103, 262–271. [Google Scholar] [CrossRef] [PubMed]
  93. Deng, X.; Zhang, P.; Liang, T.; Deng, S.; Chen, X.; Zhu, L. Ovarian cancer stem cells induce the M2 polarization of macrophages through the PPARγ and NF-κB pathways. Int. J. Mol. Med. 2015, 36, 449–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Raggi, C.; Correnti, M.; Sica, A.; Andersen, J.B.; Cardinale, V.; Alvaro, D.; Chiorino, G.; Forti, E.; Glaser, S.; Alpini, G.; et al. Cholangiocarcinoma stem-like subset shapes tumor-initiating niche by educating associated macrophages. J. Hepatol. 2017, 66, 102–115. [Google Scholar] [CrossRef] [Green Version]
  95. Lu, C.S.; Shiau, A.L.; Su, B.H.; Hsu, T.S.; Wang, C.T.; Su, Y.C.; Tsai, M.S.; Feng, Y.H.; Tseng, Y.L.; Yen, Y.T.; et al. Oct4 promotes M2 macrophage polarization through upregulation of macrophage colony-stimulating factor in lung cancer. J. Hematol. Oncol. 2020, 13, 62. [Google Scholar] [CrossRef]
  96. Zhang, M.; Liu, Z.Z.; Aoshima, K.; Cai, W.L.; Sun, H.; Xu, T.; Zhang, Y.; An, Y.; Chen, J.F.; Chan, L.H.; et al. CECR2 drives breast cancer metastasis by promoting NF-κB signaling and macrophage-mediated immune suppression. Sci. Transl. Med. 2022, 14, eabf5473. [Google Scholar] [CrossRef]
  97. Sami, E.; Paul, B.T.; Koziol, J.A.; ElShamy, W.M. The Immunosuppressive Microenvironment in BRCA1-IRIS-Overexpressing TNBC Tumors Is Induced by Bidirectional Interaction with Tumor-Associated Macrophages. Cancer Res. 2020, 80, 1102–1117. [Google Scholar] [CrossRef] [Green Version]
  98. Liu, Q.; Wu, H.; Li, Y.; Zhang, R.; Kleeff, J.; Zhang, X.; Cui, M.; Liu, J.; Li, T.; Gao, J.; et al. Combined blockade of TGf-β1 and GM-CSF improves chemotherapeutic effects for pancreatic cancer by modulating tumor microenvironment. Cancer Immunol. Immunother. CII 2020, 69, 1477–1492. [Google Scholar] [CrossRef]
  99. Tao, W.; Chu, C.; Zhou, W.; Huang, Z.; Zhai, K.; Fang, X.; Huang, Q.; Zhang, A.; Wang, X.; Yu, X.; et al. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat. Commun. 2020, 11, 3015. [Google Scholar] [CrossRef]
  100. Tsukamoto, H.; Mishra, L.; Machida, K. Alcohol, TLR4-TGF-β antagonism, and liver cancer. Hepatol. Int. 2014, 8 (Suppl. S2), 408–412. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, Q.; Cai, D.J.; Li, B. Ovarian cancer stem-like cells elicit the polarization of M2 macrophages. Mol. Med. Rep. 2015, 11, 4685–4693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yin, J.; Kim, S.S.; Choi, E.; Oh, Y.T.; Lin, W.; Kim, T.H.; Sa, J.K.; Hong, J.H.; Park, S.H.; Kwon, H.J.; et al. ARS2/MAGL signaling in glioblastoma stem cells promotes self-renewal and M2-like polarization of tumor-associated macrophages. Nat. Commun. 2020, 11, 2978. [Google Scholar] [CrossRef] [PubMed]
  103. Heiler, S.; Wang, Z.; Zöller, M. Pancreatic cancer stem cell markers and exosomes—The incentive push. World J. Gastroenterol. 2016, 22, 5971–6007. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Correction: Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kγ to Promote Pancreatic Cancer Metastasis. Cancer Res. 2020, 80, 922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M.; et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef] [PubMed]
  106. Li, B.; Song, T.N.; Wang, F.R.; Yin, C.; Li, Z.; Lin, J.P.; Meng, Y.Q.; Feng, H.M.; Jing, T. Tumor-derived exosomal HMGB1 promotes esophageal squamous cell carcinoma progression through inducing PD1(+) TAM expansion. Oncogenesis 2019, 8, 17. [Google Scholar] [CrossRef] [Green Version]
  107. Yin, Z.; Ma, T.; Lin, Y.; Lu, X.; Zhang, C.; Chen, S.; Jian, Z. IL-6/STAT3 pathway intermediates M1/M2 macrophage polarization during the development of hepatocellular carcinoma. J. Cell. Biochem. 2018, 119, 9419–9432. [Google Scholar] [CrossRef]
  108. Myers, K.V.; Amend, S.R.; Pienta, K.J. Targeting Tyro3, Axl and MerTK (TAM receptors): Implications for macrophages in the tumor microenvironment. Mol. Cancer 2019, 18, 94. [Google Scholar] [CrossRef] [Green Version]
  109. Gomez, K.E.; Wu, F.; Keysar, S.B.; Morton, J.J.; Miller, B.; Chimed, T.S.; Le, P.N.; Nieto, C.; Chowdhury, F.N.; Tyagi, A.; et al. Cancer Cell CD44 Mediates Macrophage/Monocyte-Driven Regulation of Head and Neck Cancer Stem Cells. Cancer Res. 2020, 80, 4185–4198. [Google Scholar] [CrossRef]
  110. Huang, R.; Wang, S.; Wang, N.; Zheng, Y.; Zhou, J.; Yang, B.; Wang, X.; Zhang, J.; Guo, L.; Wang, S.; et al. CCL5 derived from tumor-associated macrophages promotes prostate cancer stem cells and metastasis via activating β-catenin/STAT3 signaling. Cell Death Dis. 2020, 11, 234. [Google Scholar] [CrossRef]
  111. Jeong, M.; Wang, Y.Y.; Choi, J.Y.; Lim, M.C.; Choi, J.H. CC Chemokine Ligand 7 Derived from Cancer-Stimulated Macrophages Promotes Ovarian Cancer Cell Invasion. Cancers 2021, 13, 2745. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, X.; Chen, L.; Dang, W.Q.; Cao, M.F.; Xiao, J.F.; Lv, S.Q.; Jiang, W.J.; Yao, X.H.; Lu, H.M.; Miao, J.Y.; et al. CCL8 secreted by tumor-associated macrophages promotes invasion and stemness of glioblastoma cells via ERK1/2 signaling. Lab. Investig. J. Tech. Methods Pathol. 2020, 100, 619–629. [Google Scholar] [CrossRef] [PubMed]
  113. He, Z.; Chen, D.; Wu, J.; Sui, C.; Deng, X.; Zhang, P.; Chen, Z.; Liu, D.; Yu, J.; Shi, J.; et al. Yes associated protein 1 promotes resistance to 5-fluorouracil in gastric cancer by regulating GLUT3-dependent glycometabolism reprogramming of tumor-associated macrophages. Arch. Biochem. Biophys. 2021, 702, 108838. [Google Scholar] [CrossRef] [PubMed]
  114. Zhu, F.; Li, X.; Chen, S.; Zeng, Q.; Zhao, Y.; Luo, F. Tumor-associated macrophage or chemokine ligand CCL17 positively regulates the tumorigenesis of hepatocellular carcinoma. Med. Oncol. 2016, 33, 17. [Google Scholar] [CrossRef]
  115. Yeung, O.W.; Lo, C.M.; Ling, C.C.; Qi, X.; Geng, W.; Li, C.X.; Ng, K.T.; Forbes, S.J.; Guan, X.Y.; Poon, R.T.; et al. Alternatively activated (M2) macrophages promote tumour growth and invasiveness in hepatocellular carcinoma. J. Hepatol. 2015, 62, 607–616. [Google Scholar] [CrossRef]
  116. Wang, S.; Wang, N.; Huang, X.; Yang, B.; Zheng, Y.; Zhang, J.; Wang, X.; Lin, Y.; Wang, Z. Baohuoside i suppresses breast cancer metastasis by downregulating the tumor-associated macrophages/C-X-C motif chemokine ligand 1 pathway. Phytomed. Int. J. Phytother. Phytopharmacol. 2020, 78, 153331. [Google Scholar] [CrossRef]
  117. Nie, G.; Cao, X.; Mao, Y.; Lv, Z.; Lv, M.; Wang, Y.; Wang, H.; Liu, C. Tumor-associated macrophages-mediated CXCL8 infiltration enhances breast cancer metastasis: Suppression by Danirixin. Int. Immunopharmacol. 2021, 95, 107153. [Google Scholar] [CrossRef]
  118. Feng, X.; Szulzewsky, F.; Yerevanian, A.; Chen, Z.; Heinzmann, D.; Rasmussen, R.D.; Alvarez-Garcia, V.; Kim, Y.; Wang, B.; Tamagno, I.; et al. Loss of CX3CR1 increases accumulation of inflammatory monocytes and promotes gliomagenesis. Oncotarget 2015, 6, 15077–15094. [Google Scholar] [CrossRef] [Green Version]
  119. Wan, S.; Zhao, E.; Kryczek, I.; Vatan, L.; Sadovskaya, A.; Ludema, G.; Simeone, D.M.; Zou, W.; Welling, T.H. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology 2014, 147, 1393–1404. [Google Scholar] [CrossRef] [Green Version]
  120. Zhou, N.; Zhang, Y.; Zhang, X.; Lei, Z.; Hu, R.; Li, H.; Mao, Y.; Wang, X.; Irwin, D.M.; Niu, G.; et al. Exposure of tumor-associated macrophages to apoptotic MCF-7 cells promotes breast cancer growth and metastasis. Int. J. Mol. Sci. 2015, 16, 11966–11982. [Google Scholar] [CrossRef] [Green Version]
  121. Yang, L.; Dong, Y.; Li, Y.; Wang, D.; Liu, S.; Wang, D.; Gao, Q.; Ji, S.; Chen, X.; Lei, Q.; et al. IL-10 derived from M2 macrophage promotes cancer stemness via JAK1/STAT1/NF-κB/Notch1 pathway in non-small cell lung cancer. Int. J. Cancer 2019, 145, 1099–1110. [Google Scholar] [CrossRef] [PubMed]
  122. Shen, Z.; Seppänen, H.; Vainionpää, S.; Ye, Y.; Wang, S.; Mustonen, H.; Puolakkainen, P. IL10, IL11, IL18 are differently expressed in CD14+ TAMs and play different role in regulating the invasion of gastric cancer cells under hypoxia. Cytokine 2012, 59, 352–357. [Google Scholar] [CrossRef] [PubMed]
  123. Ruiz-Torres, S.J.; Bourn, J.R.; Benight, N.M.; Hunt, B.G.; Lester, C.; Waltz, S.E. Macrophage-mediated RON signaling supports breast cancer growth and progression through modulation of IL-35. Oncogene 2022, 41, 321–333. [Google Scholar] [CrossRef]
  124. Fan, Q.M.; Jing, Y.Y.; Yu, G.F.; Kou, X.R.; Ye, F.; Gao, L.; Li, R.; Zhao, Q.D.; Yang, Y.; Lu, Z.H.; et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014, 352, 160–168. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, B.; Ye, H.; Ren, X.; Zheng, S.; Zhou, Q.; Chen, C.; Lin, Q.; Li, G.; Wei, L.; Fu, Z.; et al. Macrophage-expressed CD51 promotes cancer stem cell properties via the TGF-β1/smad2/3 axis in pancreatic cancer. Cancer Lett. 2019, 459, 204–215. [Google Scholar] [CrossRef]
  126. Zhang, X.; Zhu, M.; Hong, Z.; Chen, C. Co-culturing polarized M2 Thp-1-derived macrophages enhance stemness of lung adenocarcinoma A549 cells. Ann. Transl. Med. 2021, 9, 709. [Google Scholar] [CrossRef]
  127. Liu, Q.; Hodge, J.; Wang, J.; Wang, Y.; Wang, L.; Singh, U.; Li, Y.; Yao, Y.; Wang, D.; Ai, W.; et al. Emodin reduces Breast Cancer Lung Metastasis by suppressing Macrophage-induced Breast Cancer Cell Epithelial-mesenchymal transition and Cancer Stem Cell formation. Theranostics 2020, 10, 8365–8381. [Google Scholar] [CrossRef]
  128. Ma, C.; Komohara, Y.; Ohnishi, K.; Shimoji, T.; Kuwahara, N.; Sakumura, Y.; Matsuishi, K.; Fujiwara, Y.; Motoshima, T.; Takahashi, W.; et al. Infiltration of tumor-associated macrophages is involved in CD44 expression in clear cell renal cell carcinoma. Cancer Sci. 2016, 107, 700–707. [Google Scholar] [CrossRef] [Green Version]
  129. Gulluoglu, S.; Tuysuz, E.C.; Sahin, M.; Yaltirik, C.K.; Kuskucu, A.; Ozkan, F.; Dalan, A.B.; Sahin, F.; Ture, U.; Bayrak, O.F. The role of TNF-α in chordoma progression and inflammatory pathways. Cell. Oncol. 2019, 42, 663–677. [Google Scholar] [CrossRef]
  130. Yang, J.; Liao, D.; Chen, C.; Liu, Y.; Chuang, T.H.; Xiang, R.; Markowitz, D.; Reisfeld, R.A.; Luo, Y. Tumor-associated macrophages regulate murine breast cancer stem cells through a novel paracrine EGFR/Stat3/Sox-2 signaling pathway. Stem Cells 2013, 31, 248–258. [Google Scholar] [CrossRef]
  131. Liguori, M.; Digifico, E.; Vacchini, A.; Avigni, R.; Colombo, F.S.; Borroni, E.M.; Farina, F.M.; Milanesi, S.; Castagna, A.; Mannarino, L.; et al. The soluble glycoprotein NMB (GPNMB) produced by macrophages induces cancer stemness and metastasis via CD44 and IL-33. Cell. Mol. Immunol. 2021, 18, 711–722. [Google Scholar] [CrossRef] [PubMed]
  132. Wei, X.; Yang, S.; Pu, X.; He, S.; Yang, Z.; Sheng, X.; Meng, X.; Chen, X.; Jin, L.; Chen, W.; et al. Tumor-associated macrophages increase the proportion of cancer stem cells in lymphoma by secreting pleiotrophin. Am. J. Transl. Res. 2019, 11, 6393–6402. [Google Scholar] [PubMed]
  133. Shi, Y.; Ping, Y.F.; Zhou, W.; He, Z.C.; Chen, C.; Bian, B.S.; Zhang, L.; Chen, L.; Lan, X.; Zhang, X.C.; et al. Tumour-associated macrophages secrete pleiotrophin to promote PTPRZ1 signalling in glioblastoma stem cells for tumour growth. Nat. Commun. 2017, 8, 15080. [Google Scholar] [CrossRef] [PubMed]
  134. Lv, J.; Liu, C.; Chen, F.K.; Feng, Z.P.; Jia, L.; Liu, P.J.; Yang, Z.X.; Hou, F.; Deng, Z.Y. M2-like tumour-associated macrophage-secreted IGF promotes thyroid cancer stemness and metastasis by activating the PI3K/AKT/mTOR pathway. Mol. Med. Rep. 2021, 24, 1–10. [Google Scholar] [CrossRef]
  135. Okuda, H.; Kobayashi, A.; Xia, B.; Watabe, M.; Pai, S.K.; Hirota, S.; Xing, F.; Liu, W.; Pandey, P.R.; Fukuda, K.; et al. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells. Cancer Res. 2012, 72, 537–547. [Google Scholar] [CrossRef] [Green Version]
  136. Sainz, B., Jr.; Alcala, S.; Garcia, E.; Sanchez-Ripoll, Y.; Azevedo, M.M.; Cioffi, M.; Tatari, M.; Miranda-Lorenzo, I.; Hidalgo, M.; Gomez-Lopez, G.; et al. Microenvironmental hCAP-18/LL-37 promotes pancreatic ductal adenocarcinoma by activating its cancer stem cell compartment. Gut 2015, 64, 1921–1935. [Google Scholar] [CrossRef] [Green Version]
  137. Wei, R.; Zhu, W.W.; Yu, G.Y.; Wang, X.; Gao, C.; Zhou, X.; Lin, Z.F.; Shao, W.Q.; Wang, S.H.; Lu, M.; et al. S100 calcium-binding protein A9 from tumor-associated macrophage enhances cancer stem cell-like properties of hepatocellular carcinoma. Int. J. Cancer 2021, 148, 1233–1244. [Google Scholar] [CrossRef]
  138. Xu, H.Z.; Li, T.F.; Wang, C.; Ma, Y.; Liu, Y.; Zheng, M.Y.; Liu, Z.J.; Chen, J.B.; Li, K.; Sun, S.K.; et al. Synergy of nanodiamond-doxorubicin conjugates and PD-L1 blockade effectively turns tumor-associated macrophages against tumor cells. J. Nanobiotechnol. 2021, 19, 268. [Google Scholar] [CrossRef]
  139. Jinushi, M.; Chiba, S.; Yoshiyama, H.; Masutomi, K.; Kinoshita, I.; Dosaka-Akita, H.; Yagita, H.; Takaoka, A.; Tahara, H. Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc. Natl. Acad. Sci. USA 2011, 108, 12425–12430. [Google Scholar] [CrossRef] [Green Version]
  140. Chang, J.; Li, H.; Zhu, Z.; Mei, P.; Hu, W.; Xiong, X.; Tao, J. microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol. Toxicol. 2021. [Google Scholar] [CrossRef]
  141. You, Y.; Tian, Z.; Du, Z.; Wu, K.; Xu, G.; Dai, M.; Wang, Y.; Xiao, M. M1-like tumor-associated macrophages cascade a mesenchymal/stem-like phenotype of oral squamous cell carcinoma via the IL6/Stat3/THBS1 feedback loop. J. Exp. Clin. Cancer Res. CR 2022, 41, 10. [Google Scholar] [CrossRef] [PubMed]
  142. Tabu, K.; Liu, W.; Kosaku, A.; Terashima, K.; Murota, Y.; Aimaitijiang, A.; Nobuhisa, I.; Hide, T.; Taga, T. Glioma stem cell (GSC)-derived autoschizis-like products confer GSC niche properties involving M1-like tumor-associated macrophages. Stem Cells 2020, 38, 921–935. [Google Scholar] [CrossRef] [PubMed]
  143. Mine, N.; Yamamoto, S.; Saito, N.; Sato, T.; Sakakibara, K.; Kufe, D.W.; VonHoff, D.D.; Kawabe, T. CBP501 suppresses macrophage induced cancer stem cell like features and metastases. Oncotarget 2017, 8, 64015–64031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Pradhan, R.; Chatterjee, S.; Hembram, K.C.; Sethy, C.; Mandal, M.; Kundu, C.N. Nano formulated Resveratrol inhibits metastasis and angiogenesis by reducing inflammatory cytokines in oral cancer cells by targeting tumor associated macrophages. J. Nutr. Biochem. 2021, 92, 108624. [Google Scholar] [CrossRef] [PubMed]
  145. Sawa-Wejksza, K.; Kandefer-Szerszeń, M. Tumor-Associated Macrophages as Target for Antitumor Therapy. Arch. Immunol. Ther. Exp. 2018, 66, 97–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ning, Y.; Feng, W.; Cao, X.; Ren, K.; Quan, M.; Chen, A.; Xu, C.; Qiu, Y.; Cao, J.; Li, X.; et al. Genistein inhibits stemness of SKOV3 cells induced by macrophages co-cultured with ovarian cancer stem-like cells through IL-8/STAT3 axis. J. Exp. Clin. Cancer Res. CR 2019, 38, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Hide, T.; Komohara, Y.; Miyasato, Y.; Nakamura, H.; Makino, K.; Takeya, M.; Kuratsu, J.I.; Mukasa, A.; Yano, S. Oligodendrocyte Progenitor Cells and Macrophages/Microglia Produce Glioma Stem Cell Niches at the Tumor Border. EBioMedicine 2018, 30, 94–104. [Google Scholar] [CrossRef] [Green Version]
  148. Fang, M.; Li, Y.; Huang, K.; Qi, S.; Zhang, J.; Zgodzinski, W.; Majewski, M.; Wallner, G.; Gozdz, S.; Macek, P.; et al. IL33 Promotes Colon Cancer Cell Stemness via JNK Activation and Macrophage Recruitment. Cancer Res. 2017, 77, 2735–2745. [Google Scholar] [CrossRef] [Green Version]
  149. Hu, L.; Liu, Y.; Kong, X.; Wu, R.; Peng, Q.; Zhang, Y.; Zhou, L.; Duan, L. Fusobacterium nucleatum Facilitates M2 Macrophage Polarization and Colorectal Carcinoma Progression by Activating TLR4/NF-κB/S100A9 Cascade. Front. Immunol. 2021, 12, 658681. [Google Scholar] [CrossRef]
  150. Wang, Y.; Gong, X.; Li, J.; Wang, H.; Xu, X.; Wu, Y.; Wang, J.; Wang, S.; Li, Y.; Zhang, Z. M2 macrophage microvesicle-inspired nanovehicles improve accessibility to cancer cells and cancer stem cells in tumors. J. Nanobiotechnol. 2021, 19, 397. [Google Scholar] [CrossRef]
  151. Kaneda, M.M.; Cappello, P.; Nguyen, A.V.; Ralainirina, N.; Hardamon, C.R.; Foubert, P.; Schmid, M.C.; Sun, P.; Mose, E.; Bouvet, M.; et al. Macrophage PI3Kγ Drives Pancreatic Ductal Adenocarcinoma Progression. Cancer Discov. 2016, 6, 870–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Schmid, M.C.; Khan, S.Q.; Kaneda, M.M.; Pathria, P.; Shepard, R.; Louis, T.L.; Anand, S.; Woo, G.; Leem, C.; Faridi, M.H.; et al. Integrin CD11b activation drives anti-tumor innate immunity. Nat. Commun. 2018, 9, 5379. [Google Scholar] [CrossRef] [PubMed]
  153. Atanasov, G.; Pötner, C.; Aust, G.; Schierle, K.; Dietel, C.; Benzing, C.; Krenzien, F.; Bartels, M.; Eichfeld, U.; Schmelzle, M.; et al. TIE2-expressing monocytes and M2-polarized macrophages impact survival and correlate with angiogenesis in adenocarcinoma of the pancreas. Oncotarget 2018, 9, 29715–29726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Zhang, W.J.; Wang, X.H.; Gao, S.T.; Chen, C.; Xu, X.Y.; Sun, Q.; Zhou, Z.H.; Wu, G.Z.; Yu, Q.; Xu, G.; et al. Tumor-associated macrophages correlate with phenomenon of epithelial-mesenchymal transition and contribute to poor prognosis in triple-negative breast cancer patients. J. Surg. Res. 2018, 222, 93–101. [Google Scholar] [CrossRef]
  155. Sørensen, M.D.; Dahlrot, R.H.; Boldt, H.B.; Hansen, S.; Kristensen, B.W. Tumour-associated microglia/macrophages predict poor prognosis in high-grade gliomas and correlate with an aggressive tumour subtype. Neuropathol. Appl. Neurobiol. 2018, 44, 185–206. [Google Scholar] [CrossRef] [Green Version]
  156. Wang, H.; Li, P.; Wang, L.; Xia, Z.; Huang, H.; Lu, Y.; Li, Z. High numbers of CD68+ tumor-associated macrophages correlate with poor prognosis in extranodal NK/T-cell lymphoma, nasal type. Ann. Hematol. 2015, 94, 1535–1544. [Google Scholar] [CrossRef]
  157. Zhang, Y.; Cheng, S.; Zhang, M.; Zhen, L.; Pang, D.; Zhang, Q.; Li, Z. High-infiltration of tumor-associated macrophages predicts unfavorable clinical outcome for node-negative breast cancer. PLoS ONE 2013, 8, e76147. [Google Scholar] [CrossRef]
  158. Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.J.; Gorjestani, S.; Woo, G.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P.; et al. PI3Kγ is a molecular switch that controls immune suppression. Nature 2016, 539, 437–442. [Google Scholar] [CrossRef] [Green Version]
  159. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
  160. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  161. Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.M.; Ries, C.H.; Rüttinger, D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef] [PubMed]
  162. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Wang, Q.; Lu, Y.; Li, R.; Jiang, Y.; Zheng, Y.; Qian, J.; Bi, E.; Zheng, C.; Hou, J.; Wang, S.; et al. Therapeutic effects of CSF1R-blocking antibodies in multiple myeloma. Leukemia 2018, 32, 176–183. [Google Scholar] [CrossRef] [PubMed]
  165. Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ries, C.H.; Cannarile, M.A.; Hoves, S.; Benz, J.; Wartha, K.; Runza, V.; Rey-Giraud, F.; Pradel, L.P.; Feuerhake, F.; Klaman, I.; et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014, 25, 846–859. [Google Scholar] [CrossRef] [Green Version]
  167. Tap, W.D.; Gelderblom, H.; Palmerini, E.; Desai, J.; Bauer, S.; Blay, J.Y.; Alcindor, T.; Ganjoo, K.; Martín-Broto, J.; Ryan, C.W.; et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): A randomised phase 3 trial. Lancet 2019, 394, 478–487. [Google Scholar] [CrossRef]
  168. Yan, D.; Kowal, J.; Akkari, L.; Schuhmacher, A.J.; Huse, J.T.; West, B.L.; Joyce, J.A. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene 2017, 36, 6049–6058. [Google Scholar] [CrossRef]
  169. Coniglio, S.J.; Eugenin, E.; Dobrenis, K.; Stanley, E.R.; West, B.L.; Symons, M.H.; Segall, J.E. Microglial stimulation of glioblastoma invasion involves epidermal growth factor receptor (EGFR) and colony stimulating factor 1 receptor (CSF-1R) signaling. Mol. Med. 2012, 18, 519–527. [Google Scholar] [CrossRef]
  170. Sakamoto, C.; Kohara, H.; Inoue, H.; Narusawa, M.; Ogawa, Y.; Hirose-Yotsuya, L.; Miyamoto, S.; Matsumura, Y.; Yamada, K.; Takahashi, A.; et al. Therapeutic vaccination based on side population cells transduced by the granulocyte-macrophage colony-stimulating factor gene elicits potent antitumor immunity. Cancer Gene Ther. 2017, 24, 165–174. [Google Scholar] [CrossRef]
  171. Wang, C.Y.; Hua, R.; Liu, L.; Zhan, X.; Chen, S.; Quan, S.; Chu, Q.J.; Zhu, Y.T. Immunotherapy against metastatic bladder cancer by combined administration of granulocyte macrophage-colony stimulating factor and interleukin-2 surface modified MB49 bladder cancer stem cells vaccine. Cancer Med. 2017, 6, 689–697. [Google Scholar] [CrossRef] [PubMed]
  172. Lim, S.Y.; Yuzhalin, A.E.; Gordon-Weeks, A.N.; Muschel, R.J. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget 2016, 7, 28697–28710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Sainz, B., Jr.; Carron, E.; Vallespinós, M.; Machado, H.L. Cancer Stem Cells and Macrophages: Implications in Tumor Biology and Therapeutic Strategies. Med. Inflamm. 2016, 2016, 9012369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kalbasi, A.; Komar, C.; Tooker, G.M.; Liu, M.; Lee, J.W.; Gladney, W.L.; Ben-Josef, E.; Beatty, G.L. Tumor-Derived CCL2 Mediates Resistance to Radiotherapy in Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 137–148. [Google Scholar] [CrossRef] [Green Version]
  175. Loberg, R.D.; Ying, C.; Craig, M.; Yan, L.; Snyder, L.A.; Pienta, K.J. CCL2 as an important mediator of prostate cancer growth in vivo through the regulation of macrophage infiltration. Neoplasia 2007, 9, 556–562. [Google Scholar] [CrossRef] [Green Version]
  176. Loberg, R.D.; Ying, C.; Craig, M.; Day, L.L.; Sargent, E.; Neeley, C.; Wojno, K.; Snyder, L.A.; Yan, L.; Pienta, K.J. Targeting CCL2 with systemic delivery of neutralizing antibodies induces prostate cancer tumor regression in vivo. Cancer Res. 2007, 67, 9417–9424. [Google Scholar] [CrossRef] [Green Version]
  177. Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Mayer, A.; Deshpande, A.D.; Carpenter, D.; Mitchem, J.B.; Plambeck-Suess, S.M.; Worley, L.A.; Goetz, B.D.; et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: A role for targeting the CCL2/CCR2 axis. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 3404–3415. [Google Scholar] [CrossRef] [Green Version]
  178. Wang, D.; Yue, D.L.; Wang, D.; Chen, X.F.; Yin, X.Y.; Wang, Y.P.; Yang, L.; Zhang, Y. [Aspirin inhibits cell stemness of esophageal cancer by downregulation of chemokine CCL2]. Zhonghua Zhong Liu Za Zhi [Chin. J. Oncol.] 2018, 40, 744–749. [Google Scholar] [CrossRef]
  179. Izumi, K.; Fang, L.Y.; Mizokami, A.; Namiki, M.; Li, L.; Lin, W.J.; Chang, C. Targeting the androgen receptor with siRNA promotes prostate cancer metastasis through enhanced macrophage recruitment via CCL2/CCR2-induced STAT3 activation. EMBO Mol. Med. 2013, 5, 1383–1401. [Google Scholar] [CrossRef]
  180. Udartseva, O.O.; Zhidkova, O.V.; Ezdakova, M.I.; Ogneva, I.V.; Andreeva, E.R.; Buravkova, L.B.; Gollnick, S.O. Low-dose photodynamic therapy promotes angiogenic potential and increases immunogenicity of human mesenchymal stromal cells. J. Photochem. Photobiol. B Biol. 2019, 199, 111596. [Google Scholar] [CrossRef]
  181. Eckert, F.; Schilbach, K.; Klumpp, L.; Bardoscia, L.; Sezgin, E.C.; Schwab, M.; Zips, D.; Huber, S.M. Potential Role of CXCR4 Targeting in the Context of Radiotherapy and Immunotherapy of Cancer. Front. Immunol. 2018, 9, 3018. [Google Scholar] [CrossRef] [PubMed]
  182. Burger, J.A.; Peled, A. CXCR4 antagonists: Targeting the microenvironment in leukemia and other cancers. Leukemia 2009, 23, 43–52. [Google Scholar] [CrossRef] [PubMed]
  183. Grande, F.; Giancotti, G.; Ioele, G.; Occhiuzzi, M.A.; Garofalo, A. An update on small molecules targeting CXCR4 as starting points for the development of anti-cancer therapeutics. Eur. J. Med. Chem. 2017, 139, 519–530. [Google Scholar] [CrossRef] [PubMed]
  184. Taromi, S.; Kayser, G.; Catusse, J.; von Elverfeldt, D.; Reichardt, W.; Braun, F.; Weber, W.A.; Zeiser, R.; Burger, M. CXCR4 antagonists suppress small cell lung cancer progression. Oncotarget 2016, 7, 85185–85195. [Google Scholar] [CrossRef] [Green Version]
  185. Bertolini, G.; Cancila, V.; Milione, M.; Lo Russo, G.; Fortunato, O.; Zaffaroni, N.; Tortoreto, M.; Centonze, G.; Chiodoni, C.; Facchinetti, F.; et al. A novel CXCR4 antagonist counteracts paradoxical generation of cisplatin-induced pro-metastatic niches in lung cancer. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 2963–2978. [Google Scholar] [CrossRef]
  186. Hao, Q.; Vadgama, J.V.; Wang, P. CCL2/CCR2 signaling in cancer pathogenesis. Cell Commun. Signal. 2020, 18, 82. [Google Scholar] [CrossRef]
  187. Laparidou, M.; Schlickenrieder, A.; Thoma, T.; Lengyel, K.; Schusser, B. Blocking of the CXCR4-CXCL12 Interaction Inhibits the Migration of Chicken B Cells into the Bursa of Fabricius. Front. Immunol. 2019, 10, 3057. [Google Scholar] [CrossRef]
  188. Poh, A.R.; Ernst, M. Targeting Macrophages in Cancer: From Bench to Bedside. Front. Oncol. 2018, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  189. Linger, R.M.; Cohen, R.A.; Cummings, C.T.; Sather, S.; Migdall-Wilson, J.; Middleton, D.H.; Lu, X.; Barón, A.E.; Franklin, W.A.; Merrick, D.T.; et al. Mer or Axl receptor tyrosine kinase inhibition promotes apoptosis, blocks growth and enhances chemosensitivity of human non-small cell lung cancer. Oncogene 2013, 32, 3420–3431. [Google Scholar] [CrossRef] [Green Version]
  190. Zhang, G.; Wang, M.; Zhao, H.; Cui, W. Function of Axl receptor tyrosine kinase in non-small cell lung cancer. Oncol. Lett. 2018, 15, 2726–2734. [Google Scholar] [CrossRef] [Green Version]
  191. Xie, S.; Li, Y.; Li, X.; Wang, L.; Yang, N.; Wang, Y.; Wei, H. Mer receptor tyrosine kinase is frequently overexpressed in human non-small cell lung cancer, confirming resistance to erlotinib. Oncotarget 2015, 6, 9206–9219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Schmidt, T.; Ben-Batalla, I.; Schultze, A.; Loges, S. Macrophage-tumor crosstalk: Role of TAMR tyrosine kinase receptors and of their ligands. Cell. Mol. Life Sci. 2012, 69, 1391–1414. [Google Scholar] [CrossRef] [PubMed]
  193. Cummings, C.T.; Zhang, W.; Davies, K.D.; Kirkpatrick, G.D.; Zhang, D.; DeRyckere, D.; Wang, X.; Frye, S.V.; Earp, H.S.; Graham, D.K. Small Molecule Inhibition of MERTK Is Efficacious in Non-Small Cell Lung Cancer Models Independent of Driver Oncogene Status. Mol. Cancer Ther. 2015, 14, 2014–2022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Georgoudaki, A.M.; Prokopec, K.E.; Boura, V.F.; Hellqvist, E.; Sohn, S.; Östling, J.; Dahan, R.; Harris, R.A.; Rantalainen, M.; Klevebring, D.; et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016, 15, 2000–2011. [Google Scholar] [CrossRef] [Green Version]
  195. La Fleur, L.; Boura, V.F.; Alexeyenko, A.; Berglund, A.; Pontén, V.; Mattsson, J.S.M.; Djureinovic, D.; Persson, J.; Brunnström, H.; Isaksson, J.; et al. Expression of scavenger receptor MARCO defines a targetable tumor-associated macrophage subset in non-small cell lung cancer. Int. J. Cancer 2018, 143, 1741–1752. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, H.W.; Joyce, J.A. Alternative activation of tumor-associated macrophages by IL-4: Priming for protumoral functions. Cell Cycle 2010, 9, 4824–4835. [Google Scholar] [CrossRef] [Green Version]
  197. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  198. McWhirter, S.M.; Jefferies, C.A. Nucleic Acid Sensors as Therapeutic Targets for Human Disease. Immunity 2020, 53, 78–97. [Google Scholar] [CrossRef]
  199. Adams, S.; Kozhaya, L.; Martiniuk, F.; Meng, T.C.; Chiriboga, L.; Liebes, L.; Hochman, T.; Shuman, N.; Axelrod, D.; Speyer, J.; et al. Topical TLR7 agonist imiquimod can induce immune-mediated rejection of skin metastases in patients with breast cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 6748–6757. [Google Scholar] [CrossRef] [Green Version]
  200. Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef]
  201. Vanpouille-Box, C.; Hoffmann, J.A.; Galluzzi, L. Pharmacological modulation of nucleic acid sensors—Therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 2019, 18, 845–867. [Google Scholar] [CrossRef] [PubMed]
  202. Tallerico, R.; Garofalo, C.; Carbone, E. A New Biological Feature of Natural Killer Cells: The Recognition of Solid Tumor-Derived Cancer Stem Cells. Front. Immunol. 2016, 7, 179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Donini, C.; Rotolo, R.; Proment, A.; Aglietta, M.; Sangiolo, D.; Leuci, V. Cellular Immunotherapy Targeting Cancer Stem Cells: Preclinical Evidence and Clinical Perspective. Cells 2021, 10, 543. [Google Scholar] [CrossRef] [PubMed]
  204. Guerriero, J.L.; Sotayo, A.; Ponichtera, H.E.; Castrillon, J.A.; Pourzia, A.L.; Schad, S.; Johnson, S.F.; Carrasco, R.D.; Lazo, S.; Bronson, R.T.; et al. Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature 2017, 543, 428–432. [Google Scholar] [CrossRef]
  205. Neele, A.E.; de Winther, M.P.J. Repressing the repressor: Ezh2 mediates macrophage activation. J. Exp. Med. 2018, 215, 1269–1271. [Google Scholar] [CrossRef]
  206. Qi, B.; Yang, C.; Zhu, Z.; Chen, H. EZH2-Inhibited MicroRNA-454-3p Promotes M2 Macrophage Polarization in Glioma. Front. Cell Dev. Biol. 2020, 8, 574940. [Google Scholar] [CrossRef]
  207. Hamaidia, M.; Gazon, H.; Hoyos, C.; Hoffmann, G.B.; Louis, R.; Duysinx, B.; Willems, L. Inhibition of EZH2 methyltransferase decreases immunoediting of mesothelioma cells by autologous macrophages through a PD-1-dependent mechanism. JCI Insight 2019, 4, e128474. [Google Scholar] [CrossRef]
  208. Christofides, A.; Karantanos, T.; Bardhan, K.; Boussiotis, V.A. Epigenetic regulation of cancer biology and anti-tumor immunity by EZH2. Oncotarget 2016, 7, 85624–85640. [Google Scholar] [CrossRef] [Green Version]
  209. Yamagishi, M.; Uchimaru, K. Targeting EZH2 in cancer therapy. Curr. Opin. Oncol. 2017, 29, 375–381. [Google Scholar] [CrossRef]
  210. Eich, M.L.; Athar, M.; Ferguson, J.E., 3rd; Varambally, S. EZH2-Targeted Therapies in Cancer: Hype or a Reality. Cancer Res. 2020, 80, 5449–5458. [Google Scholar] [CrossRef]
  211. Straining, R.; Eighmy, W. Tazemetostat: EZH2 Inhibitor. J. Adv. Pract. Oncol. 2022, 13, 158–163. [Google Scholar] [CrossRef] [PubMed]
  212. Italiano, A.; Soria, J.C.; Toulmonde, M.; Michot, J.M.; Lucchesi, C.; Varga, A.; Coindre, J.M.; Blakemore, S.J.; Clawson, A.; Suttle, B.; et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: A first-in-human, open-label, phase 1 study. Lancet. Oncol. 2018, 19, 649–659. [Google Scholar] [CrossRef]
  213. Ren, F.; Fan, M.; Mei, J.; Wu, Y.; Liu, C.; Pu, Q.; You, Z.; Liu, L. Interferon-γ and celecoxib inhibit lung-tumor growth through modulating M2/M1 macrophage ratio in the tumor microenvironment. Drug Des. Dev. Ther. 2014, 8, 1527–1538. [Google Scholar] [CrossRef] [Green Version]
  214. Hernández-SanMiguel, E.; Gargini, R.; Cejalvo, T.; Segura-Collar, B.; Núñez-Hervada, P.; Hortigüela, R.; Sepúlveda-Sánchez, J.M.; Hernández-Laín, A.; Pérez-Núñez, A.; Sanz, E.; et al. Ocoxin Modulates Cancer Stem Cells and M2 Macrophage Polarization in Glioblastoma. Oxidative Med. Cell. Longev. 2019, 2019, 9719730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Wang, J.; Lee, J.S.; Kim, D.; Zhu, L. Exploration of Zinc Oxide Nanoparticles as a Multitarget and Multifunctional Anticancer Nanomedicine. ACS Appl. Mater. Interfaces 2017, 9, 39971–39984. [Google Scholar] [CrossRef]
  216. Chan, K.S.; Espinosa, I.; Chao, M.; Wong, D.; Ailles, L.; Diehn, M.; Gill, H.; Presti, J., Jr.; Chang, H.Y.; van de Rijn, M.; et al. Identification, molecular characterization, clinical prognosis, and therapeutic targeting of human bladder tumor-initiating cells. Proc. Natl. Acad. Sci. USA 2009, 106, 14016–14021. [Google Scholar] [CrossRef] [Green Version]
  217. Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef] [Green Version]
  218. Grégoire, H.; Roncali, L.; Rousseau, A.; Chérel, M.; Delneste, Y.; Jeannin, P.; Hindré, F.; Garcion, E. Targeting Tumor Associated Macrophages to Overcome Conventional Treatment Resistance in Glioblastoma. Front. Pharmacol. 2020, 11, 368. [Google Scholar] [CrossRef] [Green Version]
  219. Yuan, J.; Shi, X.; Chen, C.; He, H.; Liu, L.; Wu, J.; Yan, H. High expression of CD47 in triple negative breast cancer is associated with epithelial-mesenchymal transition and poor prognosis. Oncol. Lett. 2019, 18, 3249–3255. [Google Scholar] [CrossRef] [Green Version]
  220. Cao, X.; Li, B.; Chen, J.; Dang, J.; Chen, S.; Gunes, E.G.; Xu, B.; Tian, L.; Muend, S.; Raoof, M.; et al. Effect of cabazitaxel on macrophages improves CD47-targeted immunotherapy for triple-negative breast cancer. J. Immunother. Cancer 2021, 9, e002022. [Google Scholar] [CrossRef]
  221. Zhang, X.; Wang, Y.; Fan, J.; Chen, W.; Luan, J.; Mei, X.; Wang, S.; Li, Y.; Ye, L.; Li, S.; et al. Blocking CD47 efficiently potentiated therapeutic effects of anti-angiogenic therapy in non-small cell lung cancer. J. Immunother. Cancer 2019, 7, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Zhang, X.; Fan, J.; Wang, S.; Li, Y.; Wang, Y.; Li, S.; Luan, J.; Wang, Z.; Song, P.; Chen, Q.; et al. Targeting CD47 and Autophagy Elicited Enhanced Antitumor Effects in Non-Small Cell Lung Cancer. Cancer Immunol. Res. 2017, 5, 363–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Ye, P.; Chi, X.; Cha, J.H.; Luo, S.; Yang, G.; Yan, X.; Yang, W.H. Potential of E3 Ubiquitin Ligases in Cancer Immunity: Opportunities and Challenges. Cells 2021, 10, 3309. [Google Scholar] [CrossRef] [PubMed]
  224. Chin, A.R.; Wang, S.E. Cytokines driving breast cancer stemness. Mol. Cell. Endocrinol. 2014, 382, 598–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Agliano, A.; Calvo, A.; Box, C. The challenge of targeting cancer stem cells to halt metastasis. Semin. Cancer Biol. 2017, 44, 25–42. [Google Scholar] [CrossRef]
  226. Gao, Y.Y.; Tao, T.; Wu, D.; Zhuang, Z.; Lu, Y.; Wu, L.Y.; Liu, G.J.; Zhou, Y.; Zhang, D.D.; Wang, H.; et al. MFG-E8 attenuates inflammation in subarachnoid hemorrhage by driving microglial M2 polarization. Exp. Neurol. 2021, 336, 113532. [Google Scholar] [CrossRef]
  227. Shimagaki, T.; Yoshio, S.; Kawai, H.; Sakamoto, Y.; Doi, H.; Matsuda, M.; Mori, T.; Osawa, Y.; Fukai, M.; Yoshida, T.; et al. Serum milk fat globule-EGF factor 8 (MFG-E8) as a diagnostic and prognostic biomarker in patients with hepatocellular carcinoma. Sci. Rep. 2019, 9, 15788. [Google Scholar] [CrossRef]
  228. Zhang, H.; Zhang, W.; Sun, X.; Dang, R.; Zhou, R.; Bai, H.; Ben, J.; Zhu, X.; Zhang, Y.; Yang, Q.; et al. Class A1 scavenger receptor modulates glioma progression by regulating M2-like tumor-associated macrophage polarization. Oncotarget 2016, 7, 50099–50116. [Google Scholar] [CrossRef] [Green Version]
  229. Gao, L.; Li, F.; Dong, B.; Zhang, J.; Rao, Y.; Cong, Y.; Mao, B.; Chen, X. Inhibition of STAT3 and ErbB2 suppresses tumor growth, enhances radiosensitivity, and induces mitochondria-dependent apoptosis in glioma cells. Int. J. Radiat. Oncol. Biol. Phys. 2010, 77, 1223–1231. [Google Scholar] [CrossRef]
  230. Singh, J.K.; Simões, B.M.; Clarke, R.B.; Bundred, N.J. Targeting IL-8 signalling to inhibit breast cancer stem cell activity. Expert Opin. Ther. Targets 2013, 17, 1235–1241. [Google Scholar] [CrossRef] [Green Version]
  231. Singh, J.K.; Farnie, G.; Bundred, N.J.; Simões, B.M.; Shergill, A.; Landberg, G.; Howell, S.J.; Clarke, R.B. Targeting CXCR1/2 significantly reduces breast cancer stem cell activity and increases the efficacy of inhibiting HER2 via HER2-dependent and -independent mechanisms. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 643–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. 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 2018, 3, e122375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Mondal, S.; Adhikari, N.; Banerjee, S.; Amin, S.A.; Jha, T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: A minireview. Eur. J. Med. Chem. 2020, 194, 112260. [Google Scholar] [CrossRef] [PubMed]
  234. Liang, Y.; Yang, N.; Pan, G.; Jin, B.; Wang, S.; Ji, W. Elevated IL-33 promotes expression of MMP2 and MMP9 via activating STAT3 in alveolar macrophages during LPS-induced acute lung injury. Cell. Mol. Biol. Lett. 2018, 23, 52. [Google Scholar] [CrossRef] [PubMed]
  235. Venkatesh, V.; Nataraj, R.; Thangaraj, G.S.; Karthikeyan, M.; Gnanasekaran, A.; Kaginelli, S.B.; Kuppanna, G.; Kallappa, C.G.; Basalingappa, K.M. Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investig. 2018, 5, 5. [Google Scholar] [CrossRef] [Green Version]
  236. Sekulic, A.; Migden, M.R.; Oro, A.E.; Dirix, L.; Lewis, K.D.; Hainsworth, J.D.; Solomon, J.A.; Yoo, S.; Arron, S.T.; Friedlander, P.A.; et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N. Engl. J. Med. 2012, 366, 2171–2179. [Google Scholar] [CrossRef] [Green Version]
  237. Yao, Z.; Zhang, J.; Zhang, B.; Liang, G.; Chen, X.; Yao, F.; Xu, X.; Wu, H.; He, Q.; Ding, L.; et al. Imatinib prevents lung cancer metastasis by inhibiting M2-like polarization of macrophages. Pharmacol. Res. 2018, 133, 121–131. [Google Scholar] [CrossRef]
  238. Keir, H.R.; Richardson, H.; Fillmore, C.; Shoemark, A.; Lazaar, A.L.; Miller, B.E.; Tal-Singer, R.; Chalmers, J.D.; Mohan, D. CXCL-8-dependent and -independent neutrophil activation in COPD: Experiences from a pilot study of the CXCR2 antagonist danirixin. ERJ Open Res. 2020, 6, 00583-2020. [Google Scholar] [CrossRef]
  239. Busch-Petersen, J.; Carpenter, D.C.; Burman, M.; Foley, J.; Hunsberger, G.E.; Kilian, D.J.; Salmon, M.; Mayer, R.J.; Yonchuk, J.G.; Tal-Singer, R. Danirixin: A Reversible and Selective Antagonist of the CXC Chemokine Receptor 2. J. Pharmacol. Exp. Ther. 2017, 362, 338–346. [Google Scholar] [CrossRef] [Green Version]
  240. Brown, J.R.; Chan, D.K.; Shank, J.J.; Griffith, K.A.; Fan, H.; Szulawski, R.; Yang, K.; Reynolds, R.K.; Johnston, C.; McLean, K.; et al. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020, 5, e133247. [Google Scholar] [CrossRef]
  241. Chen, B.; Cha, J.H.; Yan, M.; Cao, N.; Ye, P.; Yan, X.; Yang, W.H. ATXN7L3B promotes hepatocellular carcinoma stemness and is downregulated by metformin. Biochem. Biophys. Res. Commun. 2021, 573, 1–8. [Google Scholar] [CrossRef] [PubMed]
  242. Wang, S.; Lin, Y.; Xiong, X.; Wang, L.; Guo, Y.; Chen, Y.; Chen, S.; Wang, G.; Lin, P.; Chen, H.; et al. Low-Dose Metformin Reprograms the Tumor Immune Microenvironment in Human Esophageal Cancer: Results of a Phase II Clinical Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4921–4932. [Google Scholar] [CrossRef] [PubMed]
  243. Ejarque, M.; Ceperuelo-Mallafré, V.; Serena, C.; Pachón, G.; Núñez-Álvarez, Y.; Terrón-Puig, M.; Calvo, E.; Núñez-Roa, C.; Oliva-Olivera, W.; Tinahones, F.J.; et al. Survivin, a key player in cancer progression, increases in obesity and protects adipose tissue stem cells from apoptosis. Cell Death Dis. 2017, 8, e2802. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular crosstalk between TAMs and CSCs. CSCs promote macrophage recruitment by secreting CCL2, CCL3, CCL5, CCL8, CCL9, CXCL5, CXCL12, CXCL14, IL-10, IL-33, CSF-1, IGF-β, MIC-1, LOX, VEGF, OLFML3, Sdf1, POSTN, CX3CL1, HGF, MIF, and FGL2. CSCs promote macrophage polarization by secreting CCL2, IL-4, IL-6, IL-8, IL-10, IL-13, IL-33, IL-34, CSF1, CSF2, WISP1, TGF-β1, MIC-1, PGE2, Osteoactivin, and exosomes. TAMs produce factors to support CSC-like features, including CCL2, CCL5, CCL7, CCL8, CCL17, CCL22, CXCL1, CXCL7, CXCL8, IL-1β, IL-6, IL-8, IL-10, IL-18, IL-35, TGF-β, EGF, GPNMB, TNF-α, PTN, IGF, PDGF-BB, hCAP-18, S100A9, HMGB1, MFG-E8, and extracellular vesicles.
Figure 1. Molecular crosstalk between TAMs and CSCs. CSCs promote macrophage recruitment by secreting CCL2, CCL3, CCL5, CCL8, CCL9, CXCL5, CXCL12, CXCL14, IL-10, IL-33, CSF-1, IGF-β, MIC-1, LOX, VEGF, OLFML3, Sdf1, POSTN, CX3CL1, HGF, MIF, and FGL2. CSCs promote macrophage polarization by secreting CCL2, IL-4, IL-6, IL-8, IL-10, IL-13, IL-33, IL-34, CSF1, CSF2, WISP1, TGF-β1, MIC-1, PGE2, Osteoactivin, and exosomes. TAMs produce factors to support CSC-like features, including CCL2, CCL5, CCL7, CCL8, CCL17, CCL22, CXCL1, CXCL7, CXCL8, IL-1β, IL-6, IL-8, IL-10, IL-18, IL-35, TGF-β, EGF, GPNMB, TNF-α, PTN, IGF, PDGF-BB, hCAP-18, S100A9, HMGB1, MFG-E8, and extracellular vesicles.
Biomolecules 12 00850 g001
Figure 2. Targeting strategies of CSCs and TAMs in cancer therapy. Key pathways regulating CSC and TAM properties during tumorigenesis and progression have been identified, including TAM survival, recruitment, polarization, and phagocytosis and maintenance of CSC stemness. Targeting these pathways can help regulate CSC properties and TAMs and interfere with the interaction between them, thereby inhibiting the development of CSCs and providing more effective therapeutic strategies.
Figure 2. Targeting strategies of CSCs and TAMs in cancer therapy. Key pathways regulating CSC and TAM properties during tumorigenesis and progression have been identified, including TAM survival, recruitment, polarization, and phagocytosis and maintenance of CSC stemness. Targeting these pathways can help regulate CSC properties and TAMs and interfere with the interaction between them, thereby inhibiting the development of CSCs and providing more effective therapeutic strategies.
Biomolecules 12 00850 g002
Table 4. Drugs targeting the TAM-related pathways in clinical trials.
Table 4. Drugs targeting the TAM-related pathways in clinical trials.
TargetsDrugsCancer TypeTrial PhaseClinicalTrials.Gov ID
CSF-1/CSF-1RCM082Small cell lung cancerIINCT03904719
ABSK021Advanced solid tumorINCT04192344
AMB-051Tenosynovial giant cell tumorIINCT04731675
SNDX-6532Unresectable intrahepatic cholangiocarcinomaIINCT04301778
ChiauranibTriple-negative breast cancerIINCT05336721
Small cell lung cancerIIINCT04830813
NMS-03592088Acute myeloid leukemia
Chronic myelomonocytic leukemia
I/IINCT03922100
PexidartinibTenosynovial giant cell tumorIIINCT04488822
Q702Advanced solid cancerINCT04648254
SurufatinibNeuroendocrine tumor
Non-hematologic malignancy
I/IINCT05077384
Hepatocellular carcinomaIINCT05171439
Refractory metastatic digestive system carcinoma
Primary peritoneal cancer
IINCT05030246
TPX-0022Advanced
metastatic solid tumor
I/IINCT03993873
VorolanibRefractory thoracic tumorI/IINCT03583086
Metastatic renal cell cancerIIINCT03095040
CCL2/CCR2BMS-813160Locally advanced pancreatic ductal Adenocarcinoma
Pancreatic ductal adenocarcinoma
I/IINCT03767582
CCL5/CCR5MaravirocColorectal cancer metastaticINCT04721301
LeronlimabSolid tumorIINCT04504942
POL6326Advanced breast cancerI/IINCT04826016
Metastatic breast cancerIIINCT03786094
MotixafortidePancreatic cancerIINCT04543071
CD40/CD40LABBV-368Advanced solid tumorsINCT04196283
Triple-negative breast cancer
Non small cell lung cancer
Metastatic solid tumor
INCT03893955
ABBV-927Advanced solid tumor
Triple-negative breast cancer
Non-small cell lung cancer
Metastatic solid tumor
INCT03893955
Pancreatic cancerIINCT04807972
CDX-1140Pancreatic cancerIINCT04536077
Solid tumor
Diffuse large B-cell lymphoma
Mantle cell lymphoma
INCT03329950
Ovarian clear cell adenocarcinomaIINCT05231122
MelanomaI/IINCT04364230
Metastatic triple negative breast cancerINCT05029999
Malignant epithelial neoplasmINCT04520711
GEN-1042Malignant solid tumor
Non-small cell lung cancer
Colorectal cancer
Melanoma
Head and Neck squamous cell carcinoma
Pancreatic ductal adenocarcinoma
I/IINCT04083599
MitazalimabMetastatic pancreatic ductal adenocarcinomaI/IINCT04888312
NG-350AEpithelial tumor
Metastatic cancer
INCT05165433
SelicrelumabColorectal cancerI/IINCT03555149
Triple-negative breast cancerI/IINCT03424005
Pancreatic adenocarcinomaI/IINCT03193190
YH-003Advanced solid tumorI/IINCT04481009
Melanoma
Pancreatic ductal adenocarcinoma
IINCT05031494
SL-172154Ovarian cancer
Fallopian tube cancer
Primary peritoneal carcinoma
INCT04406623
Cutaneous squamous cell carcinoma
Head and Neck squamous cell carcinoma
INCT04502888
Acute myeloid leukemiaINCT05275439
BDB-001Solid tumorIINCT04819373
Pancreatic cancer
Virus-associated tumor
Non-small cell lung cancer
Melanoma
Bladder cancer
Triple-negative breast cancer
IINCT03915678
BDC-1001HER2 positive solid tumorI/IINCT04278144
BNT-411Extensive-stage small cell lung cancerI/IINCT04101357
DSP-0509NeoplasmI/IINCT03416335
ImiquimodMetastatic melanomaINCT03276832
Oral cancerINCT04883645
Squamous cell carcinomaINCT03370406
ResiquimodLocally advanced solid tumor
Metastatic solid tumor
I/IINCT04799054
Anaplastic astrocytomaIINCT01204684
SBT-6050HER2 positive solid tumorINCT04460456
TLR9AST008Advanced or Metastatic merkel cell carcinomaI/IINCT03684785
CMP-001Head and Neck squamous cell carcinomaIINCT04633278
Metastatic melanomaIINCT04698187
Merkel cell carcinomaIINCT04916002
Malignant colorectal neoplasmINCT03507699
Advanced malignancy
Non-small cell lung cancer
Ovarian cancer
Urothelial cancer
Solid tumor
IIINCT05059522
TilsotolimodAdvanced solid tumorINCT04196283
Malignant melanomaIINCT04126876
SD-101Metastatic uveal melanoma in the LiverINCT04935229
Hepatocellular carcinoma
Intrahepatic cholangiocarcinoma
I/IINCT05220722
B-Cell Non-Hodgkin lymphomaINCT03410901
Metastatic pancreatic adenocarcinomaINCT04050085
TLR3Poly-ICLCHepatocellular carcinomaINCT05281926
Glioblastoma multiformeI/IINCT03665545
RintatolimodHematopoietic and lymphoid cell neoplasmI/IINCT04379518
Prostate adenocarcinomaIINCT03899987
PI3Kγ signal pathwayCopanlisibNon-small cell lung cancerINCT04895579
Diffuse large B-cell lymphomaIINCT04433182
Refractory/Recurrent primary central system lymphomaI/IINCT03581942
Follicular lymphoma
Endometrial cancer
IINCT04750941
Mantle cell lymphomaI/IINCT03877055
Head and Neck squamous cell carcinoma
Hepatocellular carcinoma
INCT03735628
DuvelisibNon-Hodgkin lymphomaINCT05065866
Unresectable melanomaI/IINCT04688658
Recurrent diffuse large B-Cell lymphomaINCT04890236
EganelisibBladder cancer
Urothelial carcinoma
IINCT03980041
Non-small cell lung cancer
Melanoma
Head and Neck squamous cell cancer
Triple-negative breast cancer
Adrenocortical carcinoma
Mesothelioma
High-circulating myeloid-derived suppressor cells
INCT02637531
Breast cancer
Renal cell carcinoma
IINCT03961698
GedatolisibHER2-positive breast cancer
Metastatic breast cancer
IINCT03698383
Triple-negative breast cancerI/IINCT03911973
Squamous cell lung cancer
Solid tumor
Head and Neck cancer
Pancreatic cancer
INCT03065062
TenalisibLocally advanced breast cancer
Metastatic breast cancer
IINCT05021900
CD47/SIRPα pathwayALX-148Microsatellite stable metastatic colorectal cancerIINCT05167409
Aggressive B-Cell Non-Hodgkin lymphomaI/IINCT05025800
Head and Neck squamous cell carcinomaIINCT04675294
Acute myeloid leukemiaI/IINCT04755244
Gastroesophageal junction adenocarcinomaII/IIINCT05002127
Non-Hodgkin lymphomaINCT03013218
AO-176Multiple myelomaI/IINCT04445701
Solid tumorI/IINCT03834948
HX-009Advanced solid tumorIINCT04886271
Relapsed/Refractory lymphomaI/IINCT05189093
IBI-188Acute myeloid leukemiaI/IINCT04485052
Lung adenocarcinoma
Osteosarcoma
INCT04861948
IBI-322Advanced malignant tumor
Lymphoma
INCT04338659
Non-small cell lung cancerIINCT05296278
Hematologic malignancyINCT04795128
Advanced malignancyINCT04328831
Myeloid tumorINCT05148442
IMC-002Advanced cancerINCT05276310
Solid tumor
Lymphoma
INCT04306224
IMM-01Acute myeloid leukemiaI/IINCT05140811
IMM-0306B-cell non-Hodgkin’s lymphomaINCT04746131
LemzoparlimabAcute myeloid leukemia
Myelodysplastic syndrome
INCT04912063
Multiple myelomaINCT04895410
MagrolimabMalignant brain tumorINCT05169944
Recurrent neuroblastomaINCT04751383
Solid tumorIINCT04827576
Recurrent acute myeloid leukemiaI/IINCT04435691
Metastatic colorectal cancerIINCT05330429
Multiple myelomaIINCT04892446
Head and neck squamous cell carcinomaIINCT04854499
Metastatic colorectal cancerIINCT05330429
Multiple myelomaIINCT04892446
Urothelial carcinoma
Bladder cancer
I/IINCT03869190
RRx-001Central nervous system neoplasmINCT04525014
Small cell lung carcinomaIIINCT03699956
Small cell carcinoma
Non-small cell lung carcinoma
Neuroendocrine tumor
Ovarian epithelial cancer
IINCT02489903
TTI-622Multiple myelomaINCT05139225
Fallopian tube cancer
Primary peritoneal carcinoma
I/IINCT05261490
Acute myeloid leukemia
Diffuse large B-Cell lymphoma
INCT03530683
STING pathwaySNX-281Advanced solid tumor
Advanced lymphoma
INCT04609579
BMS-986301Advanced solid cancerINCT03956680
GSK-3745417NeoplasmINCT03843359
E−7766Lymphoma
Advanced solid tumor
INCT04144140
SYNB-1891Metastatic solid neoplasm
lymphoma
INCT04167137
TAK-676Non-small cell lung carcinoma
Triple-negative breast neoplasm
Head and Neck squamous cell carcinoma
INCT04879849
Solid neoplasmINCT04420884
MK-2118LymphomaINCT03249792
IMSA101Solid tumorI/IINCT04020185
CDK-002Advanced solid tumorI/IINCT04592484
MK-1454Head and neck squamous cell carcinomaIINCT04220866
NOX66Late-stage prostate cancerINCT03307629
Metastatic soft-tissue sarcomaINCT05100628
Metastatic castration-resistant prostate cancerI/IINCT04957290
SB-11285Melanoma
Head and neck squamous cell carcinoma
INCT04096638
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Luo, S.; Yang, G.; Ye, P.; Cao, N.; Chi, X.; Yang, W.-H.; Yan, X. Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells. Biomolecules 2022, 12, 850. https://0-doi-org.brum.beds.ac.uk/10.3390/biom12060850

AMA Style

Luo S, Yang G, Ye P, Cao N, Chi X, Yang W-H, Yan X. Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells. Biomolecules. 2022; 12(6):850. https://0-doi-org.brum.beds.ac.uk/10.3390/biom12060850

Chicago/Turabian Style

Luo, Shahang, Guanghui Yang, Peng Ye, Nengqi Cao, Xiaoxia Chi, Wen-Hao Yang, and Xiuwen Yan. 2022. "Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells" Biomolecules 12, no. 6: 850. https://0-doi-org.brum.beds.ac.uk/10.3390/biom12060850

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop