Open Access

Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review)

  • Authors:
    • Mingkai Yu
    • Haixia Yu
    • Hongmei Wang
    • Xiaoya Xu
    • Zhaoqing Sun
    • Wenshuai Chen
    • Miaomiao Yu
    • Chunhua Liu
    • Mingchun Jiang
    • Xiaowei Zhang
  • View Affiliations

  • Published online on: September 5, 2024     https://doi.org/10.3892/ijo.2024.5688
  • Article Number: 100
  • Copyright: © Yu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Hepatocellular carcinoma (HCC) tissue is rich in dendritic cells, T cells, B cells, macrophages, natural killer cells and cellular stroma. Together they form the tumor microenvironment (TME), which is also rich in numerous cytokines. Tumor‑associated macrophages (TAMs) are involved in the regulation of tumor development. TAMs in HCC receive stimuli in different directions, polarize in different directions and release different cytokines to regulate the development of HCC. TAMs are mostly divided into two cell phenotypes: M1 and M2. M1 TAMs secrete pro‑inflammatory mediators, and M2 TAMs secrete a variety of anti‑inflammatory and pro‑tumorigenic substances. The TAM polarization in HCC tumors is M2. Both direct and indirect methods for TAMs to regulate the development of HCC are discussed. TAMs indirectly support HCC development by promoting peripheral angiogenesis and regulating the immune microenvironment of the TME. In terms of the direct regulation between TAMs and HCC cells, the present review mainly focuses on the molecular mechanism. TAMs are involved in both the proliferation and apoptosis of HCC cells to regulate the quantitative changes of HCC, and stimulate the related invasive migratory ability and cell stemness of HCC cells. The present review aims to identify immunotherapeutic options based on the mechanisms of TAMs in the TME of HCC.

Introduction

Multidimensional analysis has indicated that a special tumor microenvironment (TME) exists in liver cancer tissues (1). The study of treatment protocols for tumors in modern medicine from the perspective of cancer biology no longer considers cancer cells as the central link, but evolved from the study of cancer cells to the study of the TME in which cancer cells are located, categorizing cancer cells in the stromal cell network (2). The molecular mechanisms involved in tumor development can be elucidated by analyzing the characteristics of different cell types in the TME (3): Tumor-associated macrophages (TAMs), CD4+ cells, cytotoxic T lymphocytes (CD8+ cells), regulatory T cells (Tregs), dendritic cells, natural killer cells (NK cells), tumor-associated endothelial cells, cancer-associated fibroblasts (CAFs) and myeloid-derived immunosuppressive cells (MDSCs) (4). Kupffer cells (KCs) and monocyte-derived hepatic macrophages are among the innate immune-responsive cells that are widely present in the human body (5). Macrophages exert immunomodulatory effects through phagocytosis, exogenous antigen presentation, and cytokine and growth factor secretion (6).

TAM description

Classical macrophage phenotypes

Macrophage polarization is a process that occurs in response to changes in microenvironmental stimuli and signals. Functionally polarized subpopulations acquire substantially different functional attributes after the action of different stimuli (7). Lipopolysaccharides (LPS) and interferon-γ direct macrophage polarization towards the M1 phenotype by activating the Janus kinase (JAK)/STAT, toll-like receptor 4 (TLR4)/NF-κB and NOTCH signaling pathways (8). The types of macrophage polarization are classified into two groups according to their functions (classically activated M1-polarized macrophages and alternatively activated M2-polarized macrophages), with different expression levels of proteins on the cell surface (9). Proinflammatory cytokines, including IL-1β, IL-6, IL-12, IL-18, IFN-γ, inducible nitric oxide synthase (iNOS) and TNF-α, are highly expressed in M1-polarized macrophages. These cytokines also function as tumor suppressors in hepatocellular carcinoma (HCC) (10). By contrast, M2 macrophages secrete anti-inflammatory cytokines such as IL-4, TGF-β and IL-10 (11). M2 macrophages exert complex immunomodulatory functions depending on these cytokines, which possess cancer-promoting and inflammation-inhibiting effects by triggering immunosuppression (12). M2 polarization is induced by factors that activate the TGF-β/Smads and peroxisome proliferator-activated receptor γ (PPARγ) signaling pathways, such as TGF-β and growth differentiation factor 3 (13). M2 macrophages induced by diverse stimulatory factors are further subdivided into four types: M2a, M2b, M2c and M2d (14).

The M4 macrophage phenotype is induced by chemokine C-X-C motif ligand (CXCL)4 in atherosclerosis, and characterized by MMP7, CD68 and S100 calcium binding protein A8 expression (15). Hemoglobin and oxidized phospholipids polarize M0 macrophages to hemoglobin-stimulated macrophages and Mox macrophages (16). Mantovani et al (17) hypothesized that regulatory M3 macrophages are a bridge for the mutual transformation of M1 and M2 macrophages. Fig. 1 shows a summary of the directions of macrophage polarization.

Figure 1

Classical macrophage phenotypes. Macrophages have M1, M2, M3, M4, M(Hb) and Mox polarization phenotypes. The M1 and M2 phenotypes are the two main macrophage polarization phenotypes. M3 macrophages are an intermediate phenotype that bridges the interconversion between M1 and M2 macrophages. Macrophages are referred to as TAMs in the tumor microenvironment. TAMs can also be divided into M1 and M2 subtypes. Pre-tumor macrophages polarize to the M1 subtype and inhibit tumorigenesis and progression, while more macrophages polarize to the M2 subtype in the later stages and exert pro-tumorigenic effects. M1 macrophages are mainly induced by LPS and IFN-γ. M2 macrophages are stimulated by different regulatory factors and differentiate into four subtypes: M2a, M2b, M2c and M2d. IL-4 and CCL22 are responsible for M2a macrophage polarization, and M2a macrophages focus on tissue remodeling and reducing inflammation. Fc-γ receptor and LPS/IL-1β cause M2b macrophage polarization, and M2b macrophages serve a role in antigen presentation and promoting HCC progression. M2c macrophages are polarized by TGF and IL-10 and have functions in matrix deposits and immunoregulation. TLR and IL-6 drive M2d macrophage polarization, and M2d macrophages contribute to angiogenesis, and tumor growth and progression. CXCL4 can drive macrophage polarization toward the M4 phenotype. In Hb and OxPL pathological tissue, macrophages are polarised to M(Hb) and Mox, respectively. CCL22, C-C motif chemokine ligand 22; CXCL4, chemokine C-X-C motif ligand 4; M(Hb), hemoglobin-stimulated macrophage; HCC, hepatocellular carcinoma; LPS, lipopolysaccharide; OxPL, oxidized phospholipid; TAMs, tumor-associated macrophages; TLR, toll-like receptor.

Regulation of macrophage polarization by HCC cells

Hypoxic conditions in the HCC TME inhibit the xanthine oxidoreductase (XOR)/isocitrate dehydrogenase (NAD (+)) 3 catalytic subunit α (IDH3α) axis of TAMs and increase the ratio of M2 TAMs to total TAMs (18). Hematopoietic stem cells (HSCs) are induced to differentiate into CAFs in HCC (4). The expression levels of IL-6, which is associated with the polarization of the M0 cell phenotype to the M2 phenotype, are increased in CAFs-conditioned medium compared with HSCs. In addition, high levels of CXCL12 expression by CAFs regulate the expression of plasminogen activator (PA) inhibitor-1 (PAI-1) in polarized M2 TAMs by specifically binding to C-X-C chemokine receptor type 4 (CXCR4) on the macrophage surface (19). Angiopoietin (Ang)-like protein 8 (ANGPTL8) is expressed in HCC cells and ANGPTL8 expression is positively associated with the degree of malignancy of HCC. The interaction of ANGPTL8 with the leukocyte Ig-like receptor subfamily B/paired Ig-like receptor results in the inhibition of the M1 polarization and promotes the polarization towards the M2 phenotype of TAMs (20). A related study using HCC as a model has indicated that iron restriction due to HCC activates TAM polarization towards the M2 phenotype. TAMs in HCC exhibit upregulated expression levels of the apolipoprotein C1 (APOC1) gene. APOC1 inhibition activates apoptosis in TAMs, reverses the M2 phenotype to the M1 phenotype and alters the phenotypic proportion of TAMs in HCC (21). Metabolism-related RNA expression in HCC is an important regulator of the activation of TAM polarization. Long non-coding RNAs (lncRNAs/lncs), microRNAs (miRNAs/miRs) and circular RNAs (circRNAs/circs) are regulated by gene expression to alter the polarization of TAMs (22,23). lncRNA is a type of RNA that is >200 nucleotides long and does not encode proteins (24). ZNNT1 is a lncRNA, and its high expression is associated with the infiltration of M2-polarized macrophages, promoting HCC cell proliferation, migration and invasion, and inhibiting HCC cell apoptosis (25). S100A9 secreted by M2 macrophages upregulates ZNNT1 expression in HCC cells through the activation of advanced glycosylation end-product specific receptor (AGER)/NF-κB signaling, a process that is related to the regulation of ZNNT1 transcript stability by m6A modification (26). MEG3 lncRNA is widely distributed in a range of solid tumors. Its silencing promotes drug resistance in various malignant tumors (27,28). Human antigen R (HuR), also known as embryonic lethal anomalous visual-like 1, is an RNA-binding protein (29). Chemokine (C-C motif) ligand (CCL)5, also known as RANTES, participates in inflammatory and immune responses as a chemokine. HuR is a target RNA of MEG3 and inhibits CCL5 expression in bone marrow-derived macrophages by suppressing CCL5 transcription and promoting M2 macrophage polarization. MEG3 inhibits HuR expression in M1 and M2 macrophages, leading to a change in the direction of macrophage polarization (30,31). Thus, MEG3 regulates HuR at the post-transcriptional level in HCC, being a potential therapeutic target for liver disease (32). Furthermore, lncRNA MEG3 is linked to miR-145-5p in HCC cells, and MEG3 overexpression leads to a decrease in miR-145-5p mRNA expression. High miR-145-5p expression is positively associated with M2 macrophage polarization. DAB adaptor protein 2 (DAB2) is a downstream target of miR-145-5p, and DAB2 upregulation is a hallmark of M1 polarization. MEG3 inhibits M2 macrophage polarization through the upregulation of DAB2 (33). Insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3) is highly expressed in human solid tumors and promotes the differentiation, proliferation, invasion and metastasis of HCC cells (34). When highly expressed, it binds to the mRNAs of CCL5 or TGF-β1 in HCC cells, promotes the secretion of cytokines, and induces macrophage M2 polarization. IGF2BP3 knockdown combined with anti-CD47 therapy slows down tumor growth (35). A combination of in vivo and in vitro experiments revealed the role of IGF2BP3 as a specific migration factor in HCC (36). Evidence indicates that circRNAs absorb miRNAs via the 'sponge adsorption' effect and interact with RNA-binding proteins. circ_0010882 is an upstream regulatory factor of miR-382. circ_0010882 silencing inhibits M2 macrophage polarization by targeting miR-382 (37). miR-21-5p in HCC cell exosomes affects SP1/X-box binding protein 1 (XBP1) protein expression and promotes M2 polarization in TAMs (38).

The upregulation of tripartite motif containing 65 (TRIM65) is associated with the high grading of HCC tumors (39). The JAK1/STAT1 signaling pathway is activated and the expression of pro-inflammatory M1 markers is increased in TAMs after culturing with conditioned medium from the supernatant of Hep1-6 cells with TRIM65 knockdown. This suggests that TRIM65 could serve as a unique tumor immunotherapeutic target (40). Although in vivo and in vitro experiments have validated the therapeutic efficacy of targeting TRIM65 in the treatment of HCC, there is no evidence that has associated the effect of TRIM65 knockdown on HCC with its distribution in macrophages (41). Zinc-fingers and homeoboxes 2 is a regulator of a number of liver-enriched genes and it maintains macrophage polarization by regulating interferon regulatory factor (IRF)1 transcription, which could therefore be a potential target for macrophage-based cancer immunotherapy (42). CCL16 is a factor with strong chemotactic properties. C-C chemokine receptor type 1 (CCR1) is a receptor that positively regulates the extent of M2 TAM infiltration in HCC. CCL16 produced by hepatocytes in the normal liver binds to CCR1 expressed by human KCs. Cancer cells bind to the CCR1 receptor on macrophages and release CCL16 to increase the number of CD68+ CCR1+ cells, which induces M2 polarization in TAMs (43). Auxiliary proteins are involved in IL-4-induced M2 polarization. Major vault protein (MVP) is the main component in vault nanoparticles. Its expression is increased in HCC compared with that in the normal liver (44). IL-4 activates the dimerization of IL-4 receptor (IL-4R) α and γ chains by binding to the macrophage surface receptor IL-4R. Activation of the JAK1/STAT6 signaling pathway increases the M2 polarized subtype of TAMs (45). MVP interacts with JAK1 and recruits STAT6 to form the ternary complex JAK1/MVP/STAT6, which enhances IL-4-induced STAT6 activity. The degree of infiltration of M2 TAMs is increased in HCC with high MVP expression (46). xCT (encoded by SLC7A11) is a heavy chain subunit of the system x that provides a nutrient environment for tumor development by reducing glutathione biosynthesis and antioxidant defense in tumor cells. xCT activates the suppressor of cytokine signaling 3/STAT6/PPARγ signaling axis and regulates the pro-tumorigenic M2-like phenotypic shift in the TME caused by IL-4 (47). Therefore, xCT knockdown in macrophages inhibits the pro-tumorigenic phenotypic activation of IL-4. In addition to IL-4, STAT6 mediates M2 cell polarization via different mechanisms of activation (48). Myeloid differentiation factor 88 (MyD88) is a ligand for TLR and activates a pro-inflammatory cascade, while it promotes CCL9/CCL15 secretion in HSCs, which enhances M2 polarization in macrophages via the activation of the STAT6/PPARβ signaling pathway in HCC cells (49). Specific deletion of MyD88 in myofibroblasts reduces the secretion of CCL9, a macrophage inflammatory protein, in nonalcoholic fatty liver disease-associated HCC (50). STAT3, another component of the STAT family, is also involved in the regulation of TAM polarization in HCC (51). The proteasome subunit α (PSMA) family of proteins mediates the synthesis of the 20S proteasome core complex and is present in exosomes secreted by metastatic HCC tissues (52). PSMA5 is a member of the PSMA family of proteins, and differences in its expression are critical for tumor growth and development. PSMA5 is activated after the activation of JAK2 and JAK1 in TAMs (53). M2 macrophages receive exosomes from HCC tissues (54). Formimidoyltransferase cyclodeaminase (FTCD) mediates enzymatic reactions and cellular biofilm ligation in tumor cells. As a downstream target of hypoxia-inducible factor-1α (HIF-1α), FTCD-stimulated macrophages are polarized toward the M1 type, and M1 TAMs secrete pyruvate kinase M2 (PKM2) to inhibit HCC cell proliferation (55). Macrophage glucose metabolism is also a factor inducing M2 polarization in TAMs in the TME. XOR mediates the regulation of α-ketoglutarate (α-KG) synthesis and mediates the activation of an M1 promoter in macrophages (NLR family containing pyrin structural domain 3), leading to increased IL-1β expression (56). Tumor cells suppress XOR mRNA and protein expression in macrophages. In macrophages, XOR binds to the active structural domain of IDH3α. IDH3 is a regulatory enzyme in the tricarboxylic acid cycle and is involved in the catalytic synthesis of α-KG (57). XOR knockdown in macrophages mediates the regulation of IDH3 activity, leading to an increase in α-KG, and activation of IDH3α/α-KG/jumonji domain containing 3 signaling, thus enhancing M2 polarization (18) There are numerous factors that activate macrophage polarization in HCC. Table I shows the classification of polarization factors based on whether they also promote macrophage differentiation in other tumor tissues.

Table I

Rough classification of factors that activate macrophage polarization in HCC tissues according to specificity.

Table I

Rough classification of factors that activate macrophage polarization in HCC tissues according to specificity.

Polarization factors unique to HCCCo-polarizing factors in tumors
lncRNA ZNNT1, miR-21-5P, miR-382, IRF1, CCL9, ANGPTL8, IGF2BP3lncRNA MEG, IL-6, IL-4, IL-10, IL-12, IL-18, CXCL12, TGF-β, IFN-γ

[i] ANGPTL8, angiopoietin-like protein 8; CCL9, chemokine (C-C motif) ligand 9; CXCL12, chemokine C-X-C motif ligand 12; HCC, hepatocellular carcinoma; IGF2BP3, insulin like growth factor 2 mRNA binding protein 3; IRF1, interferon regulatory factor 1; lncRNA, long non-coding RNA; miR, microRNA.

Direct modulation of the HCC TME by TAMs

Role of TAMs in regulating cancer stem cell (CSC) differentiation in HCC

Primary liver cancer includes HCC, intrahepatic cholangiocarcinoma (iCCA) and combined HCC-CCA (cHCC-CCA). IgGFc-binding protein+ secreted phosphoprotein 1 (SPP1)+ TAMs are mainly observed in primary HCC and are commonly distributed in areas of HCC and iCCA, and the poorly differentiated areas in cHCC-CCA. The necroptotic apoptotic microenvironment is associated with HCC differentiation, and IgGFc-binding protein+ SPP1+ TAMs promote HCC to gradually acquire CCA features, which are important for intrahepatic biliary metastasis of HCC cells (58).

CSCs in the TME are a type of self-renewing undifferentiated cells with tumorigenic and stem cell-like characteristics (59). CSCs are essential for tumor development and migration (60). Stemness markers, including NANOG, OCT4, SOX2, MYC oncogene and krüppel-like factor 4 (KLF4), are essential transcription factors in the pluripotent stemness of CSCs (61). M2 TAMs act as a 'HCC stemness regulatory complex' by secreting VEGFA, integrin subunit β3 binding protein and ADAM metallopeptidase domain 9 ligands (62,63). The CXCL12-integrin subunit β1 and VEGFA-integrin subunit αV pairs in M2 TAMs are involved in the initiation of tumor cells and the stem-like properties of OV6 CSCs, which are important assistants in the stemness of HCC (64).

TAMs produce IL-6, which promotes the expansion of these CSCs and tumorigenesis (65). Exploiting the interaction between TAMs and CSCs in HCC represents a novel approach for the development of tumor suppressors. S100 calcium binding protein A4 (S100A4) and collagen I enhance HCC stem cell properties by relying on the receptor for advanced glycation endproducts/β-catenin signaling pathway. S100A4+ macrophages are a subtype of M2 macrophages in HCC tissues (66). The stemness markers Oct-4, Nanog, CD133, SOX2 and CD44 are upregulated in HuH7 liver cancer cells after stimulation by S100A4. In vivo experiments suggest that S100A4 knockdown in a mouse model of HCC decreases the level of CSC markers (67). S100A9, also belonging to the S100 family, is a common factor expressed in exosomes of TAMs and CSCs. S100A9 stimulates CCL2 and tumor stemness-related gene production via the AGER/NF-κB signaling pathway in HepG2 and MHCC-97H cells. In addition to the effect of S100A9 expressed by TAMs in malignancy development, S100A9 in the TME serves a role in the recruitment and polarization of TAMs (68).

A sharp drop in the size of subcutaneous tumors in a tumor mouse model was observed after intracardiac injection of HCC cells co-cultured with M2 TAMs. Furthermore, co-culture of HCC cells with M2 TAMs resulted in a decrease in the expression levels of characteristic stem cell surface markers of HCC cells, including CD133, master transcription factors Oct-4 and SOX-2, N-cadherin, and vimentin. The in vitro approach revealed that TNF-α secreted by M2 TAMs activated the Wnt/β-catenin signaling pathways, leading to epithelial-to-mesenchymal transition (EMT) and the stemness features of HCC (69).

TGF-β1 is a central regulator of inflammation and liver cancer (70). RAW264.7 macrophages are induced to differentiate into M2 macrophages by IL-4, resulting in an increase in TGF-β1 expression, which is linked to the action of the TGF-β1/Smad signaling pathway for EMT and the levels of Bmi1 and KLF4, which are two genes characterizing HCC stem cells (71,72). TGF-β type II receptor gene (TGFBR2) is the receptor of TGF-β1 and TGF-β2 (73). TAMs increase lncRNA H19 expression by regulating TGFBR2, downregulating miR-193b expression in HCC cells. Finally, MAPK1 signaling is activated, regulating the EMT pathway and the conversion of non-CSCs to CSCs (74).

Role of TAMs in regulating the proliferation and apoptosis of HCC cells

The properties of HCC cells depend on the stimulation of various components in the TME, especially cytokines, chemotactic cytokines and exosomes. IL-6 is a critical cytokine for HCC cell proliferation (75). There are numerous cells in the TME secreting the inflammatory factor IL-6 (76). By contrast, the scavenger receptor cysteine-rich type 1 protein M130 activates CD163+ TAMs, a type of M2 TAMs expressing IL-6. This effect in the HCC TME involves the IL-6/STAT3 signaling pathway activated by M2 TAMs and their exosomes (77).

Autophagy protects the viability of cells and maintains the steady state of cells (78). Macrophage autophagy has the dual function of inhibiting and promoting tumor growth in the TME. The PI3K/Akt/mTOR signaling pathway is a classical pathway in autophagy. M2 TAMs are involved in the activation of this pathway in HCC cells, resulting in the inhibition of M2 TAM autophagy (79). miRNA-210 is an autophagy agonist acting on the downregulation of PI3K/Akt/mTOR signaling in macrophages, as a consequence of which, M2 TAMs increase the expression levels of IL-10 and TGF-β1, promoting HCC cell proliferation (80).

The proliferation of HCC cells is also associated with the cell cycle. M2 TAMs induced by CAFs regulate the HCC cell cycle directly by inducing PAI-1 expression. PAI-1 downregulates FAS and Fas ligand, resulting in the decrease of the antiapoptotic abilities of liver cancer cells and in the promotion of HCC (19). Extracellular ubiquitin (eUb) regulates cell apoptosis with the assistance of TAMs. eUb is not a regulator of TAM activation and polarization. The absence of TAMs affects the effect of eUb in regulating HCC cell apoptosis. After inhibition by the pro-inflammatory factor TNF-α released by M1 TAMs, HCC cells receive the signal from eUb. and activate the Akt/mTOR signaling pathway and suppress apoptosis (81). In addition, large tumor suppressor kinase 1 activates the Hippo signaling pathway in HCC cells. The Hippo signaling pathway is critical for cell apoptosis and mitochondrial damage (82). Arsenate induces M2-type polarization of macrophages by inducing miR-15b expression to inhibit HCC cell apoptosis activated by LAST1 (83). Upregulation of the EZH2-associated lncRNA HEIH is associated with poor prognosis in patients with HCC. HEIH promotes macrophage M2 polarization by targeting the miR-98-5p/STAT3 axis, and promotes tumor growth and metastasis induced by M2 TAMs (84). Lysosomal acid lipase (LAL) inhibition leads to the accumulation of cellular lipid content and reduces the expression levels of CD36 and ATP binding cassette subfamily a member 1 in macrophages. LAL inhibition slows down monocyte cholesterol metabolism, which reduces the number of M2 TAMs and decreases the HCC tumor growth-promoting axis in which M2 TAMs are involved (85).

M1 TAMs polarized by the signal NORCH-recombination signal binding protein for immunoglobulin κJ region (RBPJ) release exosomes with high RBPJ expression. Hsa-cir-004658 in RBPJ+/+exosome secreted by M1 TAMs is involved in the miR499b-5p/junctional adhesion molecule 3 signaling pathway and promotes the apoptosis of liver cancer cells, while it inhibits the increase in the number of HCC cells (86). Under the stimulation of hepatoma cells, macrophages increase the expression of the classical M1 TAM chemotactic cytokine interleukin-8 (CXCL8) (87). CXCL8 facilitates HCC cell proliferation by changing the percentage of miRNA-17, which is a potent proliferative molecule in the miRNA clusters (88). TAM cell glycolysis in HCC is involved in the proliferative ability of HCC (89). The glycolytic enzymes 6-phosphofructo-2-kinase, hexokinase 2, triose-phosphate isomerase, PKM2, glucose transporter type 1 and lactate dehydrogenase A negatively regulate the apoptosis of H22 tumor cells. Dectin3, a C-type lectin-like receptor, increases HCC cell proliferation by activating cell glycolysis (90,91). HCC cells undergo circ fucosyltransferase 8 (FUT8) m6A modification mediated by METTL14, which promotes the translocation of circFUT8 to the cytoplasm and the recognition of the specific protein YTH N6-methyladenosine RNA binding protein C1, and positively regulates the tumor growth cycle. M1 TAMs competitively bind cytoplasmic circFUT8 through the exosomal miR-628-5p to inhibit HCC progression (92).

Role of TAMs in regulating the invasion and metastasis of HCC cells

According to several studies, the polarized orientation of TAMs in the TME is closely associated with the invasive and metastatic ability of HCC cells through the regulation of multiple signaling pathways (5,43,45,93). M2 TAMs activate the ability of HCC cells to invade and migrate (5). M1 type macrophages antagonize the action of M2 type macrophages by inhibiting HCC progression (9). EMT is an important process in cancer progression, and the prediction of cancer cell metastasis based on the expression of EMT markers is a well-established and effective method (11).

M2 TAM secretion of the cytokines CXCL12, CXCL16, epidermal growth factor (EGF), CCL18 and CCL22 enhances the invasion of HCC cells (94,95). The level of macrophage-derived chemokine (MDC)/CCL22 secretion is increased in M2 TAMs (96). CCL22 selectively interacts with C-C motif chemokine receptor 4, and is involved in the direct activation of EMT in HCC cells (95). In a previous study, the further away the colony-stimulating factor (CSF)-1 receptor (CSF-1R) was from the peri-carcinoma tissue, the less expressed it was. The main drawback of the study was that it did not specifically investigate the mechanisms associated with CSF-1 affecting intrahepatic metastasis of tumors (97). High CSF-1R expression in peritumoral HCC tissues results in an increase in macrophage infiltration in HCC through CSF-1, affecting the intrahepatic metastasis of tumors. Specifically, CSF-1 mediates the upregulation of AIF1 in M0 macrophages, induces M2 macrophage polarization and increases the secretion of the cytokine CXCL16, thereby leading to the migration of Hepa1-6 cells (98). M2 TAMs secrete IL-8, activate the JAK2/STAT3/Snail signaling pathway, upregulate the expression of the mesenchymal marker N-calmodulin and are involved in the induction of EMT in the HepG2 cell line (99). TGF-β-treated M2 TAMs upregulate the expression of T cell immunoglobulin and mucin structural domain-containing protein-3, activate the NF-κB signaling pathway, upregulate the expression of the classical cytokines IL-6, colony stimulating factor 2 (CSF2) and IL-10, and promote tumor migration (100,101). GSK3β is upregulated in macrophages, which in turn activates the NF-κB pathway-mediated direction of M2 TAM polarization, and upregulates the secretion of STAT1, CCL5, IL-6 and CSF2 in macrophages, promoting HCC metastasis and invasion (102). The activation of the STAT3 signaling pathway in HCC cells mediated by the secretion of IL-6 by M2 TAMs is necessary for HCC invasion and metastasis (103). Clinicopathological analysis has revealed that CD163-positive macrophages aggregate around STAT3-positive HCC cells. M2 TAMs activate the STAT3 tyrosine phosphorylation (p-STAT3) signaling pathway through IL-6 upregulation and participate in HCC cell migration and invasion (104,105). The expression of the homologous heterotrimeric transcription factor sine oculis homeobox homolog 1 (SIX1) is increased in tumor tissue TAMs (106). Co-culture of macrophages with high SIX1 expression with HCC cells increases the levels of p-STAT3, HCC cell migration and invasion, and the extent of EMT. Specifically, in HCC, macrophages alter the transcriptional level of the p65 gene in the nucleus, increase IL-6 secretion, activate the STAT3 signaling pathway in HCC cells, and lead to an increase in MMP-9 expression through upregulation of SIX1 expression, regulating the migration, invasion and EMT of HCC cells (107). A study has demonstrated that MMP-9 degrades the extracellular matrix (ECM) at an early tumor stage and is a classical pro-metastatic invasive factor for HCC cells (108). Furthermore, M2 TAM exosomes participate in HCC metastasis by activating the TLR4/STAT3 signaling pathway (109,110).

Exosomes work as a messaging link for TAMs to regulate the extent of the invasive migration of HCC cells (111). The ratio of CD11b/CD18 is increased in M2 macrophage exosomes. CD11b/CD18 upregulates MMP9 expression in HCC cells, and mediates the metastatic invasion of HepG2 and Huh7 cells (112). Integrins are proteins that regulate the function of HCC cells. Upregulation of integrins in HCC regulates tumor cell proliferation, metastasis, invasion and survival (113). S100A4 secretion by M2 TAMs regulates ECM remodeling in the TME via the ERK signaling pathway (65). The heterogeneous nuclear ribonucleoprotein A1 in M2 TAMs is a component of miR-23a-3p-containing exosomes, and the formed exosomes mainly act on the targets PTEN and tight junction protein 1 in HCC, leading to the secretion of CSF2, VEGF, granulocyte-colony stimulating factor, monocyte chemoattractant protein-1 (MCP-1) and IL-4, accelerating the EMT process in HCC cells (114).

By contrast, miR326 expression is abundant in the exosomes of M1 TAMs and is involved in the tumor-suppressive effects of M1 TAM exosomes. Specifically, HCC cells receive exosomes from M1 macrophages, and miR326 in the exosomes decreases the expression levels of twist family BHLH transcription factor 1 in HCC cells, hindering the metastatic invasion of HCC (115). lnc-Ma301 expression is higher in M1 TAMs than in M2 TAMs, but lower in HCC tissues. lnc-Ma301 is an inhibitor of the invasive migration of HCC cells in mouse HCC lung metastasis. lnc-Ma301 regulates the Akt/Erk1 signaling pathway downstream proteins by silencing caprin1, and inhibits the migration and invasion HCC cells, as well as the EMT (116). M2 TAMs secrete the cytokines IL-6, TNF-α, CSF2 and intercellular cell adhesion molecule-1 to mediate the Akt/proline-rich Akt substrate of 40 kDa signaling pathway and activate EMT in interstitial cells of Cajal. Akt inhibitor VII has a reversal effect on the aforementioned cytokines, upregulating EMT markers (117). This mechanism provides a potential approach for Akt-targeted therapy, which might turn into a main method to cure HCC. Fig. 2 shows the mechanistic process of the crosstalk of HCC cells and TAMs.

Indirect control of HCC development by TAMs

Role of TAMs in promoting angiogenesis of HCC

The angiogenesis of new blood vessels is necessary to supply oxygen and nutrition, and remove carbon dioxide and other metabolic waste during the proliferation and migration of tumor cells. The formation of new blood vessels and recruitment of pre-existing vessels are two main causes of vascular hyperplasia in the tumor (118). There is a positive relationship between the increase in macrophage numbers and high micro-vessel density. The recruitment of TAMs stimulated by hypoxia is associated with a wide range of roles in the formation of new blood vessels. After stimulation by hypoxia, TAMs secrete vascular endothelial growth factor, which binds to receptors, and promote the movement and direct migration of endothelial cells (114). Subsequently, TAMs, regulated by HIF-1α in the hypoxic lactate-enriched TME, express multiple genes for the fine-tuning of the polarized M2 macrophage phenotype and secrete several proangiogenic factors to activate pro-angiogenic signaling pathways in HCC (119). M2 TAMs produce miR-23a-3p-containing exosomes that are taken up by HUVECs, leading to their migration to form new blood vessels (114).

M2d macrophages are the main pro-angiogenic phenotype in HCC. They form numerous new blood vessels through the secretion of several vasoactive substances, such as VEGF-A, EGF, placenta growth factor (PlGF), TGF-β, TNF-α, IL-1β, IL-8, CCL2, CXCL8, CXCL12, MMP2, MMP9, MMP12, thymidine phosphorylase (TP), cathepsins and Pas (120,121). HCC TAM infiltration into tissues inhibits the antiangiogenic factors endostatin and angiostatin, while it upregulates the expression of angiogenic factors. VEGFs serve a dominant role in promoting tumor angiogenesis (122,123). The VEGF family is comprises five members: VEGF-A, VEGF-B, VEGF-C, VEGF-D and PlGF. Increased lncRNA-cyclooxygenase-2 (COX2), H3-AS1 and MALAT1 levels are present in malignant liver tumors (124). These lncRNAs are the messengers for the increase in the proportion of M2-like TAMs, and the density and growth of tumor vessels (125). TAMs activate the STAT3 signaling pathway to stimulate VEGF expression in liver cancer cells through the regulation of B7-H3 expression (126). VEGFs are involved in tumor angiogenesis after being secreted, and bind to the cognate receptors VEGFR-1, VEGFR-2, VEGFR-3 and recombinant neuropilin protein (127). The proteolytic modification of the ECM, contributing to the degradation of the vascular basement membrane, relies on MMPs, TP, cathepsins and PA, which promote liver fibrosis and angiogenesis (128). Urokinase-type PA and TP found in the TAM tumor-promoting secretions increase the perivascular space (129).

In addition to the classical angiogenic factor VEGF, the TME contains other angiogenetic factors, including the S100 protein family, semaphorin (SEMA) family, COX-2, SPP1, secreted protein acidic and rich in cysteine, chitinase-like proteins found mainly in cartilage and connective tissues [chitinase 3-like-2 and chitinase 3-like-1 (YKL-40)] and Tie-2. These cytokines mainly target TAMs, increase their infiltration in HCC and are involved in the increase of proangiogenic factor expression (130-132). For example, S100 calcium binding protein, frequently identified at the site of tissue injury, is associated with the recruitment of inflammatory cells, regulating the inflammatory microenvironment and remodeling vessels (133). In addition, inhibition of the SEMA3A/neuropilin 1 signaling pathway inhibits the high level of TAM infiltration response to the hypoxic microenvironment in HCC (134). Furthermore, the increase in the number of TAMs results in increased YKL-40 protein expression in HCC precancerous tissue sections (135). Tie-2-expressing TAMs, a special subtype of TAMs, are involved in the sustenance of the tumor vasculature by regulating the Ang-2/Ang-1 ratio, thus exerting pro-angiogenic effects (136). However, VEGF-A takes part in the functional transformation of the pro-angiogenic effects of Ang-2 that leads to the degeneration of blood vessels (137).

C-C motif chemokine receptor 2 (CCR2), the chemokine receptor of CCL2, is essential for the recruitment of TAMs. The inhibition of the MCP-1-CCR2 (CCL2-CCR2) axis reduces the TAM (CCR2+) response and suppresses angiogenesis, controlling the development of HCC. CCR2+ TAMs, which are deprived of most of the properties of M2-type macrophages, are a novel M1-type macrophage. The high level of CCR2+ TAM infiltration in the boundaries of HCC increases the liver blood volume and pathogenic neoangiogenesis (138). In addition, M1 TAMs are involved in the NF-κB, STAT3 and activator protein-1 (AP-1) signaling pathways in HCC cells through the production of inflammatory cytokines (IL-1β and IL-6). M1 TAMs activate an angiogenic response, which is one of the antitumoral proinflammatory effects (139). Fig. 3 shows the effect of TAMs on angiogenesis in HCC. Table II shows the key differences in the molecular mechanisms by which TAMs directly vs. indirectly regulate HCC progression.

Table II

Direct and indirect molecular mechanisms by which tumor-associated macrophages affect HCC development, and the cell types regulated.

Table II

Direct and indirect molecular mechanisms by which tumor-associated macrophages affect HCC development, and the cell types regulated.

First author/s, yearDirect/indirect mechanismsCellsMolecular signals involved in regulationEffects(Refs.)
Ganjalikhani Hakemi et al, 2020; Liu et al, 2022DirectHCC cells TGF-β/Tim-3/NF-κBPromoting tumor migration(101,103)
Wei et al, 2021 S100A9/AGBR/NF-κBDevelopment of malignancies(68)
Qi et al, 2022S100A4/ERKPromoting the expansion of CSCs andtumorigenesis(65)
Li et al, 2020 S100A4/RAGE/β-cateninDevelopment of new tumor suppressors(66)
Chen et al, 2019 TNF-α/Wnt/β-cateninEMT and stemness features of HCC(69)
Liu et al, 2023TGF-β1/SmadHCC stem cell-like characteristics(73)
Keawvilai et al, 2024LAL/CD36/ACBAInactivation of the HCC tumor growth-promoting axis(85)
Wu et al, 2021CD11b/CD18/MMPInvasive migration of HCC cells(113)
Wan et al, 2014
Dong et al, 2022;
SIX/P65/IL-6/STAT3/MMP9Regulation of the migration, invasion and EMT of HCC cells(77,106)
Peng et al, 2019 IL-8/JAK2/STAT3/SnailInduction of EMT in the HepG2 cell line(99)
Xu et al, 2022TLR4/STAT3Invasive migration of HCC cells(111)
Yeung et al, 2015CCL22/CCR4Direct activation of EMT in HCC cells(95)
Zhang et al, 2024CXCL2/ITGB1Initiation condition of tumor cells and stem-like properties of OV6 CSCs(64)
Qu et al, 2022; Ji et al, 2023CXCL8/miR-17HCC cell proliferation(90,91)
Ye et al, 2020lncRNA H19/miR-193b/MAPK1EMT pathway and the conversion of non-CSCs to CSCs(74)
Wu et al, 2020 hsa-circ-004658/miRNA-99b-5p/JAM3Apoptosis of liver cancer cells(87)
Lu et al, 2023 miRNA-32a-3p/PTEN/TJP1EMT process in HCC cells(114)
Bi et al, 2023PI3K/AKT/mTORHCC cell proliferation(80)
Cai et al, 2020eUB/AKT/mTORHCC cell apoptosis(81)
Chen et al, 2021 PAI-1/FAS/FASLGAntiapoptotic abilities of liver cancer cells(19)
She et al, 2023LATS1/HippoHCC cell apoptosis and mitochondrial damage(82)
Wang et al, 2022 METTL14/cirfut8/YTHBC1HCC progression(92)
Cheng et al, 2021IndirectHUVECs B7-H3/STAT3/VEGFPromoting tumor angiogenesis(126)
Lu et al, 2023miRNA-23a-3pMigration of HUVECs(114)
Duran et al, 2021Tie-2/Ang-1Degenerati on of blood vessels(137)
Xiang et al, 2023 IL-6/NF-κB/STAT3/AP-1Angiogenic response(139)
Wen et al, 2023Immune cells OIT3/NF-κB/PD-L1CD4+ and CD8+ cell infiltration(148)
Wang et al, 2023 MyD88/CX3CR1/CX3CL1Therapeutic efficacy of anti-PD-1 antibodies in HCC(149)
Lu et al, 2021CD39/CD73CD8+ cell activity inhibition(155)
Szefel et al, 2019PGE2/EP2Modulating PD-1 levels(157)
Szefel et al, 2019PGE2/EP4(157)
Wang et al, 2023 MISP/IQGAP/STAT3Upregulati on of the levels of PD-L1 secretion(159)
Goswami et al, 2021 CCL17/CCL18/CCL5/CCL20Recruitment of T helper 2 cells(160)
Liu et al, 2024Arg-1/IL-10T cell depletion and NK cell inactivation(141)
Liu et al, 2022TGF-β/PGE2/IDOCytotoxicity and killing effect of NK cells on tumor cells(162)
Ai et al, 2024CXCL2/CXCR4Recruiting B cells(84)

[i] circ, circular RNA; CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; HCC, hepatocellular carcinoma; lncRNA, long non-coding RNA; miR/miRNA, microRNA; NK, natural killer; PD-1, programmed cell death protein-1; PD-L1, programmed death ligand 1.

Role of TAMs in regulating the immune environment of HCC cells

TAMs inhibit effector T cell activation and proliferation through bidirectional effects. Mitochondrial autophagy regulates cellular energy metabolism by changing the mitochondrial DNA copy number (5). Autophagy maintains the basic cellular function of keeping oxidative phosphorylation nutrient levels stable, which is essential for tumor maintenance and progression (140). Mitochondrial function and β-oxidation production are the main energy sources for the pro-tumorigenic effects of Tregs or M2 TAMs. Upregulation of arginase 1 and IL-10 in M2 TAMs enhances mitochondrial autophagy due to polyamines, which in turn enhances the metabolic energy supply of the cell and promotes the formation of an immune-suppressive HCC microenvironment, leading to T cell depletion and NK cell inactivation, thus inhibiting the progression of HCC (141). Liu et al (142) investigated a novel immunotherapeutic mechanism. It was based on a reductive reactive nanoplatform consisting of poly (disulfide amide) and a lipid-poly (ethylene glycol) shell, which repolarizes TAMs to M1 TAMs via IFNγ and sialic acid binding Ig like lectin 15 (Siglec15) small interfering RNA. Siglec15 silencing in the repolarized TAMs enhanced CXCL9 secretion, increased T cell proliferation and infiltration, and created a tumor immunosuppressive microenvironment. Resiquimod (R848) is a novel TLR7/8 agonist, which repolarizes M2 TAMs to M1 TAMs via the TLR7 MyD88-dependent signaling pathway. Cell microparticles (MPs) are extracellular vesicles and AFP is a classical tumor marker for HCC widely used in HCC vaccine-related investigations. Targeting R848 using MPs as vectors has been used for reprogramming of TAMs after the recognition of AFP in M2 TAMs. The use of R848@ M2pep-MPsAFP to improve the immunosuppressive microenvironment is a novel approach for tumor immunotherapy (143).

On the one hand, TAMs positively inhibit cellular immunity by mediating the secretion of multiple cytokines that act directly on CD8+ cells (144). Immune checkpoint ligand programmed death ligand 1 (PD-L1) is the main acting protein that inhibits CD4+ and CD8+ cell infiltration (145). The binding of programmed cell death protein-1 (PD-1) to PD-L1 leads to CD8+ cell depletion, promoting the expression of regulatory molecules such as B7-H4 and Ig inhibitor of the V structural domain of T cell activation in T cells, which effectively attenuates antitumor effects due to CD8+ cell infiltration (146). IL-10 in the TME induces high PD-L1 and PD-1 expression in monocytes (147). Oncoprotein induced transcript 3 is upregulated in M2 TAMs and involved in the activation of the NF-κB/PD-L1 axis (148). Calcyclin binding protein (CacyBP) competes for the E3 ubiquitin ligase binding site on MyD88 in HCC cells, preventing the degradation of MyD88 via the classical E3 ubiquitin ligase siah E3 ubiquitin protein ligase 1, leading to increased C-X3-C motif chemokine ligand 1 (CX3CL1) expression. The activation of C-X3-C motif chemokine receptor 1/CX3CL1 signaling is critical for the infiltration of TAMs in HCC tissues. The inhibition of CacyBP has become an emerging idea to maintain the therapeutic effect of anti-PD-1 antibodies in HCC (149). In addition, mucosal-associated invariant T (MAIT) cells are MR1-restricted, innate-like T cells. There is mutual crosstalk of CD163 TAMs and MAIT cells via cell contact and a PD-L1-dependent mechanism. PD-L1 blockade and depletion of CSF-1R TAMs were involved in the functional inhibition of MAIT cells in an HCC mouse model (150).

TGF-β1, IL-10 and prostaglandin E2 (PGE2) block Th1 cell differentiation, and CD4+ and CD8+ cell infiltration to exert an antitumor effect (151). Th1 cells assist CD8+ cells to participate in antitumor cellular immunity (152). In vivo and in vitro experiments have demonstrated that M2 TAMs depleted the already activated CD8 T cells, reduced the secretion of the related inflammatory factors IFN-γ and granzyme B in T cells, and activated the immune escape and immunosuppression in HCC (153). Adenosine in the HCC TME produced by the ATP-adenosine pathway is a CD8+ cell exhaustion promoter that interferes with CD8+ cell proliferation. CD39, secreted by M2 TAMs, is the key enzyme for the activation of the ATP-adenosine pathway (154). CD39 together with CD73 secreted by HCC cells increases the proportion of ATP converted to adenosine and is involved in CD8 T cell activity inhibition (155). Activation of the PGE2/prostaglandin E receptor 2 and PGE2/prostaglandin E receptor 4 signaling pathways modulates PD-1 expression during CD8+ cell infiltration, leading to immune tolerance in the TME (156). T cells interact with tumor antigens through the T cell receptor ζ, which is associated with L-arginine (157). M1 TAMs express iNOS and M2 TAMs express arginase 1, which metabolizes arginine to nitric oxide and urea (158).

On the other hand, TAMs exert antitumor effects on T cells through the interaction with other relevant cells in the TME. In addition to the immunosuppressive effect of PD-L1 secreted directly by TAMs, these cells also activate the mitotic spindle positioning/IQ motif containing GTPase activating protein 1/STAT3 axis in HCC cells, upregulate the levels of PD-L1 secretion and promote the immune escape of HCC (159). Regulatory T cells can be divided into naturally occurring natural regulatory T cells (nTregs) and adaptive regulatory T cells. TAMs upregulate IL-10 and TGF-β expression, and promote the expression of the chemokines thymic and activating regulatory chemokine (CCL17), CCL18, CCL5, macrophage inflammatory protein 3α (CCL20) and small inducible cytokine subfamily A (Cys-Cys) to recruit T helper 2 cells, induced Tregs and nTregs (160). Tregs migrate toward tumors and antagonize CD8+ cell-mediated antitumor cellular immune functions (161). High protein expression levels of TGF-β, PGE2, indoleamine 2,3-dioxygenase and B7 in TAMs reduces the migration, proliferation and apoptotic ability of NK cells, reduces IFN-γ and TNF-α expression in the TME, and reduces the cytotoxicity and killing effect of NK cells on tumor cells (162). In addition, CAFs recruit TAMs from the peripheral blood through the macrophage migration inhibitory factor/CD74 axis and induce differentiation of TAMs into CXCL2+ TAMs via the activation of SMAD3. This was demonstrated in vivo and in vitro, and the binding was strong enough to explain the activation of TAMs in relation to CAFs (19). Activated TAMs possess the ability to recruit B cells via the CXCL12/CXCR4 signaling pathway and enhance cytotoxic T cell depletion through the expression of PD-L1 regulated by CXCR4 (84). Fig. 4 shows how TAMs enhance immune protection against HCC by regulating the function of other immune cells.

Treatment of HCC targeting TAMs

TAMs are an important type of innate immune cells in HCC. Studies have identified an inextricable relationship among polarization, recruitment of TAMs and tumor development (163-167). Therapies targeting TAMs effectively inhibit the invasion of HCC and the proliferation of tumor cells. Targeted TAM therapy is mainly carried out as follows: i) Controlling the number of TAMs in HCC, preventing the generation of the HCC TME by reducing the infiltration of TAMs and their polarized phenotypes; ii) restoring the tumor-resistant effects of TAMs by altering the function of TAMs in the HCC TME, activating the antitumor mechanism and the synthesis of its products in TAMs; and iii) inhibition of immune escape in HCC. It has been confirmed that HCC uses the 'do not kill me' mechanism to evade the phagocytosis of tumor cells by TAMs. Therefore, tumor cells can be eliminated by cutting off the signaling pathway of HCC that interferes with the phagocytosis of TAMs (3,47,119,168-171). Fig. 5 shows the direction of treatment options targeting TAMs in HCC.

Targeting the infiltrated TAMs and their polarized subtypes Removal of TAMs that are already present

The control of HCC by the specific depletion of TAMs is the most direct immunotherapeutic approach to target TAMs. Sorafenib is a common therapeutic agent used to treat HCC (172). CSF-1, stromal cell-derived factor 1α (SDF-1α) and VEGF are important chemokines for the proliferation of macrophages. Studies have indicated that plasma CSF-1 expression is increased after sorafenib treatment. CSF-1 activates macrophages, promoting macrophage-tumor interactions, ultimately leading to tumor progression. By contrast, sorafenib in combination with chlorolipids or zoledronic acid inhibits macrophage infiltration (173,174). Tumor progression and tumor angiogenesis are inhibited after the depletion of macrophages by chlorolipids or zoledronic acid. Furthermore, the combination of these drugs controls lung metastasis of HCC (175). In vivo experiments were only performed using mice, and the in vitro experiments were performed using RAW264.7 mouse macrophages, while human macrophages such as THP-1 cells were not used; thus, the results are less representative than those using human macrophages (176). Microtubule depolymerization is specifically induced by 2'-carboxy-D-arabinitol 1-phosphate (CA1P) through the inhibition of the p150-AKT-GSK3β signaling pathway (177). Downregulation of Akt phosphorylation activates GSK-3β, which in turn inhibits the Wnt/β-catenin signaling pathway in HepG2 cells (178). This signaling is abnormally activated during HCC development; thus, CA1P induces apoptosis in HCC cells in vitro and in vivo. In addition, CA1P induces apoptosis in tumor cells and TAMs via the same pathway and alters the TME by disrupting the crosstalk between tumor cells and tumor-associated immune cells. The effectiveness of CA1P in the treatment of HCC and the effect of CA1P on macrophages are mentioned in a study by Mao et al (179); however, to the best of our knowledge, the relationship between the two has not been confirmed in any particular study. Since TAMs have multiple polarized subtypes in HCC, their widespread clearance leads to an imbalance in immune regulation. Current therapeutic regimens targeting the removal of TAMs fail to specifically target M2 TAMs, resulting in the unavoidable removal of antitumor M1 TAMs (180).

Inhibition of monocyte/macrophage recruitment

CCL2 is a feature of inflammatory monocytes, and its only known receptor CCR2 is expressed in hepatic macrophages. CCL2-CCR2 signaling is involved in the infiltration of TAMs and the polarization of M2 TAMs. HCC can be treated at a biomacromolecular level by targeting CCR2 signaling blockade. A natural product called 747 has been found to act as a CCR2 antagonist. It reduces macrophage infiltration by activating the CD8 T cell population (181). Genipin, also derived from herbal origins, is a natural cyclic enol ether terpene glycoside commonly used in clinical practice for the treatment of inflammatory diseases and heat-associated HCC. Genipin directly binds to PPARγ and activates PPAR signaling in postoperative hepatic macrophages in mice (182,183). Activated PPARγ triggers the degradation of p65/RelA by acting as an E3 ligase (184). p65/RelA is a transcription factor for CCL2, and its inhibition results in the blockade of the CCL2-CCR2 signaling pathway. These effects in turn inhibit the recruitment of the hepatic macrophage population, as well as macrophage pro-inflammatory cytokine and chemokine expression, and postoperative HCC recurrence, resulting in a reduction in postoperative patient survival (5). Cenicriviroc (CVC) is a novel oral dual CCR2/CCR5 antagonist. CVC reduces macrophage infiltration in the liver of patients with autoimmune liver disease. CVC treatment blocks the release of LPS-induced macrophage cytokines. The oral administration of genipin after CVC treatment in an experimental mouse model of HCC reduced the risk of lung metastasis of HCC cells. In addition, CVC prevents pro-inflammatory signaling and macrophage activation in the liver, blocking the TAM polarization pathway in the treatment of HCC (185). A total of 10 CCL2 inhibitors (MK0812, PF04634817, PF04136309, INCB3344, cenicriviroc, JNJ2714149, CCR2-ra-[R], CAS-445479-97-0, RS504393 and RS102895) are commercially available at present and are used in clinical HCC therapy (186). Pharmacological experiments have confirmed that MK0812 inhibits tumor metastasis in humanized mice, with the lowest IC50 (1.73 nM) being the most potent human CCR2 inhibitor. Oral administration of MK0812 in human CCR2B knock-in mice did not result in weight loss or other serious side effects (187). The cytidine analog gemcitabine (GEM) is a first-line chemotherapeutic agent in the treatment of parenchymal tumors, functioning by inhibiting DNA replication. However, GEM has some limitations such as poor delivery and susceptibility to cytidine deaminase-induced degradation, which hamper its clinical therapeutic application. The poor delivery of GEM was addressed by constructing GEM-conjugated polymers (PGEMs) showing high tumor penetration in several pancreatic ductal adenocarcinoma models. PGEM has a reduced particle size, being a small and acceptable polymer drug complex. A study has demonstrated that PGEM induces double-stranded DNA damage in tumor cells and activates the cyclic GMP-AMP synthase (cGAS)-interferon gene-stimulating factor (STING) pathway (188). STING is an endoplasmic reticulum transmembrane protein that activates CCL2 and CCL7 expression, leading to a marked increase in the percentage of MDSCs and TAMs, thus regulating tumor innate immunity. PF-6309 is an optimal CCR2 inhibitor for loading PGEM micelles. It was also evident that free PF-6309 and PGEM/PF-6309 treatments led to a decrease in the percentage of MDSCs and TAMs, the latter also leading to a decrease in the percentage of M2-type macrophages and an increase in the percentage of M1-type macrophages and the M1/M2 ratio, suggesting that macrophages infiltrating the tumors are polarized from a tumor-promoting to a tumor-suppressing phenotype (189). Hypoxic conditions in HCC lead to the upregulation of SDF-1α expression, leading to the recruitment of M2 macrophages and the promotion of tumor progression (19,190). The expression levels of CXCR4 and VEGF in TAMs, as well as the expression levels of M2 TAM markers CCL22 and arginase 1 are more than 2-fold higher in TAMs after sorafenib treatment than in TAMs without sorafenib treatment. AMD3100 is a classical inhibitor of the SDF-1α receptor (CrXrC receptor type 4 or CXCR4), which blocks the SDF-1α/CXCR4 axis that arrests the recruitment of TAMs, prevents polarization to an immunosuppressive microenvironment after sorafenib treatment, inhibits tumor growth, reduces lung metastasis and improves survival. AMD3100 alone or in combination with sorafenib reduces the expression levels of M2 type markers in TAMs but does not affect the expression of M1 type markers. This combined therapeutic approach exerts an anticancer effect not through reprogramming M2 TAMs but by cutting off the recruitment channel of M2 TAMs (191). CXCL17 is a selective inhibitor of CXCR4 signaling with low potency. Binding of CXCL17 to neuropilin-1, a VEGFR2 co-receptor containing glycosaminoglycans, blocks endogenous CXCL12 binding to CXCR4, thereby inhibiting ligand binding to NanoLuc/CXCR4 and inhibiting β-arrestin 2 recruitment to CXCR4 (192). The differentiation of human M2 macrophages results in the upregulation of the expression levels of P2Y11, a G protein-coupled ATP receptor that activates the IL-1 receptor in a cyclic AMP-dependent manner, upregulating the expression levels of CECR7 through increased EGFR expression, along with the upregulation of CXCR4 expression. The antagonist NF340 is a specific inhibitor of P2Y11, and this inhibition controls macrophage infiltration by suppressing CXCR4 expression (193).

Restoration of tumor-resistant effects of TAMs
Inhibition of the polarization pathway from monocytes to M2 TAMs

Some chemotherapeutic agents, such as Adriamycin (Dox) and oxaliplatin are effective in tumor immunotherapy regimens. These drugs applied in combination therapy are effective in treating the development of HCC (194,195). Manganese (Mn2+) is an important intracellular ion that activates cGAS-STING signaling. Cell membrane-derived hybrid nanovesicles (Mn2+/Dox loaded-hybrid vesicles) have been constructed and applied in antitumor therapy. Cell-membrane vesicles were derived from M1-like macrophages and fused with liposomes carrying Mn2+ and Dox. Finally, the fused vesicles were modified with CXCR4-binding peptide to target macrophages highly expressing CXCR4. Furthermore, Mn2+-mediated activation of cGAS leading to an increased production of cyclic GMP-AMP and activation of STING resulted in the upregulation of IRF3 transcription. i-type interferon and cytokines for antitumor immunity were controlled by STING, upregulating the expression levels of CD80 and CD86, leading to M1 phenotypic polarization (196). CSF2-derived macrophages preferentially produce IL-6, IL-12 and TNFα in response to lipopolysaccharide, whereas CSF1-derived macrophages produce IL-10 and CCL2 but not IL-12. CSF-1R signaling promotes a tumor-promoting macrophage phenotype, whereas its inhibition polarizes TAMs to an antitumor phenotype. The inhibition of CSF1/CSF-1R signaling depletes M2 TAMs to reduce tumor cell infiltration and reprogram the TME (197). CCAAT-enhancer-binding protein α (C/EBPα) is known for its ability to inhibit the tumor polarization of M2 macrophages; however, its role in regulating immunosuppressive myeloid cells is unclear. Application of therapeutic agents associated with C/EBPα mediated by small activating RNA represents a good immunotherapeutic option. The transcription of the CEBPA gene is specifically regulated by CEBPA-51. The encapsulation of CEBPA-51 in SMARTICLES liposomal nanoparticles composed of MTL-CEBPA results in the promotion of C/EBPα expression. In addition, the upregulation of CEBPA expression in monocytes after the action of MTL-CEBPA leads to an increase in the synthesis of PGE2, which is a potent immunosuppressive mediator, resulting in the rapid downregulation of genes and proteins involved in MDSC inhibitory activity, as well as the activation of classical monocytes and granulocytes. Furthermore, MTL-CEBPA and sorafenib induce the transition from M2-type polarized TAMs to M1-type polarized TAMs (198).

Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase that promotes M2 polarization of macrophages through the regulation of multiple signaling pathways, including the PI3K/Akt/JAK/STAT3 and p38/JNK/ERK signaling pathways. Phellinus linteus is an anticancer herbal medicine. Upon binding to FAK, the combination of DBL and hispolon blocks the action of FAK and inhibits cell proliferation (199).

PLX3397 is a competitive inhibitor targeting CSF-1R and the affinity for CSF-1R tyrosine kinase is higher than that of CSF-1R. Bone marrow-derived monocytes are stimulated to polarize toward an M2-like or M1-like phenotype by either CSF1 or CSF2, the latter protecting macrophages from PLX3397 treatment; thus, TAMs from PLX3397-treated tumors show an M1-like phenotype, and PLX3397 inhibits tumor growth without depleting TAM infiltration in vivo (200).

Blocking the change of already polarized M1 TAMs to M2 TAMs is a novel line of research. CSF-1 regulates the repolarization process from M1 macrophages to M2 macrophages through MAPK/ERK/AP-1 signaling pathway activation. CSF-1 mediates the high expression of c-Jun, which belongs to the AP-1 family proteins and is involved in the repolarization process of M1 macrophages with the assistance of NF-κB (201). The triggering receptor expressed on myeloid cell (TREM) family mainly exerts effects in the regulation of the inflammatory response through the receptor form. TREM2 regulates macrophage polarization, and its inhibition leads to the activation of the PI3K/Akt/NF-κB axis and the increase of CXCL3 secretion in macrophages. High TREM2 expression is a hallmark of the repolarizing metabolic reprogramming of M1 macrophages to M2 macrophages (202,203). Echinacoside is a phenyl ethanol glycoside involved in hepatoprotection, anti-inflammation, neuroprotection and tumor therapy. A study found that it reduced TREM2 expression in HCC, while activating PI3K/Akt signaling, acting as an antitumor agent (204). Oxaliplatin is a commonly used clinical anticancer drug. TREM2-IN-1 (OPA), a platinum (IV) complex made from oxaliplatin and artesunate, is a potent TREM2 inhibitor. OPA is also involved in the cytotoxicity induced by oxidative damage to nuclear DNA. It serves an antitumor role in both chemotherapy and immune activation (205). The use of Fc structural domain effector-enhanced antibodies against TREM2 demonstrated that the pro-tumor function of TAMs was reversed in the TME after treatment (206-208). Furthermore, the humanized anti-TREM2 monoclonal antibody PY314 is already used in a clinical trial (209).

Reprogramming of TAM polarization

The use of cellular autophagy to block the immunoregulation of HCC involving TAMs has received unanimous confirmation from experts and scholars (163-165,172,173,180). Baicalin inhibits in situ HCC growth through the reprogramming of macrophages. This process requires the involvement of RelB and p52, which are highly expressed in M1-like macrophages compared with M2 TAMs. Baicalin is an activator of the autophagic degradation of TNF receptor associated factor (TRAF)2 in TAMs, and TRAF2/TRAF3 has a limiting effect on intracellular IKKα/RelB/p52 signaling. The indirect activation of the RelB/p52 signaling pathway by baicalin causes the expression of the RelB/p52-specific target genes CXCL12 and CCL9, leading to higher expression of macrophage-secreted IL-6 and TNF-α, which are cytokines with typical pro-inflammatory effects, and causes transformation of M1 TAMs to M2 TAMs (210). Activation of the NF-κB signaling pathway for macrophage autophagy therapy is widely recognized since it is involved in M1 macrophage polarization. A study has also identified the role of the tumor vaccine Lmdd-MPFG in targeting TAMs. The activation of the NF-κB signaling pathway by the Lmdd-MPFG vaccine induces M1 polarization through macrophage autophagy (211). Sirtuin 1 (SIRT1) is a deacetylase that regulates transcriptional silencing and cell viability. Overexpression of SIRT1 in macrophages enhances the phosphorylation of p65 and IKK, and promotes M1 macrophage polarization. IKKβ inhibition increases tumor suppressor polarization of macrophages (212).

lncRNA COX-2 expression is higher in M1 macrophages than in M2 macrophages. Its inhibition in turn inhibits macrophage polarization to the M1 type, decreases the ability of M1 macrophages to inhibit HCC cell proliferation and increases the ability of M2 macrophages to promote proliferation (213).

Targeting multiple receptors is an essential part of the treatment of HCC, and relevant therapeutic regimens around TAMs take this into account. Use of TLR inhibitors for the treatment of HCC has been widely emphasized (210,212,214). The role of TLR in the activation of the non-specific immunity of the body cannot be ignored. The non-specific immune system promotes the inflammatory response by recruiting immune cells such as macrophages and mast cells to secrete inflammatory factors (215). The most important protein for mitochondrial fission, dynamin-related protein 1, is involved in HCC progression by regulating cell survival and metastasis. The dynamics of mitochondrial fusion and fission are necessary for mitochondrial DNA (mtDNA) distribution and mitochondrial homeostasis. Increased mitochondrial fission induces cytoplasmic mtDNA stress in HCC cells, which in turn promotes CCL2 secretion via the TLR9-mediated NF-κB signaling pathway, leading to TAM infiltration in HCC tissues (216). In HCC treatment, motolimod (TLR7 agonist), GS9620 (TLR8 agonist) and R848 (resiquimod; dual TLR7/8 agonist) produce an M1 increase. Certain dextran nanoparticles have natural macrophage affinity and are effective carriers for targeting TAMs with drugs. Among them, β-cyclodextrin has a similar chemical composition to linear dextran, and easily activates the phagocytic effect of macrophages to exert drug effects (214,217-221). Cyclodextrin nanoparticles (CDNPs) have a high macrophage affinity and a high drug-carrying dose. After combining with CDNPs, the so-called nano R484 inhibitor enhances the reprogramming ability of TAMs (217). In addition, PI3Kγ is a classical therapeutic target for reprogramming TAMs and is a key signaling molecule required for macrophage accumulation in inflammation. PI3Kγ blocks pro-inflammatory responses of stimulating macrophages. Macrophages deficient in PI3Kγ exhibit a reduced ability to respond to immunotropism through G protein coupling (222), upregulation of M1 polarization-associated genes and proteins, and downregulation of M2 polarization-associated cytokine expression. PI3Kγ promotes immunosuppression through the activation of mTOR/S6Kα/C/EBPβ signaling and the inhibition of NF-κB (223). Indirect promotion of Th1 cells and cytotoxicity reduce immune responses (224). A pan-PI3K inhibitor has recently been identified as an enhancer of pro-inflammatory cytokine transcription in TAMs (3). The metabolism of substances in TAMs serves an integral role in the polarization of cellular phenotypes, and the inhibition of certain pathways of macrophage substance metabolism activates the reprogramming of M2 TAMs (225). Macrophages are a relevant source of active insulin-like growth factor (IGF-1). IGF-1 is a key growth factor for macrophages to drive the growth of HCC cells from macrophages. Sorafenib inhibits the release of IGF-1 (226). IGF-binding protein 4 (IGBP-4) binds IGF-1 to inhibit its activity (227). Low expression of IGBP-4 in polarized macrophages leads to active secretion of IGF-1, which ultimately promotes the proliferation of HCC cells. Targeted inhibition of the IGF/IGF-1R signaling axis is a promising approach to modulate the HCC environment (228). Receptor interacting serine/threonine kinase 3 (RIPK3) is downregulated in HCC-associated TAMs. The extent of the distribution of lipid droplet deposition is increased in TAMs after RIPK3 knockdown. Reactive oxygen species/caspase 1 signaling mediates the inhibition of PPAR expression by RIPK3. The lack of RIPK3 in TAMs results in an increase in the expression levels of PPARα and PPARγ, which regulate fatty acid metabolism, promote TAM infiltration and M2 polarization, and facilitate the development of HCC. The upregulation of RIPK3 reverses TAM polarization and attenuates HCC. The RIPK3 inhibitor GSK872 and fatty acid oxidation blockers are two factors mediating reprogramming of M2 TAMs (229).

Regulation of TAM products

TAM products are protein molecules in TAMs that can act in the tumor microenvironment to regulate tumor development, such as TGF-β and Wnt1. TAMs accelerate tumor progression by releasing tumor growth factors. LPS-induced activation of PI3K/Akt increases the levels of miR-101, which in turn binds to dual specificity phosphatase 1 (DUSP1) mRNA and directly represses its expression (230). DUSP1 is a negative regulator of the activity of MAPKs. MAPKs are highly conserved serine/threonine protein kinases that include ERK, JNK/stress-activated protein kinases and p38 (231). Sorafenib inhibits PI3K/Akt activation and decreases miR-101 expression, and subsequently inactivates MAPK via the induction of DUSP1 expression in HCC. Sorafenib inhibits the release of TGF-β and CD206 in M2 macrophages (232). TGF-β is a key growth factor for macrophage-driven HCC cell proliferation and metastasis (217). Wnt/β-catenin signaling is a classically conserved signaling pathway, and its activation is associated with the degree of HCC. M2 macrophages specifically release Wnt1, which stimulates the activation of β-catenin signaling in tumor cells, leading to HCC cell proliferation and malignant transformation in a Wnt1-dependent manner. Bufferin targets M2 macrophages and inhibits HCC growth by specifically blocking Wnt1/β-catenin signaling (222). Isoproterenol stimulates the secretion of microvesicles by TAMs, which deliver miR-142-3p from macrophages to HCC cells. miR-142-3p is taken up by HCC cells and directly inhibits their viability, proliferation and invasiveness in vitro by downregulating Rac family small GTPase 1 (224). Compound kosher injection (CKI; also known as Yanshu injection) is used in clinical practice in the treatment of a variety of solid tumors, including HCC, gastric carcinoma, breast carcinoma, lung carcinoma and colorectal carcinoma. CKI increases the therapeutic effect of low-dose sorafenib. It also reduces the M2 TAM levels in the TME and increases the ratio of M1 TAMs to CD8 T cells by targeting TNF receptor superfamily member 1A and its downstream NF-κB p65 and MAPK p38 signaling cascades, reducing tumor recurrence and triggering an effective antitumor memory response against HCC (3).

Inhibition of immune escape in HCC

HCC avoids phagocytosis by TAMs mainly through the 'do not eat me' signaling pathway. Signal-regulated proteins (SIRPs) have extracellular immunoglobulin-like domains, and SIRPα and SIRPγ bind to the widely expressed cell-surface transmembrane glycoprotein CD47 (also known as integrin-associated protein). The binding of SIRPα to macrophages reduces the production of the pro-inflammatory cytokine TNF and reduces polarization in the M1 direction. The binding of CD47-SIRPα to macrophages leads to the inhibition of the 'do not eat me' signaling, negatively regulating the activation of macrophages and phagocytosis (233). Activation of SIRPα activates the phosphorylation of tyrosine-based inhibitory motifs of the immune receptor, preventing the recruitment of Src homologous phosphatase (SHP)-1 and SHP-2 phosphatases in cells. Two phosphatases are recruited to prevent myosin-IIa accumulation at phagocytic synapses in macrophages. The disruption of the CD47-SIRPα axis in preclinical models leads to enhanced phagocytosis and tumor reduction, and it is a means of cross-presenting tumor antigens to T cells (234). Two therapeutic options are available based on CD47-SIRPα axis inhibition, anti-CD47 therapy and anti-SIRPα therapy. At present, scholars generally choose anti-CD47 therapy as the main therapy to restore macrophage capacity through the use of surface-engineered exosomes equipped with membrane-bound proteins. Researchers have used exosome models containing SIRPα variants specifically competing for SIRPA binding sites on CD47 on the surface of tumor cells. Newly developed SIRPα variants have an improved affinity for human CD47 compared with wild-type SIRPα. SIRPα exosomes block CD47-SIRPα interactions, inducing increased phagocytosis of various cancer cells by bone marrow-derived macrophages (235). Treatment of macrophages with B6H12 or CD47 monoclonal antibody 400 antibodies, which are two specific CD47 blocking antibodies, enhances phagocytosis of HCC cells and reduces tumor size in mice with ectopic tumors. Furthermore, CD47 inhibition leads to an increase in the migration of macrophages to HCC in vivo, which is more conducive to the phagocytosis of macrophages. CD47 blocking antibodies have great advantages in maintaining the normal physiological functions of the liver, and the use of the antibodies does not cause direct in vivo damage to the liver and normal hepatocyte viability (236). In a recent study, a high affinity CD47 blocker (modified SIRPα D1 structural domain) was added to the inactive IgG Fc region, resulting in the engineered protein evorpacept (also known as ALX148). ALX148 is recognized by macrophages through the active Fc structural domain and binds to its Fcγ receptor, and the resulting CD47 inhibitor targets the CD47/SIRPα signaling pathway, increasing autoimmune processes in the TME. Evorpacept had a good safety profile both when administered alone and in combination with the standard regimen of pembrolizumab and trastuzumab (237). In addition, CD47 expression in tumor cells is positively regulated by STAT3 phosphorylation levels. IL-6 reverses the suppressed CD47 expression in HCC cells after blocking STAT3 and enhances the ability of tumor cells to evade phagocytosis. Tocilizumab, a humanized monoclonal antibody directed against the IL-6 receptor, is used to inhibit the inflammation in rheumatoid arthritis, but it also blocks TAM-mediated anti-phagocytosis, making it a potential drug for HCC treatment (238). Histone deacetylase 6 (HDAC6) is involved in autophagy signaling and degradation of ubiquitinated proteins. It directly inhibits the expression of let-7i-5p, which regulates the expression of thrombospondin-1 (TSP1), which competes with SIRPα. Activation of the HDAC6/let-7i-5p-TSP1 regulatory axis converts CD47-SIRPα to CD47-TSP1 interactions between HCC and macrophages, thereby altering the 'do not eat me' self-protection mechanism of tumors. Therefore, targeting of the HDAC6/let-7i-5p/TSP1 axis could represent a novel immunotherapeutic strategy to treat human HCC in the future (239). B6H12.2 is a CD47 blocking antibody that enhances CD47-SIRPα expression in CCA, thus enhancing the phagocytosis of CCA cells highly expressing CD47 (238). Glypican-3 (GPC3) is one of the most well-characterized HCC-associated antigens. Bispecific antibodies co-acting with GPC3 and CD47 have excellent antitumor effects with minimal toxicity. GPC3/CD47 bispecific antibody readily binds preferentially to bi-antigen-expressing tumor cells, and efficiently blocks SIRPα/CD47 (240). Anti-SIRPα treatment regimens have progressed more slowly compared with CD47 antibody therapy (241). CRISPRed macrophages can be used for cell-based cancer immunotherapy. The CRISPR-CRISPR associated protein 9 (Cas9) nuclease system is commonly used to target modifications of the genome in cells to block gene expression (242). Cationic arginine-coated gold nanoparticles (ArgNPs) bind Cas9 proteins, and CRISPR-Cas9 encapsulated with ArgNPs can knock down SIRPα in RAW264.7 cells, activating phagocytosis and eliminating cancer cells (243). In contrast to therapeutic measures targeting CD47, therapeutic regimens targeting SIRPα are still missing (244).

Drug resistance, efficacy and potential risks

In response to a study of tumor-targeted macrophage therapeutic regimens, current studies have found that the use of CSF-1R may lead to the development of drug resistance (97,150,197,200). The activation of PI3K is involved in the resistance of HCC after treatment with the CSF-1R inhibitor BLZ945. After the treatment with BLZ945, IGF-1 drives the activation of PI3K signaling (245). Therefore, IGF-1R and PI3K may serve as biomarkers for predicting drug resistance. Combined inhibition of IGF-1R and PI3K enhances the efficacy of CSF-1R in treating tumors by reducing drug resistance. Novel advances in CSF-1R antagonistic therapy in combination with other receptor tyrosine kinase inhibitors have been reported (246). The generation of numerous tumors is inextricably linked to the neogenesis of blood vessels in their tissues (247). The complications associated with treatments in patients in whom the angiogenic pathway is blocked are a difficult problem to resolve. Monocytes/macrophages expressing the Ang receptor TIE-2 regulate interstitial angiogenesis in tumors after receiving CXCL12/CXCR4 recruitment messages in combination with Ang (248-250). Targeted therapies are often applied with TIE-2-expressing monocytes/macrophages (TEMs) as a therapeutic focus to avoid the development of complications while determining efficacy. By blocking TEM depletion, targeted therapies, by avoiding angiogenic mechanisms in tumors, avoid issues in human physiological functions triggered by extensive angiogenic inhibition (251). There are potential risks associated with the regulation of angiogenesis in tumors, as hypoxia due to TEM inhibition may activate strong vascular remodeling and maturation in the periphery of the tumor, leading to a failure of the tumor-suppressive effect of the drug (252).

Discussion

The present review focuses on the crosstalk between tumor cells and macrophages; however, to the best of our knowledge, there is no in-depth research finding on the mechanism regulating the crosstalk between macrophages and specific immune cells. Furthermore, information on the joint crosstalk between multiple cells is still lacking, and there is no systematic mechanism to regulate the tumor immune microenvironment. The study of the effect of HCC on the polarization direction of macrophages revealed the possibility that some juvenile macrophages may already have shifted their polarization direction when they are not affected by HCC, a factor mostly ignored by other studies (3-6,8,13,15). At present, TAMs are roughly classified into M1 TAMs and M2 TAMs, but more and more polarized phenotypes of macrophages have been identified, with no clear and distinct boundary between the macrophage subtypes that have been investigated. Furthermore, some TAMs have the characteristics of both the M1 subtype and the M2 subtype of macrophages, suggesting that the balance of the physiological functions of macrophages in HCC should be studied in the future. More and more subtypes of TAMs have been identified, immunotherapy measures targeting macrophages in HCC are too homogeneous at present, and the proportions of macrophage subtypes in tumors of different patients with HCC vary greatly, leading to the conclusion that it is necessary to study the signature biomarkers of multiple subtypes of TAMs, and use these biomarkers for the detection of macrophages in tumors as well as the selection of targeted therapeutic regimens. The TRP ion pathway is a tumor-related signaling pathway that has received increasing attention from experts in tumor targeted therapy (253). The TRP ion pathway is a classical pain target, and high TRP ion pathway activation is present in liver cancer. The specific functions and regulatory factors of the TRP ion pathway have not yet been conclusively determined (254). Thus, the TRP ion pathway in liver cancer should be investigated in the future, potentially representing a novel development direction (255).

Conclusion

Chinese and international scientists have investigated the role of TAMs in the microenvironment of HCC and found that TAMs are an important factor in controlling HCC development. The present review is divided into two parts, focusing on the role of TAMs in the HCC microenvironment: Direct control of HCC cells and indirect promotion of HCC development. M2 TAMs positively regulate the migration, invasion and proliferation of tumor cells through the secretion of related cytokines, but inflammatory factors produced by M1 TAMs also contribute to the formation of blood vessels, leading to the development of tumors.

Existing scientific studies tend to analyze the pro-tumor direction of M2 TAMs and study the effect of TAMs on advanced HCC. Since the polarization of TAMs in the early stages of a tumor is biased towards the M1 phenotype, there is still a gap in the study of the pro-tumorigenic role of M1 TAMs and the repolarization process. After early diagnosis, the inhibition of the early polarization orientation by M1 TAMs can predictably target early-stage HCC compared with the treatment of HCC by inhibiting the pro-tumor effect of M2 TAMs. Early relevant diagnosis and treatment by enhanced polarization of TAMs towards M1 TAMs can improve the high lethality of advanced HCC. Most studies macroscopically replace tumor characteristics with only the tumor cell characteristics, ignoring the characteristics of the immune cells in HCC. Therefore, more studies on signaling crosstalk between TAMs and HCC cells should be carried out to investigate the differences between HCC and other tumor types.

Availability of data and materials

Not applicable.

Authors' contributions

MinY and HY contributed to the preparation, creation and description of the work for publication, in particular writing a first draft (including substantive translation). MinY, HY and HW contributed to presentation of research ideas and the development and formation of overall research objectives. CL, XZ and ZS contributed to the development and design of methods, and created models. WC and HY contributed to the application of statistical, mathematical, computer and other forms of techniques to analyze and integrate research data. XZ, MJ and XX contributed to implementing research and data/evidence collection. MJ, CL and XZ provided research materials, reagents, patients, experimental samples, animals, instruments, computational resources or other analytical tools. MiaY and MJ carried out metadata management, data cleansing and data maintenance (including software code needed to interpret the data) for initial use and subsequent reuse. HW revised the manuscript. MJ carried out review and revision (both pre- and post-publication phases). MinY and HW revised the manuscript. CL and MJ oversaw and led the planning and execution of research activities. CL and XZ assumed responsibility for the management and coordination of the planning and execution of research activities. CL and XZ obtained financial support for this publication project. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

The authors would like to thank Professor Chunhua Liu, Dr Xiaowei Zhang, Dr Mingchun Jiang, Mrs. Guozheng Xu and Mr. Changwei Shi [Pharmacy College, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong, China] for their guidance and assistance during the writing of the draft. The authors would like to thank Mr. Yonglin Lu and Mr. Haochen Yang [School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong, China] for taking care of the team during the writing process. The authors would like to thank Mr. Jiaju Zhang (College of Electromechanial Engineering, Qingdao University of Science and Technology, Qingdao, Shandong, China) for his guidance and encouragement during the drawing process. The authors would also like to thank Professor Hongmei Wang (Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China) who contributed to the formulation of the title and the arrangement of the content at the early stage of writing the manuscript.

Funding

The present study was supported by Projects of Medical and Health Technology Development Program, Shandong Province (grant no. 202303030702), Shandong Province Medical Health Science and Technology Development Plan (grant no. 202105010417), Shandong Provincial Natural Science Foundation (grant no. ZR2021MH151), and Tai'an Science and Technology Innovation Development Project (grant nos. 2020NS267, 2022NS149 and 2022NS174).

References

1 

Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K and Yang JL: Lipid metabolic reprogramming in tumor microenvironment: From mechanisms to therapeutics. J Hematol Oncol. 16:1032023. View Article : Google Scholar : PubMed/NCBI

2 

Xue R, Zhang Q, Cao Q, Kong R, Xiang X, Liu H, Feng M, Wang F, Cheng J, Li Z, et al: Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature. 612:141–147. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Cheng K, Cai N, Zhu J, Yang X, Liang H and Zhang W: Tumor-associated macrophages in liver cancer: From mechanisms to therapy. Cancer Commun (Lond). 42:1112–1140. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Bilotta MT, Antignani A and Fitzgerald DJ: Managing the TME to improve the efficacy of cancer therapy. Front Immunol. 13:9549922022. View Article : Google Scholar : PubMed/NCBI

5 

Tacke F: Targeting hepatic macrophages to treat liver diseases. J Hepatol. 66:1300–1312. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Minciuna I, Taru MG, Procopet B and Stefanescu H: The inter-play between liver sinusoidal endothelial cells, platelets, and neutrophil extracellular traps in the development and progression of metabolic dysfunction-associated steatotic liver disease. J Clin Med. 13:14062024. View Article : Google Scholar

7 

McQuiston TJ and Williamson PR: Paradoxical roles of alveolar macrophages in the host response to Cryptococcus neoformans. J Infect Chemother. 18:1–9. 2012. View Article : Google Scholar

8 

Yao Z, Bai R, Liu W, Liu Y, Zhou W, Xu Z and Sheng J: Activation of angiogenin expression in macrophages by lipopolysaccharide via the TLR4/NF-κB pathway in colitis. Acta Biochim Biophys Sin (Shanghai). 56:857–865. 2024.PubMed/NCBI

9 

Murray PJ: Macrophage Polarization. Annu Rev Physiol. 79:541–566. 2017. View Article : Google Scholar

10 

Mehla K and Singh PK: metabolic regulation of macrophage polarization in cancer. Trends Cancer. 5:822–834. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Locati M, Curtale G and Mantovani A: Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 15:123–147. 2020. View Article : Google Scholar

12 

Boutilier AJ and Elsawa SF: Macrophage polarization states in the tumor microenvironment. Int J Mol Sci. 22:69952021. View Article : Google Scholar : PubMed/NCBI

13 

Wang L, Li Y, Wang X, Wang P, Essandoh K, Cui S, Huang W, Mu X, Liu Z, Wang Y, et al: GDF3 protects mice against sepsis-induced cardiac dysfunction and mortality by suppression of macrophage pro-inflammatory phenotype. Cells. 9:1202020. View Article : Google Scholar : PubMed/NCBI

14 

Liu L, Stokes JV, Tan W and Pruett SB: An optimized flow cytometry panel for classifying macrophage polarization. J Immunol Methods. 511:1133782022. View Article : Google Scholar : PubMed/NCBI

15 

Domschke G and Gleissner CA: CXCL4-induced macrophages in human atherosclerosis. Cytokine. 122:1541412019. View Article : Google Scholar

16 

Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber F, Han J, Chen W, et al: Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 107:737–746. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A and Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25:677–686. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Lu Y, Sun Q, Guan Q, Zhang Z, He Q, He J, Ji Z, Tian W, Xu X, Liu Y, et al: The XOR-IDH3α axis controls macrophage polarization in hepatocellular carcinoma. J Hepatol. 79:1172–1184. 2023. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Gao Y, Yuan Y, Wen S, Chen Y, Zhang Z, Feng Y, Jiang B, Ma S, Hu R, Fang C, et al: Dual role of ANGPTL8 in promoting tumor cell proliferation and immune escape during hepatocarcinogenesis. Oncogenesis. 12:262023. View Article : Google Scholar : PubMed/NCBI

21 

Hao X, Zheng Z, Liu H, Zhang Y, Kang J, Kong X, Rong D, Sun G, Sun G, Liu L, et al: Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing. Redox Biol. 56:1024632022. View Article : Google Scholar : PubMed/NCBI

22 

Zhao S, Mi Y, Guan B, Zheng B, Wei P, Gu Y, Zhang Z, Cai S, Xu Y, Li X, et al: Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J Hematol Oncol. 13:1562020. View Article : Google Scholar : PubMed/NCBI

23 

Kuo CC, Hanzelmann S, Senturk Cetin N, Frank S, Zajzon B, Derks JP, Akhade VS, Ahuja G, Kanduri C, Grummt I, et al: Detection of RNA-DNA binding sites in long noncoding RNAs. Nucleic Acids Res. 47:e322019. View Article : Google Scholar : PubMed/NCBI

24 

Li P, He J, Yang Z, Ge S, Zhang H, Zhong Q and Fan X: ZNNT1 long noncoding RNA induces autophagy to inhibit tumorigenesis of uveal melanoma by regulating key autophagy gene expression. Autophagy. 16:1186–1199. 2020. View Article : Google Scholar :

25 

Wang H, Liu Y, Tang A and Zhang X: Molecular subtypes of clear cell renal carcinoma based on PCD-related long non-coding RNAs expression: Insights into the underlying mechanisms and therapeutic strategies. Eur J Med Res. 29:2922024. View Article : Google Scholar : PubMed/NCBI

26 

Wei H, Li W, Yang M, Fang Q, Nian J, Huang Y, Wei Q, Huang Z, Liu G, Xu Z, et al: METTL3/16-mediated m(6)A modification of ZNNT1 promotes hepatocellular carcinoma progression by activating ZNNT1/osteopontin/S100A9 positive feedback loop-mediated crosstalk between macrophages and tumour cells. Clin Immunol. 261:1099242024. View Article : Google Scholar : PubMed/NCBI

27 

Huang X, Wu W, Jing D, Yang L, Guo H, Wang L, Zhang W, Pu F and Shao Z: Engineered exosome as targeted lncRNA MEG3 delivery vehicles for osteosarcoma therapy. J Control Release. 343:107–117. 2022. View Article : Google Scholar : PubMed/NCBI

28 

Zhang J, Lin Z, Gao Y and Yao T: Downregulation of long noncoding RNA MEG3 is associated with poor prognosis and promoter hypermethylation in cervical cancer. J Exp Clin Cancer Res. 36:52017. View Article : Google Scholar : PubMed/NCBI

29 

Wu X and Xu L: The RNA-binding protein HuR in human cancer: A friend or foe? Adv Drug Deliv Rev. 184:1141792022. View Article : Google Scholar : PubMed/NCBI

30 

Majumder M, Chakraborty P, Mohan S, Mehrotra S and Palanisamy V: HuR as a molecular target for cancer therapeutics and immune-related disorders. Adv Drug Deliv Rev. 188:1144422022. View Article : Google Scholar : PubMed/NCBI

31 

Brauss TF, Winslow S, Lampe S, Scholz A, Weigert A, Dehne N, von Stedingk K, Schmid T and Brüne B: The RNA-binding protein HuR inhibits expression of CCL5 and limits recruitment of macrophages into tumors. Mol Carcinog. 56:2620–2629. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Wei H, Wu X, Huang L, Long C, Lu Q, Huang Z, Huang Y, Li W and Pu J: LncRNA MEG3 reduces the ratio of M2/M1 Macrophages Through the HuR/CCL5 axis in hepatocellular carcinoma. J Hepatocell Carcinoma. 11:543–562. 2024. View Article : Google Scholar : PubMed/NCBI

33 

Wei Q, Liu G, Huang Z, Huang Y, Huang L, Huang Z, Wu X, Wei H and Pu J: LncRNA MEG3 inhibits tumor progression by modulating macrophage phenotypic polarization via miR-145-5p/DAB2 axis in hepatocellular carcinoma. J Hepatocell Carcinoma. 10:1019–1035. 2023. View Article : Google Scholar : PubMed/NCBI

34 

Su T, Zhang N, Wang T, Zeng J, Li W, Han L and Yang M: Super Enhancer-Regulated LncRNA LINC01089 induces alternative splicing of DIAPH3 to drive hepatocellular carcinoma metastasis. Cancer Res. 83:4080–4094. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Peng Y, Yin D, Li X, Wang K, Li W, Huang Y, Liu X, Ren Z, Yang X, Zhang Z, et al: Integration of transcriptomics and metabolomics reveals a novel gene signature guided by FN1 associated with immune response in oral squamous cell carcinoma tumorigenesis. J Cancer Res Clin Oncol. 149:6097–6113. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Ma L, Jiang J, Si Q, Chen C and Duan Z: IGF2BP3 enhances the growth of hepatocellular carcinoma tumors by regulating the properties of macrophages and CD8(+) T cells in the tumor microenvironment. J Clin Transl Hepatol. 11:1308–1320. 2023.PubMed/NCBI

37 

Yang M, Yu T and Han L: Hsa_circ_0010882 facilitates hepatocellular carcinoma progression by modulating M1/M2 macrophage polarization. J Viral Hepat. 31:189–196. 2024. View Article : Google Scholar : PubMed/NCBI

38 

Hu Z, You L, Hu S, Yu L, Gao Y, Li L and Zhang S: Hepatocellular carcinoma cell-derived exosomal miR-21-5p promotes the polarization of tumor-related macrophages (TAMs) through SP1/XBP1 and affects the progression of hepatocellular carcinoma. Int Immunopharmacol. 126:1111492024. View Article : Google Scholar

39 

Bian Z, Xu C, Wang X, Zhang B, Xiao Y, Liu L, Zhao S, Huang N, Yang F, Zhang Y, et al: TRIM65/NF2/YAP1 signaling coordinately orchestrates metabolic and immune advantages in hepatocellular carcinoma. Adv Sci (Weinh). Jul 15–2024.Epub ahead of print. View Article : Google Scholar

40 

Zhu Y, Chen M, Xu D, Li TE, Zhang Z, Li JH, Wang XY, Yang X, Lu L, Jia HL, et al: The combination of PD-1 blockade with interferon-α has a synergistic effect on hepatocellular carcinoma. Cell Mol Immunol. 19:726–737. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Jiang M, Wang D, Su N, Lou W, Chen Y, Yang H, Chen C, Xi F, Chen Y, Deng L and Tang X: TRIM65 knockout inhibits the development of HCC by polarization tumor-associated macrophages towards M1 phenotype via JAK1/STAT1 signaling pathway. Int Immunopharmacol. 128:1114942024. View Article : Google Scholar : PubMed/NCBI

42 

Tan S, Wang Z, Li N, Guo X, Zhang Y, Ma H, Peng X, Zhao Y, Li C, Gao L, et al: Transcription factor Zhx2 is a checkpoint that programs macrophage polarization and antitumor response. Cell Death Differ. 30:2104–2119. 2023. View Article : Google Scholar : PubMed/NCBI

43 

Dai Z, Wang Y, Sun N and Zhang C: Characterizing ligand-receptor interactions and unveiling the pro-tumorigenic role of CCL16-CCR1 axis in the microenvironment of hepatocellular carcinoma. Front Immunol. 14:12999532024. View Article : Google Scholar : PubMed/NCBI

44 

Wu XM, Liao YW, Wang HY, Ji KQ, Li GF and Zang B: Integrin alphavbeta6 is involved in measles protein-induced airway immune suppression. Cytokine. 59:59–64. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Hu WM, Liu SQ, Zhu KF, Li W, Yang ZJ, Yang Q, Zhu ZC and Chang J: The ALOX5 inhibitor Zileuton regulates tumor-associated macrophage M2 polarization by JAK/STAT and inhibits pancreatic cancer invasion and metastasis. Int Immunopharmacol. 121:1105052023. View Article : Google Scholar : PubMed/NCBI

46 

Yu C, Zhu Q, Ma C, Luo C, Nie L, Cai H, Wang Q, Wang F, Ren H, Yan H, et al: Major vault protein regulates tumor-associated macrophage polarization through interaction with signal transducer and activator of transcription 6. Front Immunol. 14:12897952024. View Article : Google Scholar : PubMed/NCBI

47 

Tang B, Zhu J, Wang Y, Chen W, Fang S, Mao W, Xu Z, Yang Y, Weng Q, Zhao Z, et al: Targeted xCT-mediated ferroptosis and protumoral polarization of macrophages is effective against HCC and enhances the efficacy of the Anti-PD-1/L1 Response. Adv Sci (Weinh). 10:e22039732023. View Article : Google Scholar

48 

Savchenko VL: Regulation of NADPH oxidase gene expression with PKA and cytokine IL-4 in neurons and microglia. Neurotox Res. 23:201–213. 2013. View Article : Google Scholar

49 

Liu Y, Chen H, Yan X and Zhang J, Deng Z, Huang M, Gu J and Zhang J: MyD88 in myofibroblasts enhances nonalcoholic fatty liver disease-related hepatocarcinogenesis via promoting macrophage M2 polarization. Cell Commun Signal. 22:862024. View Article : Google Scholar : PubMed/NCBI

50 

Ravindran C, Cheng YC and Liang SM: CpG-ODNs induces up-regulated expression of chemokine CCL9 in mouse macrophages and microglia. Cell Immunol. 260:113–118. 2010. View Article : Google Scholar

51 

Chen S, Wang M, Lu T, Liu Y, Hong W, He X, Cheng Y, Liu J, Wei Y and Wei X: JMJD6 in tumor-associated macrophage regulates macrophage polarization and cancer progression via STAT3/IL-10 axis. Oncogene. 42:2737–2750. 2023. View Article : Google Scholar : PubMed/NCBI

52 

Silswal N, Reis J, Qureshi AA, Papasian C and Qureshi N: Of mice and men: Proteasome's Role in LPS-Induced inflammation and tolerance. Shock. 47:445–454. 2017. View Article : Google Scholar

53 

Lu F, Zhou J, Chen Q, Zhu J, Zheng X, Fang N and Qiao L: PSMA5 contributes to progression of lung adenocarcinoma in association with the JAK/STAT pathway. Carcinogenesis. 43:624–634. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Xie S, Li X, Yan J, Yu H, Chen S and Chen K: Knockdown of liver cancer cell-secreted exosomal PSMA5 controls macrophage polarization to restrain cancer progression by blocking JAK2/STAT3 signaling. Immun Inflamm Dis. 12:e11462024. View Article : Google Scholar : PubMed/NCBI

55 

Liu Y, Tang Y, Jiang H, Zhang X, Chen X, Guo J, Jin C and Wu M: Exosome-Related FTCD Facilitates M1 macrophage polarization and impacts the prognosis of hepatocellular carcinoma. Biomolecules. 14:412023. View Article : Google Scholar

56 

Liu L, Zhang W, Liu T, Tan Y, Chen C, Zhao J, Geng H and Ma C: The physiological metabolite α-ketoglutarate ameliorates osteoarthritis by regulating mitophagy and oxidative stress. Redox Biol. 62:1026632023. View Article : Google Scholar

57 

Zhang D, Wang Y, Shi Z, Liu J, Sun P, Hou X, Zhang J, Zhao S, Zhou BP and Mi J: Metabolic reprogramming of cancer-associated fibroblasts by IDH3α downregulation. Cell Rep. 10:1335–1348. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Wang C, Chen C, Hu W, Tao L and Chen J: Revealing the role of necroptosis microenvironment: FCGBP + tumor-associated macrophages drive primary liver cancer differentiation towards cHCC-CCA or iCCA. Apoptosis. 29:460–481. 2024. View Article : Google Scholar

59 

Mai Y, Su J, Yang C, Xia C and Fu L: The strategies to cure cancer patients by eradicating cancer stem-like cells. Mol Cancer. 22:1712023. View Article : Google Scholar : PubMed/NCBI

60 

Zhao Q, Zong H, Zhu P, Su C, Tang W, Chen Z and Jin S: Crosstalk between colorectal CSCs and immune cells in tumorigenesis, and strategies for targeting colorectal CSCs. Exp Hematol Oncol. 13:62024. View Article : Google Scholar : PubMed/NCBI

61 

Liu X, Ye Y, Zhu L, Xiao X, Zhou B, Gu Y, Si H, Liang H, Liu M, Li J, et al: Niche stiffness sustains cancer stemness via TAZ and NANOG phase separation. Nat Commun. 14:2382023. View Article : Google Scholar : PubMed/NCBI

62 

Liang T, Tao T, Wu K, Liu L, Xu W, Zhou D, Fang H, Ding Q, Huang G and Wu S: Cancer-Associated fibroblast-induced remodeling of tumor microenvironment in recurrent bladder cancer. Adv Sci (Weinh). 10:e23032302023. View Article : Google Scholar : PubMed/NCBI

63 

Wang L, Zhang L, Zhao L, Shao S, Ning Q, Jing X, Zhang Y, Zhao F, Liu X, Gu S, et al: VEGFA/NRP-1/GAPVD1 axis promotes progression and cancer stemness of triple-negative breast cancer by enhancing tumor cell-macrophage crosstalk. Int J Biol Sci. 20:446–463. 2024. View Article : Google Scholar : PubMed/NCBI

64 

Zhang Q, Tsui YM, Zhang VX, Lu AJ, Lee JM, Lee E, Cheung GC, Li PM, Cheung ET, Chia NH, et al: Reciprocal interactions between malignant cells and macrophages enhance cancer stemness and M2 polarization in HBV-associated hepatocellular carcinoma. Theranostics. 14:892–910. 2024. View Article : Google Scholar : PubMed/NCBI

65 

Qi Y, Zhao T, Li R and Han M: Macrophage-Secreted S100A4 supports breast cancer metastasis by remodeling the extracellular matrix in the premetastatic niche. Biomed Res Int. 2022:98955042022. View Article : Google Scholar : PubMed/NCBI

66 

Li Y, Wang J, Song K, Liu S, Zhang H, Wang F, Ni C, Zhai W, Liang J, Qin Z and Zhang J: S100A4 promotes hepatocellular carcinogenesis by intensifying fibrosis-associated cancer cell stemness. Oncoimmunology. 9:17253552020. View Article : Google Scholar : PubMed/NCBI

67 

Huang A, Lv B, Zhang Y, Yang J, Li J, Li C, Yu Z and Xia J: Construction of a tumor immune infiltration macrophage signature for predicting prognosis and immunotherapy response in liver cancer. Front Mol Biosci. 9:9838402022. View Article : Google Scholar : PubMed/NCBI

68 

Wei R, Zhu WW, Yu GY, Wang X, Gao C, Zhou X, Lin ZF, Shao WQ, Wang SH, Lu M and Qin LX: S100 calcium-binding protein A9 from tumor-associated macrophage enhances cancer stem cell-like properties of hepatocellular carcinoma. Int J Cancer. 148:1233–1244. 2021. View Article : Google Scholar

69 

Chen Y, Wen H, Zhou C, Su Q, Lin Y, Xie Y, Huang Y, Qiu Q, Lin J, Huang X, et al: TNF-alpha derived from M2 tumor-associated macrophages promotes epithelial-mesenchymal transition and cancer stemness through the Wnt/beta-catenin pathway in SMMC-7721 hepatocellular carcinoma cells. Exp Cell Res. 378:41–50. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Chen J, Feng W, Sun M, Huang W, Wang G, Chen X, Yin Y, Chen X, Zhang B, Nie Y, et al: TGF-β1-Induced SOX18 elevation promotes hepatocellular carcinoma progression and metastasis through transcriptionally upregulating PD-L1 and CXCL12. Gastroenterology. 167:264–280. 2024. View Article : Google Scholar : PubMed/NCBI

71 

Ye S, Huang H, Xiao Y, Han X, Shi F, Luo W, Chen J, Ye Y, Zhao X, Huang W, et al: Macrophage Dectin-1 mediates Ang II renal injury through neutrophil migration and TGF-β1 secretion. Cell Mol Life Sci. 80:1842023. View Article : Google Scholar

72 

Yao Y, Chen R, Wang G, Zhang Y and Liu F: Exosomes derived from mesenchymal stem cells reverse EMT via TGF-β1/Smad pathway and promote repair of damaged endometrium. Stem Cell Res Ther. 10:2252019. View Article : Google Scholar

73 

Liu X, Xu J, Shen B, Xu J and Jiang J: USP33 promotes pancreatic cancer malignant phenotype through the regulation of TGFBR2/TGFβ signaling pathway. Cell Death Dis. 14:3622023. View Article : Google Scholar

74 

Ye Y, Guo J, Xiao P, Ning J, Zhang R, Liu P, Yu W, Xu L, Zhao Y and Yu J: Macrophages-induced long noncoding RNA H19 up-regulation triggers and activates the miR-193b/MAPK1 axis and promotes cell aggressiveness in hepatocellular carcinoma. Cancer Lett. 469:310–322. 2020. View Article : Google Scholar

75 

Chang CH, Chang YT, Tseng TH and Wang CJ: Mulberry leaf extract inhibit hepatocellular carcinoma cell proliferation via depressing IL-6 and TNF-α derived from adipocyte. J Food Drug Anal. 26:1024–1032. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Yang H, Kang B, Ha Y, Lee SH, Kim I, Kim H, Lee WS, Kim G, Jung S, Rha SY, et al: High serum IL-6 correlates with reduced clinical benefit of atezolizumab and bevacizumab in unresectable hepatocellular carcinoma. JHEP Rep. 5:1006722023. View Article : Google Scholar : PubMed/NCBI

77 

Wan S, Zhao E, Kryczek I, Vatan L, Sadovskaya A, Ludema G, Simeone DM, Zou W and Welling TH: Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 147:1393–1404. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Wen JH, Li DY, Liang S, Yang C, Tang JX and Liu HF: Macrophage autophagy in macrophage polarization, chronic inflammation and organ fibrosis. Front Immunol. 13:9468322022. View Article : Google Scholar : PubMed/NCBI

79 

Ma L, Zhang R, Li D, Qiao T and Guo X: Fluoride regulates chondrocyte proliferation and autophagy via PI3K/AKT/mTOR signaling pathway. Chem Biol Interact. 349:1096592021. View Article : Google Scholar : PubMed/NCBI

80 

Bi S, Zhang Y, Zhou J, Yao Y, Wang J, Fang M, Li B, Wu C and Ren C: miR-210 promotes hepatocellular carcinoma progression by modulating macrophage autophagy through PI3K/AKT/mTOR signaling. Biochem Biophys Res Commun. 662:47–57. 2023. View Article : Google Scholar : PubMed/NCBI

81 

Cai J, Qian X, Qi Q, Han J, Zhu X, Zhang Q and Xia R: Extracellular ubiquitin inhibits the apoptosis of hepatoma cells via the involvement of macrophages. Transl Cancer Res. 9:2855–2864. 2020. View Article : Google Scholar : PubMed/NCBI

82 

She G, Du JC, Wu W, Pu TT, Zhang Y, Bai RY, Zhang Y, Pang ZD, Wang HF, Ren YJ, et al: Hippo pathway activation mediates chemotherapy-induced anti-cancer effect and cardiomyopathy through causing mitochondrial damage and dysfunction. Theranostics. 13:560–577. 2023. View Article : Google Scholar : PubMed/NCBI

83 

Fang S, Wan X, Zou X, Sun S, Hao X, Liang C, Zhang Z, Zhang F, Sun B, Li H and Yu B: Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway. Cell Death Dis. 12:882021. View Article : Google Scholar : PubMed/NCBI

84 

Ai JH, Wen YZ, Dai SJ, Zhang LD, Huang ZJ and Shi J: Exosomal lncRNA HEIH, an essential communicator for hepatocellular carcinoma cells and macrophage M2 polarization through the miR-98-5p/STAT3 axis. J Biochem Mol Toxicol. 38:e236862024. View Article : Google Scholar : PubMed/NCBI

85 

Keawvilai P, Kueanjinda P, Klomsing J and Palaga T: Coculturing liver cancer cells and monocytes in spheroids conditions monocytes to adopt tumor-associated macrophage phenotypes that favor tumor growth via cholesterol metabolism. J Leukoc Biol. 115:344–357. 2024. View Article : Google Scholar

86 

Zhang L, Zhang J, Li P, Li T, Zhou Z and Wu H: Exosomal hsa_circ_0004658 derived from RBPJ overexpressed-macrophages inhibits hepatocellular carcinoma progression via miR-499b-5p/JAM3. Cell Death Dis. 13:322022. View Article : Google Scholar : PubMed/NCBI

87 

Wu H, Zhang X, Han D, Cao J and Tian J: Tumour-associated macrophages mediate the invasion and metastasis of bladder cancer cells through CXCL8. PeerJ. 8:e87212020. View Article : Google Scholar : PubMed/NCBI

88 

Zou S, Chen S, Rao G, Zhang G, Ma M, Peng B, Du X, Huang W, Lin W, Tian Y and Fu X: Extrachromosomal circular MiR-17-92 amplicon promotes HCC. Hepatology. 79:79–95. 2024. View Article : Google Scholar

89 

Yu S, Su S, Wang P, Li J, Chen C, Xin H, Gong Y, Wang H, Ye X, Mao L, et al: Tumor-associated macrophage-induced circMRCK-alpha encodes a peptide to promote glycolysis and progression in hepatocellular carcinoma. Cancer Lett. 591:2168722024. View Article : Google Scholar

90 

Qu W, Qiao S, Liu L, Chen Y, Peng C, Hou Y, Xu Z, Lv M and Wang T: Dectin3 protects against hepatocellular carcinoma by regulating glycolysis of macrophages. Int Immunopharmacol. 113(Pt A): 1093842022. View Article : Google Scholar : PubMed/NCBI

91 

Ji W, Bai J and Ke Y: Exosomal ZFPM2-AS1 contributes to tumorigenesis, metastasis, stemness, macrophage polarization, and infiltration in hepatocellular carcinoma through PKM mediated glycolysis. Environ Toxicol. 38:1332–1346. 2023. View Article : Google Scholar : PubMed/NCBI

92 

Wang L, Yi X, Xiao X, Zheng Q, Ma L and Li B: Exosomal miR-628-5p from M1 polarized macrophages hinders m6A modification of circFUT8 to suppress hepatocellular carcinoma progression. Cell Mol Biol Lett. 27:1062022. View Article : Google Scholar : PubMed/NCBI

93 

Xie Q, Zeng Y, Zhang X and Yu F: The significance of lipid metabolism reprogramming of tumor-associated macrophages in hepatocellular carcinoma. Cancer Immunol Immunother. 73:1712024. View Article : Google Scholar : PubMed/NCBI

94 

Xie M, Lin Z, Ji X, Luo X, Zhang Z, Sun M, Chen X, Zhang B, Liang H, Liu D, et al: FGF19/FGFR4-mediated elevation of ETV4 facilitates hepatocellular carcinoma metastasis by upregulating PD-L1 and CCL2. J Hepatol. 79:109–125. 2023. View Article : Google Scholar : PubMed/NCBI

95 

Yeung OW, Lo CM, Ling CC, Qi X, Geng W, Li CX, Ng KT, Forbes SJ, Guan XY, Poon RT, et al: Alternatively activated (M2) macrophages promote tumour growth and invasiveness in hepatocellular carcinoma. J Hepatol. 62:607–616. 2015. View Article : Google Scholar

96 

Chen J, Zhao D, Zhang L, Zhang J, Xiao Y, Wu Q, Wang Y and Zhan Q: Tumor-associated macrophage (TAM)-derived CCL22 induces FAK addiction in esophageal squamous cell carcinoma (ESCC). Cell Mol Immunol. 19:1054–1066. 2022. View Article : Google Scholar : PubMed/NCBI

97 

Jiang Q, Zeng Y, Xu Y, Xiao X, Liu H, Zhou B, Kong Y, Saw PE and Luo B: Ultrasound molecular imaging as a potential non-invasive diagnosis to detect the margin of hepatocarcinoma via CSF-1R Targeting. Front Bioeng Biotechnol. 8:7832020. View Article : Google Scholar : PubMed/NCBI

98 

Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V, et al: CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 19:1264–1272. 2013. View Article : Google Scholar : PubMed/NCBI

99 

Peng S, Chen Y, Gong Y, Li Z, Xie R, Lin Y, Zou B, Li J and Zeng L: Predictive value of intratumour inflammatory cytokine mRNA levels of hepatocellular carcinoma patients and activation of two distinct pathways govern IL-8 induced epithelial-mesenchymal transition in human hepatic cancer cell lines. Cytokine. 119:81–89. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Yan W, Liu X, Ma H, Zhang H, Song X, Gao L, Liang X and Ma C: Tim-3 fosters HCC development by enhancing TGF-β-mediated alternative activation of macrophages. Gut. 64:1593–1604. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Ganjalikhani Hakemi M, Jafarinia M, Azizi M, Rezaeepoor M, Isayev O and Bazhin AV: The Role of TIM-3 in hepatocellular carcinoma: A promising target for immunotherapy? Front Oncol. 10:6016612020. View Article : Google Scholar

102 

Sun G, Liu H, Zhao J, Zhang J, Huang T, Sun G, Zhao S, Zhang Z, Cao H, Rong D, et al: Macrophage GSK3β-deficiency inhibits the progression of hepatocellular carcinoma and enhances the sensitivity of anti-PD1 immunotherapy. J Immunother Cancer. 10:e0056552022. View Article : Google Scholar

103 

Liu D, Luo X, Xie M, Zhang T, Chen X, Zhang B, Sun M, Wang Y, Feng Y, Ji X, et al: HNRNPC downregulation inhibits IL-6/STAT3-mediated HCC metastasis by decreasing HIF1A expression. Cancer Sci. 113:3347–3361. 2022. View Article : Google Scholar : PubMed/NCBI

104 

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

105 

Johnson DE, O'Keefe RA and Grandis JR: Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15:234–248. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Dong S, Yang Z, Xu P, Zheng W, Zhang B, Fu F, Mao Z, Yuan J, Chen H and Yu W: Mutually exclusive epigenetic modification on SIX6 with hypermethylation for precancerous stage and metastasis emergence tracing. Signal Transduct Target Ther. 7:2082022. View Article : Google Scholar : PubMed/NCBI

107 

Liu C, Yao Z, Wang J, Zhang W, Yang Y, Zhang Y, Qu X, Zhu Y, Zou J, Peng S, et al: Macrophage-derived CCL5 facilitates immune escape of colorectal cancer cells via the p65/STAT3-CSN5-PD-L1 pathway. Cell Death Differ. 27:1765–1781. 2020. View Article : Google Scholar :

108 

Chang M and Nguyen TT: Strategy for treatment of infected diabetic foot ulcers. Acc Chem Res. 54:1080–1093. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Yao RR, Li JH, Zhang R, Chen RX and Wang YH: M2-polarized tumor-associated macrophages facilitated migration and epithelial-mesenchymal transition of HCC cells via the TLR4/STAT3 signaling pathway. World J Surg Oncol. 16:92018. View Article : Google Scholar : PubMed/NCBI

110 

Lin A, Wang G, Zhao H, Zhang Y, Han Q, Zhang C, Tian Z and Zhang J: TLR4 signaling promotes a COX-2/PGE(2)/STAT3 positive feedback loop in hepatocellular carcinoma (HCC) cells. Oncoimmunology. 5:e10743762016. View Article : Google Scholar : PubMed/NCBI

111 

Xu M, Zhou C, Weng J, Chen Z, Zhou Q, Gao J, Shi G, Ke A, Ren N, Sun H and Shen Y: Tumor associated macrophages-derived exosomes facilitate hepatocellular carcinoma malignance by transferring lncMMPA to tumor cells and activating glycolysis pathway. J Exp Clin Cancer Res. 41:2532022. View Article : Google Scholar : PubMed/NCBI

112 

Zhou J, Liu M, Sun H, Feng Y, Xu L, Chan AWH, Tong JH, Wong J, Chong CCN, Lai PBS, et al: Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut. 67:931–944. 2018. View Article : Google Scholar

113 

Wu J, Gao W, Tang Q, Yu Y, You W, Wu Z, Fan Y, Zhang L, Wu C, Han G, et al: M2 macrophage-derived exosomes facilitate HCC metastasis by transferring αM β2 Integrin to Tumor Cells. Hepatology. 73:1365–1380. 2021. View Article : Google Scholar

114 

Lu Y, Han G, Zhang Y, Zhang L, Li Z, Wang Q, Chen Z, Wang X and Wu J: M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma. Cell Commun Signal. 21:2992023. View Article : Google Scholar : PubMed/NCBI

115 

Bai ZZ, Li HY, Li CH, Sheng CL and Zhao XN: Retraction Note: M1 Macrophage-Derived Exosomal MicroRNA-326 Suppresses Hepatocellular Carcinoma Cell Progression Via Mediating NF-κB Signaling Pathway. Nanoscale Res Lett. 17:1222022. View Article : Google Scholar

116 

Luo HL, Luo T, Liu JJ, Wu FX, Bai T, Ou C, Chen J, Li LQ and Zhong JH: Macrophage polarization-associated lnc-Ma301 interacts with caprin-1 to inhibit hepatocellular carcinoma metastasis through the Akt/Erk1 pathway. Cancer Cell Int. 21:4222021. View Article : Google Scholar : PubMed/NCBI

117 

Sun D, Luo T, Dong P, Zhang N, Chen J and Zhang S, Dong L, Janssen HLA and Zhang S: M2-polarized tumor-associated macrophages promote epithelial-mesenchymal transition via activation of the AKT3/PRAS40 signaling pathway in intrahepatic cholangiocarcinoma. J Cell Biochem. 121:2828–2838. 2020. View Article : Google Scholar

118 

Baeriswyl V and Christofori G: The angiogenic switch in carcinogenesis. Semin Cancer Biol. 19:329–337. 2009. View Article : Google Scholar : PubMed/NCBI

119 

Dong S, Guo X, Han F, He Z and Wang Y: Emerging role of natural products in cancer immunotherapy. Acta Pharm Sin B. 12:1163–1185. 2022. View Article : Google Scholar : PubMed/NCBI

120 

Pollard JW: Trophic macrophages in development and disease. Nat Rev Immunol. 9:259–270. 2009. View Article : Google Scholar : PubMed/NCBI

121 

Palazon A, Tyrakis PA, Macias D, Veliça P, Rundqvist H, Fitzpatrick S, Vojnovic N, Phan AT, Loman N, Hedenfalk I, et al: An HIF-1α/VEGF-A Axis in Cytotoxic T Cells regulates tumor progression. Cancer Cell. 32:669–683 e5. 2017. View Article : Google Scholar

122 

Xu K, Wu CL, Wang ZX, Wang HJ, Yin FJ, Li WD, Liu CC and Fan HN: VEGF family gene expression as prognostic biomarkers for Alzheimer's disease and primary liver cancer. Comput Math Methods Med. 2021:34223932021. View Article : Google Scholar : PubMed/NCBI

123 

Simons M, Gordon E and Claesson-Welsh L: Mechanisms and regulation of endothelial VEGF receptor signalling. Nat Rev Mol Cell Biol. 17:611–625. 2016. View Article : Google Scholar : PubMed/NCBI

124 

Apte RS, Chen DS and Ferrara N: VEGF in signaling and disease: Beyond discovery and development. Cell. 176:1248–1264. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Pu J, Li W, Wang A, Zhang Y, Qin Z, Xu Z, Wang J, Lu Y, Tang Q and Wei H: Long non-coding RNA HOMER3-AS1 drives hepatocellular carcinoma progression via modulating the behaviors of both tumor cells and macrophages. Cell Death Dis. 12:11032021. View Article : Google Scholar : PubMed/NCBI

126 

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

127 

Mabeta P and Steenkamp V: The VEGF/VEGFR axis revisited: Implications for cancer therapy. Int J Mol Sci. 23:155852022. View Article : Google Scholar : PubMed/NCBI

128 

Grillet B, Pereira RVS, Van Damme J, Abu El-Asrar A, Proost P and Opdenakker G: Matrix metalloproteinases in arthritis: Towards precision medicine. Nat Rev Rheumatol. 19:363–377. 2023. View Article : Google Scholar : PubMed/NCBI

129 

Riabov V, Gudima A, Wang N, Mickley A, Orekhov A and Kzhyshkowska J: Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front Physiol. 5:752014. View Article : Google Scholar : PubMed/NCBI

130 

Peacock JW, Takeuchi A, Hayashi N, Liu L, Tam KJ, Al Nakouzi N, Khazamipour N, Tombe T, Dejima T, Lee KC, et al: SEMA3C drives cancer growth by transactivating multiple receptor tyrosine kinases via Plexin B1. EMBO Mol Med. 10:219–238. 2018. View Article : Google Scholar : PubMed/NCBI

131 

Korbecki J, Siminska D, Gassowska-Dobrowolska M, Listos J, Gutowska I, Chlubek D and Baranowska-Bosiacka I: Chronic and cycling hypoxia: Drivers of cancer chronic inflammation through HIF-1 and NF-kappaB Activation: A review of the molecular mechanisms. Int J Mol Sci. 22:107012021. View Article : Google Scholar

132 

He H, Chen S, Fan Z, Dong Y, Wang Y, Li S, Sun X, Song Y, Yang J, Cao Q, et al: Multi-dimensional single-cell characterization revealed suppressive immune microenvironment in AFP-positive hepatocellular carcinoma. Cell Discov. 9:602023. View Article : Google Scholar : PubMed/NCBI

133 

Zhou Y, Zha Y, Yang Y, Ma T, Li H and Liang J: S100 proteins in cardiovascular diseases. Mol Med. 29:682023. View Article : Google Scholar : PubMed/NCBI

134 

Casazza A, Laoui D, Wenes M, Rizzolio S, Bassani N, Mambretti M, Deschoemaeker S, Van Ginderachter JA, Tamagnone L and Mazzone M: Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell. 24:695–709. 2013. View Article : Google Scholar : PubMed/NCBI

135 

Shao R: YKL-40 acts as an angiogenic factor to promote tumor angiogenesis. Front Physiol. 4:1222013. View Article : Google Scholar : PubMed/NCBI

136 

Qi M, Zhou Y, Liu J, Ou X, Li M, Long X, Ye J and Yu G: AngII induces HepG2 cells to activate epithelial-mesenchymal transition. Exp Ther Med. 16:3471–3477. 2018.PubMed/NCBI

137 

Duran CL, Borriello L, Karagiannis GS, Entenberg D, Oktay MH and Condeelis JS: Targeting Tie2 in the tumor microenvironment: From angiogenesis to dissemination. Cancers (Basel). 13:57302021. View Article : Google Scholar : PubMed/NCBI

138 

Bartneck M, Schrammen PL, Mockel D, Govaere O, Liepelt A, Krenkel O, Ergen C, McCain MV, Eulberg D, Luedde T, et al: The CCR2(+) macrophage subset promotes pathogenic angiogenesis for tumor vascularization in fibrotic livers. Cell Mol Gastroenterol Hepatol. 7:371–390. 2019. View Article : Google Scholar : PubMed/NCBI

139 

Xiang X, Pathak JL, Wu W, Li J, Huang W, Wu Q, Xin M, Wu Y, Huang Y, Ge L and Zeng S: Human serum-derived exosomes modulate macrophage inflammation to promote VCAM1-mediated angiogenesis and bone regeneration. J Cell Mol Med. 27:1131–1143. 2023. View Article : Google Scholar : PubMed/NCBI

140 

Rabinowitz JD and White E: Autophagy and metabolism. Science. 330:1344–1348. 2010. View Article : Google Scholar : PubMed/NCBI

141 

Liu Q, Yan X, Li R, Yuan Y, Wang J, Zhao Y, Fu J and Su J: Polyamine Signal through HCC Microenvironment: A key regulator of mitochondrial preservation and turnover in TAMs. Int J Mol Sci. 25:9962024. View Article : Google Scholar : PubMed/NCBI

142 

Liu X, Zhang Q, Liang Y, Xiong S, Cai Y, Cao J, Xu Y, Xu X, Wu Y, Lu Q, et al: Nanoparticles (NPs)-mediated Siglec15 silencing and macrophage repolarization for enhanced cancer immunotherapy. Acta Pharm Sin B. 13:5048–5059. 2023. View Article : Google Scholar : PubMed/NCBI

143 

Zhang X, Wei Z, Yong T, Li S, Bie N, Li J, Li X, Liu H, Xu H, Yan Y, et al: Cell microparticles loaded with tumor antigen and resiquimod reprogram tumor-associated macrophages and promote stem-like CD8(+) T cells to boost anti-PD-1 therapy. Nat Commun. 14:56532023. View Article : Google Scholar : PubMed/NCBI

144 

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

145 

Caraban BM, Matei E, Cozaru GC, Aşchie M, Deacu M, Enciu M, Bălţătescu GI, Chisoi A, Dobrin N, Petcu L, et al: PD-L1, CD4+, and CD8+ Tumor-Infiltrating Lymphocytes (TILs) expression profiles in melanoma tumor microenvironment cells. J Pers Med. 13:2212023. View Article : Google Scholar : PubMed/NCBI

146 

Yuan L, Tatineni J, Mahoney KM and Freeman GJ: VISTA: A mediator of quiescence and a promising target in cancer immunotherapy. Trends Immunol. 42:209–227. 2021. View Article : Google Scholar : PubMed/NCBI

147 

Shiri AM, Zhang T, Bedke T, Zazara DE, Zhao L, Lücke J, Sabihi M, Fazio A, Zhang S, Tauriello DVF, et al: IL-10 dampens antitumor immunity and promotes liver metastasis via PD-L1 induction. J Hepatol. 80:634–644. 2024. View Article : Google Scholar : PubMed/NCBI

148 

Wen J, Yang S, Yan G, Lei J, Liu X, Zhang N, Zhang J, Deng H, Wu L and Li Y: Increased OIT3 in macrophages promotes PD-L1 expression and hepatocellular carcinogenesis via NF-κB signaling. Exp Cell Res. 428:1136512023. View Article : Google Scholar

149 

Wang J, Zhang X, Ma X, Chen D, Cai M, Xiao L, Li J, Huang Z, Huang Y and Lian Y: Blockage of CacyBP inhibits macrophage recruitment and improves anti-PD-1 therapy in hepatocellular carcinoma. J Exp Clin Cancer Res. 42:3032023. View Article : Google Scholar : PubMed/NCBI

150 

Ruf B, Bruhns M, Babaei S, Kedei N, Ma L, Revsine M, Benmebarek MR, Ma C, Heinrich B, Subramanyam V, et al: Tumor-associated macrophages trigger MAIT cell dysfunction at the HCC invasive margin. Cell. 186:3686–3705 e32. 2023. View Article : Google Scholar : PubMed/NCBI

151 

Saadey AA, Yousif A, Osborne N, Shahinfar R, Chen YL, Laster B, Rajeev M, Bauman P, Webb A and Ghoneim HE: Rebalancing TGFβ1/BMP signals in exhausted T cells unlocks responsiveness to immune checkpoint blockade therapy. Nat Immunol. 24:280–294. 2023. View Article : Google Scholar

152 

Wang J, Zhou J, Zhou Q, Qi Y, Zhang P, Yan C and Ren X: Dysregulated Th1 cells in lung squamous cell carcinoma. J Leukoc Biol. 112:1567–1576. 2022. View Article : Google Scholar : PubMed/NCBI

153 

Xun X, Zhang C, Wang S, Hu S, Xiang X, Cheng Q, Li Z, Wang Y and Zhu J: Cyclooxygenase-2 expressed hepatocellular carcinoma induces cytotoxic T lymphocytes exhaustion through M2 macrophage polarization. Am J Transl Res. 13:4360–4375. 2021.PubMed/NCBI

154 

Vignali PDA, DePeaux K, Watson MJ, Ye C, Ford BR, Lontos K, McGaa NK, Scharping NE, Menk AV, Robson SC, et al: Hypoxia drives CD39-dependent suppressor function in exhausted T cells to limit antitumor immunity. Nat Immunol. 24:267–279. 2023. View Article : Google Scholar

155 

Lu JC, Zhang PF, Huang XY, Guo XJ, Gao C, Zeng HY, Zheng YM, Wang SW, Cai JB, Sun QM, et al: Amplification of spatially isolated adenosine pathway by tumor-macrophage interaction induces anti-PD1 resistance in hepatocellular carcinoma. J Hematol Oncol. 14:2002021. View Article : Google Scholar : PubMed/NCBI

156 

Wang J, Zhang L, Kang D, Yang D and Tang Y: Activation of PGE2/EP2 and PGE2/EP4 signaling pathways positively regulate the level of PD-1 in infiltrating CD8(+) T cells in patients with lung cancer. Oncol Lett. 15:552–558. 2018.

157 

Szefel J, Danielak A and Kruszewski WJ: Metabolic pathways of L-arginine and therapeutic consequences in tumors. Adv Med Sci. 64:104–110. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Kumari A, Syeda S, Rawat K, Kumari R and Shrivastava A: Melatonin modulates L-arginine metabolism in tumor-associated macrophages by targeting arginase 1 in lymphoma. Naunyn Schmiedebergs Arch Pharmacol. 397:1163–1179. 2024. View Article : Google Scholar

159 

Wang X, Ye X, Chen Y and Lin J: Mechanism of M2 type macrophage-derived extracellular vesicles regulating PD-L1 expression via the MISP/IQGAP1 axis in hepatocellular carcinoma immunotherapy resistance. Int Immunopharmacol. 124(Pt A): 1108482023. View Article : Google Scholar : PubMed/NCBI

160 

Goswami KK, Bose A and Baral R: Macrophages in tumor: An inflammatory perspective. Clin Immunol. 232:1088752021. View Article : Google Scholar : PubMed/NCBI

161 

Moreno Ayala MA, Campbell TF, Zhang C, Dahan N, Bockman A, Prakash V, Feng L, Sher T and DuPage M: CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8(+) T cell antitumor immunity. Immunity. 56:1613–1630 e5. 2023. View Article : Google Scholar

162 

Liu Y, Zhang Q, Xing B, Luo N, Gao R, Yu K, Hu X, Bu Z, Peng J, Ren X and Zhang Z: Immune phenotypic linkage between colorectal cancer and liver metastasis. Cancer Cell. 40:424–437 e5. 2022. View Article : Google Scholar : PubMed/NCBI

163 

Wu K, Lin K, Li X, Yuan X, Xu P, Ni P and Xu D: Redefining tumor-associated macrophage subpopulations and functions in the tumor microenvironment. Front Immunol. 11:17312020. View Article : Google Scholar : PubMed/NCBI

164 

Wang H, Tian T and Zhang J: Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): From mechanism to therapy and prognosis. Int J Mol Sci. 22:84702021. View Article : Google Scholar : PubMed/NCBI

165 

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

166 

Nowak M and Klink M: The role of tumor-associated macrophages in the progression and chemoresistance of ovarian cancer. Cells. 9:12992020. View Article : Google Scholar : PubMed/NCBI

167 

Li C, Xu X, Wei S, Jiang P, Xue L and Wang J: Senior Correspondence; Tumor-associated macrophages: Potential therapeutic strategies and future prospects in cancer. J Immunother Cancer. 9:e0013412021. View Article : Google Scholar

168 

Zheng H, Peng X, Yang S, Li X, Huang M, Wei S, Zhang S, He G, Liu J, Fan Q, et al: Targeting tumor-associated macrophages in hepatocellular carcinoma: biology, strategy, and immunotherapy. Cell Death Discov. 9:652023. View Article : Google Scholar

169 

Li Z, Wu T, Zheng B and Chen L: Individualized precision treatment: Targeting TAM in HCC. Cancer Lett. 458:86–91. 2019. View Article : Google Scholar

170 

Argentiero A, Delvecchio A, Fasano R, Andriano A, Caradonna IC, Memeo R and Desantis V: The complexity of the tumor microenvironment in hepatocellular carcinoma and emerging therapeutic developments. J Clin Med. 12:74692023. View Article : Google Scholar

171 

Agirre-Lizaso A, Huici-Izagirre M, Urretabizkaia-Garmendia J, Rodrigues PM, Banales JM and Perugorria MJ: Targeting the heterogeneous tumour-associated macrophages in hepatocellular carcinoma. Cancers (Basel). 15:49772023. View Article : Google Scholar

172 

Mantovani A, Marchesi F, Malesci A, Laghi L and Allavena P: Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 14:399–416. 2017. View Article : Google Scholar

173 

Cassetta L, Fragkogianni S, Sims AH, Swierczak A, Forrester LM, Zhang H, Soong DYH, Cotechini T, Anur P, Lin EY, et al: Human tumor-associated macrophage and monocyte transcriptional landscapes reveal cancer-specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell. 35:588–602 e10. 2019. View Article : Google Scholar : PubMed/NCBI

174 

Wen J, Wang S, Guo R and Liu D: CSF1R inhibitors are emerging immunotherapeutic drugs for cancer treatment. Eur J Med Chem. 245(Pt 1): 1148842023. View Article : Google Scholar

175 

Tkach M, Thalmensi J, Timperi E, Gueguen P, Névo N, Grisard E, Sirven P, Cocozza F, Gouronnec A, Martin-Jaular L, et al: Extracellular vesicles from triple negative breast cancer promote pro-inflammatory macrophages associated with better clinical outcome. Proc Natl Acad Sci USA. 119:e21073941192022. View Article : Google Scholar : PubMed/NCBI

176 

Zhang W, Zhu XD, Sun HC, Xiong YQ, Zhuang PY, Xu HX, Kong LQ, Wang L, Wu WZ and Tang ZY: Depletion of tumor-associated macrophages enhances the effect of sorafenib in metastatic liver cancer models by antimetastatic and antiangiogenic effects. Clin Cancer Res. 16:3420–3430. 2010. View Article : Google Scholar : PubMed/NCBI

177 

Jo H, Loison F and Luo HR: Microtubule dynamics regulates Akt signaling via dynactin p150. Cell Signal. 26:1707–1716. 2014. View Article : Google Scholar : PubMed/NCBI

178 

Liu Y, Ma J, Lu S, He P and Dong W: USP25 promotes hepatocellular carcinoma progression by interacting with TRIM21 via the Wnt/β-catenin signaling pathway. Chin Med J (Engl). 136:2229–2242. 2023. View Article : Google Scholar : PubMed/NCBI

179 

Mao J, Wang D, Wang Z, Tian W, Li X, Duan J, Wang Y, Yang H, You L, Cheng Y, et al: Combretastatin A-1 phosphate, a microtubule inhibitor, acts on both hepatocellular carcinoma cells and tumor-associated macrophages by inhibiting the Wnt/β-catenin pathway. Cancer Lett. 380:134–143. 2016. View Article : Google Scholar : PubMed/NCBI

180 

Xiang X, Wang J, Lu D and Xu X: Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther. 6:752021. View Article : Google Scholar : PubMed/NCBI

181 

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

182 

Wu J, Chan YT, Lu Y, Feng Z, Yuan H, Xu X, Xu L, Zhang C, Feng Y, Tan HY and Wang N: Genipin-activating PPARgamma impedes CCR2-mediated macrophage infiltration into postoperative liver to suppress recurrence of hepatocellular carcinoma. Int J Biol Sci. 19:5257–5274. 2023. View Article : Google Scholar :

183 

Hong M, Lee S, Clayton J, Yake W and Li J: Genipin suppression of growth and metastasis in hepatocellular carcinoma through blocking activation of STAT-3. J Exp Clin Cancer Res. 39:1462020. View Article : Google Scholar : PubMed/NCBI

184 

Gallage S, Ali A, Barragan Avila JE, Seymen N, Ramadori P, Joerke V, Zizmare L, Aicher D, Gopalsamy IK, Fong W, et al: A 5:2 intermittent fasting regimen ameliorates NASH and fibrosis and blunts HCC development via hepatic PPARα and PCK1. Cell Metab. 36:1371–1393 e7. 2024. View Article : Google Scholar

185 

Ambade A, Lowe P, Kodys K, Catalano D, Gyongyosi B, Cho Y, Iracheta-Vellve A, Adejumo A, Saha B, Calenda C, et al: Pharmacological Inhibition of CCR2/5 signaling prevents and reverses alcohol-induced liver damage, steatosis, and inflammation in mice. Hepatology. 69:1105–1121. 2019. View Article : Google Scholar

186 

Pozzi S and Satchi-Fainaro R: The role of CCL2/CCR2 axis in cancer and inflammation: The next frontier in nanomedicine. Adv Drug Deliv Rev. 209:1153182024. View Article : Google Scholar : PubMed/NCBI

187 

Sugiyama S, Yumimoto K, Fujinuma S and Nakayama KI: Identification of effective CCR2 inhibitors for cancer therapy using humanized mice. J Biochem. 175:195–204. 2024. View Article : Google Scholar

188 

Yuan JM, Grouls M, Carmella SG, Wang R, Heskin A, Jiang Y, Tan YT, Adams-Haduch J, Gao YT, Hecht SS, et al: Prediagnostic levels of urinary 8-epi-prostaglandin F2α and prostaglandin E2 metabolite, biomarkers of oxidative damage and inflammation, and risk of hepatocellular carcinoma. Carcinogenesis. 40:989–997. 2019. View Article : Google Scholar : PubMed/NCBI

189 

Wan Z, Huang H, West RE III, Zhang M, Zhang B, Cai X, Zhang Z, Luo Z, Chen Y, Zhang Y, et al: Overcoming pancreatic cancer immune resistance by codelivery of CCR2 antagonist using a STING-activating gemcitabine-based nanocarrier. Mater Today (Kidlington). 62:33–50. 2023. View Article : Google Scholar

190 

Sattiraju A, Kang S, Giotti B, Chen Z, Marallano VJ, Brusco C, Ramakrishnan A, Shen L, Tsankov AM, Hambardzumyan D, et al: Hypoxic niches attract and sequester tumor-associated macrophages and cytotoxic T cells and reprogram them for immunosuppression. Immunity. 56:1825–1843 e6. 2023. View Article : Google Scholar : PubMed/NCBI

191 

Chen Y, Ramjiawan RR, Reiberger T, Ng MR, Hato T, Huang Y, Ochiai H, Kitahara S, Unan EC, Reddy TP, et al: CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology. 61:1591–1602. 2015. View Article : Google Scholar

192 

White CW, Platt S, Kilpatrick LE, Dale N, Abhayawardana RS, Dekkers S, Kindon ND, Kellam B, Stocks MJ, Pfleger KDG and Hill SJ: CXCL17 is an allosteric inhibitor of CXCR4 through a mechanism of action involving glycosaminoglycans. Sci Signal. 17:eabl37582024. View Article : Google Scholar : PubMed/NCBI

193 

Klaver D, Gander H, Frena B, Amato M and Thurnher M: Crosstalk between purinergic receptor P2Y11 and chemokine receptor CXCR7 is regulated by CXCR4 in human macrophages. Cell Mol Life Sci. 81:1322024. View Article : Google Scholar

194 

Ashrafizadeh M, Zarrabi A, Hashemi F, Zabolian A, Salek i H, Bagher ian M, Azam i N, Bejandi A K, Hushmandi K, Ang HL, et al: Polychemotherapy with curcumin and doxorubicin via biological nanoplatforms: enhancing antitumor activity. Pharmaceutics. 12:10842020. View Article : Google Scholar : PubMed/NCBI

195 

Ma X, Yang S, Zhang T, Wang S, Yang Q, Xiao Y, Shi X, Xue P, Kang Y, Liu G, et al: Bioresponsive immune-booster-based prodrug nanogel for cancer immunotherapy. Acta Pharm Sin B. 12:451–466. 2022. View Article : Google Scholar : PubMed/NCBI

196 

Jang Y, Cho YS, Kim A, Zhou X, Kim Y, Wan Z, Moon JJ and Park H: CXCR4-Targeted macrophage-derived biomimetic hybrid vesicle nanoplatform for enhanced cancer therapy through codelivery of manganese and doxorubicin. ACS Appl Mater Interfaces. 16:17129–17144. 2024. View Article : Google Scholar : PubMed/NCBI

197 

Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V, Rey-Giraud F, Pradel LP, Feuerhake F, Klaman I, et al: Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 25:846–859. 2014. View Article : Google Scholar : PubMed/NCBI

198 

Hashimoto A, Sarker D, Reebye V, Jarvis S, Sodergren MH, Kossenkov A, Sanseviero E, Raulf N, Vasara J, Andrikakou P, et al: Upregulation of C/EBPα inhibits suppressive activity of myeloid cells and potentiates antitumor response in mice and patients with cancer. Clin Cancer Res. 27:5961–5978. 2021. View Article : Google Scholar : PubMed/NCBI

199 

Chen F, Gong M, Weng D, Jin Z, Han G, Yang Z, Han J and Wang J: Phellinus linteus activates Treg cells via FAK to promote M2 macrophage polarization in hepatocellular carcinoma. Cancer Immunol Immunother. 73:182024. View Article : Google Scholar : PubMed/NCBI

200 

Ao JY, Zhu XD, Chai ZT, Cai H, Zhang YY, Zhang KZ, Kong LQ, Zhang N, Ye BG, Ma DN and Sun HC: Colony-Stimulating Factor 1 receptor blockade inhibits tumor growth by altering the polarization of tumor-associated macrophages in hepatocellular carcinoma. Mol Cancer Ther. 16:1544–1554. 2017. View Article : Google Scholar : PubMed/NCBI

201 

Yang Y, Qin J, Lan L, Li N, Wang C, He P, Liu F, Ni H and Wang Y: M-CSF cooperating with NFκB induces macrophage transformation from M1 to M2 by upregulating c-Jun. Cancer Biol Ther. 15:99–107. 2014. View Article : Google Scholar

202 

Fang C, Zhong R, Lu S, Yu G, Liu Z, Yan C, Gao J, Tang Y, Wang Y, Zhao Q and Feng X: TREM2 promotes macrophage polarization from M1 to M2 and suppresses osteoarthritis through the NF-κB/CXCL3 axis. Int J Biol Sci. 20:1992–2007. 2024. View Article : Google Scholar :

203 

Zhu D, Huang M, Shen P, Zhang B, Chen G, Chen J, Duan L and Duan Y: TREM2 expression promotes liver and peritoneal M2 macrophage polarization in mice infected with Schistosoma japonicum. J Cell Mol Med. 27:2261–2269. 2023. View Article : Google Scholar : PubMed/NCBI

204 

Ye Y, Song Y, Zhuang J, Wang G, Ni J and Xia W: Anticancer effects of echinacoside in hepatocellular carcinoma mouse model and HepG2 cells. J Cell Physiol. 234:1880–1888. 2019. View Article : Google Scholar

205 

Yang T, Zhang S, Yuan H, Wang Y, Cai L, Chen H and Wang X, Song D and Wang X, Guo Z and Wang X: Platinum-Based TREM2 inhibitor suppresses tumors by remodeling the immunosuppressive microenvironment. Angew Chem Int Ed Engl. 62:e2022133372023. View Article : Google Scholar

206 

Zhou L, Wang M, Guo H, Hou J, Zhang Y, Li M, Wu X, Chen X and Wang L: Integrated analysis highlights the immunosuppressive role of TREM2(+) macrophages in hepatocellular carcinoma. Front Immunol. 13:8483672022. View Article : Google Scholar : PubMed/NCBI

207 

Tan J, Fan W, Liu T, Zhu B, Liu Y, Wang S, Wu J, Liu J, Zou F, Wei J, et al: TREM2(+) macrophages suppress CD8(+) T-cell infiltration after transarterial chemoembolisation in hepatocellular carcinoma. J Hepatol. 79:126–140. 2023. View Article : Google Scholar : PubMed/NCBI

208 

Esparza-Baquer A, Labiano I, Sharif O, Agirre-Lizaso A, Oakley F, Rodrigues PM, Zhuravleva E, O'Rourke CJ, Hijona E, Jimenez-Agüero R, et al: TREM-2 defends the liver against hepatocellular carcinoma through multifactorial protective mechanisms. Gut. 70:1345–1361. 2021. View Article : Google Scholar

209 

Binnewies M, Pollack JL, Rudolph J, Dash S, Abushawish M, Lee T, Jahchan NS, Canaday P, Lu E, Norng M, et al: Targeting TREM2 on tumor-associated macrophages enhances immunotherapy. Cell Rep. 37:1098442021. View Article : Google Scholar : PubMed/NCBI

210 

Tan HY, Wang N, Man K, Tsao SW, Che CM and Feng Y: Autophagy-induced RelB/p52 activation mediates tumour-associated macrophage repolarisation and suppression of hepatocellular carcinoma by natural compound baicalin. Cell Death Dis. 6:e19422015. View Article : Google Scholar : PubMed/NCBI

211 

Xu G, Feng D, Yao Y, Li P, Sun H, Yang H, Li C, Jiang R, Sun B and Chen Y: Listeria-based hepatocellular carcinoma vaccine facilitates anti-PD-1 therapy by regulating macrophage polarization. Oncogene. 39:1429–1444. 2020. View Article : Google Scholar

212 

Zhou B, Yang Y and Li C: SIRT1 inhibits hepatocellular carcinoma metastasis by promoting M1 macrophage polarization via NF-κB pathway. Onco Targets Ther. 12:2519–2529. 2019. View Article : Google Scholar :

213 

Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, Luo X, Chen R and Chen T: Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem. 119:2951–2963. 2018. View Article : Google Scholar

214 

Vyas AK, Jindal A, Hissar S, Ramakrishna G and Trehanpati N: Immune balance in Hepatitis B Infection: Present and future therapies. Scand J Immunol. 86:4–14. 2017. View Article : Google Scholar : PubMed/NCBI

215 

Anderson NR, Minutolo NG, Gill S and Klichinsky M: Macrophage-Based approaches for cancer immunotherapy. Cancer Res. 81:1201–1208. 2021. View Article : Google Scholar

216 

Bao D, Zhao J, Zhou X, Yang Q, Chen Y, Zhu J, Yuan P, Yang J, Qin T, Wan S and Xing J: Mitochondrial fission-induced mtDNA stress promotes tumor-associated macrophage infiltration and HCC progression. Oncogene. 38:5007–5020. 2019. View Article : Google Scholar : PubMed/NCBI

217 

Rodell CB, Arlauckas SP, Cuccarese MF, Garris CS, Li R, Ahmed MS, Kohler RH, Pittet MJ and Weissleder R: TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat Biomed Eng. 2:578–588. 2018. View Article : Google Scholar :

218 

Powers N, Massena C, Crouse B, Smith M, Hicks L, Evans JT, Miller S, Pravetoni M and Burkhart D: Self-Adjuvanting TLR7/8 agonist and fentanyl hapten co-conjugate achieves enhanced protection against fentanyl challenge. Bioconjug Chem. 34:1811–1821. 2023. View Article : Google Scholar : PubMed/NCBI

219 

Kim H, Khanna V, Kucaba TA, Zhang W, Sehgal D, Ferguson DM, Griffith TS and Panyam J: TLR7/8 Agonist-Loaded Nanoparticles Augment NK Cell-Mediated Antibody-Based Cancer Immunotherapy. Mol Pharm. 17:2109–2124. 2020. View Article : Google Scholar : PubMed/NCBI

220 

Kim H, Griffith TS and Panyam J: Poly(d,l-lactide-co-glycolide) Nanoparticles as delivery platforms for TLR7/8 agonist-based cancer vaccine. J Pharmacol Exp Ther. 370:715–724. 2019. View Article : Google Scholar : PubMed/NCBI

221 

Davenne T, Bridgeman A, Rigby RE and Rehwinkel J: Deoxyguanosine is a TLR7 agonist. Eur J Immunol. 50:56–62. 2020. View Article : Google Scholar :

222 

Hirsch E, Katanaev VL, Garlanda C, Azzolino O, Pirola L, Silengo L, Sozzani S, Mantovani A, Altruda F and Wymann MP: Central role for G protein-coupled phosphoinositide 3-kinase gamma in inflammation. Science. 287:1049–1053. 2000. View Article : Google Scholar : PubMed/NCBI

223 

Zhang Y, Long X, Ruan X, Wei Q, Zhang L, Wo L, Huang D, Lin L, Wang D, Xia L, et al: SIRT2-mediated deacetylation and deubiquitination of C/EBPbeta prevents ethanol-induced liver injury. Cell Discov. 7:932021. View Article : Google Scholar

224 

Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, Woo G, Nguyen AV, Figueiredo CC, Foubert P, et al: PI3Kgamma is a molecular switch that controls immune suppression. Nature. 539:437–442. 2016. View Article : Google Scholar : PubMed/NCBI

225 

Rao J, Wang H, Ni M, Wang Z, Wang Z, Wei S, Liu M, Wang P, Qiu J, Zhang L, et al: FSTL1 promotes liver fibrosis by reprogramming macrophage function through modulating the intracellular function of PKM2. Gut. 71:2539–2550. 2022. View Article : Google Scholar : PubMed/NCBI

226 

Yang XM, Wang XQ, Hu LP, Feng MX, Zhou YQ, Li DX, Li J, Miao XC, Zhang YL, Yao LL, et al: Nucleolar HEAT Repeat Containing 1 up-regulated by the mechanistic target of rapamycin complex 1 signaling promotes hepatocellular carcinoma growth by dominating ribosome biogenesis and proteome homeostasis. Gastroenterology. 165:629–646. 2023. View Article : Google Scholar : PubMed/NCBI

227 

O'Grady S, Crown J and Duffy MJ: Statins inhibit proliferation and induce apoptosis in triple-negative breast cancer cells. Med Oncol. 39:1422022. View Article : Google Scholar : PubMed/NCBI

228 

Sprinzl MF, Puschnik A, Schlitter AM, Schad A, Ackermann K, Esposito I, Lang H, Galle PR, Weinmann A, Heikenwälder M and Protzer U: Sorafenib inhibits macrophage-induced growth of hepatoma cells by interference with insulin-like growth factor-1 secretion. J Hepatol. 62:863–870. 2015. View Article : Google Scholar

229 

Wu L, Zhang X, Zheng L, Zhao H, Yan G, Zhang Q, Zhou Y, Lei J, Zhang J, Wang J, et al: RIPK3 Orchestrates Fatty Acid Metabolism in Tumor-Associated Macrophages and Hepatocarcinogenesis. Cancer Immunol Res. 8:710–721. 2020. View Article : Google Scholar : PubMed/NCBI

230 

Hu Q, Li Y, Chen H, Liao H, He Y and Zheng Q: CCDC88A Post-Transcriptionally Regulates VEGF via miR-101 and subsequently regulates hepatocellular carcinoma. Front Immunol. 13:8593312022. View Article : Google Scholar : PubMed/NCBI

231 

Xin Y, Tang L, Chen J, Chen D, Wen W and Han F: Inhibition of miR-101-3p protects against sepsis-induced myocardial injury by inhibiting MAPK and NF-κB pathway activation via the upregulation of DUSP1. Int J Mol Med. 47:202021. View Article : Google Scholar

232 

Delire B and Starkel P: The Ras/MAPK pathway and hepatocarcinoma: pathogenesis and therapeutic implications. Eur J Clin Invest. 45:609–623. 2015. View Article : Google Scholar : PubMed/NCBI

233 

Takizawa H and Manz MG: Macrophage tolerance: CD47-SIRP-alpha-mediated signals matter. Nat Immunol. 8:1287–1289. 2007. View Article : Google Scholar : PubMed/NCBI

234 

McCracken MN, Cha AC and Weissman IL: Molecular Pathways: Activating T Cells after Cancer Cell Phagocytosis from Blockade of CD47 'Don't Eat Me' Signals. Clin Cancer Res. 21:3597–3601. 2015. View Article : Google Scholar : PubMed/NCBI

235 

Koh E, Lee EJ, Nam GH, Hong Y, Cho E, Yang Y and Kim IS: Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials. 121:121–129. 2017. View Article : Google Scholar : PubMed/NCBI

236 

Xiao Z, Chung H, Banan B, Manning PT, Ott KC, Lin S, Capoccia BJ, Subramanian V, Hiebsch RR, Upadhya GA, et al: Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma. Cancer Lett. 360:302–309. 2015. View Article : Google Scholar : PubMed/NCBI

237 

Lakhani NJ, Chow LQM, Gainor JF, LoRusso P, Lee KW, Chung HC, Lee J, Bang YJ, Hodi FS, Kim WS, et al: Evorpacept alone and in combination with pembrolizumab or trastuzumab in patients with advanced solid tumours (ASPEN-01): A first-in-human, open-label, multicentre, phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 22:1740–1751. 2021. View Article : Google Scholar : PubMed/NCBI

238 

Vaeteewoottacharn K, Kariya R, Pothipan P, Fujikawa S, Pairojkul C, Waraasawapati S, Kuwahara K, Wongkham C, Wongkham S and Okada S: Attenuation of CD47-SIRPα signal in cholangiocarcinoma potentiates tumor-associated macrophage-mediated phagocytosis and suppresses intrahepatic metastasis. Transl Oncol. 12:217–225. 2019. View Article : Google Scholar

239 

Yang HD, Kim HS, Kim SY, Na MJ, Yang G, Eun JW, Wang HJ, Cheong JY, Park WS and Nam SW: HDAC6 Suppresses Let-7i-5p to Elicit TSP1/CD47-Mediated anti-tumorigenesis and phagocytosis of hepatocellular carcinoma. Hepatology. 70:1262–1279. 2019. View Article : Google Scholar : PubMed/NCBI

240 

Du K, Li Y, Liu J, Chen W, Wei Z, Luo Y, Liu H, Qi Y, Wang F and Sui J: A bispecific antibody targeting GPC3 and CD47 induced enhanced antitumor efficacy against dual antigen-expressing HCC. Mol Ther. 29:1572–1584. 2021. View Article : Google Scholar : PubMed/NCBI

241 

Kamber RA, Nishiga Y, Morton B, Banuelos AM, Barkal AA, Vences-Catalán F, Gu M, Fernandez D, Seoane JA, Yao D, et al: Inter-cellular CRISPR screens reveal regulators of cancer cell phagocytosis. Nature. 597:549–554. 2021. View Article : Google Scholar : PubMed/NCBI

242 

Chen M, Mao A, Xu M, Weng Q, Mao J and Ji J: CRISPR-Cas9 for cancer therapy: Opportunities and challenges. Cancer Lett. 447:48–55. 2019. View Article : Google Scholar : PubMed/NCBI

243 

Mout R and Rotello VM: Cytosolic and nuclear delivery of CRISPR/Cas9-ribonucleoprotein for gene editing using arginine functionalized gold nanoparticles. Bio Protoc. 7:e25862017. View Article : Google Scholar : PubMed/NCBI

244 

Ray M, Lee YW, Hardie J, Mout R, Yeşilbag Tonga G, Farkas ME and Rotello VM: CRISPRed macrophages for cell-based cancer immunotherapy. Bioconjug Chem. 29:445–450. 2018. View Article : Google Scholar : PubMed/NCBI

245 

Quail DF, Bowman RL, Akkari L, Quick ML, Schuhmacher AJ, Huse JT, Holland EC, Sutton JC and Joyce JA: The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science. 352:aad30182016. View Article : Google Scholar : PubMed/NCBI

246 

Patwardhan PP, Surriga O, Beckman MJ, de Stanchina E, Dematteo RP, Tap WD and Schwartz GK: Sustained inhibition of receptor tyrosine kinases and macrophage depletion by PLX3397 and rapamycin as a potential new approach for the treatment of MPNSTs. Clin Cancer Res. 20:3146–3158. 2014. View Article : Google Scholar : PubMed/NCBI

247 

Welford AF, Biziato D, Coffelt SB, Nucera S, Fisher M, Pucci F, Di Serio C, Naldini L, De Palma M, Tozer GM and Lewis CE: TIE2-expressing macrophages limit the therapeutic efficacy of the vascular-disrupting agent combretastatin A4 phosphate in mice. J Clin Invest. 121:1969–1973. 2011. View Article : Google Scholar : PubMed/NCBI

248 

De Palma M and Naldini L: Angiopoietin-2 TIEs up macrophages in tumor angiogenesis. Clin Cancer Res. 17:5226–5232. 2011. View Article : Google Scholar : PubMed/NCBI

249 

De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi M and Naldini L: Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell. 8:211–226. 2005. View Article : Google Scholar : PubMed/NCBI

250 

De Palma M, Murdoch C, Venneri MA, Naldini L and Lewis CE: Tie2-expressing monocytes: Regulation of tumor angiogenesis and therapeutic implications. Trends Immunol. 28:519–524. 2007. View Article : Google Scholar : PubMed/NCBI

251 

Quail DF and Joyce JA: Molecular pathways: Deciphering mechanisms of resistance to macrophage-targeted therapies. Clin Cancer Res. 23:876–884. 2017. View Article : Google Scholar :

252 

Bergers G and Hanahan D: Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 8:592–603. 2008. View Article : Google Scholar : PubMed/NCBI

253 

Koivisto AP, Belvisi MG, Gaudet R and Szallasi A: Advances in TRP channel drug discovery: From target validation to clinical studies. Nat Rev Drug Discov. 21:41–59. 2022. View Article : Google Scholar

254 

Wang W, Liu P, Zhang Y, Yan L, Zhu MX, Wang J and Yu Y: Expression and functions of transient receptor potential channels in liver diseases. Acta Pharm Sin B. 13:445–459. 2023. View Article : Google Scholar : PubMed/NCBI

255 

Liu Y, Lyu Y, Zhu L and Wang H: Role of TRP channels in liver-related diseases. Int J Mol Sci. 24:125092023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 65 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu M, Yu H, Wang H, Xu X, Sun Z, Chen W, Yu M, Liu C, Jiang M, Zhang X, Zhang X, et al: Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review). Int J Oncol 65: 100, 2024.
APA
Yu, M., Yu, H., Wang, H., Xu, X., Sun, Z., Chen, W. ... Zhang, X. (2024). Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review). International Journal of Oncology, 65, 100. https://doi.org/10.3892/ijo.2024.5688
MLA
Yu, M., Yu, H., Wang, H., Xu, X., Sun, Z., Chen, W., Yu, M., Liu, C., Jiang, M., Zhang, X."Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review)". International Journal of Oncology 65.4 (2024): 100.
Chicago
Yu, M., Yu, H., Wang, H., Xu, X., Sun, Z., Chen, W., Yu, M., Liu, C., Jiang, M., Zhang, X."Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review)". International Journal of Oncology 65, no. 4 (2024): 100. https://doi.org/10.3892/ijo.2024.5688