Skip to main content
Cancer Medicine logoLink to Cancer Medicine
. 2023 Dec 14;12(24):22109–22129. doi: 10.1002/cam4.6717

The role of tumor‐associated macrophages in hepatocellular carcinoma progression: A narrative review

Xinyi Zhang 1, Chao Yu 1, Siqi Zhao 1, Min Wang 1, Longcheng Shang 1,, Jin Zhou 1,, Yong Ma 1,
PMCID: PMC10757104  PMID: 38098217

Abstract

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, with complex etiology and mechanism, and a high mortality rate. Tumor‐associated macrophages (TAMs) are an important part of the HCC tumor microenvironment. Studies in recent years have shown that TAMs are involved in multiple stages of HCC and are related to treatment and prognosis in HCC. The specific mechanisms between TAMs and HCC are gradually being revealed. This paper reviews recent advances in the mechanisms associated with TAMs in HCC, concentrating on an overview of effects of TAMs on drug resistance in HCC and the signaling pathways linked with HCC, providing clues for the treatment and prognosis determination of HCC.

Keywords: drug resistance, hepatocellular carcinoma, signaling pathway, tumor microenvironment, tumor‐associated macrophages

1. BACKGROUND

Tumor‐associated macrophages (TAMs) are macrophages differentiated from monocytes that are recruited to tumor tissues through chemokines in the tumor microenvironment. 1 An increasing number of studies have found that TAMs play an important role in a variety of tumors, such as liver cancer, 2 breast cancer, 3 gastrointestinal cancer, 4 ovarian cancer. 5 Therefore, TAMs have become a hot spot in cancer research in recent years. Among organs in the body, the liver contains the most macrophages after the brain and lung. 6 TAMs are engaged in liver diseases ranging from acute injury to chronic inflammation, fibrosis and tumor. 7 It is a worldwide health challenge that liver cancer is expected to affect more than 1 million people annually by 2025. 8 About 90% of instances of primary liver cancer are hepatocellular carcinoma (HCC), which is also the fourth most common cause of cancer‐related deaths worldwide; HCC is concerned with the interactions of multiple factors, consisting of susceptibility genes, viral and non‐viral risk factors such as fatty liver, immune cells, and tumor microenvironment. 8 The current treatments for liver cancer include surgical resection, immunotherapy, targeted therapy, transcatheter arterial chemoembolization (TACE), liver transplantation, etc. In terms of drug therapy, drug resistance is still a challenge. Current research has shown that TAMs influence the occurrence, proliferation, invasion, and metastasis of tumors in different ways. With in‐depth studies of the mechanism of TAMs, it is worth looking forward to alleviating drug resistance and improving the effect of monotherapy through regulating TAMs.

2. CHARACTERISTICS OF TAMS

Yolk sac, fetal liver, and bone marrow are recognized as three sources of tissue‐resident macrophages differentiated from progenitor cells and monocytes; cancer induces bone marrow generation and mobilization of hematopoietic stem and progenitor cells (HSPC) located in the spleen. 2 , 9 Then, monocytes generated in the spleen and bone marrow infiltrate the tumor further and differentiate into TAMs. 2 , 9 Macrophages can be classified as M1 and M2 according to phenotype, and markers comprise transmembrane glycoproteins, growth factors, hormones, cytokines, and cytokine receptors. M1, activated by toll‐like receptor (TLR), tumor necrosis factor (TNF)‐α, interferon‐gamma (IFN‐γ), and CSF2, has pro‐inflammatory, anti‐tumor, and bactericidal functions, with markers such as CD86, CD11c, HLA‐DR, iNOS, and pSTAT1. 10 M2, activated by IL‐4, IL‐10, IL‐13, transforming growth factor (TGF)‐β, and PGE2, plays an important role in anti‐inflammatory, pro‐tumor processes, and tissue repair, with markers such as CD206, CD163, CD204, Arginase‐1, Ym1, MGL‐1, Dectin‐1, vascular endothelial growth factor (VEGF), and cMAF. 10 , 11 The infiltration level of M2 polarized macrophages is elevated in patients with hepatocellular carcinoma, and M2 can secrete IL‐6, VEGF, matrix metalloprotease (MMP)‐9, GM‐colony‐stimulating factor (CSF), IL‐10, and monocyte chemoattractant protein‐1 (MCP‐1). 12 Six macrophage clusters were identified in comprehensive analysis of immune cell composition in HCC patients from five immune‐related sites with full‐length and 3′scRNA‐Seq technology, and mutually exclusive signals of S100 calcium‐binding protein A8 (S100A8) and SLC40A1 were detected on different cells in CD68 macrophages, indicating the existence of two distinct macrophage states in HCC, namely the coexistence of M1 and M2. 13 M1 and M2 display opposite effects, the polarization of which represents the two extremes of macrophages, and their imbalance is connected to various diseases and inflammations, the M2 phenotype predominant in TAMs in many studies. However, the two are inseparable throughout the cancer process. M1 macrophages generate inflammatory precancerous ecological niches and stimulate early oncogenic mutations, while M2 is reprogrammed to release various growth factors and provide an immunosuppressive state in tumor microenvironment (TME), prompting cancer cells to establish a new vascular system. 14 Kupffer cells (KCs) are resident macrophages in the liver, considered as specific type of TAMs and secrete relevant cytokines to promote HCC development. 15 , 16 There are interactions, consisting of both direct crosstalk and indirect crosstalk via cytokines/chemokines, NK cells, T cells, and neutrophils, between TAMs and hepatocellular carcinoma cells. 17 More and more studies have demonstrated that TAMs are metabolically heterogeneous and phenotypically plastic, with different impacts on tumor progression and immune function in different contexts. 18 , 19

3. ROLE OF TAMS IN HCC

3.1. Impact of TAMs on the development of HCC

The development of cancer requires a suitable tumor microenvironment that attenuates the killing ability of immune cells. Under hypoxia, triggering receptor expressed on myeloid cells‐1 (TREM‐1) is highly expressed in TAMs, upregulating C‐C motif ligand (CCL)20 via the ERK/nuclear factor kappa B (NF‐κB) pathway and recruiting CD4+CD25+CCR6+Foxp3+ Tregs, then inducing CD8+T cell apoptosis and dysfunction, thus creating an immunosuppressive environment. 20 Importantly, blocking programmed death ligand 1 (PD‐L1) does not reverse immunosuppression despite the high expression of PD‐L1 in TREM‐1 TAM, and blocking TREM‐1 signaling is required to reduce resistance to anti‐PD‐L1 therapy. 20 The expression of cyclooxygenase‐2 (COX‐2) is associated with the M2 macrophage marker CD163/CD206 in tissue chips and paraffin sections of HCC patients and inhibits the synthesis of IFN‐γ and granzyme B+ from activated CD8+T cells via the TGF‐β pathway, which results in the loss of CD8+T cells' ability to fight tumors. 21 TAMs also participate in immunosuppression and immune escape by expressing other substances, such as Siglec‐10 22 and MARCKS, 23 leading to poor prognosis. In addition to the immunosuppressive environment, cancer cells demand certain nutrients to support growth and proliferation. The number of TAM infiltration and MMP‐9 expression is positively relevant to tumor vascular density, and TAM increases MMP‐9 expression to promote angiogenesis in HCC, involved in type 1 insulin‐like growth factor (IGF‐1) signaling via phosphatidylinositol3‐kinase (PI3‐K) and mitogen‐activated protein kinase (MAPK) pathways. 24 One study has discovered that zoledronic acid (ZA) impedes TAMs infiltration and secretion of VEGF in a rat HCC model, thereby unfavorable for tumor angiogenesis. 25

3.2. Impact of TAMs on the invasion and metastasis of HCC

Epithelial‐mesenchymal transition (EMT) and cancer stem cell (CSC) are significant pathways for tumor invasion and metastasis. EMT is the conversion of polarized epithelial cancer cells into mesenchymal cells by disassembling adhesion and tight junctions and benefits the separation of mesenchymal cells from the initial site, then passing through the dismantled basement membrane and reaching distant organs, finally returning to the epithelial cancer cell phenotype through mesenchymal‐epithelial transformation and regaining the competence to proliferate and differentiate. 26 EMT not only expands cell invasion, but also aggravates resistance to cell death, senescence, and therapy and confers stem cell properties on cells. 27 , 28 As a driver of tumor invasion and metastasis, CSC can maintain tumor heterogeneity, possess the capability of immune escape, and perform in the formation of immunosuppressive tumor microenvironment. 29 Activation of EMT and CSC is often influenced by the local microenvironment. TAMs can produce cytokines, induce EMT in HCC cells, and enhance CSC characteristics, thus improving the ability of invasion and metastasis. In the margins of human HCC, there is a positive correlation between the number of TAM and the density of CSC. TAM induces HCC cells to display CSC‐like features, undergo EMT, and acquire more invasion ability by secreting more TGF‐β1, which are linked to a bad prognosis for patients. 30 TNF‐α can be produced by M2, and through the Wnt/β‐catenin pathway, cancer cells undergo EMT and obtain stemness. 31 Under hypoxia conditions, TAMs increase IL‐6 secretion and accelerate EMT. 32 The effects of hypoxia on TAM and HCC are described below. Cancer‐associated fibroblast (CAF), one of the major stromal cells in the HCC tumor microenvironment, encourages the polarization of TAMs into M2 phenotype in vitro simulation experiments while producing CXCL12 to stimulate M2 to secrete plasminogen activator inhibitor‐1 (PAI‐1), which mediates EMT to strengthen the malignant behavior of HCC cells. 33 The TAM‐mediated angiogenesis described above is important in tumor development, as well as in tumor invasion and metastasis. It not only provides nutrients, but also is one of the good choices of metastasis.

3.3. Impact of TAMs on the drug resistance of HCC

Tumor‐associated macrophages are involved in every stage of the development of a tumor, and their infiltration may be used to target cancer prevention or treatment or as a predictive marker for clinical outcomes in a variety of cancers. 34 Based on numerous research on mechanisms of TAMs affecting HCC, TAMs play an irreplaceable role in HCC, so it is considered meaningful and promising to treat HCC by targeting TAMs. As many drugs targeting TAMs are still in clinical trials at present, main mechanisms are to eliminate the existence of TAMs, block the recruitment of TAMs, reprogram the polarization of TAMs, regulate products of TAMs, and restore the phagocytic ability of TAMs. 35 TAMs can result in drug resistance in HCC, and hence targeting TAMs may alleviate drug resistance and improve the efficacy of anti‐tumor therapy (Table 1).

TABLE 1.

Impact of targeting tumor‐associated macrophages (TAMs) on drug resistance in hepatocellular carcinoma (HCC).

Gene/compound/drug Result Reference
Adeno‐associated virus 8 Mediate hepatic IRF8 rescue, inhibiting TAMs infiltration and decreasing expression of CCL20, and significantly inhibit HCC progression, enhancing the response to anti‐PD‐1 therapy 36
Carbonic anhydrase XII (CA12) inhibitor Reduce TAM infiltration and CCL8 production and attenuate tumor growth and metastasis, promoting anti‐PD1 therapy 37
Celecoxib Downregulate the expression of Foxp3Tregs, CD68 TAM, and PD‐L1 and increase the infiltration of CD8 CTL, augmenting the therapeutic efficacy of epirubicin 38
Competitive binding antibody Siglec‐10 Fc Block Siglec‐10 expressed by TAM, decrease expression of immunosuppressive molecules, and increase the cytotoxic effects of CD8+ T cells against HCC cells, leading to promoting the anti‐tumor efficacy of the PD‐1 inhibitor pembrolizumab 22
COX‐2 inhibitor (celecoxib) Reduce the inhibitory effect on CD8+ T cells through regulating TAMs in TIME, thus enhancing the efficacy of T cell‐based immunotherapy 21
CSF1/CSF1R inhibitor PLX3397 (PFH@LSLP) Activate the immune responses via inhibiting the CSF1/CSF1R pathway in TAMs, further enhance CD8+ T cell infiltration to reverse immunosuppression in tumors, thus overcoming sorafenib resistance 39
CSF1R inhibitor BZL945/IL‐1R1 antagonist anakinra Attenuate SLC7A11‐mediated intratumoral TAM and MDSC infiltration, enhancing the immune response to anti‐PD‐L1 therapy 40
IFN‐α Increase sorafenib's therapeutic efficacy by shifting TAM polarization to an M1‐like phenotype, increasing and activating intratumoral CD8+ T cells in HCC 41
Inhibition of APOC1 Promote the transformation of M2 into M1 via the ferroptosis pathway, thereby reshaping the tumor immune microenvironment, resulting in enhancing sensitivity to anti‐PD1 therapy in HCC 42
Lmdd‐MPFG (Listeria‐based HCC vaccine) Promote anti‐PD‐1 therapy through skewing the TAMs from M2 into M1 43
MTL‐CEBPA (small activating RNA) Reverse the immunosuppressive activity of M‐MDSCs and TAMs, promoting the anti‐tumor effect of checkpoint inhibitors or PMN‐MDSC‐targeted immunotherapy 44
RNA interference of autophagy‐related 5 homolog (ATG5) Suppress autophagy activated by co‐culturing with macrophages in HCC cells, promoting the oxaliplatin cytotoxicity 45
Sorafenib Upregulate IL‐12 production in TAMs at a sub‐pharmacologic, increasing the anti‐tumor effect of mCART cells therapy 46
Triggering receptor expressed on myeloid cells‐1 (TREM‐1) inhibitor GF9 Abrogate immunosuppression mediated by TREM‐1TAM, strengthening the effect of anti‐PD‐1 therapy 20
TREM2 knockdown Remodel TAMs to an immune‐stimulating status greatly improve the therapeutic effect of PD‐1 blockade probably through increasing the infiltration of immune cells and enhancing the toxicity of infiltrated CD8, CD4, and NK cells 47
xCT knockout Mediate ferroptosis to significantly increase PD‐L1 expression in macrophages and improve the anti‐tumor efficacy of anti‐PD‐L1 therapy 48
Zoledronic acid (ZA) Enhance the effects of transcatheter arterial chemoembolization through inhibiting TAM infiltration and tumor angiogenesis in rat HCC models 25
Reduce PD‐L1+ TAMs infiltration and alleviate CD8+ T cell suppression, enhancing the efficacy of anti‐PD‐L1 therapy in HCC 49

Sorafenib, used in the first‐line systemic therapy approved by the U.S. Food and Drug Administration (FDA) and the standard of care for advanced hepatocellular carcinoma, is affected by different factors in many studies, and the percentage of beneficiaries is unable to reach a satisfactory standard with decreased sensitivity and increased resistance to sorafenib in HCC patients. 50 Accordingly, it is particularly vital to take combination therapy with other inhibitors. M2 TAMs offer hepatocyte growth factor (HGF) to activate the HGF/c‐Met, ERK1/2/MAPK, and PI3K/AKT pathways, enlisting more macrophages to tumor tissue and intensifying HCC resistance to sorafenib in a pre‐feedback manner. 51 Studies have presented that targeting TAMs probably overcomes resistance to sorafenib and raises therapeutic efficacy. In H22 and PDX mice model of HCC, PLX3397 further enhances CD8 T‐cell infiltration and activates intracellular immune responses by inhibiting the CSF1/CSF1R pathway in TAMs and reducing TAMs recruitment and M2 polarization, which is more effective and comprehensive to conquer sorafenib resistance in synergy with alleviating hypoxia. 39 IFN‐α triggers the transition from M2 to M1 to neutralize the proliferative and migratory effects on HCC, simultaneously expanding CD8+T‐cell infiltration in HCC and enhancing the therapeutic effect of sorafenib. 41 Furthermore, sorafenib acts on TAMs in turn. Sorafenib augments the anti‐tumor effect of mouse chimeric antigen receptor (mCAR) T‐cell therapy through stepping up IL‐12 produced by TAMs at a sub‐pharmacologic dose. 46

The ascent of PD‐L1 is a critical mechanism of tumor immune escape. TAMs are able to express PD‐L1, and regulation of TAMs is significantly meaningful for PD‐1/PD‐L1 immunotherapy. CA12 inhibitor dwindles TAMs infiltration and CCL8 production, attenuating tumor growth and metastasis with growing proportion of CD8+T cells, thus showing more notable efficacy in combination with anti‐PD‐1 therapy than monotherapy. 37 Lmdd‐MPFG, a Listeria‐based HCC vaccine, promotes PD‐L1 expression in HCC cells but restores tumor local T‐cell sensitivity and gains the response to anti‐PD‐1 therapy by switching TAMs from M2 to M1 polarization. 43 Zoledronic acid restrains PD‐L1 TAM infiltration and attenuates CD8 T‐cell suppression, then enhancing the effect of anti‐PD‐L1 therapy in hepatocytes, especially in patients with Golgi membrane protein 1 (GOLM1) overexpression. 49 CSF1R inhibitor BZL945 40 and TREM‐1 inhibitor GF9 20 facilitate anti‐tumor curative effect of anti‐PD‐L1 in HCC as well.

Moreover, oxaliplatin, 45 epirubicin, 38 TACE, 25 and other relevant treatments for liver cancer may be more efficient by modulation of TAMs. The above results suggest that the regulation of TAMs will reverse the immunosuppressive environment and regain the position of T cells in the immune response. Compared with single therapy, combination therapy is a direction in the treatment of HCC, worthy of consideration and application.

4. SIGNALING PATHWAY/AXIS INVOLVED IN HCC VIA TAMS

In recent years, a growing number of studies have found that TAMs are considered to occupy an irreplaceable position in the pathogenesis of HCC, including immunosuppression, angiogenesis, tumor invasion and metastasis, metabolic support, drug resistance, EMT, and malignant transformation of HCC stem cells. 35 , 52 TAMs are either active to function on HCC or stimulated by factors secreted from hepatocellular carcinoma cells or foreign factors to act on tumor tissue (Table 2 and Table 3), bringing about the promotion or inhibition of HCC progression by means of recruitment, infiltration, polarization, secretion of chemokines, and autophagy. Multiple signaling pathways/axes play a key role in the crosstalk between TAMs and HCC (Figure 1).

TABLE 2.

Impact of regulators on tumor‐associated macrophages (TAMs) in hepatocellular carcinoma (HCC).

Gene/compound/drug/axis Mechanism Influence on TAMs Influence on HCC Reference
APOBEC3B (A3B) Depress global H3K27me3 abundance via interaction with PRC2 and reduces an occupancy of H3K27me3 on promoters of the chemokine CCL2 to recruit massive TAMs and MDSCs Promote M2 + 53
Astragalus polysaccharin (APS) Repress M2 polarization of TAMs Promote M1 54
Inhibit M2
Arsenite Increase miR‐15b levels and induce M2 polarization of THP‐1 cells Promote M2 + 55
B7 homolog 3 (B7‐H3) Mediate STAT3 signaling pathway to induce M2‐type polarization of TAMs Promote M2 + 56
Baicalin Autophagy‐induced RelB/p52 activation mediates repolarization of TAM to M1‐like phenotype M2 → M1 57
Blocking of the CCL2/CCR2 axis Inhibit the recruitment of inflammatory monocytes, infiltration, and M2‐polarization of TAMs Inhibit M2 58
8‐bromo‐7‐methoxychrysin (BrMC) Reverse M2 polarization of TAMs due to inhibition of NF‐κB activation Reverse M2 59
CCAAT/enhancer‐binding protein alpha (C/EBPα) Reverse the immunosuppressive activity of M‐MDSCs and TAMs M2 → M1 44
Combretastatin A‐1 phosphate (CA1P) Induce TAM apoptosis in vitro and eliminate TAMs in the TME in vivo through GSK‐3β activation, Wnt/β‐catenin pathway inhibition and Mcl‐1 downregulation Eliminate TAM 60
Cancer‐associated fibroblasts (CAFs) Induce the M2 polarization of TAMs by upregulating the mRNA expression levels of CD163 and CD206 and downregulating IL‐6 mRNA expression and secretion in the macrophages, and induce PAI_x005f‐1 secretion via CXCL12 Promote M2 + 33
Cholestyramine Reverse the effect of Sirt5 deficiency in promoting M2‐like polarized TAMs and liver tumor growth Inhibit M2 61
Cyclooxygenase‐2 (COX‐2) Induce anti‐tumor abilities exhaustion in activated CD8+ T cell through M2 TAMs polarization and transforming growth factor (TGF) beta pathway Promote M2 + 21
DNaseI/TLR9 antagonist By depleting cytosolic mtDNA or blocking TLR9 pathway, respectively, siRNA for TLR9 or p65 in HCC cells with Drp1 overexpression significantly decrease the recruitment and polarization of TAMs Inhibit M2 62
Downregulation of TREM1 Shift M2 macrophages toward a M1 phenotype via inhibiting PI3K/AKT signaling M2 → M1 63
17β‐estradiol (E2) Function as a suppressor for macrophage alternative activation and tumor progression by keeping ERβ away from interacting with ATP5J, thus inhibiting the JAK1‐STAT6 signaling pathway Inhibit M2 64
Ethyl pyruvate and N‐acetylcysteine amide Block HMGB1 and ROS respectively, significantly reducing M2 macrophage recruitment Inhibit M2 65
Fatty acid oxidation (FAO) Contributes to IL‐1β secretion in M2 macrophages and the pro‐migratory effect in M2 MDMs Promote M2 + 66
Forkhead box Q1 (FoxQ1) Transactivate ZEB2 and VersicanV1 expression, resulting in the induction of EMT and the recruitment of macrophage infiltration Promote recruitment + 67
Gadolinium chloride (GdCl3) Downregulate the expression of CD206 in TAMs Inhibit M2 68
Genipin Suppress IRE1α‐mediated infiltration and priming of TAMs Inhibit infiltration 69
Granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) Enhance A2A receptor expression on Mϕ and function synergistically with adenosine to elicit Mϕ proliferation Promote M2 + 70
Golgi membrane protein 1 (GOLM1) Induces CD8+ T cells suppression through promoting PD‐L1 stabilization and transporting PD‐L1 into TAMs with exosome dependent Promote M2 + 49
Golgi protein 73 (GP73) Stimulate endoplasmic reticulum (ER) stress activation in neighboring macrophages, which then release cytokines and chemokines involved in the TAM phenotype Promote M2 + 71
Histone deacetylase 2 (HDAC2) Upregulate atypical chemokine receptor 3 (ACKR3) via STAT1 to induce migration of M2 macrophages and immune escape in HCC Promote M2 + 72
Hemozoin (HZ) Inhibit IGF‐1 signaling through the PI3‐K and MAPK signaling pathways and thereby decrease the expression of MMP‐9 in TAMs, suppressing tumor angiogenesis Inhibit M2 24
High‐mobility group A1 (HMGA1) Promote macrophage recruitment by activating NF‐κB‐CCL2 signaling Promote recruitment + 73
HMGA2 Promote the migrating abilities of both M0‐Mφs and TAMs‐Mφs Promote M2 + 74
High‐mobility group box 1 (HMGB1) Produced by hepatoma cells in a HIF‐1α‐dependent manner under hypoxia, inducing the infiltration and reprogramming of macrophages to augment the expression of IL‐6 Promote M2 + 32
Trigger M2 macrophage polarization through HMGB1/TLR2/NOX2/autophagy axis Promote M2 + 65
Heat shock transcription factor 1 (HSF1) Regulate individually monocarboxylate transporter 1 (MCT1) and MCT4 expressions not only in HCC cells but also in TAMs, decreasing glucose consumption rate, lactate production rate and intercellular ROS level Promote M2 + 75
Interferon gamma (IFNG) Induce M2 polarization and chemotactic migration of macrophages through IFNG/IFR1/HHLA2 axis in HCC Promote M2 + 76
IFN‐α Shift TAM polarization to an M1‐like phenotype, increase and activate intratumoral CD8+ T cells in HCCs M2 → M1 41
IL‐1β Induce SLC7A11 overexpression to upregulate PD‐L1 and CSF1 through the αKG/HIF1α axis, promoting TAM and MDSC infiltration Promote recruitment + 40
IL‐2 Modulate exosomal miRNAs from TAMs to ameliorate hepatocellular carcinoma development 77
IL‐37 Promote TAMs polarization from M2 to M1 subtype through inhibiting the IL‐6/STAT3 signaling M2 → M1 78
IL‐6 Decrease PTPRO expression through the STAT3/c‐MYC/miR‐25‐3 p axis, promoting PD‐L1 secretion in both monocytes and macrophages Promote M2 + 79
Interleukin‐1 receptor‐associated kinase 1 (IRAK1) silencing Reverse the TAM‐induced increase in expression of NLRP3 and pro‐inflammatory factors in HCC cells Inhibit M2 80
Interferon regulatory factor 8 (IRF8) Mediate repression of c‐fos transcription resulting in decreased expression of CCL20 to regulate recruitment of TAMs 36
Nanoliposome‐loaded C6‐ceremide (LipC6) Reduce the number of TAMs and the ability of TAMs to suppress the anti‐tumor immune response and induce TAMs to differentiate into an M1 phenotype Promote M1 81
Long non‐coding RNA GAS5 Inhibit M2‐like polarization of TAMs by enhancing PTEN expression Inhibit M2 82
Myristoylated alanine‐rich C‐kinase substrate (MARCKS) Influence the M2 polarization and immune escape Promote M2 + 23
Mesoporous Fe3O4 nanoparticles (mFe NPs) Reprogram TAMs into M1 phenotypes to synergistically amplify anti‐tumor immunity after TAM phagocytosis Promote M1 83
miR‐125a/b Inhibit tumor‐associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90 Inhibit M2 84
miR‐144/miR‐451a cluster Promote macrophage M1 polarization and anti‐tumor activity by targeting HGF and MIF Promote M1 85
miR‐28‐5p deficiency Upregulate IL‐34 and promote TAM infiltration Promote M2 + 86
miR‐98 Regulate expression of inflammatory cytokines in HCC‐conditioned TAMs and modulated the capacity of HCC‐conditioned TAMs to regulate HepG2 cell migration and invasion by targeting IL‐10 Inhibit M2 87
Modulate macrophage polarization from M2 to M1 M2 → M1 88
miR‐99b Promote M1 while suppressing M2 macrophage polarization by targeting κB‐Ras2 and/or mTOR respectively Promote M1 89
Inhibit M2
Mitochondrial fission Induce the cytosolic mtDNA stress to enhance the CCL2 secretion from HCC cells by TLR9‐mediated NF‐κB signaling pathway, and thus promote the TAM recruitment and polarization Promote M2 + 62
MYC and Twist1 Elicit a transcriptional program associated with the activation of innate immunity through secretion of a cytokinome that elicits recruitment and polarization of TAMs Promote M2 + 90
Norcantharidin (NCTD) Modulate a shift from M2 to M1 polarization through miR‐214 M2 → M1 91
Nogo‐B Promote TAMs M2 polarization by inducing Yap/Taz pathway Promote M2 + 92
NOTCH blockade Impede the differentiation of moTAMs, but upregulates Wnt/β‐catenin signaling to promote the proliferation and pro‐tumor cytokine production of kclTAMs Inhibit moTAM + 16
Promote kclTAM
Osteopontin (OPN) Induce chemotactic migration and M2‐like polarization of macrophages and promote the PD‐L1 expression in HCC via activation of the CSF1‐CSF1R pathway in macrophages Promote M2 + 93
Proprotein convertase subtilisin/kexin type 9 (PCSK9) Inhibit M2 polarization of TAMs in HCC by promoting OX40L expression Inhibit M2 94
Propofol Stimulate TAMs to secrete microvesicles, mediating transfer of miR‐142‐3p from macrophages to cancer cells in vivo Promote M1 95
Inhibit M2
Retinoic acid‐inducible gene I (RIG‐I) Promote the polarization of M1 through the RIG‐I/MAVS/TRAF2/NF‐κB pathway in mice peritoneal macrophages Promote M1 96
Receptor‐interacting protein 140 (RIP140) Inhibit M2 polarization and the activation of NF‐κB/IL‐6 axis in the TAMs Inhibit M2 97
Inhibit the alternative activation of macrophages by inhibiting the NF‐κB/IL‐6 axis in TAMs Promote M1 98
Inhibit M2
Receptor‐interacting protein kinase 3 (RIPK3) deficiency Reduce reactive oxygen species and significantly inhibit caspase1‐mediated cleavage of PPAR, facilitating fatty acid metabolism and inducing M2 polarization in the tumor microenvironment Promote M2 + 99
S100 calcium‐binding protein A9 (S100A9) Recruit more macrophages via CCL2 Promote M2 + 100
Semaphorin3A Increase TAM infiltration Promote M2 + 101
Sirtuin 1 (SIRT1) Enhance NF‐κB stimulation, promoting phosphorylation of p65, IκB, and IκB kinase, to reinforce M1‐like macrophage infiltration Promote M1 102
SIRT4 silencing Facilitate M2 polarization via the FAO‐PPARδ‐STAT3 axis Promote M2 + 103
SIRT5 deficiency Elevate bile acids abnormally and promote M2‐like macrophage polarization Promote M2 + 61
Signaling lymphocyte activation molecule family member 6 (SLAMF6/Ly108) Promote macrophage M2 polarization Promote M2 + 104
Sorafenib Inhibit miR‐101 expression, enhance DUSP1 expression and lower TGF‐β and CD206 release in M2 cells Inhibit M2 105
SPP1 Trigger the polarization of macrophages to M2‐phenotype TAMs via SPP1‐CD44 and SPP1‐PTGER4 association Promote M2 + 106
Statins (inhibitors of hydroxymethylglutaryl‐CoA reductase) Inhibit YAP‐induced IL‐6 expression to block the recruitment of TAMs Inhibit M2 107
Tannin fraction of Terminalia bellirica (TB‐TF) Inhibit orthotopic tumor growth and promote the polarization of M2‐TAMs toward the anti‐tumor M1 phenotype in vivo M2 → M1 108
T cell immunoglobulin and mucin‐domain containing protein‐3 (Tim‐3) Enhance TGF‐β‐mediated alternative activation of macrophages Promote M2 + 109
TLR9 agonist CpGODN Downregulate the level of glycolysis and inhibit the M2 polarization of HCC‐TAMs Inhibit M2 110
Tocilizumab Block IL6 signaling to inhibit TAM‐stimulated activity of CD44 (+) cells Inhibit M2 111
Glycyrrhetinic acid‐tetramethylpyrazine conjugate (TOGA) Inhibit IL‐1β‐induced activation of the IL‐1R1/IκB/IKK/NF‐κB signaling pathway, effectively preventing the support of TAMs from fueling tumorigenesis Inhibit M2 112
Ubiquitin‐like with PHD and ring finger domains 1 (UHRF1) Induce DNA hypomethylation of the CSF1 promoter, promoting CSF1 expression, thereby leading to TAM recruitment and activation Promote recruitment/activation + 113
Verteporfin Block Nogo‐B‐Yap/Taz‐mediated macrophages M2 polarization Inhibit M2 92
Wnt/β‐catenin Stimulate M2‐like polarization of TAMs Promote M2 + 114
Wnt2b Promote the polarization of TAMs to M2‐like macrophages by activating Wnt2b/β‐catenin/c‐Myc signaling Promote M2 + 110
Yes‐associated protein (YAP) Induce IL‐6 secretion, promoting the TAMs recruitment Promote M2 + 107
ZA Inhibit infiltration of TAMs and tumor angiogenesis Inhibit M2 25
Zip9 Enhance phosphorylated STAT6 to promote M2 macrophage polarization but suppress the phosphorylation of IκBα/β to inhibit M1 macrophage polarization Promote M2 + 115
Inhibit M1

TABLE 3.

Impact of tumor‐associated macrophages (TAMs) on hepatocellular carcinoma (HCC).

Gene/compound Mechanism Influence on HCC Reference
xCT Derived from TAM, facilitates carcinogenesis though increasing TAM recruitment and infiltration, modulating M2 polarization and reducing ferroptosis + 48
CXCL1 and CXCL2 Secreted by M2 TAM, increasing HCC CSC activity, decreasing SOR‐induced apoptosis by affecting BCL‐2 family gene expression and upregulating SOR resistance in HCC cells via CXCR2/ERK + 116
Dectin3 Expressed by macrophages, contributing to the apoptosis of tumor cells and inhibiting the proliferation of tumor cells by regulating the glycolysis of macrophages 117
Deletion of IL‐6 Inhibit IL‐6/signal transducer and activator of transcription 3 signaling in monocytes/KCs 15
Gal‐1 Secreted by TAM via secretory autophagy + 118
GM‐CSF Secreted by TAM, enhancing A2A receptor expression on Mϕ and functioning synergistically with adenosine to elicit Mϕ proliferation in HCC + 70
IL‐6 Produced by TAM, promoting expansion of these CSCs and tumorigenesis via STAT3 signaling + 111
Produced by TAM, promoting EMT of HCC cells and enhancing the invasiveness and metastasis of murine HCC cells under hypoxia + 32
Inhibition of APOC1 Expressed by TAM, promoting the transformation of M2 macrophages into M1 macrophages via the ferroptosis pathway, thereby reshaping the tumor immune microenvironment and inhibiting HCC progression 42
lncMMPA Transmitted by exosome derived from TAM, interacting with miR‐548 s and increasing the mRNA level of ALDH1A3, then further promoting glucose metabolism and cell proliferation in HCC; polarize M2 macrophage + 119
lncRNAH19 Induced by TAM, triggering and activating the miR‐193b/MAPK1 axis + 120
microRNA (miR)‐17–92 cluster Originating from the EVs of M2‐TAMs, stimulating the imbalance of TGF‐β1/BMP‐7 pathways in HCC cells and increasing inhibitor of differentiation 1 (ID1) expression + 121
miR‐628‐5p Transferred by exosomes derived from M1 macrophages to HCC cells to inhibit human methyltransferase‐like 14 (METTL14) expression, hindering the m6A modification of circFUT8 competitively binding to miR‐552‐3p to increase CHMP4B expression 122
NOR1 Expressed by TAM, promote M2 alternative polarization + 123
PGE2 Derived from TAM, stimulating UHRF1 expression by repressing miR‐520d that targets the 3′‐UTR of UHRF1 mRNA + 113
S100A9 Derived from TAM, enhances stem cell traits of HCC cells via AGER/NF‐κB axis + 100
Siglec‐10 Expressed by TAM, exerting immunosuppressive function during HCC progression + 22
TGF‐β1 Produced by TAM, promoting CSC‐like properties via inducing EMT + 30
Tim‐3 Expressed by TAM, promoting tumor growth via NF‐κB‐IL‐6 axis + 109
TLR4 M2‐polarized macrophages facilitate the migration and EMT of HCC cells via the TLR4/STAT3 signaling pathway + 124
Tumor necrosis factor‐α Derived from M2‐TAMs, promoting EMT and cancer stemness cells via the Wnt/β‐catenin pathway + 31
TREM‐1 Expressed by TAM under hypoxia, elevating CCL20 expression through the extracellular signal‐regulated kinase/nuclear factor kappa B pathway and initiating the onset of tumor immunosuppression through attracting CCR6+ Foxp3+ Tregs + 20
TREM2 knockdown Expressed by TAM, remodeling TAMs to an immune‐stimulating status and suppressing the growth of hepatocellular carcinoma 47
β2‐AR Expressed by TAM, downregulating GRK2 and activating the downstream cyclic adenosine monophosphate (cAMP)/protein kinase A/cAMP‐response element binding protein and cAMP/interleukin‐6/signal transducer and the activator of transcription 3 signaling pathways + 12

FIGURE 1.

FIGURE 1

The signaling pathways associated with tumor‐associated macrophages (TAMs) in hepatocellular carcinoma (HCC).

4.1. STAT signaling pathway

Signal transducers and activators of transcription (STAT) function complexly and importantly in controlling normal physiological cellular processes such as angiogenesis, differentiation, proliferation, apoptosis, and immune system, and meanwhile are used for the epigenetic makeup of immune cells. At the same time, abnormal STAT management may occur, giving rise to pathological events about cancer‐containing occurrence, progression, metastasis, survival, and treatment resistance. To date, seven STAT genes have been identified in the human genome, and among them, STAT3 and STAT5 may be more important in cancer development. STAT3 is the most studied gene and is closely associated with tumor growth and immune escape in most tumors, while STAT5 is mainly in hematologic tumors. STAT1 is considered as a tumor suppressor, linked with the M1 phenotype, 125 , 126 whereas STAT3 is thought as a tumor promoter. Currently, it is primarily STAT3 and STAT6 that act between HCC and TAMs according to lots of research.

IL‐6 is a classic inflammatory factor and the most decisive activator of STAT3 that has a core role in transcription factors driving IL‐6‐induced transcriptome alterations in macrophages. 127 The experiments on patients and xenograft mice demonstrate that TAMs secrete IL‐6, which provokes CSC in HCC through STAT3 signaling. 111 IL‐6 upgrades miR‐25‐3p through STAT3/c‐MYC signaling to downgrade PTPRO in HCC monocytes, ascending PD‐L1 expression and tumor growth in vivo. 79 IL‐37 hinders IL‐6/STAT3 signaling to promote polarization of TAMs from M2 to M1 subtypes, constricting proliferation, migration, and invasion of HCC cells. 78 In Mdr2‐deficient mice that spontaneously develop HCC, loss of IL‐6 in monocytes/KCs contributes to suppression of IL‐6/STAT3 signaling and delayed tumorigenesis. 15 In addition, there are other factors influencing the role of STAT3 between HCC and TAMs. B7‐H3, a co‐stimulatory molecule involved in the regulation of non‐immune functions, favors PMA‐induced differentiation of THP‐1 cells to the M2 phenotype when overexpressing in HCC, and partial blockade of the STAT3 signaling pathway may inhibit the elevation of B7‐H3 expression on THP‐1 cells. 56 SIRT4 silencing facilitates M2 TAM polarization through the fatty acid oxidation (FAO)‐PPARδ‐STAT3 signaling pathway. 103 Toll‐like receptor 4 (TLR4) is a molecular biomarker of aggressive tumors and unfavorable prognoses. Human HCC cells undergo migration and EMT when M2‐polarized macrophages stimulate the STAT3 signaling pathway downstream of TLR4 and amplify TLR4 expression in HCC cells. 124

The JAK/STAT pathway is a critical pathway to mediate inflammatory response and tumorigenesis. Expression of PTPRO suppresses PD‐L1 expression in HCC macrophages or monocytes through JAK2/STAT1 and JAK2/STAT3/c‐MYC activation. 79 In a BALB/c mouse ectopic tumor model, E2 is found to restrict the Jak1‐Stat6 signaling pathway by keeping estrogen receptor beta (ERβ) away from interaction with ATPase‐coupling factor 6 (ATP5J), thus acting as an suppressor of macrophage alternative activation and tumor progression, whereas androgen has no significant role in HCC progression. 64 Furthermore, STAT6 signaling is crucial for invasion of EMT and CRC cells induced by IL‐4 and IL‐13. 128 Zip9 decreases phosphorylation of the IκBα/β pathway to inhibit M1 polarization and simultaneously increases phosphorylation of STAT6 to stimulate M2 polarization. 115 Exosome‐mediated ASO suppresses STAT6 expression in TAM and causes effective reprogramming of TAMs to the M1 phenotype in CT26 and Hepa1‐6 tumor model. 129 Basic leucine zipper ATF‐like transcription factor (BATF), involved in the synergistic induction of target gene expression, is further induced by co‐binding of STAT3 and STAT6, and high levels of BATF expressed from macrophages may contribute to tumor progression. 127 Consequently, targeting STAT3 and STAT6 pathways is an option for the treatment of HCC.

4.2. NF‐κB signaling pathway

NF‐κB is an important nuclear transcription factor. The NF‐κB family consists of five proteins, including RelA, c‐Rel, RelB, p50, and p52, which assemble into multiple homodimers and heterodimers, each with unique function in regulation of transcription in immune system cells and many other cell types. 130 It not only plays a specific part in inflammation, innate immunity, cancer, and apoptosis, 131 , 132 but also promotes hepatocarcinogenesis through liver inflammation, hepatocyte death, and compensatory proliferation. 133 NF‐κB activation is linked to the induction of carcinogenesis in a number of experimental types of inflammation‐associated malignancies, as it is a fundamental regulator of inflammation, particularly in TAM. 134 Through the study of HCC peritoneal tissue, the downregulation of SIRT4 can activate the NF‐κB pathway, leading to the downstream upregulation of MCP‐1 gene expression and increasing the infiltration of TAMs. 103 High‐mobility group A1 (HMGA1) elevates CCL2 expression in an NF‐κB‐dependent manner and induces the recruitment of macrophages in HCC. 73 Through the TLR9‐mediated NF‐κB signaling pathway, mitochondrial fission causes cytosolic mtDNA stress and increases CCL2 release in HCC cells, which in turn causes TAM recruitment and polarization. 62 RIP140 overexpression inhibits NF‐κB/IL‐6 axis activation, thereby suppressing M2 polarization to hinder hepatocellular carcinoma cell growth and proliferation. 97 , 98 S100A9 enhances the stem cell traits of HCC cells through the AGER/NF‐κB axis, 100 and Tim‐3 adds IL‐6 production by activating NF‐κB in macrophages, thereby promoting the growth of liver cancer cells. 109 Experiments have been conducted to reduce expression/activation of this transcription factor with different pharmacological approaches, thus restoring chemosensitivity. 135 Accordingly, transcription factors can be considered as effective drug targets for oncological diseases. However, the function of NF‐κB activation in TAM may differ based on the tumor microenvironment and tumor development stage. On the one hand, NF‐κB makes for cancer, and on the other hand, it hinders cancer. 132 TAMs frequently exhibit an anti‐inflammatory phenotype associated with immunosuppression that does not necessarily coincide with the pro‐inflammatory function of NF‐κB in TAMs. 134 Sirtuin 1 (SIRT1) enhances stimulation of NF‐κB pathway in macrophages and promotes TAM polarization to M1 tumor suppressor phenotype. 102 Through a positive feedback regulatory loop, MiR‐99b may enhance M1 macrophage function via NF‐κB, leading to elevated antigen presentation and phagocytosis while attenuating M2 polarization. 89 Due to the contradiction of NF‐κB between cancer inhibition and promotion, the complexity of the whole disease process should be taken into account, which means treatment strategies should be formulated carefully to avoid opposite effects.

4.3. Wnt signaling pathway

Hepatocytes that receive Wnt/β‐catenin signaling from the microenvironment have high tumor potential, and the activation of this signaling pathway plays an important role throughout liver regeneration and hepatocarcinogenesis during chronic liver injury. 136 , 137 Wnt/β‐catenin signaling pathway can also affect HCC through different impacts on TAMs. After THP‐1‐derived macrophages (THP‐1‐M) are incubated with 50% HCC‐TCM, M2‐type macrophage markers CD163, IL‐10, and CCR2 are upregulated, and further studies reveal that HCC‐TCM promotes M2 polarization through Wnt2b/β‐catenin signaling, inducing HCC to undergo EMT facilitating proliferation and migration. 110 Orthotopically inoculated hepatic Hepa1‐6 tumors in mice are accelerated by myeloid‐specific NOTCH blockade by conditional disruption of recombination signal binding protein Jκ (RBPj cKO), and NOTCH signaling is negatively correlated with WNT activation in CD68+ macrophages in patient‐derived HCC biopsies, while positively relevant to advanced HCC stages. 16 Consequently, NOTCH inhibition prevents moTAM differentiation while elevating Wnt/β‐catenin signaling to encourage kclTAM proliferation and pro‐tumor cytokine release, which speeds up the development of HCC and colorectal cancer's liver metastasis. 16 Besides, CA1P‐induced microtubule depolymerization mediates AKT inactivation, then activates GSK‐3β and downregulates Wnt/β‐Catenin signaling pathway and Mcl‐1, leading to the apoptosis of HepG2 cells; by the same mechanism, TAM apoptosis is induced, and secondary metastasis of TAMs partially rescues the growth of CA1P‐suppressed tumors. 60 In SMMC‐7721 hepatocellular carcinoma cells and nude mice subcutaneous tumor models, TNF‐α produced by TAMs promotes EMT and CSC via Wnt/β‐catenin pathway. 31 Another study finds that nuclear accumulation of β‐catenin is positively correlated with CD68 TAM in biopsies from HCC patients; in addition to β‐catenin, levels of Axin2 and c‐Myc are increased in M2, and Wnt/β‐catenin activation stimulates M2 macrophage polarization via c‐Myc. 114

4.4. CSF1/CSF1R axis

CSF1R is the core of many diseases and is expressed at a high level on TAMs. The CSF1/CSF1R axis helps to increase recruitment and infiltration of TAMs and promote the progression and metastasis of HCC. Overexpression of solute carrier family 7 member 11 (SLC7A11) in HCC cells causes metastasis of HCC by upregulating PD‐L1 and CSF1 via the αKG/HIF1α axis, simultaneously supporting infiltration of TAMs and MDSCs in tumors via the CSF1/CSF1R axis. 40 MiRNA is dysregulated in many types of malignant diseases. MiR‐148b deficiency induces CSF1 expression, which then binds to its receptor CSF1R to further induce macrophage infiltration into TME, thereby benefiting HCC metastasis and indicating poor prognosis. 138 Osteopontin (OPN), a prominent tumor‐maintaining inflammatory mediator linked with tumor progression, metastasis, and immunosuppression, contributes to chemotactic migration, M2 polarization of macrophages, and PD‐L1 expression in HCC by activating the CSF1‐CSF1R axis in macrophages. 93 In the H22 tumor‐bearing mouse model, the CSF1/CSF1R inhibitor PLX3397 diminishes macrophage recruitment and M2 polarization and cooperates with immunotherapy by reshaping the tumor immune microenvironment and enhancing infiltration of CD8+T cells and mature DC cells. 39 In a word, effective blockade of CSF1/CSF1R axis is the strong assistor of immunotherapy against tumors, and the synergistic effect of both will achieve better therapeutic effect.

4.5. Other signaling pathways/axes

Yes‐associated protein (YAP), lncRNA, and TAM are closely related, and their individuality and interaction are significantly meaningful for tumorigenesis, metastasis, treatment, and prognosis. 139 Nogo‐B is widely expressed in most tissues and is the only subtype of the family expressed in the liver. The tumor microenvironment assists Nogo‐B expression on macrophages to promote TAMs M2 polarization through initiating the Yap/Taz pathway, and verteporfin, an inhibitor of Yap, blocks Nogo‐B‐Yap/Taz‐mediated M2 polarization to inhibit HCC progression. 92 High expression of YAP in human HCC cells induces hepatocytes to secrete IL‐6 recruiting macrophages to the tumor, so suppression of YAP pathway blocks macrophage chemotaxis and infiltration in HepG2 xenograft tumors. 107 SPON2‐α4β1 integrin signaling activates RhoA and Rac1, adds F‐actin reorganization, and stimulates M1‐like macrophage recruitment to repress tumor metastasis, and at the same time, F‐actin buildup promotes YAP nuclear translocation, prevents LATS1 phosphorylation, and starts the production of downstream genes to activate the Hippo pathway. 140 What's more, lncRNA, in close contact with TAMs, can be expressed in TAMs and affect TAMs. LncRNA H19, positively correlated with in situ CD68+ TAM and induced by TAMs, motivates EMT and stem cells and accelerates HCC cell invasion through triggering the miR‐193b/MAPK1 axis. 120 Long non‐coding RNA GAS5 overexpression prevents M2‐like polarization of TAMs in SMCC‐7721 cells by regulating PTEN expression. 82 For this reason, further research on the oncogenic mechanisms of lncRNA, YAP, and TAM and the realization of multi‐target therapy will be conducive to alleviating tumor progression and improving patient survival.

Other signaling pathways/axes, such as PI3‐K and MAPK signaling pathway, 24 , 63 HMGB1/TLR2/NOX2/autophagy axis 65 and IFNG/IFR1/HHLA2 axis, 76 also regulate TAMs to act on HCC, offering a new direction for future anti‐tumor therapy.

5. IMPACT OF TUMOR MICROENVIRONMENT ON TAMS IN HCC

5.1. Hypoxia/metabolism

Tumor‐associated macrophages are influenced by different factors in the tumor microenvironment, facilitating tumor development, and in the meantime, developing resistance to anti‐tumor therapy. First of all, hypoxia is a hallmark of the solid tumor microenvironment, and tumor hypoxia is considered a major detrimental factor in cancer treatment. The median PO2 of normal liver tissue is 30 mmHg, while that of liver tumor tissue is 6 mmHg, 141 so liver cancer is one of the malignant tumors with severe hypoxia. Hypoxia‐inducible factor‐1 (HIF‐1), one of the primary mediators of the hypoxic response, can activate hypoxia‐responsive genes. High expression of HIF‐1 in the HCC microenvironment facilitates HCC cell growth and metastasis and is also connected with worse prognosis for hepatocellular carcinoma. 142 , 143 A study has demonstrated that under sustained and severe hypoxic conditions, M2 secretes more IL‐1β, which upregulates HIF‐1 via cyclooxygenase 2, thus leading to EMT and metastasis in HCC. 144 Hypoxia produces HMGB1 expression in human and mouse hepatocellular tumor cells in a HIF‐1α‐dependent manner and expands macrophage infiltration and reprogramming to elevate IL‐6 secretion, subsequently promoting EMT, invasion, and metastasis of HCC cells. 32 Moreover, TREM‐1 TAMs are abundant in hypoxic tissues of HCC, especially in advanced stage and impair CD8+T‐cell function, connected with poor prognosis, because TREM‐1+ TAMs increase chemokine (CC motif) ligand 20 expression through the extracellular signal‐regulated kinase/NF‐κβ pathway and recruit CCR6+Foxp3+Treg, causing immunosuppression. 20 TREM‐1+ TAMs have higher expression of programmed cell death ligand 1 (PD‐L1) under hypoxia as well. 20 Hypoxia can induce apoptosis, limit tumor size of HCC, and even make it disappear, but in the tumor microenvironment, it can also enhance the adaptive capacity of tumor cells in different ways toward malignant acceleration.

Hypoxia brings alterations in a range of metabolic pathways through HIF, consisting of glycolysis. In hypoxia, metabolism shifts from oxidative to glycolytic metabolism through HIF‐dependent upregulation of pyruvate dehydrogenase kinase 1 (PDK1) and lactate dehydrogenase A (LDHA). 145 However, even under aerobic conditions, tumor cells break down glucose into lactate via glycolysis to rapidly meet the energy requirements for cell proliferation, which is called the Warburg effect. In HCC cells, lactate, a byproduct of glycolysis, augments VEGF and arginase 1 (Arg1) expression through HIF‐1α and motivates M2 polarization. 146 An experiment in vivo has confirmed that high expression of ECT2 is a proven independent prognostic risk factor for HCC and may also promote M2 macrophage polarization by enhancing aerobic glycolysis and inhibiting immune cell function, which will cause HCC cells to proliferate and migrate. 147 Aerobic glycolysis motivates CA12 upregulation in macrophages through HIF1α and autocrine cytokine‐dependent pathways, which not only mediates macrophage survival in a relatively acidic tumor microenvironment, but also induces macrophages to generate large amounts of CCL8, thereby facilitating EMT and the metastasis of cancer cells. 37

In addition to glycolysis, fatty acid metabolism is another significant metabolic change in cancer cells. The dysregulated FA oxidation, sometimes referred to β‐oxidation, is another manifestation of dysregulated fatty acid metabolism in various cancers. 148 It has been shown that FAO inhibition is associated with the development of HCC, 149 , 150 and yet fatty acid oxidation can supply energy to tumors, playing a certain role in pro‐tumor function of TAM. In a mimic model in vitro, FAO is responsible for the upregulation of IL‐1β secretion in a reactive oxygen species and NLRP3‐dependent manner, mediating the migration of M2 MDM and boosting the proliferation, migration, and invasion of HCC cells. 66 Receptor‐interacting protein kinase 3 (RIPK3) is a central factor in necroptosis, and hence, RIPK3 deficiency not only decreases reactive oxygen species but also significantly inhibits caspase1‐mediated PPAR cleavage, promoting fatty acid metabolism containing fatty acid oxidation (FAO), inducing M2 polarization in the tumor microenvironment. 99 Metabolism in the tumor microenvironment provides energy for tumor growth and also plays a role of a facilitator in TAMs‐induced tumor progression.

5.2. Cytokine

Cytokines are also one of the essential elements of tumor microenvironment, which can be categorized as tumor necrosis factors, interferons, interleukins, colony‐stimulating factors, and more. There are various types and different functions, all of which are irreplaceable in the development of tumor. IL‐6, closely connected with STAT3 pathway, not only upregulates miR‐25‐3p through STAT3/c‐MYC signaling and targets 3′UTR to downregulate PTPRO expression in HCC monocytes, thus promoting PD‐L1 expression in macrophages to magnify T‐cell exhaustion in HCC, but also controls PTPRO expression through IFN‐γ‐dependent mechanism. 79 In contrast, IL‐37 hinders tumor growth by promoting the polarization of TAMs from M2 to M1 by blocking IL‐6/STAT3 signaling. 78 Furthermore, IFN‐α suppresses the M2 phenotype of TAM induced by IL‐13, making a decline in M2 markers and a significant rise in M1 markers and neutralizing the positive function on proliferation and invasion of HCC cells, and as a result, IFN‐α acts as an adjuvant therapy to sorafenib to develop its anti‐tumor capacity. 41 In conclusion, cytokines involved are important potential targets in the treatment of HCC, providing theoretical support for the generation of new treatment options.

5.3. Non‐coding RNA

Non‐coding RNAs have also been discovered to participate in the development of HCC by affecting TAMs. MicroRNA (miRNAs) are a large family of small molecule non‐coding single‐stranded RNAs, the universality and diversity of which suggest that they have many important biological functions. MiR‐98 mimics notably elevate the levels of IL‐1β and TNF‐α in TAM under hepatocellular carcinoma conditions, but significantly lower IL‐10 and TGF‐β, meaning that M2 turns into M1, thus repressing EMT, invasion, and migration of SMMC7721 and HepG2 cells. 88 Further study has revealed that IL‐10 is the target gene of miRNA‐98, and miRNA‐98 is directly linked to the 3′UTR of IL‐10 to inhibit HCC. 87 MiR‐144 and miR‐451a, respectively, target HGF and MIF to stimulate M1 polarization and form a negative feedback regulatory circuit with EZH2 in HCC, revealing the criticality of chromatin remodeling involved in DNA methylation‐induced gene silencing at the same time, regulating miR‐144/miR‐451a cluster expression and further controlling plasticity of TAM. 85 Long non‐coding RNA (lncRNA) GAS5, which is also involved in TAM polarization, constricts the proliferation and invasion of SMCC‐7721 cells by enhancing the expression of PTEN to suppress M2 and promote M1. 82

5.4. Exogenous drug

Current studies have discovered that some drugs originally applied in other diseases are also relevant for TAMs in HCC. Cholestyramine, a BA chelator for hyperlipidemia, reverses the polarization of M2 TAMs due to SIRT5 deficiency which demonstrates that metabolic dysregulation can cause the formation of an immunosuppressive tumor microenvironment conducive to the development of HCC. 61 Propofol, an intravenous anesthetic, has been injected into the abdominal cavity of tumor‐bearing mice at different doses, and the tumor size is significantly reduced in a dose‐dependent manner. 95 Low and medium doses mainly exert anti‐tumor activity, while high doses directly inhibit tumor growth. 95 It has been further found that propofol stimulates TAMs to secrete microvesicles (MVs) that transport miR‐142‐3p into HCC cells, then inhibiting tumor cell migration. 95 Statins, inhibitors of HMG‐CoA reductase, block the YAP pathway that stimulates IL‐6 expression, strongly impeding chemotaxis of TAMs. 107 Genipin inactivates inosital‐requiringenzyme‐1α (IRE1α), a key factor in macrophage initiation, which declines the initiation of TAMs and secretion of pro‐inflammatory factors, significantly reduces the migration and infiltration of TAMs and effectively constrains the in situ growth of HCC in vivo. 69 In short, more and more drugs have been testified to possess anti‐tumor function and may become candidates for tumor treatment in the future, but more research is still needed to verify in order to ensure their safety and efficacy.

6. SUMMARY AND FUTURE PERSPECTIVE

Tumor‐associated macrophages are gradually occupying an important place in studies on HCC. In this review, we summarize new developments in the mechanism of TAMs in HCC, as well as exploring implications of modulating TAMs in HCC drug resistance. We have demonstrated the influence of several major signaling pathways and multiple substances in the tumor microenvironment in the oncogenesis of TAMs, and at the same time, we have discussed the combination of multiple HCC therapies through modulating TAMs, which effectively mitigates drug resistance and enhances curative efficacy, with the promise of improved prognosis and better survival. There have been several reviews talking about origin, pathogenesis, or therapeutic strategies between TAM and HCC, 151 , 152 , 153 , 154 , 155 and this paper both completes some parts of them and detailing the crosstalk between TAM and HCC from a different perspective. This article complements them by making the relationship between TAM and HCC clearer, providing a more complete understanding of TAM and HCC, and offering valuable information for the future discovery of effective strategies for targeting TAM for the treatment of HCC. As the conclusions are mainly argued on various models, whether similar results can be obtained in humans needs further confirmation.

Most studies have shown that M2 TAMs are a major factor in carcinogenesis, with higher M2 indicating high drug resistance and predicting poor prognosis, while M1 exhibits anti‐tumor effects. Therefore, in terms of therapeutic strategies, research will focus on how to inhibit M2 recruitment, reduce the number of M2, or convert M2 into M1. However, is it true that the lower the density of M2 is, the greater the anti‐tumor effect will be? Does a higher M1/M2 ratio imply worse drug resistance and better prognosis? Nowadays, there are evidences that M1 has pro‐tumor function in HCC. 156 , 157 The function of M1 deserves further in‐depth study. Furthermore, the M1/M2 ratio should be in relative equilibrium like Th1/Th2 in order to maintain normal physiological function, and there may be maximum and minimum thresholds of the ratio for making a difference. To sum up, functional mechanisms of TAMs still require us to explore continually to furnish a theoretical basis for future cancer treatment, and meanwhile, the strategy of combination therapy is supposed to be widely carried out to achieve more satisfactory efficacy of anti‐tumor therapy.

AUTHOR CONTRIBUTIONS

Xinyi Zhang: Conceptualization (equal); investigation (lead); writing – original draft (lead). Chao Yu: Investigation (supporting); resources (equal). Siqi Zhao: Conceptualization (equal); resources (equal). Min Wang: Resources (equal). Longcheng Shang: Conceptualization (equal); supervision (equal). Jin Zhou: Supervision (equal); writing – review and editing (equal). Yong Ma: Supervision (equal); writing – review and editing (equal).

FUNDING INFORMATION

This work was supported by the National Natural Science Foundation of China (82203762), the Chen Xiaoping foundation for the development of science and technology of Hubei Province (CXPJJH12000009‐08), and the Xinghuo Talent Program of Nanjing First Hospital.

CONFLICT OF INTEREST STATEMENT

The authors declare that they have no competing interests.

Zhang X, Yu C, Zhao S, et al. The role of tumor‐associated macrophages in hepatocellular carcinoma progression: A narrative review. Cancer Med. 2023;12:22109‐22129. doi: 10.1002/cam4.6717

Xinyi Zhang and Chao Yu contributed equally to this work.

Contributor Information

Longcheng Shang, Email: njushanglc@126.com.

Jin Zhou, Email: georgenjmu@163.com.

Yong Ma, Email: yma0917@163.com.

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  • 1. Ugel S, de Sanctis F, Mandruzzato S, Bronte V. Tumor‐induced myeloid deviation: when myeloid‐derived suppressor cells meet tumor‐associated macrophages. J Clin Invest. 2015;125(9):3365‐3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Arvanitakis K, Koletsa T, Mitroulis I, Germanidis G. Tumor‐associated macrophages in hepatocellular carcinoma pathogenesis, prognosis and therapy. Cancers (Basel). 2022;14(1):226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Munir MT, Kay MK, Kang MH, et al. Tumor‐associated macrophages as multifaceted regulators of breast tumor growth. Int J Mol Sci. 2021;22(12):6526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Wang B, Tan B. Noncoding RNAs: regulating the crosstalk between tumor‐associated macrophages and gastrointestinal cancer. Biomed Pharmacother. 2022;153:113370. [DOI] [PubMed] [Google Scholar]
  • 5. Nowak M, Klink M. The role of tumor‐associated macrophages in the progression and chemoresistance of ovarian cancer. Cell. 2020;9(5):1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Guilliams M, Dutertre CA, Scott CL, et al. Unsupervised high‐dimensional analysis aligns dendritic cells across tissues and species. Immunity. 2016;45(3):669‐684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Huang H, Jiang J, Chen R, Lin Y, Chen H, Ling Q. The role of macrophage TAM receptor family in the acute‐to‐chronic progression of liver disease: from friend to foe? Liver Int. 2022;42(12):2620‐2631. [DOI] [PubMed] [Google Scholar]
  • 8. Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):7. [DOI] [PubMed] [Google Scholar]
  • 9. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Jayasingam SD, Citartan M, Thang TH, Mat Zin AA, Ang KC, Ch'ng ES. Evaluating the polarization of tumor‐associated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol. 2019;9:1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Roszer T. Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediators Inflamm. 2015;2015:816460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Wu JJ, Yang Y, Peng WT, Sun JC, Sun WY, Wei W. G protein‐coupled receptor kinase 2 regulating beta2‐adrenergic receptor signaling in M2‐polarized macrophages contributes to hepatocellular carcinoma progression. Onco Targets Ther. 2019;12:5499‐5513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Li XY, Shen Y, Zhang L, Guo X, Wu J. Understanding initiation and progression of hepatocellular carcinoma through single cell sequencing. Biochim Biophys Acta Rev Cancer. 2022;1877(3):188720. [DOI] [PubMed] [Google Scholar]
  • 14. Nasrollahzadeh E, Razi S, Keshavarz‐Fathi M, Mazzone M, Rezaei N. Pro‐tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother. 2020;69(9):1673‐1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kong L, Zhou Y, Bu H, Lv T, Shi Y, Yang J. Deletion of interleukin‐6 in monocytes/macrophages suppresses the initiation of hepatocellular carcinoma in mice. J Exp Clin Cancer Res. 2016;35(1):131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ye YC, Zhao JL, Lu YT, et al. NOTCH signaling via WNT regulates the proliferation of alternative, CCR2‐independent tumor‐associated macrophages in hepatocellular carcinoma. Cancer Res. 2019;79(16):4160‐4172. [DOI] [PubMed] [Google Scholar]
  • 17. Sung PS. Crosstalk between tumor‐associated macrophages and neighboring cells in hepatocellular carcinoma. Clin Mol Hepatol. 2022;28(3):333‐350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Wang J, Mi S, Ding M, Li X, Yuan S. Metabolism and polarization regulation of macrophages in the tumor microenvironment. Cancer Lett. 2022;543:215766. [DOI] [PubMed] [Google Scholar]
  • 19. Hasan MN, Capuk O, Patel SM, Sun D. The role of metabolic plasticity of tumor‐associated macrophages in shaping the tumor microenvironment immunity. Cancers (Basel). 2022;14(14):3331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Wu Q, Zhou W, Yin S, et al. Blocking triggering receptor expressed on myeloid Cells‐1‐positive tumor‐associated macrophages induced by hypoxia reverses immunosuppression and anti‐programmed cell death ligand 1 resistance in liver cancer. Hepatology. 2019;70(1):198‐214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Xun X, Zhang C, Wang S, et al. Cyclooxygenase‐2 expressed hepatocellular carcinoma induces cytotoxic T lymphocytes exhaustion through M2 macrophage polarization. Am J Transl Res. 2021;13(5):4360‐4375. [PMC free article] [PubMed] [Google Scholar]
  • 22. Xiao N, Zhu X, Li K, et al. Blocking siglec‐10(hi) tumor‐associated macrophages improves anti‐tumor immunity and enhances immunotherapy for hepatocellular carcinoma. Exp Hematol Oncol. 2021;10(1):36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ren X, Ju Y, Wang C, Wei R, Sun H, Zhang Q. MARCKS on tumor‐associated macrophages is correlated with immune infiltrates and poor prognosis in hepatocellular carcinoma. Cancer Invest. 2021;39(9):756‐768. [DOI] [PubMed] [Google Scholar]
  • 24. Wang B, Li Q, Wang J, et al. Plasmodium infection inhibits tumor angiogenesis through effects on tumor‐associated macrophages in a murine implanted hepatoma model. Cell Commun Signal. 2020;18(1):157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zhou DY, Qin J, Huang J, et al. Zoledronic acid inhibits infiltration of tumor‐associated macrophages and angiogenesis following transcatheter arterial chemoembolization in rat hepatocellular carcinoma models. Oncol Lett. 2017;14(4):4078‐4084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Chaffer CL, Thompson EW, Williams ED. Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs. 2007;185(1–3):7‐19. [DOI] [PubMed] [Google Scholar]
  • 27. Song W, Mazzieri R, Yang T, Gobe GC. Translational significance for tumor metastasis of tumor‐associated macrophages and epithelial‐mesenchymal transition. Front Immunol. 2017;8:1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 2018;13:395‐412. [DOI] [PubMed] [Google Scholar]
  • 29. Prager BC, Xie Q, Bao S, Rich JN. Cancer stem cells: the architects of the tumor ecosystem. Cell Stem Cell. 2019;24(1):41‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Fan QM, Jing YY, Yu GF, et al. Tumor‐associated macrophages promote cancer stem cell‐like properties via transforming growth factor‐beta1‐induced epithelial‐mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014;352(2):160‐168. [DOI] [PubMed] [Google Scholar]
  • 31. Chen Y, Wen H, Zhou C, 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. 2019;378(1):41‐50. [DOI] [PubMed] [Google Scholar]
  • 32. Jiang J, Wang GZ, Wang Y, Huang HZ, Li WT, Qu XD. Hypoxia‐induced HMGB1 expression of HCC promotes tumor invasiveness and metastasis via regulating macrophage‐derived IL‐6. Exp Cell Res. 2018;367(1):81‐88. [DOI] [PubMed] [Google Scholar]
  • 33. Chen S, Morine Y, Tokuda K, et al. Cancerassociated fibroblastinduced M2polarized macrophages promote hepatocellular carcinoma progression via the plasminogen activator inhibitor1 pathway. Int J Oncol. 2021;59(2):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Aras S, Zaidi MR. TAMeless traitors: macrophages in cancer progression and metastasis. Br J Cancer. 2017;117(11):1583‐1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Xu W, Cheng Y, Guo Y, Yao W, Qian H. Targeting tumor associated macrophages in hepatocellular carcinoma. Biochem Pharmacol. 2022;199:114990. [DOI] [PubMed] [Google Scholar]
  • 36. Wu H, Li Y, Shi G, et al. Hepatic interferon regulatory factor 8 expression suppresses hepatocellular carcinoma progression and enhances the response to anti‐programmed cell death protein‐1 therapy. Hepatology. 2022;76(6):1602‐1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Ning WR, Jiang D, Liu XC, et al. Carbonic anhydrase XII mediates the survival and prometastatic functions of macrophages in human hepatocellular carcinoma. J Clin Invest. 2022;132(7):e153110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Chu TH, Chan HH, Hu TH, et al. Celecoxib enhances the therapeutic efficacy of epirubicin for Novikoff hepatoma in rats. Cancer Med. 2018;7(6):2567‐2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wang Y, Wang Z, Jia F, et al. CXCR4‐guided liposomes regulating hypoxic and immunosuppressive microenvironment for sorafenib‐resistant tumor treatment. Bioact Mater. 2022;17:147‐161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. He Q, Liu M, Huang W, et al. IL‐1β‐induced elevation of solute carrier family 7 member 11 promotes hepatocellular carcinoma metastasis through up‐regulating programmed death ligand 1 and colony‐stimulating factor 1. Hepatology. 2021;74(6):3174‐3193. [DOI] [PubMed] [Google Scholar]
  • 41. Zhang Z, Zhu Y, Xu D, et al. IFN‐α facilitates the effect of sorafenib via shifting the M2‐like polarization of TAM in hepatocellular carcinoma. Am J Transl Res. 2021;13(1):301‐313. [PMC free article] [PubMed] [Google Scholar]
  • 42. Hao X, Zheng Z, Liu H, 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. 2022;56:102463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Xu G, Feng D, Yao Y, et al. Listeria‐based hepatocellular carcinoma vaccine facilitates anti‐PD‐1 therapy by regulating macrophage polarization. Oncogene. 2020;39(7):1429‐1444. [DOI] [PubMed] [Google Scholar]
  • 44. Hashimoto A, Sarker D, Reebye V, et al. Upregulation of C/EBPalpha inhibits suppressive activity of myeloid cells and potentiates antitumor response in mice and patients with cancer. Clin Cancer Res. 2021;27(21):5961‐5978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Fu XT, Song K, Zhou J, et al. Tumor‐associated macrophages modulate resistance to oxaliplatin via inducing autophagy in hepatocellular carcinoma. Cancer Cell Int. 2019;19:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Wu X, Luo H, Shi B, et al. Combined antitumor effects of Sorafenib and GPC3‐CAR T cells in mouse models of hepatocellular carcinoma. Mol Ther. 2019;27(8):1483‐1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Wang Q, Zheng K, Tan D, Liang G. TREM2 knockdown improves the therapeutic effect of PD‐1 blockade in hepatocellular carcinoma. Biochem Biophys Res Commun. 2022;636(Pt 1):140‐146. [DOI] [PubMed] [Google Scholar]
  • 48. Tang B, Zhu J, Wang Y, 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). 2022;10:e2203973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Chen J, Lin Z, Liu L, et al. GOLM1 exacerbates CD8(+) T cell suppression in hepatocellular carcinoma by promoting exosomal PD‐L1 transport into tumor‐associated macrophages. Signal Transduct Target Ther. 2021;6(1):397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Fan G, Wei X, Xu X. Is the era of sorafenib over? A review of the literature. Ther Adv Med Oncol. 2020;12:1758835920927602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Dong N, Shi X, Wang S, et al. M2 macrophages mediate sorafenib resistance by secreting HGF in a feed‐forward manner in hepatocellular carcinoma. Br J Cancer. 2019;121(1):22‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Deng L, He K, Pan Y, Wang H, Luo Y, Xia Q. The role of tumor‐associated macrophages in primary hepatocellular carcinoma and its related targeting therapy. Int J Med Sci. 2021;18(10):2109‐2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Wang D, Li X, Li J, et al. APOBEC3B interaction with PRC2 modulates microenvironment to promote HCC progression. Gut. 2019;68(10):1846‐1857. [DOI] [PubMed] [Google Scholar]
  • 54. Li C, Pan XY, Ma M, Zhao J, Zhao F, Lv YP. Astragalus polysacharin inhibits hepatocellular carcinoma‐like phenotypes in a murine HCC model through repression of M2 polarization of tumour‐associated macrophages. Pharm Biol. 2021;59(1):1533‐1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Li J, Xue J, Ling M, et al. MicroRNA‐15b in extracellular vesicles from arsenite‐treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1‐mediated Hippo pathway. Cancer Lett. 2021;497:137‐153. [DOI] [PubMed] [Google Scholar]
  • 56. Kang F‐B, Wang L, Li D, Zhang Y‐G, Sun D‐X. Hepatocellular carcinomas promote tumor‐associated macrophage M2‐polarization via increased B7‐H3 expression. Oncol Rep. 2015;33(1):274‐282. [DOI] [PubMed] [Google Scholar]
  • 57. Tan HY, Wang N, Man K, Tsao SW, Che CM, Feng Y. Autophagy‐induced RelB/p52 activation mediates tumour‐associated macrophage repolarisation and suppression of hepatocellular carcinoma by natural compound baicalin. Cell Death Dis. 2015;6(10):e1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Li X, Yao W, Yuan Y, et al. Targeting of tumour‐infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 2017;66(1):157‐167. [DOI] [PubMed] [Google Scholar]
  • 59. Sun S, Cui Y, Ren K, et al. 8‐bromo‐7‐methoxychrysin reversed M2 polarization of tumor‐associated macrophages induced by liver cancer stem‐like cells. Anticancer Agents Med Chem. 2017;17(2):286‐293. [DOI] [PubMed] [Google Scholar]
  • 60. Mao J, Wang D, Wang Z, 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. 2016;380(1):134‐143. [DOI] [PubMed] [Google Scholar]
  • 61. Sun R, Zhang Z, Bao R, et al. Loss of SIRT5 promotes bile acid‐induced immunosuppressive microenvironment and hepatocarcinogenesis. J Hepatol. 2022;77(2):453‐466. [DOI] [PubMed] [Google Scholar]
  • 62. Bao D, Zhao J, Zhou X, et al. Mitochondrial fission‐induced mtDNA stress promotes tumor‐associated macrophage infiltration and HCC progression. Oncogene. 2019;38(25):5007‐5020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Chen M, Lai R, Lin X, Chen W, Wu H, Zheng Q. Downregulation of triggering receptor expressed on myeloid cells 1 inhibits invasion and migration of liver cancer cells by mediating macrophage polarization. Oncol Rep. 2021;45(4):37. [DOI] [PubMed] [Google Scholar]
  • 64. Yang W, Lu Y, Xu Y, et al. Estrogen represses hepatocellular carcinoma (HCC) growth via inhibiting alternative activation of tumor‐associated macrophages (TAMs). J Biol Chem. 2012;287(48):40140‐40149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Shiau DJ, Kuo WT, Davuluri GVN, et al. Hepatocellular carcinoma‐derived high mobility group box 1 triggers M2 macrophage polarization via a TLR2/NOX2/autophagy axis. Sci Rep. 2020;10(1):13582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Zhang Q, Wang H, Mao C, et al. Fatty acid oxidation contributes to IL‐1beta secretion in M2 macrophages and promotes macrophage‐mediated tumor cell migration. Mol Immunol. 2018;94:27‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Xia L, Huang W, Tian D, et al. Forkhead box Q1 promotes hepatocellular carcinoma metastasis by transactivating ZEB2 and VersicanV1 expression. Hepatology. 2014;59(3):958‐973. [DOI] [PubMed] [Google Scholar]
  • 68. Zhu F, Li X, Jiang Y, et al. GdCl3 suppresses the malignant potential of hepatocellular carcinoma by inhibiting the expression of CD206 in tumor‐associated macrophages. Oncol Rep. 2015;34(5):2643‐2655. [DOI] [PubMed] [Google Scholar]
  • 69. Tan HY, Wang N, Tsao SW, Che CM, Yuen MF, Feng Y. IRE1alpha inhibition by natural compound genipin on tumour associated macrophages reduces growth of hepatocellular carcinoma. Oncotarget. 2016;7(28):43792‐43804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Wang J, Wang Y, Chu Y, et al. Tumor‐derived adenosine promotes macrophage proliferation in human hepatocellular carcinoma. J Hepatol. 2021;74(3):627‐637. [DOI] [PubMed] [Google Scholar]
  • 71. Wei C, Yang X, Liu N, et al. Tumor microenvironment regulation by the endoplasmic reticulum stress transmission mediator Golgi protein 73 in mice. Hepatology. 2019;70(3):851‐870. [DOI] [PubMed] [Google Scholar]
  • 72. Chen F, Yang L, Peng X, et al. Histone deacetylase 2 regulates STAT1‐dependent upregulation of atypical chemokine receptor 3 to induce M2 macrophage migration and immune escape in hepatocellular carcinoma. Mol Immunol. 2022;151:204‐217. [DOI] [PubMed] [Google Scholar]
  • 73. Chen J, Ji K, Gu L, Fang Y, Pan M, Tian S. HMGA1 promotes macrophage recruitment via activation of NF‐kappaB‐CCL2 signaling in hepatocellular carcinoma. J Immunol Res. 2022;2022:4727198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Deng L, Huang S, Chen B, et al. Tumor‐linked macrophages promote HCC development by mediating the CCAT1/let‐7b/HMGA2 signaling pathway. Onco Targets Ther. 2020;13:12829‐12843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Liu HT, Huang DA, Li MM, Liu HD, Guo K. HSF1: a mediator in metabolic alteration of hepatocellular carcinoma cells in cross‐talking with tumor‐associated macrophages. Am J Transl Res. 2019;11(8):5054‐5064. [PMC free article] [PubMed] [Google Scholar]
  • 76. Wang R, Guo H, Tang X, et al. Interferon gamma‐induced interferon regulatory factor 1 activates transcription of HHLA2 and induces immune escape of hepatocellular carcinoma cells. Inflammation. 2022;45(1):308‐330. [DOI] [PubMed] [Google Scholar]
  • 77. Chen H, Tang C, Tan C, et al. IL‐2 modulates TAMs derived exosomal MiRNAs to ameliorate hepatocellular carcinoma development and progression. J Oncol. 2022;2022:3445350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Zhang Z, Zhang J, He P, Han J, Sun C. Interleukin‐37 suppresses hepatocellular carcinoma growth through inhibiting M2 polarization of tumor‐associated macrophages. Mol Immunol. 2020;122:13‐20. [DOI] [PubMed] [Google Scholar]
  • 79. Zhang W, Liu Y, Yan Z, et al. IL‐6 promotes PD‐L1 expression in monocytes and macrophages by decreasing protein tyrosine phosphatase receptor type O expression in human hepatocellular carcinoma. J Immunother Cancer. 2020;8(1):e000285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Chen W, Hu M, Wei T, et al. IL‐1 receptor‐associated kinase 1 participates in the modulation of the NLRP3 inflammasome by tumor‐associated macrophages in hepatocellular carcinoma. J Gastrointest Oncol. 2022;13(3):1317‐1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Li G, Liu D, Kimchi ET, et al. Nanoliposome C6‐ceramide increases the anti‐tumor immune response and slows growth of liver tumors in mice. Gastroenterology. 2018;154(4):1024‐1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Wang X, Li FY, Zhao W, et al. Long non‐coding RNA GAS5 overexpression inhibits M2‐like polarization of tumour‐associated macrophages in SMCC‐7721 cells by promoting PTEN expression. Int J Exp Pathol. 2020;101(6):215‐222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Luo Y, Wang J, Xu L, et al. A theranostic metallodrug modulates immunovascular crosstalk to combat immunosuppressive liver cancer. Acta Biomater. 2022;154:478‐496. [DOI] [PubMed] [Google Scholar]
  • 84. Wang Y, Wang B, Xiao S, Li Y, Chen Q. miR‐125a/b inhibits tumor‐associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J Cell Biochem. 2019;120(3):3046‐3055. [DOI] [PubMed] [Google Scholar]
  • 85. Zhao J, Li H, Zhao S, et al. Epigenetic silencing of miR‐144/451a cluster contributes to HCC progression via paracrine HGF/MIF‐mediated TAM remodeling. Mol Cancer. 2021;20(1):46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Zhou SL, Hu ZQ, Zhou ZJ, et al. miR‐28‐5p‐IL‐34‐macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology. 2016;63(5):1560‐1575. [DOI] [PubMed] [Google Scholar]
  • 87. Li L, Sun P, Zhang C, Li Z, Zhou W. MiR‐98 suppresses the effects of tumor‐associated macrophages on promoting migration and invasion of hepatocellular carcinoma cells by regulating IL‐10. Biochimie. 2018;150:23‐30. [DOI] [PubMed] [Google Scholar]
  • 88. Li L, Sun P, Zhang C, Li Z, Cui K, Zhou W. MiR‐98 modulates macrophage polarization and suppresses the effects of tumor‐associated macrophages on promoting invasion and epithelial‐mesenchymal transition of hepatocellular carcinoma. Cancer Cell Int. 2018;18:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Wang L, Hu YY, Zhao JL, et al. Targeted delivery of miR‐99b reprograms tumor‐associated macrophage phenotype leading to tumor regression. J Immunother Cancer. 2020;8(2):e000517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Dhanasekaran R, Baylot V, Kim M, et al. MYC and Twist1 cooperate to drive metastasis by eliciting crosstalk between cancer and innate immunity. Elife. 2020;9:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Lu S, Gao Y, Huang X, Wang X. Cantharidin exerts anti‐hepatocellular carcinoma by miR‐214 modulating macrophage polarization. Int J Biol Sci. 2014;10(4):415‐425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zhao X, Wang X, You Y, et al. Nogo‐B fosters HCC progression by enhancing Yap/Taz‐mediated tumor‐associated macrophages M2 polarization. Exp Cell Res. 2020;391(1):111979. [DOI] [PubMed] [Google Scholar]
  • 93. Zhu Y, Yang J, Xu D, et al. Disruption of tumour‐associated macrophage trafficking by the osteopontin‐induced colony‐stimulating factor‐1 signalling sensitises hepatocellular carcinoma to anti‐PD‐L1 blockade. Gut. 2019;68(9):1653‐1666. [DOI] [PubMed] [Google Scholar]
  • 94. Hu J, Zhang M, Gui L, et al. PCSK9 suppresses M2‐like tumor‐associated macrophage polarization by regulating the secretion of OX40L from hepatocellular carcinoma cells. Immunol Invest. 2022;51(6):1678‐1693. [DOI] [PubMed] [Google Scholar]
  • 95. Zhang J, Shan WF, Jin TT, et al. Propofol exerts anti‐hepatocellular carcinoma by microvesicle‐mediated transfer of miR‐142‐3p from macrophage to cancer cells. J Transl Med. 2014;12:279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Zhou B, Li C, Yang Y, Wang Z. RIG‐I promotes cell death in hepatocellular carcinoma by inducing M1 polarization of perineal macrophages through the RIG‐I/MAVS/NF‐kappaB pathway. Onco Targets Ther. 2020;13:8783‐8794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Yi ZJ, Zhou Y, Zhang WF, et al. Over‐expression of receptor‐interacting protein 140 in tumor‐associated macrophages suppresses invasion and proliferation of hepatoma cells in vitro. Nan Fang Yi Ke Da Xue Xue Bao. 2016;36(11):1461‐1467. [PubMed] [Google Scholar]
  • 98. Hu YC, Yi ZJ, Zhou Y, et al. Overexpression of RIP140 suppresses the malignant potential of hepatocellular carcinoma by inhibiting NFkappaBmediated alternative polarization of macrophages. Oncol Rep. 2017;37(5):2971‐2979. [DOI] [PubMed] [Google Scholar]
  • 99. Wu L, Zhang X, Zheng L, et al. RIPK3 orchestrates fatty acid metabolism in tumor‐associated macrophages and hepatocarcinogenesis. Cancer Immunol Res. 2020;8(5):710‐721. [DOI] [PubMed] [Google Scholar]
  • 100. Wei R, Zhu WW, Yu GY, et al. S100 calcium‐binding protein A9 from tumor‐associated macrophage enhances cancer stem cell‐like properties of hepatocellular carcinoma. Int J Cancer. 2021;148(5):1233‐1244. [DOI] [PubMed] [Google Scholar]
  • 101. Hu ZQ, Zhou SL, Zhou ZJ, et al. Overexpression of semaphorin 3A promotes tumor progression and predicts poor prognosis in hepatocellular carcinoma after curative resection. Oncotarget. 2016;7(32):51733‐51746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Zhou B, Yang Y, Li C. SIRT1 inhibits hepatocellular carcinoma metastasis by promoting M1 macrophage polarization via NF‐kappaB pathway. Onco Targets Ther. 2019;12:2519‐2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Li Z, Li H, Zhao ZB, et al. SIRT4 silencing in tumor‐associated macrophages promotes HCC development via PPARdelta signalling‐mediated alternative activation of macrophages. J Exp Clin Cancer Res. 2019;38(1):469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Meng Q, Duan X, Yang Q, et al. SLAMF6/Ly108 promotes the development of hepatocellular carcinoma via facilitating macrophage M2 polarization. Oncol Lett. 2022;23(3):83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Wei X, Tang C, Lu X, et al. MiR‐101 targets DUSP1 to regulate the TGF‐beta secretion in sorafenib inhibits macrophage‐induced growth of hepatocarcinoma. Oncotarget. 2015;6(21):18389‐18405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Liu L, Zhang R, Deng J, et al. Construction of TME and identification of crosstalk between malignant cells and macrophages by SPP1 in hepatocellular carcinoma. Cancer Immunol Immunother. 2022;71(1):121‐136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Zhou TY, Zhou YL, Qian MJ, et al. Interleukin‐6 induced by YAP in hepatocellular carcinoma cells recruits tumor‐associated macrophages. J Pharmacol Sci. 2018;138(2):89‐95. [DOI] [PubMed] [Google Scholar]
  • 108. Chang Z, Zhang Q, Hu Q, Liu Y, Zhang L, Liu R. Tannins in Terminalia bellirica inhibits hepatocellular carcinoma growth via re‐educating tumor‐associated macrophages and restoring CD8(+)T cell function. Biomed Pharmacother. 2022;154:113543. [DOI] [PubMed] [Google Scholar]
  • 109. Yan W, Liu X, Ma H, et al. Tim‐3 fosters HCC development by enhancing TGF‐beta‐mediated alternative activation of macrophages. Gut. 2015;64(10):1593‐1604. [DOI] [PubMed] [Google Scholar]
  • 110. Jiang Y, Han Q, Zhao H, Zhang J. Promotion of epithelial‐mesenchymal transformation by hepatocellular carcinoma‐educated macrophages through Wnt2b/beta‐catenin/c‐Myc signaling and reprogramming glycolysis. J Exp Clin Cancer Res. 2021;40(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Wan S, Zhao E, Kryczek I, et al. Tumor‐associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 2014;147(6):1393‐1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Wang X, Tan Y, Zhang Y, et al. The novel glycyrrhetinic acid‐tetramethylpyrazine conjugate TOGA induces anti‐hepatocarcinogenesis by inhibiting the effects of tumor‐associated macrophages on tumor cells. Pharmacol Res. 2020;161:105233. [DOI] [PubMed] [Google Scholar]
  • 113. Zhang J, Zhang H, Ding X, et al. Crosstalk between macrophage‐derived PGE(2) and tumor UHRF1 drives hepatocellular carcinoma progression. Theranostics. 2022;12(8):3776‐3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Yang Y, Ye YC, Chen Y, et al. Crosstalk between hepatic tumor cells and macrophages via Wnt/beta‐catenin signaling promotes M2‐like macrophage polarization and reinforces tumor malignant behaviors. Cell Death Dis. 2018;9(8):793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Gou Y, Yang D, Tian T, et al. The transcription of ZIP9 is associated with the macrophage polarization and the pathogenesis of hepatocellular carcinoma. Front Immunol. 2022;13:725595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Wang HC, Haung LY, Wang CJ, et al. Tumor‐associated macrophages promote resistance of hepatocellular carcinoma cells against sorafenib by activating CXCR2 signaling. J Biomed Sci. 2022;29(1):99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Qu W, Qiao S, Liu L, et al. Dectin3 protects against hepatocellular carcinoma by regulating glycolysis of macrophages. Int Immunopharmacol. 2022;113(Pt A):109384. [DOI] [PubMed] [Google Scholar]
  • 118. Davuluri GVN, Chen CC, Chiu YC, et al. Autophagy drives galectin‐1 secretion from tumor‐associated macrophages facilitating hepatocellular carcinoma progression. Front Cell Dev Biol. 2021;9:741820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Xu M, Zhou C, Weng J, et al. Tumor associated macrophages‐derived exosomes facilitate hepatocellular carcinoma malignance by transferring lncMMPA to tumor cells and activating glycolysis pathway. J Exp Clin Cancer Res. 2022;41(1):253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Ye Y, Guo J, Xiao P, et al. 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. 2020;469:310‐322. [DOI] [PubMed] [Google Scholar]
  • 121. Ning J, Ye Y, Bu D, et al. Imbalance of TGF‐beta1/BMP‐7 pathways induced by M2‐polarized macrophages promotes hepatocellular carcinoma aggressiveness. Mol Ther. 2021;29(6):2067‐2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Wang L, Yi X, Xiao X, Zheng Q, Ma L, Li B. Exosomal miR‐628‐5p from M1 polarized macrophages hinders m6A modification of circFUT8 to suppress hepatocellular carcinoma progression. Cell Mol Biol Lett. 2022;27(1):106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Chen S, Zheng P, Wang W, et al. Abberent expression of NOR1 protein in tumor associated macrophages contributes to the development of DEN‐induced hepatocellular carcinoma. J Cell Physiol. 2018;233(6):5002‐5013. [DOI] [PubMed] [Google Scholar]
  • 124. Yao RR, Li JH, Zhang R, Chen RX, 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. 2018;16(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Medvedeva GF, Kuzmina DO, Nuzhina J, Shtil AA, Dukhinova MS. How macrophages become transcriptionally dysregulated: a hidden impact of antitumor therapy. Int J Mol Sci. 2021;22(5):2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Zhu X, Guo Q, Zou J, et al. MiR‐19a‐3p suppresses M1 macrophage polarization by inhibiting STAT1/IRF1 pathway. Front Pharmacol. 2021;12:614044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Gupta S, Jain A, Syed SN, et al. IL‐6 augments IL‐4‐induced polarization of primary human macrophages through synergy of STAT3, STAT6 and BATF transcription factors. Onco Targets Ther. 2018;7(10):e1494110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Verhoeven Y, Tilborghs S, Jacobs J, et al. The potential and controversy of targeting STAT family members in cancer. Semin Cancer Biol. 2020;60:41‐56. [DOI] [PubMed] [Google Scholar]
  • 129. Kamerkar S, Leng C, Burenkova O, et al. Exosome‐mediated genetic reprogramming of tumor‐associated macrophages by exoASO‐STAT6 leads to potent monotherapy antitumor activity. Sci Adv. 2022;8(7):eabj7002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Smale ST. Dimer‐specific regulatory mechanisms within the NF‐κB family of transcription factors. Immunol Rev. 2012;246(1):193‐204. [DOI] [PubMed] [Google Scholar]
  • 131. Haefner B. NF‐κB arresting a major culprit in cancer. Drug Discov Today. 2002;7(12):653‐663. [DOI] [PubMed] [Google Scholar]
  • 132. Vainer GW, Pikarsky E, Ben‐Neriah Y. Contradictory functions of NF‐kappaB in liver physiology and cancer. Cancer Lett. 2008;267(2):182‐188. [DOI] [PubMed] [Google Scholar]
  • 133. Yang YM, Kim SY, Seki E. Inflammation and liver cancer: molecular mechanisms and therapeutic targets. Semin Liver Dis. 2019;39(1):26‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Lawrence T. Macrophages and NF‐kappaB in cancer. Curr Top Microbiol Immunol. 2011;349:171‐184. [DOI] [PubMed] [Google Scholar]
  • 135. Labbozzetta M, Notarbartolo M, Poma P. Can NF‐kappaB be considered a valid drug target in neoplastic diseases? Our point of view. Int J Mol Sci. 2020;21(9):3070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Kurosaki S, Nakagawa H, Hayata Y, et al. Cell fate analysis of zone 3 hepatocytes in liver injury and tumorigenesis. JHEP Rep. 2021;3(4):100315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Wang W, Smits R, Hao H, He C. Wnt/beta‐catenin signaling in liver cancers. Cancers (Basel). 2019;11(7):926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Ke M, Zhang Z, Cong L, et al. MicroRNA‐148b‐colony‐stimulating factor‐1 signaling‐induced tumor‐associated macrophage infiltration promotes hepatocellular carcinoma metastasis. Biomed Pharmacother. 2019;120:109523. [DOI] [PubMed] [Google Scholar]
  • 139. Xu J, Liu XY, Zhang Q, et al. Crosstalk among YAP, LncRNA, and tumor‐associated macrophages in tumorigenesis development. Front Oncol. 2021;11:810893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Zhang YL, Li Q, Yang XM, et al. SPON2 promotes M1‐like macrophage recruitment and inhibits hepatocellular carcinoma metastasis by distinct integrin‐rho GTPase‐hippo pathways. Cancer Res. 2018;78(9):2305‐2317. [DOI] [PubMed] [Google Scholar]
  • 141. McKeown SR. Defining normoxia, physoxia and hypoxia in tumours‐implications for treatment response. Br J Radiol. 2014;87(1035):20130676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Luo D, Wang Z, Wu J, Jiang C, Wu J. The role of hypoxia inducible factor‐1 in hepatocellular carcinoma. Biomed Res Int. 2014;2014:409272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Chen H, Chen J, Yuan H, Li X, Li W. Hypoxia‐inducible factor‐1alpha: a critical target for inhibiting the metastasis of hepatocellular carcinoma. Oncol Lett. 2022;24(2):284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Zhang J, Zhang Q, Lou Y, et al. Hypoxia‐inducible factor‐1alpha/interleukin‐1beta signaling enhances hepatoma epithelial‐mesenchymal transition through macrophages in a hypoxic‐inflammatory microenvironment. Hepatology. 2018;67(5):1872‐1889. [DOI] [PubMed] [Google Scholar]
  • 145. Bao MH, Wong CC. Hypoxia, metabolic reprogramming, and drug resistance in liver cancer. Cell. 2021;10(7):1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Colegio OR, Chu NQ, Szabo AL, et al. Functional polarization of tumour‐associated macrophages by tumour‐derived lactic acid. Nature. 2014;513(7519):559‐563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Xu D, Wang Y, Wu J, et al. ECT2 overexpression promotes the polarization of tumor‐associated macrophages in hepatocellular carcinoma via the ECT2/PLK1/PTEN pathway. Cell Death Dis. 2021;12(2):162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Wang M, Han J, Xing H, et al. Dysregulated fatty acid metabolism in hepatocellular carcinoma. Hepat Oncol. 2016;3(4):241‐251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Huang J, Viswakarma N, Yu S, et al. Progressive endoplasmic reticulum stress contributes to hepatocarcinogenesis in fatty Acyl‐CoA oxidase 1–deficient mice. Am J Pathol. 2011;179(2):703‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Li J, Huang Q, Long X, et al. CD147 reprograms fatty acid metabolism in hepatocellular carcinoma cells through Akt/mTOR/SREBP1c and P38/PPARalpha pathways. J Hepatol. 2015;63(6):1378‐1389. [DOI] [PubMed] [Google Scholar]
  • 151. Zhou D, Luan J, Huang C, Li J. Tumor‐associated macrophages in hepatocellular carcinoma: friend or foe? Gut Liver. 2021;15(4):500‐516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor‐associated macrophages in tumor progression: implications on therapeutic strategies. Exp Hematol Oncol. 2022;11(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Cheng K, Cai N, Zhu J, Yang X, Liang H, Zhang W. Tumor‐associated macrophages in liver cancer: from mechanisms to therapy. Cancer Commun (Lond). 2022;42(11):1112‐1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Zheng H, Peng X, Yang S, et al. Targeting tumor‐associated macrophages in hepatocellular carcinoma: biology, strategy, and immunotherapy. Cell Death Discov. 2023;9(1):65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Huang Y, Ge W, Zhou J, Gao B, Qian X, Wang W. The role of tumor associated macrophages in hepatocellular carcinoma. J Cancer. 2021;12(5):1284‐1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Zong Z, Zou J, Mao R, et al. M1 macrophages induce PD‐L1 expression in hepatocellular carcinoma cells through IL‐1beta signaling. Front Immunol. 2019;10:1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Wang H, Wang X, Li X, et al. CD68(+)HLA‐DR(+) M1‐like macrophages promote motility of HCC cells via NF‐kappaB/FAK pathway. Cancer Lett. 2014;345(1):91‐99. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from Cancer Medicine are provided here courtesy of Wiley

RESOURCES