Open Access

Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review)

  • Authors:
    • Chenglin Lu
    • Ying Liu
    • Linxuan Miao
    • Xiangle Kong
    • Huili Li
    • Haoran Chen
    • Xu Zhao
    • Bin Zhang
    • Xiaonan Cui
  • View Affiliations

  • Published online on: December 7, 2023     https://doi.org/10.3892/ijo.2023.5599
  • Article Number: 11
  • Copyright: © Lu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The tumor microenvironment (TME) is a complex system composed mainly of tumor cells, mesenchymal cells and immune cells. Macrophages, also known as tumor‑associated macrophages (TAMs), among innate immune cells, are some of the most abundant components of the TME. They may influence tumor growth and metastasis through interactions with other cell populations in the TME and have been associated with poor prognosis in a variety of tumors. Therefore, a better understanding of the role of TAMs in the TME may provide new insight into tumor therapy. In the present review, the origin and classification of TAMs in the TME were outlined and their polarization and dual effects on tumor cells, as well as emerging strategies for cancer therapies targeting TAMs, were discussed.

1. Introduction

Malignant tumor is a major health problem worldwide and a leading cause of death, ranking second only to cardiovascular disease. Immunotherapy based on the tumor microenvironment (TME) has become a promising cancer treatment strategy (1). The TME is a complex system composed mainly of tumor cells, mesenchymal cells, innate immune cells [macrophages, monocytes, natural killer (NK) cells, dendritic cells and myeloid derived suppressor cells] and adaptive immune cells (T cells and B cells). Macrophages, also known as tumor-associated macrophages (TAMs), among innate immune cells [macrophages, monocytes, natural killer (NK) cells, dendritic cells and myeloid derived suppressor cells], which are found in almost all tissues and organs, are the first line of defense against exogenous and endogenous injury- or pathogen-associated molecular patterns (2). Macrophages that infiltrate tumor tissue or congregate in the microenvironment of solid tumors are defined as tumor-associated macrophages (TAMs). As an important part of the TME, macrophages have a complex role in tumorigenesis and tumor progression. They can not only inhibit tumor growth by releasing pro-inflammatory cytokines and exerting cytotoxic activities, but also promote tumor progression by affecting the occurrence and growth of tumor cells, participating in tumor angiogenesis and metastasis, and shaping an immunosuppressive microenvironment (3). In the present review, the heterogeneity of the origin of TAMs, the factors affecting the polarization of TAMs and the complex role of TAMs in tumor progression were summarized, and therapeutic approaches targeting TAMs, such as consuming TAMs or re-educating TAMs were discussed in order to provide a reference for colleagues to gain insight for tumor immunotherapy.

2. Origin and classification of TAMs

Origin

It is generally thought that TAMs are mainly derived from monocytes produced by hematopoietic stem cells in bone marrow (4). However, recent evidence suggested that certain TAMs (such as alveolar macrophages, brain macrophages and liver macrophages) originate from pre-natal embryonic precursors (yolk sac or fetal liver) (5). These cells were recruited into the TME under the action of chemokines [such as C-C motif ligand 2 (CCL2), CCL3, CCL4 and CXCL12], colony-stimulating factor 1 (CSF-1), interleukin-6 (IL-6), IL-1β and vascular endothelial growth factor (VEGF) produced by tumor cells or stromal cells to become TAMs (6,7). As an important part of the TME, these TAMs can affect tumorigenesis and development, tumor angiogenesis and immune regulation through interactions with other cell populations in the TME. Furthermore, TAMs are also associated with poor prognosis of various tumors, such as breast cancer (8), bladder cancer (9), head and neck neoplasm (10), glioma (11), melanoma (12) and prostate cancer (13). However, more recently, it has been found that high macrophage infiltration is associated with better prognosis in colorectal and gastric cancers (14). This opposite effect may be related to the plasticity of macrophages and the resulting heterogeneity in phenotype and function of various cancers.

Classification

Macrophages are highly plastic and their phenotype and function are regulated by the surrounding microenvironment. Macrophages usually exist in two distinct subsets: Classically activated macrophages (M1) and alternately activated macrophages (M2). M1 macrophages, normally recognized and induced by type I T-helper cell (Th1) cytokines [e.g. IFN-γ and tumor necrosis factor (TNF)-α] or bacterial lipopolysaccharide (LPS), secrete pro-inflammatory cytokines such as IL-12 and TNF-α, produce high levels of nitric oxide (NO) and reactive oxygen species (ROS), and have powerful anti-microbial and anti-tumor activities (15). By contrast, M2 macrophages are activated by Th2 cytokines (IL-4 and IL-13), secrete anti-inflammatory cytokines such as IL-10, IL-13 and IL-4, and express abundant arginase-1 (Arg-1), mannose receptor (CD206) and scavenger receptor (CD163), which have the functions of removing debris, promoting angiogenesis, tissue reconstruction, damage repair, as well as promoting tumorigenesis and development (16). However, depending on the different activation stimuli, M2 macrophages can be further divided into four distinct subgroups, including M2a, M2b, M2c and M2d. The M2a subgroup is induced by IL-4 and IL-13 to produce high levels of CD206, decoy receptor IL-1 receptor II and IL-1 receptor antagonist (17,18). The M2b subgroup can be induced by immune complexes combined with IL-1β or LPS, which produces anti-inflammatory and pro-inflammatory cytokines IL-10, IL-1β, IL-6 and TNF-α (17,18). The M2c subgroup is generally induced by glucocorticoids and IL-10, releases large amounts of IL-10 and TGF-β and exhibits strong anti-inflammatory activity against apoptotic cells (17,19). Finally, the M2d subgroup, induced by Toll-like receptor (TLR) agonists via adenosine receptors, inhibits the production of pro-inflammatory cytokines and induces the secretion of anti-inflammatory cytokines and VEGFs (20-22). Overall, M2a and M2b macrophages have an immunomodulatory role and promote helper T-cell responses, while M2c macrophages are associated with immune response suppression and tissue remodeling. M2d macrophages are involved in angiogenesis and tumor progression (17,23) (Fig. 1).

3. Polarization of macrophages

The factors influencing TAM polarization are diverse and regulated by a variety of signals in tumor cells and stromal cells in the TME, mainly including the following factors, immunogenic signals, hypoxia (tumor-derived metabolic signals) and extracellular matrix (ECM) components.

Immunogenic signals

Immunogenic signaling mainly refers to the cytokines, chemokines and growth factors released by tumor cells, stromal cells and other infiltrating cells in the microenvironment, which are key determinants of TAM polarization. Among these factors, CCL2 and CSF-1 are the most well-studied stimulators. Studies have shown that tumor-derived chemokine CCL2 binds to C-C chemokine receptor 2 (CCR2) expressed on the surface of macrophages to polarize macrophages towards the pro-tumor phenotype. Blocking the CCL2-CCR2 interaction by gene ablation or antibodies was observed to significantly inhibit tumor metastasis, prolong the survival of tumor-bearing mice and reduce the expression of pro-tumor cytokines (24-26). Later, it was proved that, in addition to the influence of CCL2 derived from tumor cells on the polarization of macrophages, the chemotactic signal of CCL2 expressed by tumor-associated fibroblasts could also recruit macrophages to the tumor site and drive the polarization of macrophages towards the pro-tumor phenotype to increase the aggressiveness of cancer cells and the occurrence of breast tumors (27). CSF-1, another important factor affecting the polarization of macrophages, is generally widely overexpressed at the invasive margins of various tumors and is associated with tumor progression (15). A study on follicular lymphoma found that CSF-1 derived from tumor cells promoted the polarization of macrophages toward pro-tumor M2-like phenotypes and the use of CSF-1R inhibitor (PLX3397) resulted in the repolarization of macrophages toward the M1-type, which showed synergistic anti-tumor effects when combined with anti-CD20 rituximab (28). In addition to the above factors, there are other factors involved in the induction of macrophage polarization, such as VEGF-A, epidermal growth factor (EGF) and prostaglandin E2 (29-31).

Hypoxia

Hypoxia, as one of the key drivers of macrophage recruitment and polarization in the TME, is caused by tumor cells with vigorous metabolism and rapid growth but poor vascular organization, which is a common feature of most solid tumors (32). Hypoxia can regulate the phenotype of TAMs through various factors, particularly through lactic acid produced by glycolysis affecting macrophage polarization (33). For instance, a study on gastric cancer showed that lactic acid produced by glycolysis can induce changes in the macrophage phenotype through monocarboxylate channel transporter-hypoxia-inducible factor 1α signaling, polarizing macrophages towards an M2-like state (34). In addition, the hypoxic TME can stimulate the secretion of tumor-derived exosomes, regulate the macrophage phenotype and promote tumor development. For instance, exosomes of hypoxic tumor cells are enriched with immunomodulatory proteins and chemokines, including CSF-1, CCL2, ferritin heavy chain, ferritin light chain and TGF-β, which influence macrophage recruitment and promote macrophage polarization to the M2 phenotype (35). Therefore, the hypoxic microenvironment can shape a specific macrophage phenotype, which promotes immune escape and metastasis of tumor cells.

ECM components

Last but not least, the ECM component of the TME also has a regulatory effect on macrophage polarization. The ECM is a highly dynamic and complex macromolecular network consisting of a variety of fibrins, proteoglycans and stromal cell-associated proteins (36). The ECM molecule, elastin microfibril interfacer 2 (EMILIN-2), which belongs to the EDEN protein family, exerts a tumor-suppressive effect by affecting the polarization state of macrophages in a variety of tumors (37,38). In a study on colorectal cancer, EMILIN-2 was found to promote M1 polarization through activation of the TLR-4/myeloid differentiation factor-88/NF-κB signaling pathway. EMILIN-2 deficiency is associated with increased M2 macrophage infiltration (39). This suggests that certain components of the ECM are key regulators of the tumor-associated inflammatory environment and may represent promising prognostic biomarkers for tumor patients. In summary, the polarization of TAMs is regulated by complex biological networks, which is closely related to cancer development. Understanding the mechanisms of macrophage polarization may enable researchers to manipulate the macrophage polarization status to stimulate their anti-tumor potential for therapeutic purposes.

4. Role of TAMs in tumorigenesis and tumor progression

Inhibition of tumor development

TAM is a key player in the interaction between cancer cells and their microenvironment and has a dual potential in tumorigenesis and development. As a tumor suppressor, M1-type macrophages have high cytotoxicity and immunostimulatory effects against tumor cells, and can kill tumor cells through two different mechanisms. One is that M1-type macrophages directly mediate the cytotoxic effect of killing tumor cells, i.e., macrophages directly target infected cells or tumor cells by releasing lysosomal enzymes or cytotoxic molecules (such as ROS and NO), which is a slow process (40). Another way to kill tumors is antibody-dependent cell-mediated cytotoxicity, which usually requires the involvement of anti-tumor antibodies to kill tumor cells in a short period of time (41). Therefore, M1-type macrophages are considered anti-tumor or 'good' macrophages (Fig. 2).

Promotion of tumor development

In contrast, in most formed tumors, macrophages contribute to cancer initiation, progression and metastasis through a variety of mechanisms, including promoting cancer cell survival and proliferation, angiogenesis and suppression of immune responses (Fig. 2).

Effects on tumorigenesis and proliferation

TAMs not only provide structural support within the tumor stroma but also promote tumorigenesis and tumor cell growth by producing growth factors, cytokines and chemokines such as EGF, platelet-derived growth factor, TGF-β, hepatocyte growth factor, basic fibroblast growth factor, IL-10 and other cytokines, e.g. CXCL, CCL and VEGF (42). For example, M2-type macrophages have a promoting role in the proliferation and invasion of oral squamous cell carcinoma through the production of EGF and the number of CD206+ TAMs is positively correlated with a poorer clinical prognosis in oral squamous cell carcinoma (43). Furthermore, in a study on clear cell renal cell carcinoma (ccRCC), it was found that TAM-derived chemokine CCL5 can promote tumor cell proliferation and the formation of an immunosuppressive TME, which is closely related to the poor prognosis for patients with ccRCC (44). In addition to supporting tumor cell growth, TAM has also been found to have a role in supporting the growth of cancer stem cells (CSCs). In a recent study on breast cancer, TAM-derived IL-6 was found to regulate the enrichment of CSCs in breast cancer through the STAT-3 pathway and lead to tumor growth (45) (Fig. 2).

Effects on tumor angiogenesis

Angiogenesis is essential for tumor growth and metastasis, which is considered a 'hallmark' of cancer. An increasing amount of evidence indicates that TAMs are closely related to angiogenesis in tumors (46). On the one hand, TAMs participate in angiogenesis by secreting pro-angiogenic factors, including VEGF-A, matrix metalloproteinase (MMP), EGF, TGF-β, TNF-α, CCL2, CXCL8 and CXCL12 (47). For example, in a study on bladder cancer, TAM-derived CXCL8 was found to be highly associated with tumor migration, invasion and angiogenesis (48). In addition, the release of thymine phosphorylase (TP) and urine-stimulated plasminogen activator by TAMs can stimulate the migration of endothelial cells, increase ECM degradation and indirectly promote tumor angiogenesis (49). Studies have found that macrophage-derived TP is significantly associated with tumor angiogenesis and poor prognosis in gastric cancer (50). In addition, TAMs also promote tumor angiogenesis by secreting inflammatory mediators such as IL-1 and IL-6. A study on breast cancer found that TAM-derived IL-6 affected breast cancer cell migration and angiogenesis and induced CSC populations, leading to tumor growth (51,52). In summary, these studies suggest that TAMs promote tumor vascularization in different ways and are closely related to tumor progression (Fig. 2).

Effects on tumor metastasis

Macrophages have an important role in every step of the metastasis process, including preparation for pre-metastatic niche formation, intravasation, survival of circulating tumor cells, extravasation and invasion. In terms of forming a pre-metastatic niche, macrophages are recruited to the pre-metastatic site in response to various factors secreted by tumor cells, which provide a roadmap for the homing of circulating tumor cells to the pre-metastatic niche through enhanced expression of chemokines and secretion of molecules such as MMPs and integrins (53-55). In terms of intravasation, macrophages can decompose the surrounding matrix by secreting various proteolytic enzymes to promote the intravasation of tumor cells. For instance, Gocheva et al (56) found that IL-4 induces cathepsin activity in TAMs, promoting tumor growth and invasion. In terms of circulating tumor cell survival, macrophages secrete chemokines or cytokines to promote the successful survival of numerous tumor cells at metastatic sites. A study on breast cancer found that macrophages bind to vascular cell adhesion molecule-1 on the surface of tumor cells via α4 integrin, triggering the PI3K/Akt survival signaling pathway in cancer cells and protecting tumor cells from pro-apoptotic cytokines (57). In terms of extravasation, when tumor cells settle in the capillaries of the target organ, they attempt to attach and extrude through the vessel wall with the assistance of macrophages, and the extravasation rate decreases significantly after the loss of macrophages, indicating that macrophages have an important role in promoting the extravasation of circulating tumor cells (58,59). In terms of invasion, TAM contributes to tumor invasion and metastasis mainly through epithelial-mesenchymal transformation (EMT). A recent study found that CCL2, secreted by TAMs, promotes EMT in triple-negative breast cancer (TNBC) cells through activating the AKT/β-catenin signaling pathway, which may provide a new strategy for the diagnosis and treatment of TNBC (60) (Fig. 2).

Effects on tumor immunosuppressive microenvironment

In addition to their tumor-killing role, TAMs can also mediate immunosuppression, reshape the tumor immune microenvironment and promote tumor development. On the one hand, TAMs can express ligands of inhibitory receptors [such as programmed cell death protein-1 (PD-1) ligand 1, CD80/CD86 and death receptor ligands Fas ligand or tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)], which bind to the immune cell surface receptors PD-1, cytotoxic T-lymphocyte antigen-4, FaS and TRAIL-RI/-RII, thereby inhibiting the anti-tumor effects of immune cells (such as T cells and NK cells) (61,62). On the other hand, TAMs can also form an immunosuppressive microenvironment by producing chemokines, cytokines and enzymes. For instance, a study in ovarian cancer found that the TAM-secreted chemokine CCL22 recruited CCR4 + T-regulatory cells (Tregs) to promote an immunosuppressive microenvironment (63). It has also been observed in a mouse model of colorectal cancer that CCL20, a TAM-derived chemokine, recruited CCR6(+) Treg cells to the tumor mass, creating an immunosuppressive microenvironment that promoted tumor development (64). In addition, Xu et al (9) found that TAM-derived cytokine IL-10 was associated with the depletion of CD8+ T cells and dysfunction of NK cells in the tumor immune microenvironment, which led to poor prognosis in patients with bladder cancer. Recently, a study on pancreatic cancer found that TAM-derived Arg-1 drives immunosuppression by depleting arginine and inhibiting T-cell activation (65,66). In conclusion, these findings support the immunomodulatory role of TAM in promoting tumor progression by shaping the immunosuppressive microenvironment (Fig. 2).

5. Therapeutics targeting macrophages

TAMs are abundant in the TME of most cancer types and are commonly associated with poor clinical prognosis for cancer patients. TAMs are becoming a key target for immunotherapy and the different approaches targeting TAMs that have been explored may be broadly divided into three main categories: i) Eliminating TAMs already present in the TME; ii) inhibiting monocyte recruitment; iii) reprogramming of TAMs (67,68). These strategies have been investigated in preclinical models and some of them have been translated into clinical studies as adjuncts to immunotherapy (69). In the present review, some of the current approaches to targeting macrophages and clinical trials were outlined and the potential advantages and disadvantages of these approaches were discussed (Table I).

Table I

Clinical trials of agents targeting TAMs for cancer treatment.

Table I

Clinical trials of agents targeting TAMs for cancer treatment.

MechanismTAM-targeted agentCompoundClinical trial phaseClinical trial numbers
EliminationZoledronate acidZoledronate acidI/IINCT00588913
NCT00582790
NCT00278434
NCT03664687
TrabectedinTrabectedinIINCT02194231
NCT01339754
Recruitment inhibitionCCR2 antagonistPF04136309IbNCT02732938
NCT01413022
CCL2 antibodyCNTO888IbNCT01204996
NCT00537368
NCT00992186
CCX872IbNCT02345408
CCR2/5 inhibitorBMS-813160IINCT04123379
NCT03496662
CSF-1R antibodyRG7155I/IIINCT01494688
NCT05417789
PLX3397II/IIINCT01217229
NCT02371369
VimseltinibIIINCT05059262
ReprogrammingCD24CD24FcI/IINCT04060407
NCT04552704
CD47 antibodyHu5F9-G4I/IINCT02216409
NCT02953509
NCT03558139
NCT02953782
CC90002INCT02641002
NCT02367196
SIRP antibodyTTI-621I/IINCT02663518
NCT05507541
CC-95251INCT03783403
NCT05168202
PI3Kγ inhibitorIPI-549Ib/IINCT02637531
NCT03961698
TLR agonistImiquimodIINCT00899574
MotolimodIINCT01836029
NKTR-262I/IINCT03435640
CD40 agonistAPX005MI/IINCT03214250
RO7009789INCT02665416
SEA-CD40INCT02376699
CP-870893INCT01103635

[i] CCL2, C-C motif ligand 2; CCR2, C-C motif receptor 2; TLR, Toll-like receptor; SIRP, signal-regulatory protein; CSF, colony-stimulating factor.

Consumption of macrophages already present in the TME

Selective elimination of TAMs has been used in cancer treatment. An attractive strategy for depleting TAMs in the TME is to trigger its apoptosis and restore local immune surveillance in the TME, which can effectively inhibit tumor growth. Several compounds have been shown to induce apoptosis in macrophages, mainly including bisphosphonates and trabectedin.

Bisphosphonates are a class of anti-bone resorption drugs, which can be divided into two categories according to their structural characteristics: Non-nitrogenous bisphosphonates and nitrogenous bisphosphonates (70). Bisphosphonates exhibit direct or indirect antitumor properties. They can inhibit cancer cell proliferation, induce tumor cell apoptosis, block angiogenesis and interfere with immune surveillance. At the same time, bisphosphonates can also inhibit the proliferation, migration and invasion of macrophages, leading to the apoptosis of macrophages (71,72). For instance, in earlier studies, dichloromethylenediphosphonates (also known as clodronates) from the non-nitrogenous bisphosphonate family were often used to consume macrophages in the liver and spleen when loaded with liposomes (73); Zoledronate, the third generation of nitrogenous bisphosphonates, is selectively cytotoxic to TAMs expressing MMP9 and inhibits the progression of cervical cancer (74). In addition, a study of non-small cell lung cancer found that calcium zoledronate nanoparticles modified with biotin and mannose preferentially targeted biotin-expressing tumor cells and mannose-expressing TAMs, ultimately suppressing tumor growth and survival (75). A study about thyroid cancer found that zoledronic acid inhibits thyroid cancer stemness and metastasis by repressing M2-like TAM polarization and the Wnt/β-catenin pathway, reducing the tumor burden (76). Furthermore, a prospective phase II clinical trial found that zoledronic acid combined with radiotherapy reduced bone pain and improved quality of life in patients with bone metastases from gastrointestinal tumors (77). Trabectedin is a tetrahydroisoquinoline alkaloid that directly kills tumor cells by interfering with multiple transcription factors, DNA-binding proteins and DNA repair pathways (78). Furthermore, it also selectively consumes monocytes and macrophages in the TME by activating caspase 8 through a TNF-related apoptosis-inducing ligand-dependent mechanism (79). In a study on fibrosarcoma, Trabectedin selectively reduced macrophages in the TME and enhanced the anti-tumor response to anti-PD-1 therapy (80). In several clinical trials, Trabectedin was found to show good safety and efficacy in the treatment of soft tissue sarcoma and ovarian cancer (81,82). However, disappointing results were obtained in malignant pleural mesothelioma and pancreatic cancer tumors (83,84).

Macrophages have an important regulatory role in maintaining host defenses and tissue homeostasis, and a major problem with the depletion of macrophages is the inability to avoid the side effects that arise from non-selective macrophage depletion. Therefore, the key to minimizing potential toxic side effects is to develop drugs that preferentially target M2-like macrophages. A recent study designed M2-targeting nanoliposomes, which effectively depleted M2-type TAMs, remodeled the TME and effectively inhibited tumor growth (85). Consequently, targeted elimination of M2-like TAMs is a promising approach for cancer immunotherapy.

Inhibition of monocyte recruitment

As mentioned earlier, most TAMs originate from the production of bone marrow monocytes. TAMs are recruited to tumor sites in response to tumor-derived chemokines. Therefore, the application of monoclonal antibodies or small molecule inhibitors to interfere with chemokine signaling may be an effective way to prevent the accumulation of TAMs in the TME.

The expansion of TAMs in tumors is usually mediated by monocyte recruitment on the CCL2-CCR2 axis. Monocytes expressing the CCR-2 receptor are recruited to the tumor site by CCL-2 released by tumor cells, macrophages and stromal cells within the TME, where they further mature into TAMs (86). Therefore, reducing macrophage recruitment and infiltration into the TME by blocking the CCL2/CCR2 axis may be a promising therapeutic anti-tumor strategy. A study on esophageal cancer found that blocking the CCL2-CCR2 axis significantly reduced the tumor incidence by preventing TAM recruitment and also enhanced the anti-tumor effects of CD8+ T cells in the TME (87). Several CCL-2 antibodies are undergoing clinical trials. The two main drugs tested so far are the anti-CCL2 monoclonal antibody Carlumab (CNTO 888) and a targeted small molecule inhibitor (PF04136309), which have shown a certain benefit in tumor control (88,89).

In addition, BMS-813160, a CCR2/5 inhibitor, has been selected as a clinical candidate for its ability to inhibit the migration of inflammatory monocytes and macrophages. Clinical trials of BMS-813160 are ongoing in non-small cell carcinoma, liver cancer and pancreatic cancer (90) (Table I).

CSF-1 controls the proliferation, differentiation, recruitment, survival and function of mononuclear phagocytes (e.g., macrophages, monocytes) (91). Therefore, targeting the CSF1/CSF1R signaling pathway is considered to be another important and effective strategy for the treatment of malignant tumors. Ries et al (92) found that the use of CSF1R monoclonal antibody, RG7155, in patients with advanced diffuse giant cell tumors significantly reduced CSF1R+CD163+ macrophages in the TME. In a study of advanced solid tumors, RG7155 specifically depleted immunosuppressed M2-like macrophages and was used in combination with paclitaxel to enhance anti-tumor responses (93). In addition, in a study on endometrial cancer, it was found to promote TAM recruitment in the TME and tumor cell proliferation, which was significantly diminished by a CSF1R blocker (PLX3397) (94). A phase I clinical trial found PLX3397 to have favorable safety and tolerability in Asian patients with advanced solid tumors (95). In a rare tumor called tendonsynovial giant cell tumor, vimseltinib, a small molecule inhibitor targeting CSF1R, was found to persistently inhibit CSF1R activity in vitro and in vivo, depleting macrophages and other CSF1R-dependent cells, and inhibiting tumor growth and bone degradation in a mouse model of cancer (96).

Although CSF1/CSF1R and CCL2/CCR2 blockade are the most widely studied axes of TAM depletion, other cytokines have also been shown to have a role in this process. In a mouse model of malignant lobular tumors, monocytes are recruited into tumors through the interaction between CCL5 and CCR5. Blockade of the CCL5-CCR5 axis by CCR5 inhibitors resulted in markedly attenuated monocyte recruitment into tumors and inhibition of tumor growth (97). Therefore, actively exploring the factors that interfere with macrophage recruitment will provide a new idea for targeted macrophage therapy.

TAM reprogramming

As mentioned above, it is widely acknowledged that M2 and M1 macrophages have opposite roles in tumor growth and metastasis. Therefore, therapeutic strategies to re-educate the tumor-promoting M2 phenotype into the tumor-killing M1 phenotype have been proposed. Reprogramming macrophages involves the following two aspects: Restoration of phagocytosis and promotion of the polarization phenotype.

Role of cellular phenotypes in restoring phagocytosis
i) CD24

CD24 is a highly glycosylated glycosylphosphatidylinositol-anchored surface protein that acts as a 'don't eat me' signal. It regulates phagocytosis in macrophages through interaction with sialic-binding IG-10 (Siglec-10), an inhibitory receptor on TAMs (98). CD24 is commonly overexpressed in cancers and its overexpression is associated with poor prognosis in various cancers (99). It has been found in ovarian cancer and TNBC that CD24 expressed by tumor cells can interact with Siglec-10 on TAMs, blocking phagocytosis of macrophages. Blockade of CD24 or Siglec-10 enhanced the phagocytosis of macrophages on tumors and inhibited tumor growth (100). In addition, a recent study in mantle cell lymphoma and follicular lymphoma found that high expression of CD24 was associated with poor prognosis of patients. Treatment with CD24 monoclonal antibody significantly enhanced phagocytosis by macrophages and inhibited tumor progression (101). These studies demonstrate the therapeutic potential of CD24 blockade as a cancer immunotherapy.

ii) CD47

CD47, a transmembrane glycoprotein widely expressed on cancer cells, can block macrophage phagocytosis by binding to the signal-regulatory protein (SIRP)α on the surface of macrophages, enabling cancer cells to escape immune surveillance (102). Based on this characteristic, targeting the CD47-SIRPα axis is an effective modality. In a study of thyroid cancer, it was found that the degree of infiltration of TAMs in xenografted mice treated with anti-CD47 antibody was significantly increased and phagocytosis of tumor cells by macrophages was enhanced, which inhibited tumor growth (103). Furthermore, in small cell lung cancer (SCLC), macrophages showed increased phagocytosis and enhanced anti-tumor effects on GFP-expressing SCLC cells in mice treated with radiotherapy combined with CD47 block, compared with CD47 block alone, a finding that is particularly important for cancer patients suffering from metastatic disease (104). In addition to increasing phagocytosis of tumor cells, anti-CD47 therapy has also been shown to modulate TAM phenotypic changes. For instance, a glioblastoma study found that anti-CD47 treatment not only enhanced macrophage phagocytosis of tumor cells but also shifted the phenotype of macrophages towards the M1 subtype (105). Hence, preclinical studies of CD47-SIRPα blockade suggest its potential for clinical efficacy. To date, clinical studies have indicated that CD47 inhibitors (Hu5F9-G4) in combination with rituximab showed good anti-tumor activity in patients with non-Hodgkin lymphoma, while CD47 inhibitors (CC90002) have provided disappointing results in patients with acute myeloid leukemia (106,107). Clinical trials targeting SIRP molecules are still ongoing (Table I).

Use of polarization changes for therapy
i) PI3Kγ

As a member of the class 1B family, PI3Kγ is usually associated with G-protein-coupled receptor signaling and is abundantly expressed on a variety of immune cells, including macrophages and neutrophils, and the PI3Kγ pathway is related to the phenotypic transformation of TAMs as well as immunosuppressive states (108,109). For instance, in a mouse model carrying breast cancer, blocking PI3Kγ was also found to reduce the number of M2-like macrophages and enhance the role of cytotoxic T lymphocytes (CTLs), which significantly prevented tumor progression and prolonged survival (110). In addition, Carnevalli et al (111) found that AZD3458, a PI3Kγ inhibitor, did not alter tumor macrophage polarization, but instead promoted antigen-presenting macrophage and cytotoxic macrophage activation, which activated CD8 T cell-mediated antitumor activity of associated immune checkpoint inhibitors. A phase I clinical study found that a highly selective PI3Kγ inhibitor (IPI-549) demonstrated significant anti-tumor activity in patients with advanced solid disease (112). Therefore, the above studies have demonstrated the important role of PI3Kγ in TAMs reprogramming and recruitment of immune cells that inhibit tumor growth, suggesting that PI3Kγ may be a promising target for tumor therapy.

ii) TLRs

TLRs are innate immune pattern recognition receptors that have an important role in the activation of innate immune responses (113). In the reprogramming of macrophages, the phenotype of macrophages can change and they may be polarized in a pro-inflammatory direction. Numerous therapies aim at targeting TLR to repolarize macrophages from an M2-like activated state to an M1-like activated state and to enhance the immune response to cancer cells. For instance, local in situ inoculation of melanoma and neck tumor models with the tumor antigen (protein and peptide) adjuvant nanoemulsion loaded with TLR7/8 agonists induced recruitment and activation of innate immune cells, infiltration of lymphocytes and polarization of tumor-associated M2 macrophages, resulting in inhibition of tumor growth and prolonged tumor survival (114). Furthermore, oxaliplatin combined with TLR agonist R848 was found to reverse the polarization of macrophages and enhance the anti-tumor effects of oxaliplatin in colorectal cancer resistant to oxaliplatin (115). In addition to TLR7 and TLR8, it was found that TLR3 and TLR4 agonists also have the effect of altering the macrophage polarization status (116,117). These findings suggest the clinical relevance of TLR signaling and its potential application for cancer treatment by targeting TAMs. To date, several targeting TLRs have been tested in clinical trials with promising results (118-120).

iii) CD40

CD40 is a member of the TNF receptor superfamily and is highly expressed on antigen-presenting cells, such as macrophages. The ligand of CD40 is CD40L, which is mainly expressed by activated T cells, B cells and platelets (121). The CD40-CD40L interaction promotes the polarization of macrophages towards pro-inflammatory macrophages. For instance, in a mouse model of melanoma, it was found that myeloid-derived suppressor cells induce macrophage reprogramming by inhibiting the expression of CD40 on the surface of macrophages, thereby promoting melanoma progression (122). In addition, a study on pancreatic cancer found that CD40 agonists could drive the transformation of the macrophage phenotype towards M1, remodeling the pancreatic cancer microenvironment and inhibiting the development of pancreatic cancer (123). More recently, Frankish et al (124) found that HERA-CD40L, a novel molecule targeting CD40-mediated signal transduction, activates the signaling transduction mechanisms in dendritic cells, leading to an increase in intratumoral T cells and manipulating TME phenotypic changes to repolarize M2 macrophages to M1, thereby enhancing tumor control. In addition, a clinical trial in pancreatic ductal adenocarcinoma showed that selicrelumab, an agonist CD40 antibody, induced changes in the TME in patients with resectable pancreatic cancer, a reduction in M2-like TAMs and a significant reduction in tumor burden (125). Based on the above observations, reprogramming TAMs to anti-tumor phenotypes using CD40, rather than targeting and ablating TAMs, may be the preferred therapeutic paradigm for cancer treatment.

6. Conclusion and prospects

TAMs are one of the most important innate immune cell types in TME and have a complex regulatory role in tumor therapy. Therefore, it is crucial to reveal the exact regulatory mechanisms and specific targets of macrophages on tumors in order to optimize the effectiveness of current tumor therapies. In the present review, the heterogeneity of the origin of TAMs, the relevant regulators of recruitment and polarization and the complex roles of TAMs in tumorigenesis, progression and metastasis were discussed. Certain therapeutic approaches targeting TAMs, such as consumption of TAMs or re-education of TAMs were also outlined to provide insight for tumor immunotherapy. Specifically, targeting TAMs is a promising immunotherapy strategy. However, the clinical application of current therapeutic strategies is still very limited. For instance, the efficacy is restricted to certain patients, the anti-tumor spectrum is narrow, the adverse reactions are more frequent and drug resistance may easily occur. These defects limit the clinical application of targeting macrophages in tumor therapy. In addition, numerous questions remain regarding the nature and function of macrophages in the TME; many unknown molecular mechanisms have a crucial role in regulating tumor growth and development, and various potential targets require more research and attention. Therefore, it is necessary to further investigate the dialogue between macrophages and tumor cells. With a greater understanding of macrophage diversity through single-cell sequencing and other techniques, TAM-targeted therapies will be an important complement to cancer immunotherapy.

Availability of data and materials

Not applicable.

Authors' contributions

CL, YL and LM conceived the article. CL wrote the first version of the manuscript with critical input from all authors. XK, HL, HR and XZ performed the literature search. BZ and XN edited the manuscript for important intellectual content. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors have no competing interests to declare.

Acknowledgments

Not applicable.

Funding

This work was supported by XC, the host of the Capacity Construction Project of Major Clinical (specialized) Departments of Traditional Chinese Medicine of Liaoning Province (grant no. LNZYXZK201909 to XC) and the host of the Distinguished Professor Program of Liaoning Province (grant no. 203296 to XC). It was also supported by YL, the host of the Hospital Research Fund by Affiliated Zhongshan Hospital of Dalian University (grant no. 20230320 to YL).

References

1 

Yan S and Wan G: Tumor-associated macrophages in immunotherapy. FEBS J. 288:6174–6186. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al: Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity. 41:14–20. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Chen Y, Song Y, Du W, Gong L, Chang H and Zou Z: Tumor-associated macrophages: An accomplice in solid tumor progression. J Biomed Sci. 26:782019. View Article : Google Scholar : PubMed/NCBI

4 

Pathria P, Louis TL and Varner JA: Targeting tumor-associated macrophages in cancer. Trends Immunol. 40:310–327. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Li F, Okreglicka KM, Pohlmeier LM, Schneider C and Kopf M: Fetal monocytes possess increased metabolic capacity and replace primitive macrophages in tissue macrophage development. EMBO J. 39:e1032052020. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z, Junt T and Bentires-Alj M: Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 515:130–133. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Zhang WJ, Wang XH, Gao ST, Chen C, Xu XY, Sun Q, Zhou ZH, Wu GZ, Yu Q, Xu G, et al: Tumor-associated macrophages correlate with phenomenon of epithelial-mesenchymal transition and contribute to poor prognosis in triple-negative breast cancer patients. J Surg Res. 222:93–101. 2018. View Article : Google Scholar

9 

Xu Y, Zeng H, Jin K, Liu Z, Zhu Y, Xu L, Wang Z, Chang Y and Xu J: Immunosuppressive tumor-associated macrophages expressing interlukin-10 conferred poor prognosis and therapeutic vulnerability in patients with muscle-invasive bladder cancer. J Immunother Cancer. 10:e0034162022. View Article : Google Scholar : PubMed/NCBI

10 

Kumar AT, Knops A, Swendseid B, Martinez-Outschoom U, Harshyne L, Philp N, Rodeck U, Luginbuhl A, Cognetti D, Johnson J and Curry J: Prognostic significance of tumor-associated macrophage content in head and neck squamous cell carcinoma: A meta-analysis. Front Oncol. 9:6562019. View Article : Google Scholar : PubMed/NCBI

11 

Zhang H, Luo YB, Wu W, Zhang L, Wang Z, Dai Z, Feng S, Cao H, Cheng Q and Liu Z: The molecular feature of macrophages in tumor immune microenvironment of glioma patients. Comput Struct Biotechnol J. 19:4603–4618. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Wu Z, Lei K, Li H, He J and Shi E: Transcriptome-based network analysis related to M2-like tumor-associated macrophage infiltration identified VARS1 as a potential target for improving melanoma immunotherapy efficacy. J Transl Med. 20:4892022. View Article : Google Scholar : PubMed/NCBI

13 

Yuri P, Shigemura K, Kitagawa K, Hadibrata E, Risan M, Zulfiqqar A, Soeroharjo I, Hendri AZ, Danarto R, Ishii A, et al: Increased tumor-associated macrophages in the prostate cancer microenvironment predicted patients' survival and responses to androgen deprivation therapies in Indonesian patients cohort. Prostate Int. 8:62–69. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Cortese N, Carriero R, Laghi L, Mantovani A and Marchesi F: Prognostic significance of tumor-associated macrophages: Past, present and future. Semin Immunol. 48:1014082020. View Article : Google Scholar : PubMed/NCBI

15 

Qian BZ and Pollard JW: Macrophage diversity enhances tumor progression and metastasis. Cell. 141:39–51. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Movahedi K, Laoui D, Gysemans C, Baeten M, Stangé G, Van den Bossche J, Mack M, Pipeleers D, In't Veld P, De Baetselier P and Van Ginderachter JA: Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 70:5728–5739. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT and Sahebkar A: Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 233:6425–6440. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Martinez FO, Sica A, Mantovani A and Locati M: Macrophage activation and polarization. Front Biosci. 13:453–461. 2008. View Article : Google Scholar

19 

Zizzo G, Hilliard BA, Monestier M and Cohen PL: Efficient clearance of early apoptotic cells by human macrophages requires M2c polarization and MerTK Induction. J Immunol. 189:3508–3520. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Ferrante AW Jr: Macrophages, fat, and the emergence of immunometabolism. J Clin Invest. 123:4992–4993. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Haskó G, Pacher P, Deitch EA and Vizi ES: Shaping of monocyte and macrophage function by adenosine receptors. Pharmacol Ther. 113:264–275. 2007. View Article : Google Scholar

22 

Pinhal-Enfield G, Ramanathan M, Hasko G, Vogel SN, Salzman AL, Boons GJ and Leibovich SJ: An angiogenic switch in macrophages involving synergy between Toll-like receptors 2, 4, 7, and 9 and adenosine A(2A) receptors. Am J Pathol. 163:711–721. 2003. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Huang YH, Cai K, Xu PP, Wang L, Huang CX, Fang Y, Cheng S, Sun XJ, Liu F, Huang JY, et al: CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct Target Ther. 6:102021. View Article : Google Scholar : PubMed/NCBI

25 

Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA and Pollard JW: CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 475:222–225. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Sierra-Filardi E, Nieto C, Domínguez-Soto Á, Barroso R, Sánchez-Mateos P, Puig-Kroger A, López-Bravo M, Joven J, Ardavín C, Rodríguez-Fernández JL, et al CCL2 Shapes Macrophage Polarization by GM-CSF and M-CSF: Identification of CCL2/CCR2-dependent gene expression profile. J Immunol. 192:3858–3867. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Archer M, Bernhardt SM, Hodson LJ, Woolford L, Van der Hoek M, Dasari P, Evdokiou A and Ingman WV: CCL2-Mediated stromal interactions drive macrophage polarization to increase breast tumorigenesis. Int J Mol Sci. 24:73852023. View Article : Google Scholar : PubMed/NCBI

28 

Valero JG, Matas-Céspedes A, Arenas F, Rodriguez V, Carreras J, Serrat N, Guerrero-Hernández M, Yahiaoui A, Balagué O, Martin S, et al: The receptor of the colony-stimulating factor-1 (CSF-1R) is a novel prognostic factor and therapeutic target in follicular lymphoma. Leukemia. 35:2635–2649. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Mu G, Zhu Y, Dong Z, Shi L, Deng Y and Li H: Calmodulin 2 facilitates angiogenesis and metastasis of gastric cancer via STAT3/HIF-1A/VEGF-A mediated macrophage polarization. Front Oncol. 11:7273062021. View Article : Google Scholar : PubMed/NCBI

30 

Lian G, Chen S, Ouyang M, Li F, Chen L and Yang J: Colon cancer cell secretes EGF to Promote M2 Polarization of TAM Through EGFR/PI3K/AKT/mTOR pathway. Technol Cancer Res Treat. 18:15330338198490682019. View Article : Google Scholar : PubMed/NCBI

31 

Mazzoni M, Mauro G, Erreni M, Romeo P, Minna E, Vizioli MG, Belgiovine C, Rizzetti MG, Pagliardini S, Avigni R, et al: Senescent thyrocytes and thyroid tumor cells induce M2-like macrophage polarization of human monocytes via a PGE2-dependent mechanism. J Exp Clin Cancer Res. 38:2082019. View Article : Google Scholar : PubMed/NCBI

32 

Vaupel P and Harrison L: Tumor hypoxia: Causative factors, compensatory mechanisms, and cellular response. Oncologist. 9(Suppl 5): S4–S9. 2004. View Article : Google Scholar

33 

Zhou HC, Xin-Yan Yan, Yu WW, Liang XQ, Du XY, Liu ZC, Long JP, Zhao GH and Liu HB: Lactic acid in macrophage polarization: The significant role in inflammation and cancer. Inter Rev Immunol. 41:4–18. 2021. View Article : Google Scholar

34 

Zhang L and Li S: Lactic acid promotes macrophage polarization through MCT-HIF1α signaling in gastric cancer. Exp Cell Res. 388:1118462020. View Article : Google Scholar

35 

Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, Yeoh KW, Kon OL, Tam JP and Sze SK: Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 38:5158–5173. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Hynes RO: The extracellular matrix: Not just pretty fibrils. Science. 326:1216–1219. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Colombatti A, Spessotto P, Doliana R, Mongiat M, Bressan GM and Esposito G: The EMILIN/Multimerin family. Front Immunol. 2:932012. View Article : Google Scholar :

38 

Mongiat M, Marastoni S, Ligresti G, Lorenzon E, Schiappacassi M, Perris R, Frustaci S and Colombatti A: The extracellular matrix glycoprotein elastin microfibril interface located protein 2: A dual role in the tumor microenvironment. Neoplasia. 12:294–304. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Andreuzzi E, Fejza A, Polano M, Poletto E, Camicia L, Carobolante G, Tarticchio G, Todaro F, Di Carlo E, Scarpa M, et al: Colorectal cancer development is affected by the ECM molecule EMILIN-2 hinging on macrophage polarization via the TLR-4/MyD88 pathway. J Exp Clin Cancer Res. 41:602022. View Article : Google Scholar : PubMed/NCBI

40 

Bernsmeier C, van der Merwe S and Périanin A: Innate immune cells in cirrhosis. J Hepatol. 73:186–201. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Bruns H, Büttner M, Fabri M, Mougiakakos D, Bittenbring JT, Hoffmann MH, Beier F, Pasemann S, Jitschin R, Hofmann AD, et al: Vitamin D-dependent induction of cathelicidin in human macrophages results in cytotoxicity against high-grade B cell lymphoma. Sci Transl Med. 7:282ra472015. View Article : Google Scholar : PubMed/NCBI

42 

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

43 

Haque ASMR, Moriyama M, Kubota K, Ishiguro N, Sakamoto M, Chinju A, Mochizuki K, Sakamoto T, Kaneko N, Munemura R, et al: CD206+tumor-associated macrophages promote proliferation and invasion in oral squamous cell carcinoma via EGF production. Sci Rep. 9:146112019. View Article : Google Scholar

44 

Xu W, Wu Y, Liu W, Anwaier A, Tian X, Su J, Huang H, Wei G, Qu Y, Zhang H and Ye D: Tumor-associated macrophage-derived chemokine CCL5 facilitates the progression and immunosuppressive tumor microenvironment of clear cell renal cell carcinoma. Int J Biol Sci. 18:4884–4900. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Radharani NNV, Yadav AS, Nimma R, Kumar TVS, Bulbule A, Chanukuppa V, Kumar D, Patnaik S, Rapole S and Kundu GC: Tumor-associated macrophage derived IL-6 enriches cancer stem cell population and promotes breast tumor progression via Stat-3 pathway. Cancer Cell Int. 22:1222022. View Article : Google Scholar : PubMed/NCBI

46 

Valković T, Dobrila F, Melato M, Sasso F, Rizzardi C and Jonjić N: Correlation between vascular endothelial growth factor, angiogenesis, and tumor-associated macrophages in invasive ductal breast carcinoma. Virchows Arch. 440:583–588. 2002. View Article : Google Scholar

47 

Fu LQ, Du WL, Cai MH, Yao JY, Zhao YY and Mou XZ: The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 353:1041192020. View Article : Google Scholar : PubMed/NCBI

48 

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

49 

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

50 

Kawahara A, Hattori S, Akiba J, Nakashima K, Taira T, Watari K, Hosoi F, Uba M, Basaki Y, Koufuji K, et al: Infiltration of thymidine phosphorylase-positive macrophages is closely associated with tumor angiogenesis and survival in intestinal type gastric cancer. Oncol Rep. 24:405–415. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Hori T, Sasayama T, Tanaka K, Koma YI, Nishihara M, Tanaka H, Nakamizo S, Nagashima H, Maeyama M, Fujita Y, et al: Tumor-associated macrophage related interleukin-6 in cerebrospinal fluid as a prognostic marker for glioblastoma. J Clin Neurosci. 68:281–289. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Zhou M, Na R, Lai S, Guo Y, Shi J, Nie J, Zhang S, Wang Y and Zheng T: The present roles and future perspectives of Interleukin-6 in biliary tract cancer. Cytokine. 169:1562712023. View Article : Google Scholar : PubMed/NCBI

53 

Sceneay J, Smyth MJ and Möller A: The pre-metastatic niche: Finding common ground. Cancer Metastasis Rev. 32:449–464. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Lu X and Kang Y: Organotropism of breast cancer metastasis. J Mammary Gland Biol Neoplasia. 12:153–162. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, et al: Involvement of chemokine receptors in breast cancer metastasis. Nature. 410:50–56. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T and Joyce JA: IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 24:241–255. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Chen Q, Zhang XH and Massagué J: Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell. 20:538–549. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Qian B, Deng Y, Im JH, Muschel RJ, Zou Y, Li J, Lang RA and Pollard JW: A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS One. 4:e65622009. View Article : Google Scholar : PubMed/NCBI

59 

Genna A, Duran CL, Entenberg D, Condeelis JS and Cox D: Macrophages Promote tumor cell extravasation across an endothelial barrier through thin membranous connections. Cancers (Basel). 15:20922023. View Article : Google Scholar : PubMed/NCBI

60 

Chen X, Yang M, Yin J, Li P, Zeng S, Zheng G, He Z, Liu H, Wang Q, Zhang F and Chen D: Tumor-associated macrophages promote epithelial-mesenchymal transition and the cancer stem cell properties in triple-negative breast cancer through CCL2/AKT/β-catenin signaling. Cell Commun Signal. 20:922022. View Article : Google Scholar

61 

Li X, Shao C, Shi Y and Han W: Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 11:312018. View Article : Google Scholar : PubMed/NCBI

62 

DeNardo DG and Ruffell B: Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 19:369–382. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, et al: Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 10:942–949. 2004. View Article : Google Scholar : PubMed/NCBI

64 

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

65 

Arlauckas SP, Garren SB, Garris CS, Kohler RH, Oh J, Pittet MJ and Weissleder R: Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Theranostics. 8:5842–5854. 2018. View Article : Google Scholar

66 

Menjivar RE, Nwosu ZC, Du W, Donahue KL, Hong HS, Espinoza C, Brown K, Velez-Delgado A, Yan W, Lima F, et al: Arginase 1 is a key driver of immune suppression in pancreatic cancer. Elife. 12:e807212023. View Article : Google Scholar : PubMed/NCBI

67 

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

68 

Cassetta L and Pollard JW: Targeting macrophages: Therapeutic approaches in cancer. Nat Rev Drug Discov. 17:887–904. 2018. View Article : Google Scholar : PubMed/NCBI

69 

van der Heide D, Weiskirchen R and Bansal R: Therapeutic targeting of hepatic macrophages for the treatment of liver diseases. Front Immunol. 10:28522019. View Article : Google Scholar : PubMed/NCBI

70 

Roelofs AJ, Thompson K, Gordon S and Rogers MJ: Molecular mechanisms of action of bisphosphonates: Current status. Clin Cancer Res. 12(20 Pt 2): 6222s–6230s. 2006. View Article : Google Scholar : PubMed/NCBI

71 

Van Acker HH, Anguille S, Willemen Y, Smits EL and Van Tendeloo VF: Bisphosphonates for cancer treatment: Mechanisms of action and lessons from clinical trials. Pharmacol Ther. 158:24–40. 2016. View Article : Google Scholar

72 

Rogers TL and Holen I: Tumour macrophages as potential targets of bisphosphonates. J Transl Med. 9:1772011. View Article : Google Scholar : PubMed/NCBI

73 

Van Rooijen N, Kors N, vd Ende M and Dijkstra CD: Depletion and repopulation of macrophages in spleen and liver of rat after intravenous treatment with liposome-encapsulated dichloromethylene diphosphonate. Cell Tissue Res. 260:215–222. 1990. View Article : Google Scholar : PubMed/NCBI

74 

Giraudo E, Inoue M and Hanahan D: An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis. J Clin Invest. 114:623–633. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Zang X, Zhou J, Zhang X, Chen D, Han Y and Chen X: Dual-targeting tumor cells and tumor associated macrophages with lipid coated calcium zoledronate for enhanced lung cancer chemoimmunotherapy. Int J Pharm. 594:1201742021. View Article : Google Scholar

76 

Lv J, Chen FK, Liu C, Liu PJ, Feng ZP, Jia L, Yang ZX, Hou F and Deng ZY: Zoledronic acid inhibits thyroid cancer stemness and metastasis by repressing M2-like tumor-associated macrophages induced Wnt/β-catenin pathway. Life Sci. 256:1179252020. View Article : Google Scholar

77 

Choi J, Lee EJ, Yang SH, Im YR and Seong J: A prospective phase II study for the efficacy of radiotherapy in combination with zoledronic acid in treating painful bone metastases from gastrointestinal cancers. J Radiat Res. 60:242–248. 2019. View Article : Google Scholar :

78 

D'Incalci M and Galmarini CM: A review of trabectedin (ET-743): A unique mechanism of action. Mol Cancer Ther. 9:2157–2163. 2010. View Article : Google Scholar : PubMed/NCBI

79 

Germano G, Frapolli R, Belgiovine C, Anselmo A, Pesce S, Liguori M, Erba E, Uboldi S, Zucchetti M, Pasqualini F, et al: Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell. 23:249–262. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Belgiovine C, Frapolli R, Liguori M, Digifico E, Colombo FS, Meroni M, Allavena P and D'Incalci M: Inhibition of tumor-associated macrophages by trabectedin improves the antitumor adaptive immunity in response to anti-PD-1 therapy. Eur J Immunol. 51:2677–2686. 2021. View Article : Google Scholar : PubMed/NCBI

81 

de Sande González LM, Martin-Broto J, Kasper B, Blay JY and Le Cesne A: Real-world evidence of the efficacy and tolerability of trabectedin in patients with advanced soft-tissue sarcoma. Expert Rev Anticancer Ther. 20:957–963. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Romero I, López-Guerrero JA and Pignata S: Real-world experience with trabectedin for the treatment of recurrent ovarian cancer. Expert Rev Anticancer Ther. 21:1089–1095. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Cortinovis D, Grosso F, Carlucci L, Zucali PA, Pasello G, Tiseo M, Sperandi F, Hollander L, Galli F, Torri V, et al: Trabectedin in malignant pleural mesothelioma: Results from the multicentre, single arm, phase II ATREUS study. Clin Lung Cancer. 22:361–370.e3. 2021. View Article : Google Scholar

84 

Belli C, Piemonti L, D'Incalci M, Zucchetti M, Porcu L, Cappio S, Doglioni C, Allavena P, Ceraulo D, Maggiora P, et al: Phase II trial of salvage therapy with trabectedin in metastatic pancreatic adenocarcinoma. Cancer Chemother Pharmacol. 77:477–484. 2016. View Article : Google Scholar

85 

Cao Y, Qiao B, Chen Q, Xie Z, Dou X, Xu L, Ran H, Zhang L and Wang Z: Tumor microenvironment remodeling via targeted depletion of M2-like tumor-associated macrophages for cancer immunotherapy. Acta Biomater. 160:239–251. 2023. View Article : Google Scholar : PubMed/NCBI

86 

Kalbasi A, Komar C, Tooker GM, Liu M, Lee JW, Gladney WL, Ben-Josef E and Beatty GL: Tumor-Derived CCL2 mediates resistance to radiotherapy in pancreatic ductal adenocarcinoma. Clin Cancer Res. 23:137–148. 2017. View Article : Google Scholar

87 

Yang H, Zhang Q, Xu M, Wang L, Chen X, Feng Y, Li Y, Zhang X, Cui W and Jia X: CCL2-CCR2 axis recruits tumor associated macrophages to induce immune evasion through PD-1 signaling in esophageal carcinogenesis. Mol Cancer. 19:412020. View Article : Google Scholar : PubMed/NCBI

88 

Noel M, O'Reilly EM, Wolpin BM, Ryan DP, Bullock AJ, Britten CD, Linehan DC, Belt BA, Gamelin EC, Ganguly B, et al: Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 38:800–811. 2020. View Article : Google Scholar :

89 

Brana I, Calles A, LoRusso PM, Yee LK, Puchalski TA, Seetharam S, Zhong B, de Boer CJ, Tabernero J and Calvo E: Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: An open-label, multicenter phase 1b study. Target Onco. 10:111–123. 2015. View Article : Google Scholar

90 

Cherney RJ, Anjanappa P, Selvakumar K, Batt DG, Brown GD, Rose AV, Vuppugalla R, Chen J, Pang J, Xu S, et al: BMS-813160: A Potent CCR2 and CCR5 dual antagonist selected as a clinical candidate. ACS Med Chem Lett. 12:1753–1758. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Lelios I, Cansever D, Utz SG, Mildenberger W, Stifter SA and Greter M: Emerging roles of IL-34 in health and disease. J Exp Med. 217:e201902902020. View Article : Google Scholar : PubMed/NCBI

92 

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

93 

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

94 

Hua F, Tian Y, Gao Y, Li C and Liu X: Colony-stimulating factor 1 receptor inhibition blocks macrophage infiltration and endometrial cancer cell proliferation. Mol Med Rep. 19:3139–3147. 2019.PubMed/NCBI

95 

Lee JH, Chen TW, Hsu CH, Yen YH, Yang JC, Cheng AL, Sasaki SI, Chiu LL, Sugihara M, Ishizuka T, et al: A phase I study of pexidartinib, a colony-stimulating factor 1 receptor inhibitor, in Asian patients with advanced solid tumors. Invest New Drugs. 38:99–110. 2020. View Article : Google Scholar :

96 

Smith BD, Kaufman MD, Wise SC, Ahn YM, Caldwell TM, Leary CB, Lu WP, Tan G, Vogeti L, Vogeti S, et al: Vimseltinib: A Precision CSF1R therapy for tenosynovial giant cell tumors and diseases promoted by macrophages. Mol Cancer Ther. 20:2098–2109. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Nie Y, Huang H, Guo M, Chen J, Wu W, Li W, Xu X, Lin X, Fu W, Yao Y, et al: Breast Phyllodes Tumors Recruit and Repolarize Tumor-Associated Macrophages via Secreting CCL5 to promote malignant progression, which can be inhibited by CCR5 inhibition therapy. Clin Cancer Res. 25:3873–3886. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Altevogt P, Sammar M, Hüser L and Kristiansen G: Novel insights into the function of CD24: A driving force in cancer. Int J Cancer. 148:546–559. 2021. View Article : Google Scholar

99 

Tarhriz V, Bandehpour M, Dastmalchi S, Ouladsahebmadarek E, Zarredar H and Eyvazi S: Overview of CD24 as a new molecular marker in ovarian cancer. J Cell Physiol. 234:2134–2142. 2019. View Article : Google Scholar

100 

Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, Krishnan V, Hatakeyama J, Dorigo O, Barkal LJ and Weissman IL: CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 572:392–396. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Freile JÁ, Ustyanovska Avtenyuk N, Corrales MG, Lourens HJ, Huls G, van Meerten T, Cendrowicz E and Bremer E: CD24 Is a Potential Immunotherapeutic Target for Mantle Cell Lymphoma. Biomedicines. 10:11752022. View Article : Google Scholar : PubMed/NCBI

102 

Maute R, Xu J and Weissman IL: CD47-SIRPα-targeted therapeutics: Status and prospects. Immunooncol Technol. 13:1000702022. View Article : Google Scholar

103 

Schürch CM, Roelli MA, Forster S, Wasmer MH, Brühl F, Maire RS, Di Pancrazio S, Ruepp MD, Giger R, Perren A, et al: Targeting CD47 in anaplastic thyroid carcinoma enhances tumor phagocytosis by macrophages and is a promising therapeutic strategy. Thyroid. 29:979–992. 2019. View Article : Google Scholar : PubMed/NCBI

104 

Nishiga Y, Drainas AP, Baron M, Bhattacharya D, Barkal AA, Ahrari Y, Mancusi R, Ross JB, Takahashi N, Thomas A, et al: Radiotherapy in combination with CD47 blockade elicits a macrophage-mediated abscopal effect. Nat Cancer. 3:1351–1366. 2022. View Article : Google Scholar : PubMed/NCBI

105 

Zhang M, Hutter G, Kahn SA, Azad TD, Gholamin S, Xu CY, Liu J, Achrol AS, Richard C, Sommerkamp P, et al: Anti-CD47 treatment stimulates phagocytosis of glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages in vivo. PLoS One. 11:e01535502016. View Article : Google Scholar : PubMed/NCBI

106 

Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL, Ghosh N, Kline J, Roschewski M, LaCasce A, Collins GP, et al: CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin's Lymphoma. N Engl J Med. 379:1711–1721. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Zeidan AM, DeAngelo DJ, Palmer J, Seet CS, Tallman MS, Wei X, Raymon H, Sriraman P, Kopytek S, Bewersdorf JP, et al: Phase 1 study of anti-CD47 monoclonal antibody CC-90002 in patients with relapsed/refractory acute myeloid leukemia and high-risk myelodysplastic syndromes. Ann Hematol. 101:557–569. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Solinas G and Becattini B: The role of PI3Kγ in metabolism and macrophage activation. Oncotarget. 8:106145–106146. 2017. View Article : Google Scholar :

109 

Qiu X, Tian Y, Liang Z, Sun Y, Li Z and Bian J: Recent discovery of phosphoinositide 3-kinase γ inhibitors for the treatment of immune diseases and cancers. Future Med Chem. 11:2151–2169. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Qin H, Yu H, Sheng J, Zhang D, Shen N, Liu L, Tang Z and Chen X: PI3Kgamma inhibitor attenuates immunosuppressive effect of Poly(l-Glutamic Acid)-Combretastatin A4 conjugate in metastatic breast cancer. Adv Sci (Weinh). 6:19003272019. View Article : Google Scholar : PubMed/NCBI

111 

Carnevalli LS, Taylor MA, King M, Coenen-Stass AML, Hughes AM, Bell S, Proia TA, Wang Y, Ramos-Montoya A, Wali N, et al: Macrophage activation status rather than repolarization is associated with enhanced checkpoint activity in combination with PI3Kγ Inhibition. Mol Cancer Ther. 20:1080–1091. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Hong DS, Postow M, Chmielowski B, Sullivan R, Patnaik A, Cohen EEW, Shapiro G, Steuer C, Gutierrez M, Yeckes-Rodin H, et al: Eganelisib a first-in-class PI3Kγ inhibitor, in patients with advanced solid tumors: Results of the phase 1/1b MARIO-1 trial. Clin Cancer Res. 29:2210–2219. 2023. View Article : Google Scholar : PubMed/NCBI

113 

Brennan JJ and Gilmore TD: Evolutionary Origins of Toll-like Receptor Signaling. Mol Biol Evol. 35:1576–1587. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Kim SY, Kim S, Kim JE, Lee SN, Shin IW, Shin HS, Jin SM, Noh YW, Kang YJ, Kim YS, et al: Lyophilizable and multifaceted toll-like receptor 7/8 agonist-loaded nanoemulsion for the reprogramming of tumor microenvironments and enhanced cancer immunotherapy. ACS Nano. 13:12671–12686. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Liu Z, Xie Y, Xiong Y, Liu S, Qiu C, Zhu Z, Mao H, Yu M and Wang X: TLR 7/8 agonist reverses oxaliplatin resistance in colorectal cancer via directing the myeloid-derived suppressor cells to tumoricidal M1-macrophages. Cancer Lett. 469:173–185. 2020. View Article : Google Scholar

116 

Vidyarthi A, Khan N, Agnihotri T, Negi S, Das DK, Aqdas M, Chatterjee D, Colegio OR, Tewari MK and Agrewala JN: TLR-3 Stimulation Skews M2 Macrophages to M1 Through IFN-αβ signaling and restricts tumor progression. Front Immunol. 9:16502018. View Article : Google Scholar

117 

Sun L, Kees T, Almeida AS, Liu B, He XY, Ng D, Han X, Spector DL, McNeish IA, Gimotty P, et al: Activating a collaborative innate-adaptive immune response to control metastasis. Cancer Cell. 39:1361–1374.e9. 2021. View Article : Google Scholar : PubMed/NCBI

118 

Chow LQM, Morishima C, Eaton KD, Baik CS, Goulart BH, Anderson LN, Manjarrez KL, Dietsch GN, Bryan JK, Hershberg RM, et al: Phase Ib trial of the toll-like receptor 8 agonist, motolimod (VTX-2337), combined with cetuximab in patients with recurrent or metastatic SCCHN. Clin Cancer Res. 23:2442–2450. 2017. View Article : Google Scholar

119 

Shayan G, Kansy BA, Gibson SP, Srivastava RM, Bryan JK, Bauman JE, Ohr J, Kim S, Duvvuri U, Clump DA, et al: Phase Ib study of immune biomarker modulation with neoadjuvant cetuximab and TLR8 stimulation in head and neck cancer to overcome suppressive myeloid signals. Clin Cancer Res. 24:62–72. 2018. View Article : Google Scholar :

120 

Trutnovsky G, Reich O, Joura EA, Holter M, Ciresa-König A, Widschwendter A, Schauer C, Bogner G, Jan Z, Boandl A, et al: Topical imiquimod versus surgery for vulvar intraepithelial neoplasia: A multicentre, randomised, phase 3, non-inferiority trial. Lancet. 399:1790–1798. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y and Noelle RJ: Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev. 229:152–172. 2009. View Article : Google Scholar : PubMed/NCBI

122 

Valencia JC, Erwin-Cohen RA, Clavijo PE, Allen C, Sanford ME, Day CP, Hess MM, Johnson M, Yin J, Fenimore JM, et al: Myeloid-Derived suppressive cell expansion promotes melanoma growth and autoimmunity by inhibiting CD40/IL27 regulation in macrophages. Cancer Res. 81:5977–5990. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Lim CY, Chang JH, Lee WS, Kim J and Park IY: CD40 agonists alter the pancreatic cancer microenvironment by shifting the macrophage phenotype toward M1 and suppress human pancreatic cancer in organotypic slice cultures. Gut Liver. 16:645–659. 2022. View Article : Google Scholar :

124 

Frankish J, Mukherjee D, Romano E, Billian-Frey K, Schröder M, Heinonen K, Merz C, Redondo Müller M, Gieffers C, Hill O, et al: The CD40 agonist HERA-CD40L results in enhanced activation of antigen presenting cells, promoting an anti-tumor effect alone and in combination with radiotherapy. Front Immunol. 14:11601162023. View Article : Google Scholar : PubMed/NCBI

125 

Byrne KT, Betts CB, Mick R, Sivagnanam S, Bajor DL, Laheru DA, Chiorean EG, O'Hara MH, Liudahl SM, Newcomb C, et al: Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer. Clin Cancer Res. 27:4574–4586. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2024
Volume 64 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu C, Liu Y, Miao L, Kong X, Li H, Chen H, Zhao X, Zhang B and Cui X: Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review). Int J Oncol 64: 11, 2024
APA
Lu, C., Liu, Y., Miao, L., Kong, X., Li, H., Chen, H. ... Cui, X. (2024). Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review). International Journal of Oncology, 64, 11. https://doi.org/10.3892/ijo.2023.5599
MLA
Lu, C., Liu, Y., Miao, L., Kong, X., Li, H., Chen, H., Zhao, X., Zhang, B., Cui, X."Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review)". International Journal of Oncology 64.2 (2024): 11.
Chicago
Lu, C., Liu, Y., Miao, L., Kong, X., Li, H., Chen, H., Zhao, X., Zhang, B., Cui, X."Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review)". International Journal of Oncology 64, no. 2 (2024): 11. https://doi.org/10.3892/ijo.2023.5599