International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
C-C motif chemokine ligand 2 (CCL2), is a key member of the CC subfamily of chemokines, which was purified and identified in 1989 by Yoshimura et al (1). CCL2 exerts chemotactic effects on monocytes, macrophages and T lymphocytes (2), and is secreted by activated cells through autocrine or paracrine methods. Cytokines in the tumor microenvironment (TME), including TNFα, lipopolysaccharide, IL-1, IL-6 and transforming growth factor-β (TGF-β), stimulate tumor cells to produce CCL2 (2). C-C motif chemokine receptor 2 (CCR2), the main receptor of CCL2, is a G protein-coupled receptor expressed in both immune and tumor cells (2–4). It plays an essential role in different aspects of tumor cell biology, including the regulation of proliferation, angiogenesis, immune response and migration of cells in inflammatory environments (5). CCL2 exerts its biological effects mainly by combining with CCR2 (2,6).
Studies have found that numerous types of tumor cells, including myeloma, breast cancer, prostate cancer and melanoma cells, express CCR2 and secrete high levels of CCL2 (7–9). CCL2 thus promotes tumor cell proliferation and survival through autocrine or paracrine pathways, participating in the regulation of tumor immune tolerance, inducing tumor angiogenesis, and promoting tumor invasion and metastasis (10).
CCL2 signaling is closely associated with tumor growth and progression (11,12). The CCL2/CCR2 signaling axis serves diverse roles in the initiation and progression of cancer by regulating tumor-associated angiogenesis, recruiting tumor-associated immune cells to promote tumor growth, activating tumor-specific immune responses and stimulating tumor cell proliferation (13–15).
The ‘seed and soil’ hypothesis posited that, during metastasis, tumor cells not only adapt to the recipient microenvironment but also actively remodel it to facilitate colonization (16,17). The chemokine-receptor crosstalk represents a pivotal research frontier in tumor metastasis biology. Its multifaceted roles include tumor cell proliferation, chemoresistance, migratory/invasive capacity and organotropic metastasis, alongside regulatory effects on angiogenesis and lymphangiogenesis. Growth factors, circulatory hypoxia, antitumor drugs and radiation therapy stimulate tumor cells to secrete more CCL2, induce the establishment of an immunosuppressive TME through the CCL2/CCR2 signaling axis, and promote tumor progression and metastasis. de Visser reviewed the importance of the TME in every stage of cancer progression, including tumor initiation, progression, invasion, infiltration, metastasis, diffusion and growth (18). TME represents a highly complex and dynamic ecosystem, primarily composed of three major cellular components: myeloid cells (including tumor-associated macrophages [TAMs], myeloid-derived suppressor cells [MDSCs], dendritic cells [DCs], and tumor-associated neutrophils [TANs]), lymphoid cells (such as T cells, B cells, natural killer [NK] cells, and innate lymphoid cells [ILCs]), and stromal components (cancer-associated fibroblasts [CAFs] and endothelial cells) (18). While these host-derived cells were historically dismissed as passive bystanders in tumorigenesis, emerging evidence highlights their key roles in driving cancer pathogenesis (18). The cellular architecture and functional phenotype of the TME exhibit pronounced heterogeneity dictated by primary tumor location, cancer cell-intrinsic properties, disease stage and patient-specific factors (18). Deciphering the crosstalk among tumor cell-autonomous signals, microenvironmental cues and systemic regulatory networks is key for the rationale-driven design of next-generation cancer therapeutics. The present study summarized the role of the CCL2/CCR2 signaling axis in tumor progression and metastasis.
CCL2 orchestrates antitumor immune responses by promoting immune cell recruitment and surveillance. In vitro studies have shown that diverse tumor cell lines chemoattract CD8+ and CD4+ T lymphocytes through CCL2 secretion (19,20). For example, in a previous study of nude mice, ovarian cancer cells engineered to express CCL2 induced robust monocyte infiltration at the injection site, resulting in localized tumor growth inhibition (20) Clinically, plasma CCL2 levels in patients with pancreatic cancer have been shown to be negatively correlated with tumor proliferation markers (21). In addition, in colorectal cancer preclinical models (22–24), the targeted modulation of tumor-derived chemokines enhances immune cell infiltration and suppresses tumor progression.
CCL2 serves a pivotal role in the metastatic cascade, particularly during epithelial-mesenchymal transition (EMT). Preclinical studies have shown EMT-induced cancer cells upregulate CCL2 expression (22–25), which in turn attracts neutrophils to the TME. While neutrophils are traditionally associated with tumor-promoting inflammation, evidence has highlighted their potential to exert cytotoxic effects against cancer cells in specific contexts (26–28). Tumor-associated neutrophils (TANs) have two phenotypes. ‘N2’ is known as a pro-tumorigenic phenotype, as it exerts a pro-tumorigenic effect at the primary site by secreting oncogenic factors, promoting primary tumor growth and angiogenesis, increasing extracellular matrix degradation and suppressing immune responses (29–33). ‘N1’ is an antitumorigenic phenotype. These neutrophil phenotype switches are contingent on TGF-β signaling, as the neutralization of TGF-β has been shown to induce a shift from ‘N2’ pro-tumorigenic to ‘N1’ antitumorigenic neutrophil states (31). Tumor-entrained neutrophils suppress lung metastatic seeding via H2O2-dependent cytotoxic mechanisms, with tumor-derived CCL2 serving as a key mediator of granulocyte colony-stimulating factor-stimulated neutrophil recruitment for optimal anti-metastatic function (28). A preclinical study of mouse breast cancer models further demonstrated that neutrophils accumulate in the lungs prior to the arrival of metastatic cells, establishing a pre-metastatic surveillance niche (28).
TANs exhibit dual roles in cancer progression, with N1-polarized TANs exerting antitumor effects and N2-polarized TANs promoting tumorigenesis. In hepatocellular carcinoma (HCC), cancer-associated fibroblasts (CAFs) secrete cardiotrophin-like cytokine factor 1 (CLCF1), which drives tumor stemness via C-X-C motif chemokine ligand 6/TGF-β and recruits immunosuppressive N2 TANs, forming a pro-tumorigenic feedback loop. Conversely, in lung cancer, C-X-C chemokine receptor type 2 (CXCR2) inhibition shifts TANs toward an N1 phenotype, decreasing immunosuppressive factors (arginase 1, TGF-β) and enhancing CD8+ T-cell activity, thereby improving antitumor immunity and chemotherapy response (34,35). These findings highlight TAN polarization as a critical determinant of tumor fate, suggesting therapeutic strategies targeting N2 TANs (such asCLCF1/ERK inhibition in HCC) or promoting N1 TANs (for example via CXCR2 blockade in lung cancer) may modulate the TME.
TILs serve as both prognostic biomarkers in cancer progression and key effectors in immunotherapeutic responses (26). The CCL2/CCR2 signaling axis orchestrates TIL recruitment to the TME, while CCL2 modulates the anti-cancer functions of T lymphocytes and dendritic cells, thereby potentiating antitumor immunity (26).
A previous in vitro study has demonstrated that CCL2 triggers γδ T-cell chemotaxis toward tumor-derived extracts, a process abolished by CCL2-neutralizing antibodies (36). Mechanistic investigation using T-cell receptor Δ chain knockout and CCR2−/− murine models have revealed that the loss of γδ TILs accelerates tumor progression in vivo, implicating a protective role for the CCR2/CCL2 axis in recruiting antitumor γδ T cells (36). Human tissue analyses (37–39) have further shown that Vδ1+ (but not Vδ2+) γδ T cells selectively express CCR2 and exhibit CCL2-dependent migration, with CCR2 expression dysregulated across various types of malignancies, including lung, prostate, liver and breast cancer. These findings highlight the tumor-suppressive function of CCL2/CCR2 signaling in orchestrating γδ1 T-cell infiltration, suggesting potential therapeutic strategies targeting Vδ1+ subsets in cancer immunotherapy. The natural killer (NK) group 2D receptor is critical for immune surveillance against malignancy (40). In a murine model of metastatic liver cancer, p53 reactivation has been shown to induce CCL2 expression in tumor cells, which is key for robust NK cell recruitment to the TME (41). Mechanistically, p53-mediated CCL2 secretion establishes a chemotactic gradient that enhances NK cell infiltration, thereby facilitating tumor elimination in an NK cell-dependent manner (42). Restoring p53 function in tumor cells also upregulates chemokines with NK cell-recruiting potential, although the antibody-based neutralization of CCL2 abrogates NK cell accumulation in senescent tumors and impairs their clearance, highlighting the role of CCL2 in anti-cancer immunosurveillance (42–45).
Enhanced cell motility and survival are hallmarks of metastatic tumor cells (46). The CCL2/CCR2 signaling pathway governs tumor cell chemotaxis, migratory capacity, survival and proliferation, thereby facilitating oncogenic progression in both hematological malignancies and solid tumors (47–49). The CCL2/CCR2 signaling axis can activate downstream pathways, including the PI3K/AKT, SMAD family member 3, P42/44 MAPK and ERK1/2-MMP2/9 pathways, as well as the protein kinase C-dependent protein tyrosine phosphorylation pathway to affect the proliferation, invasion and infiltration of tumor cells (50–52).
CCL2 promotes chondrosarcoma cell migration by upregulating MMP9 expression and engaging CCR2, thereby inhibiting Ras/Raf-1/MEK/ERK and NFκB signaling cascades (53). In HCC cell model, CCL2/CCR2 engagement triggers focal adhesion kinase tyrosine phosphorylation at Y397, promoting the recruitment of Src family kinases and activation of MMP2/9 through the ERK1/2 signaling cascade (54). Concurrently, calcium ion flux mediated by CCR2 activates the calcineurin-nuclear factor of activated T cells pathway, upregulating MMP9 transcription and enhancing extracellular matrix degradation (55). Sustained CCL2 secretion also polarizes tumor-associated macrophages (TAMs) toward the M2 phenotype, fostering a pro-inflammatory milieu rich in IL-6 and TNF-α that supports cancer cell clonal expansion (56–59). This multifaceted signaling network has been implicated in metastatic progression in multiple types of cancer, including cervical and breast malignancy (56,58). In breast cancer, the CCL2/CCR2 axis regulates cellular motility and survival, thereby driving metastatic dissemination (60,61). Similarly, in prostate cancer, CCL2/CCR2 signaling governs proliferation, apoptosis resistance and invasive potential (62). In bladder cancer, the activation of this axis enhances migration and invasion via protein kinase C activation and tyrosine phosphorylation, independent of its effects on cell proliferation (63). Blockade of CCL2/CCR2 signaling markedly impairs the bone marrow homing of multiple myeloma cells, underscoring its role in hematological malignancy metastasis (64). In nasopharyngeal carcinoma (NPC), CCL2/CCR2 signaling promotes metastasis via ERK1/2-MMP2/9 pathway activation (65). In colorectal cancer, alcohol exposure upregulates CCL2/CCR2 signaling via the glycogen synthase kinase 3β/β-catenin pathway, facilitating metastatic progression (66). Epithelial ovarian cancer exploits CCL2/CCR2 signaling to promote peritoneal dissemination, as demonstrated by the enhanced migration and adhesion of ovarian cancer cells following CCL2 stimulation, a process mediated by the P38 MAPK pathway (67,68).
The CCL2/CCR2 signaling pathway drives the proliferation and migration of acute myeloid leukemia cells, as demonstrated by in vitro and in vivo studies (69). In glioblastoma, tumor-secreted CCL2 orchestrates monocyte recruitment, establishing a pro-tumorigenic microenvironment that fosters neoplastic growth (70). Leveraging this mechanistic insight, preclinical investigation have explored CCR2 antagonists as novel antitumor agents. For example, the pharmacological blockade of CCR2 in lung adenocarcinoma A549 cells attenuates their migratory and invasive capabilities (Fig. 1; Table I) (71).
Table I.C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis affects the proliferation, migration and invasion of tumor cells. |
EMT is the initial step of tumor metastasis, and is involved in the progression and metastasis of various types of cancer (72,73). The CCL2/CCR2 signaling axis promotes EMT in liver cancer through MMP2 (74). There is a growing body of data describing a direct stimulatory effect of CCL2 on tumor epithelial cells (8,75). Notably, interruption of the CCL2/CCR2/STAT3 pathway can inhibit the EMT and migration of prostate cancer cells, inhibiting the progression and metastasis of prostate cancer (75). Furthermore, the TME enhances bladder cancer metastasis by modulating estrogen receptor (ER)β/CCL2/CCR2 EMT/MMP9 signaling following mast cell recruitment (76). The CCL2/CCR2 pathway induces the invasion and EMT of HCC in vitro by activating the Hedgehog pathway (Fig. 2) (77).
Tumor angiogenesis requires the production of angiogenic factors by tumor cells and stromal components. CCL2 directly promotes angiogenesis via the activation of CCR2-expressing vascular endothelial cells (78–80). Concurrently, the CCL2/CCR2 signaling axis enhances vascular permeability, facilitating efficient extravasation of tumor cells and metastatic niche formation (81). Human endothelial cells express CCR2, which promotes tumor angiogenesis and progression after binding to CCL2. Several clinical studies have shown that CCL2 may be a biomarker of tumor angiogenesis (82–84). The activation of the CCL2/CCR2 signaling axis in the TME promotes tumor angiogenesis (83). CCL2 directly interacts with CCR2 on the surface of endothelial cells, resulting in increased vessel sprouting and angiogenesis (84). As a chemokine produced in abundance by certain types of tumor, such as hepatocellular carcinoma (HCC), glioblastoma, and Small Cell Lung Cancer (SCLC) (85), it can also directly promote tumor progression. Therefore, therapy using MCP-1 antagonists in combination with other angiogenesis inhibitors may suppress tumor growth (86).
In contrast to earlier assumptions, emerging evidence has indicated that tumor vascular endothelial cells lack CCR2 expression (87,88). Instead, CCL2 drives angiogenesis indirectly by recruiting TAMs and increasing VEGF-A production in these cells (89–92). In addition, CCL2 enhances cancer cell autonomous VEGF secretion, further contributing to neovascularization. In melanoma, this axis is amplified by autocrine/paracrine loops, wherein CCL2 and its receptor CCR2 are co-expressed (93,94). AM-derived TNFα and IL-1α synergize with VEGF to promote endothelial cell activation, thereby accelerating early-stage tumor angiogenesis and growth (9).
The TME is a dynamically complex ecosystem, where inflammatory networks composed of immune cells and their secretory products influence cancer biology and progression (95). Chemokine-chemokine receptor interactions are key to recruiting inflammatory cells into the TME, with CCL2 serving as a key driver of inflammatory monocyte accumulation (96). Inflammatory monocytes exhibit high expression of CCR2 (26), while other CCR2-expressing leukocytes, including CD8+ effector T cells and CD4+ regulatory T cells (Tregs) (19,97) and myeloid-derived suppressor cells (MDSCs) (98), are enriched in inflamed tumor tissues. The CCL2/CCR2 pathway orchestrates metastatic microenvironment formation, exerting pro-tumorigenic and pro-metastatic functions in numerous types of cancer (99,100).
In sarcoma and breast cancer, the CCL2/CCR2 axis mediates the recruitment of TAMs and MDSCs to the TME (89). T cells infiltrate tumors in an antigen-specific manner, with IFN-γ secreted by early-invading T cells inducing tissue macrophages to increase CCL2 expression (101,102). This creates a positive feedback loop wherein CCL2 recruits additional T cells and macrophages via CCR2 signaling, enhancing immune cell infiltration. CCR2 expression is detectable in tumor-infiltrating immune cells, supporting the role of active chemotactic recruitment (101,102). In NPC, this mechanism is exemplified by T cell-derived IFN-γ activating macrophages to secrete CCL2, thereby amplifying T-cell and macrophage accumulation via the CCL2/CCR2 pathway (103).
CAFs, as a key component of the TME, represent activated fibroblast populations that impact stromal compartment remodeling within the TME via collagen deposition and MMP secretion (104). CAFs constitute one of the most prevalent cell types in the tumor stroma (105), and evidence has highlighted their tumor-promoting functions, including accelerating tumor proliferation, facilitating metastatic progression and shielding tumors from therapeutic agent penetration (106–108).
Accumulating evidence has highlighted the crosstalk between CAFs and immunosuppressive cell lineages, which is primarily due to the immune-modulatory functions of CAFs (109–111). CAFs secreting CCL2 facilitate the recruitment of CCR2+ monocytes from the bloodstream into the TME, where direct cell-cell interactions drive monocyte differentiation into MDSCs. CAF-educated MDSCs suppress T-cell proliferation via upregulation of NADPH oxidase 2 and indoleamine 2,3-dioxygenase 1, leading to excessive reactive oxygen species production that inhibits immune effector function (Fig. 3) (112).
Crosstalk with cancer cells drives CCL2 upregulation in CAFs, which contributes to metastatic niche establishment during early tumor progression and modulates broader tumor functionality (113–118). In a murine liver tumor model, fibroblast STAT3/CCL2 signaling has been shown to enhance MDSC recruitment, thereby promoting tumor growth (119). In addition, the CCL2/CCR2 signaling pathway facilitates early breast cancer survival and invasion via a fibroblast-mediated mechanism (120).
A previous study demonstrated that discoidin domain receptor 1 (DDR1) orchestrates an immunosuppressive TME by activating CAFs to secrete CCL2 and IL-6. This signaling axis exhibits dual regulatory effects by recruiting immunosuppressive cells, including MDSCs and TAMs, and facilitating extracellular matrix remodeling through enhanced collagen deposition and MMP9 activation. These findings demonstrate the key role of the DDR1/CCL2/IL-6 pathway in CAF-mediated immunosuppression, providing a potential therapeutic strategy to target tumor fibrosis and immune evasion (121,122).
CCL2 primarily regulates the directional migration and invasive infiltration of reticuloendothelial system cells, with a focus on monocyte/macrophage phenotypes (123). CCL2 induces monocytes to exit the bloodstream and extravasate into peripheral tissue (71), where they differentiate into tissue-resident macrophages. The CCL2/CCR2 signaling pathway has been implicated in recruiting macrophages in various types of human cancer, including those originating in the bladder, cervix, ovary, lung and breast (124–127). TAMs, a major component of infiltrating inflammatory cells, are regulated by the CCL2/CCR2 pathway through a macrophage-dependent mechanism sustained by positive feedback loops (61,128). As a key chemokine system mediating blood cell recruitment, particularly macrophages, into tissues (11,128–130), CCL2/CCR2 signaling drives tumor progression. M2-polarized TAMs influence tumor progression and metastasis (131), and CCL2 secretion recruits TAMs that mediate metastatic phenotypes in ER-negative breast cancer (64).
TAMs drive prostate cancer metastasis via activation of the CCL2/CCR2 signaling pathway (132). Phenotypically, tumor-infiltrating macrophages can be tumor-supportive (M2) or function in tumor immune surveillance (M1). TAMs of the M1 type lead to a better prognosis, whereas TAMs of the M2 type lead to a poorer prognosis. Tumors associated with M2-type TAMs include breast, ovarian and prostate cancer (133–135). TAM recruitment is dependent on the CCL2/CCR2 signaling axis and the formation of a tumor-supportive microenvironment depends on altered cellular dynamics following the interaction of CCL2, T cells and monocytes (136).
TAMs serve a pivotal role in driving hormone resistance in prostate cancer cells (137). Within the prostate TME, M2-polarized TAMs exert pro-tumorigenic effects. Previous studies (78,138) employed a PC3 cell xenograft model to show that CCL2 increases in vivo prostate tumor growth and metastasis by enhancing TAM recruitment and angiogenesis. Investigations (139,140) have also revealed that M2-phenotype TAMs influence cancer progression and metastasis; in human lung cancer, CCL2/CCR2 signaling promotes tumor cell proliferation, migration and M2 polarization of TAMs (131). In pancreatic ductal adenocarcinoma, the CCL2/CCR2 axis recruits TAMs to establish an immunosuppressive TME (141). Conversely, anti-CCL2 antibody treatment in breast cancer xenograft models has been shown to diminish macrophage infiltration and tumor growth (142,143).
The CCL2/CCR2 pathway is indispensable for monocyte/macrophage recruitment. The therapeutic interruption of this pathway suppresses inflammatory monocyte recruitment, TAM infiltration and M2 polarization, thereby reversing the immunosuppressive state of the TME and activating antitumor CD8+ T-cell responses (39). Platelet-derived growth factor-BB-mediated autocrine signaling drives CCL2 secretion, which recruits macrophages via the CCL2/CCR2 axis to facilitate lung cancer cell invasion (144). Targeting CCL2/CCR2 signaling in tumor-infiltrating macrophages has emerged as a promising therapeutic strategy for HCC (145). Preclinical studies (26,93,146,147) have also shown that genetic knockout of CCR2 or pharmacological blockade with CCR2 antagonists inhibits malignant growth and metastasis, decreases postoperative recurrence and improves survival rates. In pancreatic adenocarcinoma, cancer cells exploit chemokine pathways, particularly CCL2/CCR2, to establish an immunosuppressive niche (146). Blocking TAM recruitment inhibits murine breast cancer growth (148). In addition, the JAK2/STAT3 signaling pathway promotes M2-like macrophage polarization, which drives gastric cancer metastasis via EMT (149).
Tregs, a specialized subset of T cells, are key for maintaining peripheral self-tolerance and preventing immunopathological responses (150). Tumors exploit immune evasion mechanisms to sustain uncontrolled growth, with high intratumoral Treg abundance contributing to the establishment of immunosuppressive microenvironments. Tregs exert suppressive effects on T-cell proliferation and cytotoxic function (151,152). The infiltration of Tregs in tumors is influenced by both in situ generation (mediated by cytokine secretion) and peripheral recruitment (driven by chemokine signaling) (153). Tumor-derived CCL2 has been implicated in inducing Treg migration into the TME. For example, in glioma local immunosuppression is enhanced by selectively recruiting CCL2/CCR2-dependent Tregs (129), while colorectal cancer cells secrete CCL2 that binds to CCR2 on cytotoxic T lymphocytes (CTLs), paradoxically promoting CTL migration to tumors (44).
Tumors employ diverse evasion strategies to circumvent immune recognition and elimination. Intratumoral immunosuppressive MDSCs represent a heterogeneous population of immature myeloid cells originating from bone marrow progenitors (154–156). MDSCs exert multifaceted tumor-promoting activities (157). The CCL2/CCR2 signaling pathway governs the recruitment of myelosuppressive cells to tumor sites. Studies have demonstrated that CCR2 is universally expressed on MDSCs, the primary drivers of tumor immune evasion, and blocking CCL2/CCR2 signaling inhibits MDSC migration and tumor growth facilitated by these cells (31,112,158–160). Collectively, these data demonstrate a key role for the CCL2/CCR2 pathway in regulating MDSC trafficking. CCL2/CCR2 interactions also recruit other immunosuppressive cells, including monocytes, to form a pro-metastatic microenvironment (26,98).
The immunosuppressive Treg-MDSC network drives immune exclusion and immune checkpoint inhibitor (ICI) resistance. In head and neck squamous cell carcinoma, semaphorin (SEMA)4D blockade using pepinemab disrupts MDSC recruitment while enhancing T-cell infiltration (KEYNOTE-B84 trial) (161). In bladder cancer, gemcitabine/BCG decreases IL-6-mediated MDSC suppression. Prostate cancer subtyping has revealed TGF-β-enriched, immune-excluded tumors (stage I) with a poor ICI response vs. inflamed subtypes (S-IV). Targeting this axis (via SEMA4D inhibition or myeloid modulation) represents a promising therapeutic strategy (162).
The CCL2/CCR2 signaling pathway serves a central role in shaping the TME by regulating both tumor progression and antitumor immunity (2). Through the activation of the PI3K/AKT, MAPK and EMT pathways (Fig. 2), the CCL2/CCR2 signaling pathway promotes tumor cell proliferation, invasion and metastasis. Paradoxically, it also mediates antitumor effects by recruiting immune cells and activating immunosurveillance mechanisms (143). This dual functionality stems from its ability to recruit diverse immune populations. While MDSCs, TAMs and Tregs establish an immunosuppressive TME that facilitates metastasis, the CCL2/CCR2 pathway simultaneously promotes TIL infiltration and enhances antitumor lymphocyte function (Fig. 4) (163). In melanoma, CCL2/CCR2 signaling drives resistance to BRAF/MEK inhibitors by expanding MDSCs and suppressing CD8+ T cells (164–168). The TNF-related apoptosis-inducing ligand-CCL2 axis further reinforces this resistance by polarizing monocytes toward MDSCs and M2-like macrophages, facilitating tumor progression (169,170).
Melanoma immunogenicity (171) highlights the complex interplay between tumors and immune evasion mechanisms (172). The CCL2/CCR2 axis represents a critical node in this interaction, making its inhibition a potential therapeutic strategy. Preclinical studies (173–175) have demonstrated that targeting this axis, particularly in combination with immunotherapy, may overcome resistance mechanisms and restore antitumor immunity.
The therapeutic targeting of the CCL2/CCR2 axis involves multiple strategies with distinct mechanisms and clinical implications. Small-molecule CCR2 antagonists (such as PF-04136309 and BMS-813160) block immunosuppressive MDSC/TAM recruitment and restore antitumor immunity in preclinical models (164,165,169,176), with ongoing clinical trials (177,178) evaluating their efficacy in combination with chemotherapy or immunotherapy (trial no. NCT03184870). While anti-CCL2 antibodies (carlumab/CNTO-888) have demonstrated partial responses in early trials, compensatory CCL2 upregulation limits their efficacy, prompting the exploration of combination strategies with programmed cell death-1 (PD-1)/CTLA-4 inhibitors to overcome resistance (172). Dual targeting approaches simultaneously inhibiting CCL2 and CCR2 (CCL2-trapping agents and CCR2 antagonists) may prevent microenvironmental bypass mechanisms, particularly in BRAF inhibitor-resistant melanoma (166–168). Furthermore, elevated CCL2/MDSC signatures could serve as predictive biomarkers for patient selection, with studies (179–181) investigating liquid biopsy-based monitoring (CCR2+ exosomes) (170).
The net biological effect of the CCL2/CCR2 axis is fundamentally context-dependent, governed by four intersecting factors: i) Temporal dynamics, where early-phase immune surveillance progressively shifts to late-stage TME remodeling; ii) spatial heterogeneity across distinct tumor niches; iii) immune cell composition ratios, particularly the M1/M2 macrophage equilibrium; and iv) host genetic background, including CCR2 isoform expression patterns. This necessitates identification of the biological threshold where pro-tumor effects supersede antitumor mechanisms. This may be regulated by three factors: i) CCL2 concentration thresholds, ii) specific immune cell infiltration ratio and iii) hypoxia-induced pathway activation states. These mechanistic insight may inform clinical strategies, as evidenced by Phase II trial (182) (trial no. NCT03184870) combining anti-CCL2 agents with PD-1 inhibitors in non-small cell lung cancer, which aim to therapeutically manipulate this balance.
While preclinical findings have provided mechanistic insights into the CCL2/CCR2 pathway in cancer progression, direct clinical validation in human malignancies remains limited. Although studies (183,184) have investigated CCR2 antagonists (PF-04136309) or CCL2 inhibitors (CNTO-888), most therapeutic strategies have focused on an individual target blockade rather than coordinated modulation of the CCL2/CCR2 interaction. Future studies should prioritize the following: i) Comprehensive CCL2/CCR2 co-expression profiling in patient-derived samples, ii) longitudinal assessment of axis activation during CCR2-targeted therapy and iii) development of novel dual-targeting approaches to elucidate the clinical relevance of this chemokine axis in human cancer.
The present study provides a rationale for the clinical evaluation of CCL2/CCR2 axis inhibitors in combination with existing immunotherapies, offering potential for treatment-resistant malignancies, including melanoma. Future studies should focus on optimizing combination strategies and identifying predictive biomarkers for patient stratification.
Not applicable.
The present study was supported by the Scientific Research Fund project of Education Department of Yunnan Province (grant no. 2025J0181).
Not applicable.
YZ, BF, HY and ZY conceived and designed the study. YZ wrote the manuscript. HY constructed figures. GC and ZH designed and implemented the comprehensive literature search strategy, established rigorous eligibility criteria for study inclusion, synthesized key findings to construct the theoretical framework, made substantial contributions to both the Introduction and Discussion sections and actively participated in critical revision of the manuscript with particular focus on ensuring the accuracy and optimal presentation of figures. Specifically, GC conceived the core visual framework for all figures, which was instrumental in interpreting the study's conceptual approach and ZH established the comparative logic and data hierarchy in figures, ensuring perfect alignment with the manuscript's analytical narrative. YiL and XM acquired full-text articles and independently screened literature (resolving discrepancies through consensus), organized references (using EndNote), revised the Discussion section for clarity and clinical relevance, and oordinated revision feedback among all co-authors to ensure consistency. TW conducted supplementary literature searches to validate results, provided visualization support (including figure and table design), revised the Discussion section with critical intellectual input and helped coordinate final revisions. WW, LC, LH, YaL, DL, XC and YY reviewed the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Yoshimura T, Robinson EA, Tanaka S, Appella E, Kuratsu J and Leonard EJ: Purification and amino acid analysis of two human glioma-derived monocyte chemoattractants. J Exp Med. 169:1449–1459. 1989. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshimura T: The production of monocyte chemoattractant protein-1 (MCP-1)/CCL2 in tumor microenvironments. Cytokine. 98:71–78. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kurihara T, Warr G, Loy J and Bravo R: Defects in macrophage recruitment and host defense in mice lacking the CCR2 chemokine receptor. J Exp Med. 186:1757–1762. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Mackay CR: Chemokines: Immunology's high impact factors. Nat Immunol. 2:95–101. 2001. View Article : Google Scholar | |
|
Strieter RM: Chemokines: Not just leukocyte chemoattractants in the promotion of cancer. Nat Immunol. 2:285–286. 2001. View Article : Google Scholar | |
|
Lim SY, Yuzhalin AE, Gordon-Weeks AN and Muschel RJ: Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget. 7:28697–28710. 2016. View Article : Google Scholar | |
|
Loberg RD, Day LL, Harwood J, Ying C, St John LN, Giles R, Neeley CK and Pienta KJ: CCL2 is a potent regulator of prostate cancer cell migration and proliferation. Neoplasia. 8:578–586. 2006. View Article : Google Scholar | |
|
Craig MJ and Loberg RD: CCL2 (Monocyte Chemoattractant Protein-1) in cancer bone metastases. Cancer Metastasis Rev. 25:611–619. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Koga M, Kai H, Egami K, Murohara T, Ikeda A, Yasuoka S, Egashira K, Matsuishi T, Kai M, Kataoka Y, et al: Mutant MCP-1 therapy inhibits tumor angiogenesis and growth of malignant melanoma in mice. Biochem Biophys Res Commun. 365:279–284. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Zhang H, Jiang X, Qian C, Liu Z and Luo D: Factors involved in cancer metastasis: A better understanding to ‘seed and soil’ hypothesis. Mol Cancer. 16:1762017. View Article : Google Scholar : PubMed/NCBI | |
|
Deshmane SL, Kremlev S, Amini S and Sawaya BE: Monocyte chemoattractant protein-1 (MCP-1): An overview. J Interferon Cytokine Res. 29:313–326. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Charo IF and Ransohoff RM: The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 354:610–621. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Raman D, Baugher PJ, Thu YM and Richmond A: Role of chemokines in tumor growth. Cancer Lett. 256:137–165. 2007. View Article : Google Scholar | |
|
Balkwill FR: The chemokine system and cancer. J Pathol. 226:148–157. 2012. View Article : Google Scholar | |
|
Frederick MJ and Clayman GL: Chemokines in cancer. Expert Rev Mol Med. 3:1–18. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Paget S: The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8:98–101. 1989.PubMed/NCBI | |
|
Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar | |
|
de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 41:374–403. 2023. View Article : Google Scholar | |
|
Brown CE, Vishwanath RP, Aguilar B, Starr R, Najbauer J, Aboody KS and Jensen MC: Tumor-derived chemokine MCP-1/CCL2 is sufficient for mediating tumor tropism of adoptively transferred T cells. J Immunol. 179:3332–3341. 2007. View Article : Google Scholar | |
|
Rollins BJ and Sunday ME: Suppression of tumor formation in vivo by expression of the JE gene in malignant cells. Mol Cell Biol. 11:3125–3131. 1991. View Article : Google Scholar | |
|
Monti P, Leone BE, Marchesi F, Balzano G, Zerbi A, Scaltrini F, Pasquali C, Calori G, Pessi F, Sperti C, et al: The CC chemokine MCP-1/CCL2 in pancreatic cancer progression: regulation of expression and potential mechanisms of antimalignant activity. Cancer Res. 63:7451–7461. 2003.PubMed/NCBI | |
|
Gunderson AJ, Yamazaki T, McCarty K, Fox N, Phillips M, Alice A, Blair T, Whiteford M, O'Brien D, Ahmad R, et al: TGFβ suppresses CD8(+) T cell expression of CXCR3 and tumor trafficking. Nat Commun. 11:17492020. View Article : Google Scholar : PubMed/NCBI | |
|
Qin J, Gong Q, Zhou C, Xu J, Cheng Y, Xu W, Zhu D, Liu Y, Zhang Y, Wang Y, et al: Differential expression pattern of CC chemokine receptor 7 guides precision treatment of hepatocellular carcinoma. Signal Transduct Target Ther. 10:2292025. View Article : Google Scholar : PubMed/NCBI | |
|
Wei XL, Zhang Y, Zhao HY, Fang WF, Luo HY, Qiu MZ, He MM, Zou BY, Xie J, Jin CL, et al: Safety and clinical activity of SHR7390 monotherapy or combined with camrelizumab for advanced solid tumor: Results from two phase I trials. Oncologist. 28:e36–e44. 2023. View Article : Google Scholar | |
|
Mestdagt M, Polette M, Buttice G, Noël A, Ueda A, Foidart JM and Gilles C: Transactivation of MCP-1/CCL2 by beta-catenin/TCF-4 in human breast cancer cells. Int J Cancer. 118:35–42. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Kitamura T, Qian BZ, Soong D, Cassetta L, Noy R, Sugano G, Kato Y, Li J and Pollard JW: CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J Exp Med. 212:1043–1059. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Granot Z, Henke E, Comen EA, King TA, Norton L and Benezra R: Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 20:300–314. 2011. View Article : Google Scholar | |
|
Pekarek LA, Starr BA, Toledano AY and Schreiber H: Inhibition of tumor growth by elimination of granulocytes. J Exp Med. 181:435–440. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Shojaei F, Singh M, Thompson JD and Ferrara N: Role of Bv8 in neutrophil-dependent angiogenesis in a transgenic model of cancer progression. Proc Natl Acad Sci USA. 105:2640–2645. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Youn JI, Nagaraj S, Collazo M and Gabrilovich DI: Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol. 181:5791–5802. 2008. View Article : Google Scholar | |
|
Nozawa H, Chiu C and Hanahan D: Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci USA. 103:12493–12498. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
De Larco JE, Wuertz BR and Furcht LT: The potential role of neutrophils in promoting the metastatic phenotype of tumors releasing interleukin-8. Clin Cancer Res. 10:4895–4900. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng Y, Mo F, Li Q, Han X, Shi H, Chen S, Wei Y and Wei X: Targeting CXCR2 inhibits the progression of lung cancer and promotes therapeutic effect of cisplatin. Mol Cancer. 20:622021. View Article : Google Scholar : PubMed/NCBI | |
|
Song M, He J, Pan QZ, Yang J, Zhao J, Zhang YJ, Huang Y, Tang Y, Wang Q, He J, et al: Cancer-associated fibroblast-mediated cellular crosstalk supports hepatocellular carcinoma progression. Hepatology. 73:1717–1735. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dutta P, Sarkissyan M, Paico K, Wu Y and Vadgama JV: MCP-1 is overexpressed in triple-negative breast cancers and drives cancer invasiveness and metastasis. Breast Cancer Res Treat. 170:477–486. 2018. View Article : Google Scholar | |
|
Parween F, Singh SP, Kathuria N, Zhang HH, Ashida S, Otaizo-Carrasquero FA, Shamsaddini A, Gardina PJ, Ganesan S, Kabat J, et al: Migration arrest and transendothelial trafficking of human pathogenic-like Th17 cells are mediated by differentially positioned chemokines. Nat Commun. 16:19782025. View Article : Google Scholar : PubMed/NCBI | |
|
Dey D, Pal S, Chakraborty BC, Baidya A, Bhadra S, Ghosh R, Banerjee S, Ahammed SKM, Chowdhury A and Datta S: Multifaceted defects in monocytes in different phases of chronic hepatitis B virus infection: Lack of restoration after antiviral therapy. Microbiol Spectr. 10:e01939222022. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Yao W, Yuan Y, Chen P, Li B, Li J, Chu R, Song H, Xie D, Jiang X and Wang H: Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 66:157–167. 2017. View Article : Google Scholar | |
|
Guerra N, Tan YX, Joncker NT, Choy A, Gallardo F, Xiong N, Knoblaugh S, Cado D, Greenberg NM and Raulet DH: NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity. 28:571–580. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V and Lowe SW: Non-cell-autonomous tumor suppression by p53. Cell. 153:449–460. 2013. View Article : Google Scholar | |
|
Iannello A, Thompson TW, Ardolino M, Lowe SW and Raulet DH: p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells. J Exp Med. 210:2057–2069. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nagai M and Masuzawa T: Vaccination with MCP-1 cDNA transfectant on human malignant glioma in nude mice induces migration of monocytes and NK cells to the tumor. Int Immunopharmacol. 1:657–664. 2001. View Article : Google Scholar | |
|
Berencsi K, Rani P, Zhang T, Gross L, Mastrangelo M, Meropol NJ, Herlyn D and Somasundaram R: In vitro migration of cytotoxic T lymphocyte derived from a colon carcinoma patient is dependent on CCL2 and CCR2. J Transl Med. 9:332011. View Article : Google Scholar : PubMed/NCBI | |
|
Nakasone Y, Fujimoto M, Matsushita T, Hamaguchi Y, Huu DL, Yanaba M, Sato S, Takehara K and Hasegawa M: Host-derived MCP-1 and MIP-1α regulate protective anti-tumor immunity to localized and metastatic B16 melanoma. Am J Pathol. 180:365–374. 2012. View Article : Google Scholar | |
|
Joyce JA and Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 9:239–252. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Sarkar S, Cua R, Zhou Y, Hader W and Yong VW: A dialog between glioma and microglia that promotes tumor invasiveness through the CCL2/CCR2/interleukin-6 axis. Carcinogenesis. 33:312–319. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Bogenrieder T and Herlyn M: Axis of evil: molecular mechanisms of cancer metastasis. Oncogene. 22:6524–6536. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Rafei M, Deng J, Boivin MN, Williams P, Matulis SM, Yuan S, Birman E, Forner K, Yuan L, Castellino C, et al: A MCP1 fusokine with CCR2-specific tumoricidal activity. Mol Cancer. 10:1212011. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson Z, Power CA, Weiss C, Rintelen F, Ji H, Ruckle T, Camps M, Wells TN, Schwarz MK, Proudfoot AE and Rommel C: Chemokine inhibition-why, when, where, which and how? Biochem Soc Trans. 32:366–377. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Mellado M, Rodríguez-Frade JM, Aragay A, del Real G, Martín AM, Vila-Coro AJ, Serrano A, Mayor F Jr and Martínez-A C: The chemokine monocyte chemotactic protein 1 triggers Janus kinase 2 activation and tyrosine phosphorylation of the CCR2B receptor. J Immunol. 161:805–813. 1998. View Article : Google Scholar | |
|
Jiménez-Sainz MC, Fast B, Mayor F Jr and Aragay AM: Signaling pathways for monocyte chemoattractant protein 1-mediated extracellular signal-regulated kinase activation. Mol Pharmacol. 64:773–782. 2003. View Article : Google Scholar | |
|
Tang CH and Tsai CC: CCL2 increases MMP-9 expression and cell motility in human chondrosarcoma cells via the Ras/Raf/MEK/ERK/NF-κB signaling pathway. Biochem Pharmacol. 83:335–344. 2012. View Article : Google Scholar | |
|
Dagouassat M, Suffee N, Hlawaty H, Haddad O, Charni F, Laguillier C, Vassy R, Martin L, Schischmanoff PO, Gattegno L, et al: Monocyte chemoattractant protein-1 (MCP-1)/CCL2 secreted by hepatic myofibroblasts promotes migration and invasion of human hepatoma cells. Int J Cancer. 126:1095–1108. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Schröer N, Pahne J, Walch B, Wickenhauser C and Smola S: Molecular pathobiology of human cervical high-grade lesions: Paracrine STAT3 activation in tumor-instructed myeloid cells drives local MMP-9 expression. Cancer Res. 71:87–97. 2011. View Article : Google Scholar | |
|
Balkwill F and Mantovani A: Inflammation and cancer: Back to Virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar | |
|
Pollard JW: Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 4:71–78. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
van Kempen LC, de Visser KE and Coussens LM: Inflammation, proteases and cancer. Eur J Cancer. 42:728–734. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Yumimoto K, Sugiyama S, Mimori K and Nakayama KI: Potentials of C-C motif chemokine 2-C-C chemokine receptor type 2 blockers including propagermanium as anticancer agents. Cancer Sci. 110:2090–2099. 2019. View Article : Google Scholar | |
|
Fang WB, Jokar I, Zou A, Lambert D, Dendukuri P and Cheng N: CCL2/CCR2 chemokine signaling coordinates survival and motility of breast cancer cells through Smad3 protein- and p42/44 mitogen-activated protein kinase (MAPK)-dependent mechanisms. J Biol Chem. 287:36593–36608. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Yao M, Fang W, Smart C, Hu Q, Huang S, Alvarez N, Fields P and Cheng N: CCR2 chemokine receptors enhance growth and cell-cycle progression of breast cancer cells through SRC and PKC Activation. Mol Cancer Res. 17:604–617. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gao J, Wang A, Zhang M, Li H, Wang K, Han Y, Wang Z, Shi C and Wang W: RNAi targeting of CCR2 gene expression induces apoptosis and inhibits the proliferation, migration, and invasion of PC-3M cells. Oncol Res. 21:73–82. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Chiu HY, Sun KH, Chen SY, Wang HH, Lee MY, Tsou YC, Jwo SC, Sun GH and Tang SJ: Autocrine CCL2 promotes cell migration and invasion via PKC activation and tyrosine phosphorylation of paxillin in bladder cancer cells. Cytokine. 59:423–432. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Vande Broek I, Asosingh K, Vanderkerken K, Straetmans N, Van Camp B and Van Riet I: Chemokine receptor CCR2 is expressed by human multiple myeloma cells and mediates migration to bone marrow stromal cell-produced monocyte chemotactic proteins MCP-1, −2 and −3. Br J Cancer. 88:855–862. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Lv X, Chen J, Xie C, Xia W, Jiang C, Zeng T, Ye Y, Ke L, Yu Y, et al: CCL2-CCR2 axis promotes metastasis of nasopharyngeal carcinoma by activating ERK1/2-MMP2/9 pathway. Oncotarget. 7:15632–15647. 2016. View Article : Google Scholar | |
|
Xu M, Wang S, Qi Y, Chen L, Frank JA, Yang XH, Zhang Z, Shi X and Luo J: Role of MCP-1 in alcohol-induced aggressiveness of colorectal cancer cells. Mol Carcinog. 55:1002–1011. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Yasui H, Kajiyama H, Tamauchi S, Suzuki S, Peng Y, Yoshikawa N, Sugiyama M, Nakamura K and Kikkawa F: CCL2 secreted from cancer-associated mesothelial cells promotes peritoneal metastasis of ovarian cancer cells through the P38-MAPK pathway. Clin Exp Metastasis. 37:145–158. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Furukawa S, Soeda S, Kiko Y, Suzuki O, Hashimoto Y, Watanabe T, Nishiyama H, Tasaki K, Hojo H, Abe M and Fujimori K: MCP-1 promotes invasion and adhesion of human ovarian cancer cells. Anticancer Res. 33:4785–4790. 2013.PubMed/NCBI | |
|
Macanas-Pirard P, Quezada T, Navarrete L, Broekhuizen R, Leisewitz A, Nervi B and Ramírez PA: The CCL2/CCR2 axis affects transmigration and proliferation but not resistance to chemotherapy of acute myeloid leukemia cells. PLoS One. 12:e01688882017. View Article : Google Scholar : PubMed/NCBI | |
|
Salacz ME, Kast RE, Saki N, Brüning A, Karpel-Massler G and Halatsch ME: Toward a noncytotoxic glioblastoma therapy: Blocking MCP-1 with the MTZ Regimen. Onco Targets Ther. 9:2535–2545. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
An J, Xue Y, Long M, Zhang G, Zhang J and Su H: Targeting CCR2 with its antagonist suppresses viability, motility and invasion by downregulating MMP-9 expression in non-small cell lung cancer cells. Oncotarget. 8:39230–39240. 2017. View Article : Google Scholar | |
|
Giannelli G, Koudelkova P, Dituri F and Mikulits W: Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol. 65:798–808. 2016. View Article : Google Scholar | |
|
van Zijl F, Zulehner G, Petz M, Schneller D, Kornauth C, Hau M, Machat G, Grubinger M, Huber H and Mikulits W: Epithelial-mesenchymal transition in hepatocellular carcinoma. Future Oncol. 5:1169–1179. 2009. View Article : Google Scholar | |
|
Li H, Li H, Li XP, Zou H, Liu L, Liu W and Duan T: C-C chemokine receptor type 2 promotes epithelial-to-mesenchymal transition by upregulating matrix metalloproteinase-2 in human liver cancer. Oncol Rep. 40:2734–2741. 2018.PubMed/NCBI | |
|
Izumi K, Fang LY, Mizokami A, Namiki M, Li L, Lin WJ and Chang C: Targeting the androgen receptor with siRNA promotes prostate cancer metastasis through enhanced macrophage recruitment via CCL2/CCR2-induced STAT3 activation. EMBO Mol Med. 5:1383–1401. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rao Q, Chen Y, Yeh CR, Ding J, Li L, Chang C and Yeh S: Recruited mast cells in the tumor microenvironment enhance bladder cancer metastasis via modulation of ERβ/CCL2/CCR2 EMT/MMP9 signals. Oncotarget. 7:7842–7855. 2016. View Article : Google Scholar | |
|
Zhuang H, Cao G, Kou C and Liu T: CCL2/CCR2 axis induces hepatocellular carcinoma invasion and epithelial-mesenchymal transition in vitro through activation of the Hedgehog pathway. Oncol Rep. 39:21–30. 2018.PubMed/NCBI | |
|
Koide N, Nishio A, Sato T, Sugiyama A and Miyagawa S: Significance of macrophage chemoattractant protein-1 expression and macrophage infiltration in squamous cell carcinoma of the esophagus. Am J Gastroenterol. 99:1667–1674. 2004. View Article : Google Scholar | |
|
Salcedo R, Ponce ML, Young HA, Wasserman K, Ward JM, Kleinman HK, Oppenheim JJ and Murphy WJ: Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression. Blood. 96:34–40. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Hong KH, Ryu J and Han KH: Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood. 105:1405–1407. 2025. View Article : Google Scholar | |
|
Wyler L, Napoli CU, Ingold B, Sulser T, Heikenwälder M, Schraml P and Moch H: Brain metastasis in renal cancer patients: Metastatic pattern, tumour-associated macrophages and chemokine/chemoreceptor expression. Br J Cancer. 110:686–694. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Valković T, Babarović E, Lučin K, Štifter S, Aralica M, Seili-Bekafigo I, Duletić-Načinović A and Jonjić N: Plasma levels of monocyte chemotactic protein-1 are associated with clinical features and angiogenesis in patients with multiple myeloma. Biomed Res Int. 2016:78705902016. | |
|
Li F, Kitajima S, Kohno S, Yoshida A, Tange S, Sasaki S, Okada N, Nishimoto Y, Muranaka H, Nagatani N, et al: Retinoblastoma inactivation induces a protumoral microenvironment via enhanced CCL2 secretion. Cancer Res. 79:3903–3915. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Loberg R, Liao J, Ying C, Snyder LA, Pienta KJ and McCauley LK: A destructive cascade mediated by CCL2 facilitates prostate cancer growth in bone. Cancer Res. 69:1685–1692. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Balkwill F: Cancer and the chemokine network. Nat Rev Cancer. 4:540–550. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Niu J, Azfer A, Zhelyabovska O, Fatma S and Kolattukudy PE: Monocyte chemotactic protein (MCP)-1 promotes angiogenesis via a novel transcription factor, MCP-1-induced protein (MCPIP). J Biol Chem. 283:14542–14551. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ohta M, Kitadai Y, Tanaka S, Yoshihara M, Yasui W, Mukaida N, Haruma K and Chayama K: Monocyte chemoattractant protein-1 expression correlates with macrophage infiltration and tumor vascularity in human esophageal squamous cell carcinomas. Int J Cancer. 102:220–224. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Kuroda T, Kitadai Y, Tanaka S, Yang X, Mukaida N, Yoshihara M and Chayama K: Monocyte chemoattractant protein-1 transfection induces angiogenesis and tumorigenesis of gastric carcinoma in nude mice via macrophage recruitment. Clin Cancer Res. 11:7629–7636. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Goede V, Brogelli L, Ziche M and Augustin HG: Induction of inflammatory angiogenesis by monocyte chemoattractant protein-1. Int J Cancer. 82:765–770. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Low-Marchelli JM, Ardi VC, Vizcarra EA, van Rooijen N, Quigley JP and Yang J: Twist1 induces CCL2 and recruits macrophages to promote angiogenesis. Cancer Res. 73:662–671. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Varney ML, Olsen KJ, Mosley RL and Singh RK: Paracrine regulation of vascular endothelial growth factor-a expression during macrophage-melanoma cell interaction: Role of monocyte chemotactic protein-1 and macrophage colony-stimulating factor. J Interferon Cytokine Res. 25:674–683. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MP and Donners MM: Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 17:109–118. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Liu L, Li Y and Li B: Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment. Biochim Biophys Acta Rev Cancer. 1880:1893442025. View Article : Google Scholar : PubMed/NCBI | |
|
Biswas SK and Mantovani A: Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat Immunol. 11:889–896. 2010. View Article : Google Scholar | |
|
Allavena P, Germano G, Marchesi F and Mantovani A: Chemokines in cancer related inflammation. Exp Cell Res. 317:664–673. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Fridlender ZG, Buchlis G, Kapoor V, Cheng G, Sun J, Singhal S, Crisanti MC, Wang LC, Heitjan D, Snyder LA and Albelda SM: CCL2 blockade augments cancer immunotherapy. Cancer Res. 70:109–118. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Huang B, Lei Z, Zhao J, Gong W, Liu J, Chen Z, Liu Y, Li D, Yuan Y, Zhang GM and Feng ZH: CCL2/CCR2 pathway mediates recruitment of myeloid suppressor cells to cancers. Cancer Lett. 252:86–92. 2007. View Article : Google Scholar | |
|
Samaniego R, Estecha A, Relloso M, Longo N, Escat JL, Longo-Imedio I, Avilés JA, del Pozo MA, Puig-Kröger A and Sánchez-Mateos P: Mesenchymal contribution to recruitment, infiltration, and positioning of leukocytes in human melanoma tissues. J Invest Dermatol. 133:2255–2264. 2013. View Article : Google Scholar | |
|
Roblek M, Strutzmann E, Zankl C, Adage T, Heikenwalder M, Atlic A, Weis R, Kungl A and Borsig L: Targeting of CCL2-CCR2-glycosaminoglycan axis using a CCL2 decoy protein attenuates metastasis through inhibition of tumor cell seeding. Neoplasia. 18:49–59. 2016. View Article : Google Scholar | |
|
Zhu Y, Yan P, Wang R, Lai J, Tang H, Xiao X, Yu R, Bao X, Zhu F, Wang K, et al: Opioid-induced fragile-like regulatory T cells contribute to withdrawal. Cell. 186:591–606.e23. 2023. View Article : Google Scholar | |
|
Kong X, Wu S, Dai X, Yu W, Wang J, Sun Y, Ji Z, Ma L, Dai X, Chen H, et al: A comprehensive profile of chemokines in the peripheral blood and vascular tissue of patients with Takayasu arteritis. Arthritis Res Ther. 24:492022. View Article : Google Scholar : PubMed/NCBI | |
|
Tang KF, Tan SY, Chan SH, Chong SM, Loh KS, Tan LK and Hu H: A distinct expression of CC chemokines by macrophages in nasopharyngeal carcinoma: implication for the intense tumor infiltration by T lymphocytes and macrophages. Hum Pathol. 32:42–49. 2001. View Article : Google Scholar | |
|
Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Gok Yavuz B, Gunaydin G, Gedik ME, Kosemehmetoglu K, Karakoc D, Ozgur F and Guc D: Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci Rep. 9:31722019. View Article : Google Scholar : PubMed/NCBI | |
|
LeBleu VS and Kalluri R: A peek into cancer-associated fibroblasts: origins, functions and translational impact. Dis Model Mech. 11:dmm0294472018. View Article : Google Scholar : PubMed/NCBI | |
|
Loeffler M, Krüger JA, Niethammer AG and Reisfeld RA: Targeting tumor-associated fibroblasts improves cancer chemotherapy by increasing intratumoral drug uptake. J Clin Invest. 116:1955–1962. 2006. View Article : Google Scholar | |
|
Valkenburg KC, de Groot AE and Pienta KJ: Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 15:366–381. 2018. View Article : Google Scholar | |
|
Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, Sirven P, Magagna I, Fuhrmann L, Bernard C, et al: Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 33:463–479.e10. 2018. View Article : Google Scholar | |
|
Cho H, Seo Y, Loke KM, Kim SW, Oh SM, Kim JH, Soh J, Kim HS, Lee H, Kim J, et al: Cancer-stimulated CAFs enhance monocyte differentiation and protumoral TAM activation via IL6 and GM-CSF secretion. Clin Cancer Res. 24:5407–5421. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mace TA, Ameen Z, Collins A, Wojcik S, Mair M, Young GS, Fuchs JR, Eubank TD, Frankel WL, Bekaii-Saab T, et al: Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 73:3007–3018. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, Afshar R, Georgiev P, Sze MA, Song XS, et al: Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res. 8:436–450. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jung DW, Che ZM and Kim J, Kim K, Kim KY, Williams D and Kim J: Tumor-stromal crosstalk in invasion of oral squamous cell carcinoma: A pivotal role of CCL7. Int J Cancer. 127:332–344. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Barbai T, Fejős Z, Puskas LG, Tímár J and Rásó E: The importance of microenvironment: the role of CCL8 in metastasis formation of melanoma. Oncotarget. 6:29111–29128. 2015. View Article : Google Scholar | |
|
Tsuyada A, Chow A, Wu J, Somlo G, Chu P, Loera S, Luu T, Li AX, Wu X, Ye W, et al: CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells. Cancer Res. 72:2768–2779. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, Nishio M, Shigeoka M, Kakeji Y and Yokozaki H: Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 99:777–792. 2019. View Article : Google Scholar | |
|
Pausch TM, Aue E, Wirsik NM, Freire Valls A, Shen Y, Radhakrishnan P, Hackert T, Schneider M and Schmidt T: Metastasis-associated fibroblasts promote angiogenesis in metastasized pancreatic cancer via the CXCL8 and the CCL2 axes. Sci Rep. 10:54202020. View Article : Google Scholar : PubMed/NCBI | |
|
Vickman RE, Broman MM, Lanman NA, Franco OE, Sudyanti PAG, Ni Y, Ji Y, Helfand BT, Petkewicz J, Paterakos MC, et al: Heterogeneity of human prostate carcinoma-associated fibroblasts implicates a role for subpopulations in myeloid cell recruitment. Prostate. 80:173–185. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, Dang Y, Chu Y, Fan J and He R: FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 76:4124–4135. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Brummer G, Acevedo DS, Hu Q, Portsche M, Fang WB, Yao M, Zinda B, Myers M, Alvarez N, Fields P, et al: Chemokine signaling facilitates early-stage breast cancer survival and invasion through fibroblast-dependent mechanisms. Mol Cancer Res. 16:296–308. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Majo S and Auguste P: The Yin and Yang of discoidin domain receptors (DDRs): Implications in tumor growth and metastasis development. Cancers (Basel). 13:17252021. View Article : Google Scholar : PubMed/NCBI | |
|
Scordamaglia D, Talia M, Cirillo F, Zicarelli A, Mondino AA, De Rosis S, Di Dio M, Silvestri F, Meliti C, Miglietta AM, et al: Interleukin-1β mediates a tumor-supporting environment prompted by IGF1 in triple-negative breast cancer (TNBC). J Transl Med. 23:6602025. View Article : Google Scholar : PubMed/NCBI | |
|
Vakilian A, Khorramdelazad H, Heidari P, Sheikh Rezaei Z and Hassanshahi G: CCL2/CCR2 signaling pathway in glioblastoma multiforme. Neurochem Int. 103:1–7. 2017. View Article : Google Scholar | |
|
Zijlmans HJ, Fleuren GJ, Baelde HJ, Eilers PH, Kenter GG and Gorter A: The absence of CCL2 expression in cervical carcinoma is associated with increased survival and loss of heterozygosity at 17q11.2. J Pathol. 208:507–517. 2006. View Article : Google Scholar | |
|
Yoshidome H, Kohno H, Shida T, Kimura F, Shimizu H, Ohtsuka M, Nakatani Y and Miyazaki M: Significance of monocyte chemoattractant protein-1 in angiogenesis and survival in colorectal liver metastases. Int J Oncol. 34:923–930. 2009. | |
|
Zhang J, Patel L and Pienta KJ: CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev. 21:41–48. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Soria G, Ofri-Shahak M, Haas I, Yaal-Hahoshen N, Leider-Trejo L, Leibovich-Rivkin T, Weitzenfeld P, Meshel T, Shabtai E, Gutman M and Ben-Baruch A: Inflammatory mediators in breast cancer: coordinated expression of TNFα & IL-1β with CCL2 & CCL5 and effects on epithelial-to-mesenchymal transition. BMC Cancer. 11:1302011. View Article : Google Scholar : PubMed/NCBI | |
|
Jin G, Kawsar HI, Hirsch SA, Zeng C, Jia X, Feng Z, Ghosh SK, Zheng QY, Zhou A, McIntyre TM and Weinberg A: An antimicrobial peptide regulates tumor-associated macrophage trafficking via the chemokine receptor CCR2, a model for tumorigenesis. PLoS One. 5:e109932010. View Article : Google Scholar : PubMed/NCBI | |
|
Vasco C, Canazza A, Rizzo A, Mossa A, Corsini E, Silvani A, Fariselli L, Salmaggi A and Ciusani E: Circulating T regulatory cells migration and phenotype in glioblastoma patients: An in vitro study. J Neurooncol. 115:353–363. 2013. View Article : Google Scholar | |
|
del Pozo MA, Cabañas C, Montoya MC, Ager A, Sánchez-Mateos P and Sánchez-Madrid F: ICAMs redistributed by chemokines to cellular uropods as a mechanism for recruitment of T lymphocytes. J Cell Biol. 137:493–508. 1997. View Article : Google Scholar | |
|
Schmall A, Al-Tamari HM, Herold S, Kampschulte M, Weigert A, Wietelmann A, Vipotnik N, Grimminger F, Seeger W, Pullamsetti SS and Savai R: Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer. Am J Respir Crit Care Med. 191:437–447. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Maolake A, Izumi K, Shigehara K, Natsagdorj A, Iwamoto H, Kadomoto S, Takezawa Y, Machioka K, Narimoto K, Namiki M, et al: Tumor-associated macrophages promote prostate cancer migration through activation of the CCL22-CCR4 axis. Oncotarget. 8:9739–9751. 2017. View Article : Google Scholar | |
|
Qian BZ and Pollard JW: Macrophage diversity enhances tumor progression and metastasis. Cell. 141:39–51. 2010. View Article : Google Scholar | |
|
DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N and Coussens LM: CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 16:91–102. 2009. View Article : Google Scholar | |
|
Reinartz S, Schumann T, Finkernagel F, Wortmann A, Jansen JM, Meissner W, Krause M, Schwörer AM, Wagner U, Müller-Brüsselbach S and Müller R: Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: Correlation of CD163 expression, cytokine levels and early relapse. Int J Cancer. 134:32–42. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
van Attekum MHA, van Bruggen JAC, Slinger E, Lebre MC, Reinen E, Kersting S, Eldering E and Kater AP: CD40 signaling instructs chronic lymphocytic leukemia cells to attract monocytes via the CCR2 axis. Haematologica. 102:2069–2076. 2017. View Article : Google Scholar | |
|
Zhu P, Baek SH, Bourk EM, Ohgi KA, Garcia-Bassets I, Sanjo H, Akira S, Kotol PF, Glass CK, Rosenfeld MG and Rose DW: Macrophage/cancer cell interactions mediate hormone resistance by a nuclear receptor derepression pathway. Cell. 124:615–629. 2006. View Article : Google Scholar | |
|
Loberg RD, Ying C, Craig M, Yan L, Snyder LA and Pienta KJ: CCL2 as an important mediator of prostate cancer growth in vivo through the regulation of macrophage infiltration. Neoplasia. 9:556–562. 2007. View Article : Google Scholar | |
|
Ye J, Yang Y, Jin J, Ji M, Gao Y, Feng Y, Wang H, Chen X and Liu Y: Targeted delivery of chlorogenic acid by mannosylated liposomes to effectively promote the polarization of TAMs for the treatment of glioblastoma. Bioact Mater. 5:694–708. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu F, Li X, Zhang Y, Ge S, Shi Z, Liu Q and Jiang S: Targeting tumor-associated macrophages to overcome immune checkpoint inhibitor resistance in hepatocellular carcinoma. J Exp Clin Cancer Res. 44:2272025. View Article : Google Scholar : PubMed/NCBI | |
|
Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, Toriola AT, Nieman RK, Worley LA, Yano M, et al: Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 17:651–662. 2016. View Article : Google Scholar | |
|
Fujimoto H, Sangai T, Ishii G, Ikehara A, Nagashima T, Miyazaki M and Ochiai A: Stromal MCP-1 in mammary tumors induces tumor-associated macrophage infiltration and contributes to tumor progression. Int J Cancer. 125:1276–1284. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Bianconi V, Sahebkar A, Atkin SL and Pirro M: The regulation and importance of monocyte chemoattractant protein-1. Curr Opin Hematol. 25:44–51. 2018. View Article : Google Scholar | |
|
Ding M, He SJ and Yang J: MCP-1/CCL2 mediated by autocrine loop of PDGF-BB promotes invasion of lung cancer cell by recruitment of macrophages via CCL2-CCR2 Axis. J Interferon Cytokine Res. 39:224–232. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Avila MA and Berasain C: Targeting CCL2/CCR2 in tumor-infiltrating macrophages: A tool emerging out of the box against hepatocellular carcinoma. Cell Mol Gastroenterol Hepatol. 7:293–294. 2019. View Article : Google Scholar | |
|
Nywening TM, Belt BA, Cullinan DR, Panni RZ, Han BJ, Sanford DE, Jacobs RC, Ye J, Patel AA, Gillanders WE, et al: Targeting both tumour-associated CXCR2(+) neutrophils and CCR2(+) macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut. 67:1112–1123. 2018. View Article : Google Scholar | |
|
Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, Mitchem JB, Plambeck-Suess SM, Worley LA, Goetz BD, et al: Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: A role for targeting the CCL2/CCR2 axis. Clin Cancer Res. 19:3404–3415. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Yin S, Wang N, Riabov V, Mossel DM, Larionova I, Schledzewski K, Trofimova O, Sevastyanova T, Zajakina A, Schmuttermaier C, et al: SI-CLP inhibits the growth of mouse mammary adenocarcinoma by preventing recruitment of tumor-associated macrophages. Int J Cancer. 146:1396–1408. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Zhang X, Wu F, Zhou Y, Bao Z, Li H, Zheng P and Zhao S: Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death Dis. 10:9182019. View Article : Google Scholar : PubMed/NCBI | |
|
Crane CA, Ahn BJ, Han SJ and Parsa AT: Soluble factors secreted by glioblastoma cell lines facilitate recruitment, survival, and expansion of regulatory T cells: Implications for immunotherapy. Neuro Oncol. 14:584–595. 2012. View Article : Google Scholar | |
|
Marangoni F, Zhakyp A, Corsini M, Geels SN, Carrizosa E, Thelen M, Mani V, Prüßmann JN, Warner RD, Ozga AJ, et al: Expansion of tumor-associated Treg cells upon disruption of a CTLA-4-dependent feedback loop. Cell. 184:3998–4015.e19. 2021. View Article : Google Scholar | |
|
Rodrigues PF, Wu S, Trsan T, Panda SK, Fachi JL, Liu Y, Du S, de Oliveira S, Antonova AU, Khantakova D, et al: Rorγt-positive dendritic cells are required for the induction of peripheral regulatory T cells in response to oral antigens. Cell. 188:2720–2737.e22. 2025. View Article : Google Scholar | |
|
Filaci G, Fenoglio D, Fravega M, Ansaldo G, Borgonovo G, Traverso P, Villaggio B, Ferrera A, Kunkl A, Rizzi M, et al: CD8+ CD28- T regulatory lymphocytes inhibiting T cell proliferative and cytotoxic functions infiltrate human cancers. J Immunol,. 179:4323–4334. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Serafini P, Borrello I and Bronte V: Myeloid suppressor cells in cancer: Recruitment, phenotype, properties, and mechanisms of immune suppression. Semin Cancer Biol. 16:53–65. 2006. View Article : Google Scholar | |
|
Serafini P, De Santo C, Marigo I, Cingarlini S, Dolcetti L, Gallina G, Zanovello P and Bronte V: Derangement of immune responses by myeloid suppressor cells. Cancer Immunol Immunother. 53:64–72. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, McDermott D, Quiceno D, Youmans A, O'Neill A, et al: Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: A mechanism of tumor evasion. Cancer Res. 65:3044–3048. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Ugel S, De Sanctis F, Mandruzzato S and Bronte V: Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 125:3365–3376. 2015. View Article : Google Scholar | |
|
Kumar V, Cheng P, Condamine T, Mony S, Languino LR, McCaffrey JC, Hockstein N, Guarino M, Masters G, Penman E, et al: CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity. 44:303–315. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Flavell RA, Sanjabi S, Wrzesinski SH and Licona-Limón P: The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol. 10:554–567. 2010. View Article : Google Scholar | |
|
Panni RZ, Sanford DE, Belt BA, Mitchem JB, Worley LA, Goetz BD, Mukherjee P, Wang-Gillam A, Link DC, Denardo DG, et al: Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol Immunother. 63:513–528. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Feigin A, vans EE, Fisher TL, Leonard JE, Smith ES, Reader A, Mishra V, Manber R, Walters KA, Kowarski L, et al: Pepinemab antibody blockade of SEMA4D in early Huntington's disease: A randomized, placebo-controlled, phase 2 trial. Nat Med. 28:2183–2193. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lin BB, Lei HQ, Xiong HY, Fu X, Shi F, Yang XW, Yang YF, Liao GL, Feng YP, Jiang DG and Pang J: MicroRNA-regulated transcriptome analysis identifies four major subtypes with prognostic and therapeutic implications in prostate cancer. Comput Struct Biotechnol J. 19:4941–4953. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Korbecki J, Kojder K, Simińska D, Bohatyrewicz R, Gutowska I, Chlubek D and Baranowska-Bosiacka I: CC chemokines in a tumor: A review of pro-cancer and anti-cancer properties of the ligands of receptors CCR1, CCR2, CCR3, and CCR4. Int J Mol Sci. 21:84122020. View Article : Google Scholar | |
|
Steinberg SM, Zhang P, Malik BT, Boni A, Shabaneh TB, Byrne KT, Mullins DW, Brinckerhoff CE, Ernstoff MS, Bosenberg MW and Turk MJ: BRAF inhibition alleviates immune suppression in murine autochthonous melanoma. Cancer Immunol Res. 2:1044–1050. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ho PC, Meeth KM, Tsui YC, Srivastava B, Bosenberg MW and Kaech SM: Immune-based antitumor effects of BRAF inhibitors rely on signaling by CD40L and IFNγ. Cancer Res. 74:3205–3217. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Cooper ZA, Reuben A, Spencer CN, Prieto PA, Austin-Breneman JL, Jiang H, Haymaker C, Gopalakrishnan V, Tetzlaff MT, Frederick DT, et al: Distinct clinical patterns and immune infiltrates are observed at time of progression on targeted therapy versus immune checkpoint blockade for melanoma. Oncoimmunology. 5:e11360442016. View Article : Google Scholar : PubMed/NCBI | |
|
Hugo W, Shi H, Sun L, Piva M, Song C, Kong X, Moriceau G, Hong A, Dahlman KB, Johnson DB, et al: Non-genomic and immune evolution of melanoma acquiring MAPKi resistance. Cell. 162:1271–1285. 2015. View Article : Google Scholar | |
|
Steinberg SM, Shabaneh TB, Zhang P, Martyanov V, Li Z, Malik BT, Wood TA, Boni A, Molodtsov A, Angeles CV, et al: Myeloid cells that impair immunotherapy are restored in melanomas with acquired resistance to BRAF inhibitors. Cancer Res. 77:1599–1610. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pavon LF, Sibov TT, de Souza AV, da Cruz EF, Malheiros SMF, Cabral FR, de Souza JG, Boufleur P, de Oliveira DM, de Toledo SRC, et al: Tropism of mesenchymal stem cell toward CD133(+) stem cell of glioblastoma in vitro and promote tumor proliferation in vivo. Stem Cell Res Ther. 9:3102018. View Article : Google Scholar : PubMed/NCBI | |
|
Tu MM, Abdel-Hafiz HA, Jones RT, Jean A, Hoff KJ, Duex JE, Chauca-Diaz A, Costello JC, Dancik GM, Tamburini BAJ, et al: Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy. Commun Biol. 3:7202020. View Article : Google Scholar | |
|
Vesely MD and Chen L: Normalization cancer immunotherapy for melanoma. J Invest Dermatol. 140:1134–1142. 2020. View Article : Google Scholar | |
|
Sanmamed MF and Chen L: A paradigm shift in cancer immunotherapy: From enhancement to normalization. Cell. 175:313–326. 2018. View Article : Google Scholar | |
|
Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA, Blackie C, Chang L, McMurtrey AE, Hebert A, et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 104:155–162. 1999. View Article : Google Scholar | |
|
Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, Cheng H, Jin K, Ni Q, Yu X and Liu C: The role of necroptosis in cancer biology and therapy. Mol Cancer. 18:1002019. View Article : Google Scholar : PubMed/NCBI | |
|
Lemke J, von Karstedt S, Zinngrebe J and Walczak H: Getting TRAIL back on track for cancer therapy. Cell Death Differ. 21:1350–1364. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Knight DA, Ngiow SF, Li M, Parmenter T, Mok S, Cass A, Haynes NM, Kinross K, Yagita H, Koya RC, et al: Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J Clin Invest. 123:1371–1381. 2013. View Article : Google Scholar | |
|
Grierson PM, Wolf C, Suresh R, Wang-Gillam A, Tan BR, Ratner L, Oppelt P, Aranha O, Frith A, Pedersen KS, et al: Neoadjuvant BMS-813160, nivolumab, gemcitabine, and nab-paclitaxel for patients with pancreatic cancer. Clin Cancer Res. 31:3644–3651. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
La Cruz TE, González-Bobes F, Eastgate MD, Sfouggatakis C, Zheng B, Kopp N, Xiao Y, Fan Y, Galindo KA, Pathirana C, et al: Scalable asymmetric synthesis of the all cis triamino cyclohexane core of BMS-813160. J Org Chem. 87:1996–2011. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Keklikoglou I, Cianciaruso C, Güç E, Squadrito ML, Spring LM, Tazzyman S, Lambein L, Poissonnier A, Ferraro GB, Baer C, et al: Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat Cell Biol. 21:190–202. 2019. View Article : Google Scholar | |
|
Lima LG, Ham S, Shin H, Chai EPZ, Lek ESH, Lobb RJ, Müller AF, Mathivanan S, Yeo B, Choi Y, et al: Tumor microenvironmental cytokines bound to cancer exosomes determine uptake by cytokine receptor-expressing cells and biodistribution. Nat Commun. 12:35432021. View Article : Google Scholar : PubMed/NCBI | |
|
Tarale P, Chaudhary S, Mukherjee S and Sarkar DK: Ethanol-activated microglial exosomes induce MCP1 signaling mediated death of stress-regulatory proopiomelanocortin neurons in the developing hypothalamus. J Neuroinflammation. 21:2792024. View Article : Google Scholar : PubMed/NCBI | |
|
Luan S, Zhao Y, Yu Y, Xu J, Xu J, Ren T, Tang X and Xie L: The relevance of B7-H3 and tumor-associated macrophages in the tumor immune microenvironment of solid tumors: Recent advances. Am J Transl Res. 17:2835–2849. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Y, Zacarías NV, de Vries H, Han GW, Gustavsson M, Dabros M, Zhao C, Cherney RJ, Carter P, Stamos D, et al: Structure of CC chemokine receptor 2 with orthosteric and allosteric antagonists. Nature. 540:458–461. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
José R, Williams A, Sulikowski M, Brealey D, Brown J and Chambers R: Regulation of neutrophilic inflammation in lung injury induced by community-acquired pneumonia. Lancet. 385 (Suppl 1):S522015. |