Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 498
1.
Mol Biol Rep ; 51(1): 720, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38824268

BACKGROUND: Tumor-associated macrophages (TAM) exert a significant influence on the progression and heterogeneity of various subtypes of breast cancer (BRCA). However, the roles of heterogeneous TAM within BRCA subtypes remain unclear. Therefore, this study sought to elucidate the role of TAM across the following three BRCA subtypes: triple-negative breast cancer, luminal, and HER2. MATERIALS AND METHODS: This investigation aimed to delineate the variations in marker genes, drug sensitivity, and cellular communication among TAM across the three BRCA subtypes. We identified specific ligand-receptor (L-R) pairs and downstream mechanisms regulated by VEGFA-VEGFR1, SPP1-CD44, and SPP1-ITGB1 L-R pairs. Experimental verification of these pairs was conducted by co-culturing macrophages with three subtypes of BRCA cells. RESULTS: Our findings reveal the heterogeneity of macrophages within the three BRCA subtypes, evidenced by variations in marker gene expression, composition, and functional characteristics. Notably, heterogeneous TAM were found to promote invasive migration and epithelial-mesenchymal transition (EMT) in MDA-MB-231, MCF-7, and SKBR3 cells, activating NF-κB pathway via P38 MAPK, TGF-ß1, and AKT, respectively, through distinct VEGFA-VEGFR1, SPP1-CD44, and SPP1-ITGB1 L-R pairs. Inhibition of these specific L-R pairs effectively reversed EMT, migration, and invasion of each cancer cells. Furthermore, we observed a correlation between ligand gene expression and TAM sensitivity to anticancer drugs, suggesting a potential strategy for optimizing personalized treatment guidance. CONCLUSION: Our study highlights the capacity of heterogeneous TAM to modulate biological functions via distinct pathways mediated by specific L-R pairs within diverse BRCA subtypes. This study might provide insights into precision immunotherapy of different subtypes of BRCA.


Breast Neoplasms , Epithelial-Mesenchymal Transition , Tumor-Associated Macrophages , Humans , Female , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Single-Cell Analysis/methods , MCF-7 Cells , Cell Movement/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Sequence Analysis, RNA/methods , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Signal Transduction/genetics , Tumor Microenvironment/genetics
2.
Immunol Lett ; 267: 106864, 2024 Jun.
Article En | MEDLINE | ID: mdl-38705481

Tumor-associated myeloid cells, including macrophages and myeloid-derived suppressor cells, can be highly prevalent in solid tumors and play a significant role in the development of the tumor. Therefore, myeloid cells are being considered potential targets for cancer immunotherapies. In this review, we focused on strategies aimed at targeting tumor-associated macrophages (TAMs). Most strategies were studied preclinically but we also included a limited number of clinical studies based on these strategies. We describe possible underlying mechanisms and discuss future challenges and prospects.


Immunotherapy , Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , Animals , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor Microenvironment/immunology , Macrophages/immunology , Macrophages/metabolism , Cellular Reprogramming/immunology , Macrophage Activation/immunology
3.
Clin Transl Sci ; 17(6): e13811, 2024 Jun.
Article En | MEDLINE | ID: mdl-38814167

Immune checkpoint inhibitors remained the standard-of-care treatment for advanced non-small cell lung cancer (NSCLC) for the past decade. In unselected patients, anti-PD-(L)1 monotherapy achieved an overall response rate of about 20%. In this analysis, we developed a pharmacokinetic and pharmacodynamic module for our previously calibrated quantitative systems pharmacology model (QSP) to simulate the effectiveness of macrophage-targeted therapies in combination with PD-L1 inhibition in advanced NSCLC. By conducting in silico clinical trials, the model confirmed that anti-CD47 treatment is not an optimal option of second- and later-line treatment for advanced NSCLC resistant to PD-(L)1 blockade. Furthermore, the model predicted that inhibition of macrophage recruitment, such as using CCR2 inhibitors, can potentially improve tumor size reduction when combined with anti-PD-(L)1 therapy, especially in patients who are likely to respond to anti-PD-(L)1 monotherapy and those with a high level of tumor-associated macrophages. Here, we demonstrate the application of the QSP platform on predicting the effectiveness of novel drug combinations involving immune checkpoint inhibitors based on preclinical or early-stage clinical trial data.


B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacokinetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Network Pharmacology/methods , Computer Simulation , Models, Biological , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
4.
J Mater Chem B ; 12(20): 4809-4823, 2024 May 22.
Article En | MEDLINE | ID: mdl-38695349

Tumor-associated macrophages (TAMs) are predominantly present in the tumor microenvironment (TME) and play a crucial role in shaping the efficacy of tumor immunotherapy. These TAMs primarily exhibit a tumor-promoting M2-like phenotype, which is associated with the suppression of immune responses and facilitation of tumor progression. Interestingly, recent research has highlighted the potential of repolarizing TAMs from an M2 to a pro-inflammatory M1 status-a shift that has shown promise in impeding tumor growth and enhancing immune responsiveness. This concept is particularly intriguing as it offers a new dimension to cancer therapy by targeting the tumor microenvironment, which is a significant departure from traditional approaches that focus solely on tumor cells. However, the clinical application of TAM-modulating agents is often challenged by issues such as insufficient tumor accumulation and off-target effects, limiting their effectiveness and safety. In this regard, nanomaterials have emerged as a novel solution. They serve a dual role: as delivery vehicles that can enhance the accumulation of therapeutic agents in the tumor site and as TAM-modulators. This dual functionality of nanomaterials is a significant advancement as it addresses the key limitations of current TAM-modulating strategies and opens up new avenues for more efficient and targeted therapies. This review provides a comprehensive overview of the latest mechanisms and strategies involving nanomaterials in modulating macrophage polarization within the TME. It delves into the intricate interactions between nanomaterials and macrophages, elucidating how these interactions can be exploited to drive macrophage polarization towards a phenotype that is more conducive to anti-tumor immunity. Additionally, the review explores the burgeoning field of TAM-associated nanomedicines in combination with tumor immunotherapy. This combination approach is particularly promising as it leverages the strengths of both nanomedicine and immunotherapy, potentially leading to synergistic effects in combating cancer.


Immunotherapy , Nanostructures , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Immunotherapy/methods , Nanostructures/chemistry , Tumor Microenvironment/drug effects , Animals , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
5.
J Clin Invest ; 134(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38690738

Targeting tumor-associated macrophages (TAMs) is an emerging approach being tested in multiple clinical trials. TAMs, depending on their differentiation state, can exhibit pro- or antitumorigenic functions. For example, the M2-like phenotype represents a protumoral state that can stimulate tumor growth, angiogenesis, metastasis, therapy resistance, and immune evasion by expressing immune checkpoint proteins. In this issue of the JCI, Vaccaro and colleagues utilized an innovative drug screen approach to demonstrate that targeting driver oncogenic signaling pathways concurrently with anti-CD47 sensitizes tumor cells, causing them to undergo macrophage-induced phagocytosis. The combination treatment altered expression of molecules on the tumor cells that typically limit phagocytosis. It also reprogrammed macrophages to an M1-like antitumor state. Moreover, the approach was generalizable to tumor cells with different oncogenic pathways, opening the door to precision oncology-based rationale combination therapies that have the potential to improve outcomes for patients with oncogene-driven lung cancers and likely other cancer types.


CD47 Antigen , Tumor-Associated Macrophages , Humans , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , CD47 Antigen/metabolism , CD47 Antigen/antagonists & inhibitors , Animals , Phagocytosis/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Macrophages/metabolism , Macrophages/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism
6.
Pathol Oncol Res ; 30: 1611586, 2024.
Article En | MEDLINE | ID: mdl-38689823

Mounting evidence suggests that the immune landscape within prostate tumors influences progression, metastasis, treatment response, and patient outcomes. In this study, we investigated the spatial density of innate immune cell populations within NOD.SCID orthotopic prostate cancer xenografts following microinjection of human DU145 prostate cancer cells. Our laboratory has previously developed nanoscale liposomes that attach to leukocytes via conjugated E-selectin (ES) and kill cancer cells via TNF-related apoptosis inducing ligand (TRAIL). Immunohistochemistry (IHC) staining was performed on tumor samples to identify and quantify leukocyte infiltration for different periods of tumor growth and E-selectin/TRAIL (EST) liposome treatments. We examined the spatial-temporal dynamics of three different immune cell types infiltrating tumors using QuPath image analysis software. IHC staining revealed that F4/80+ tumor-associated macrophages (TAMs) were the most abundant immune cells in all groups, irrespective of time or treatment. The density of TAMs decreased over the course of tumor growth and decreased in response to EST liposome treatments. Intratumoral versus marginal analysis showed a greater presence of TAMs in the marginal regions at 3 weeks of tumor growth which became more evenly distributed over time and in tumors treated with EST liposomes. TUNEL staining indicated that EST liposomes significantly increased cell apoptosis in treated tumors. Additionally, confocal microscopy identified liposome-coated TAMs in both the core and periphery of tumors, highlighting the ability of liposomes to infiltrate tumors by "piggybacking" on macrophages. The results of this study indicate that TAMs represent the majority of innate immune cells within NOD.SCID orthotopic prostate tumors, and spatial density varies widely as a function of tumor size, duration of tumor growth, and treatment of EST liposomes.


Liposomes , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms , Tumor-Associated Macrophages , Animals , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/immunology , Mice , Humans , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Xenograft Model Antitumor Assays , Apoptosis , Disease Models, Animal , TNF-Related Apoptosis-Inducing Ligand/metabolism , E-Selectin/metabolism , Tumor Microenvironment/immunology
7.
Cancer Immunol Immunother ; 73(7): 128, 2024 May 14.
Article En | MEDLINE | ID: mdl-38743074

The majority of the immune cell population in the tumor microenvironment (TME) consists of tumor-associated macrophages (TAM), which are the main players in coordinating tumor-associated inflammation. TAM has a high plasticity and is divided into two main phenotypes, pro-inflammatory M1 type and anti-inflammatory M2 type, with tumor-suppressive and tumor-promoting functions, respectively. Considering the beneficial effects of M1 macrophages for anti-tumor and the high plasticity of macrophages, the conversion of M2 TAM to M1 TAM is feasible and positive for tumor treatment. This study sought to evaluate whether the glycopeptide derived from simulated digested Codonopsis pilosula extracts could regulate the polarization of M2-like TAM toward the M1 phenotype and the potential regulatory mechanisms. The results showed that after glycopeptide dCP1 treatment, the mRNA relative expression levels of some M2 phenotype marker genes in M2-like TAM in simulated TME were reduced, and the relative expression levels of M1 phenotype marker genes and inflammatory factor genes were increased. Analysis of RNA-Seq of M2-like TAM after glycopeptide dCP1 intervention showed that the gene sets such as glycolysis, which is associated with macrophage polarization in the M1 phenotype, were significantly up-regulated, whereas those of gene sets such as IL-6-JAK-STAT3 pathway, which is associated with polarization in the M2 phenotype, were significantly down-regulated. Moreover, PCA analysis and Pearson's correlation also indicated that M2-like TAM polarized toward the M1 phenotype at the transcriptional level after treatment with the glycopeptide dCP1. Lipid metabolomics was used to further explore the efficacy of the glycopeptide dCP1 in regulating the polarization of M2-like TAM to the M1 phenotype. It was found that the lipid metabolite profiles in dCP1-treated M2-like TAM showed M1 phenotype macrophage lipid metabolism profiles compared with blank M2-like TAM. Analysis of the key differential lipid metabolites revealed that the interconversion between phosphatidylcholine (PC) and diacylglycerol (DG) metabolites may be the central reaction of the glycopeptide dCP1 in regulating the conversion of M2-like TAM to the M1 phenotype. The above results suggest that the glycopeptide dCP1 has the efficacy to regulate the polarization of M2-like TAM to M1 phenotype in simulated TME.


Codonopsis , Phenotype , Tumor Microenvironment , Tumor-Associated Macrophages , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/drug effects , Animals , Mice , Tumor Microenvironment/drug effects , Humans , Glycopeptides/metabolism , Glycopeptides/pharmacology , Macrophage Activation/drug effects , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/immunology
8.
Nat Commun ; 15(1): 4388, 2024 May 23.
Article En | MEDLINE | ID: mdl-38782901

Lung cancer is the second most frequently diagnosed cancer and the leading cause of cancer-related mortality worldwide. Tumour ecosystems feature diverse immune cell types. Myeloid cells, in particular, are prevalent and have a well-established role in promoting the disease. In our study, we profile approximately 900,000 cells from 25 treatment-naive patients with adenocarcinoma and squamous-cell carcinoma by single-cell and spatial transcriptomics. We note an inverse relationship between anti-inflammatory macrophages and NK cells/T cells, and with reduced NK cell cytotoxicity within the tumour. While we observe a similar cell type composition in both adenocarcinoma and squamous-cell carcinoma, we detect significant differences in the co-expression of various immune checkpoint inhibitors. Moreover, we reveal evidence of a transcriptional "reprogramming" of macrophages in tumours, shifting them towards cholesterol export and adopting a foetal-like transcriptional signature which promotes iron efflux. Our multi-omic resource offers a high-resolution molecular map of tumour-associated macrophages, enhancing our understanding of their role within the tumour microenvironment.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Single-Cell Analysis , Transcriptome , Tumor Microenvironment , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Single-Cell Analysis/methods , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Gene Expression Regulation, Neoplastic , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Gene Expression Profiling/methods , Macrophages/metabolism , Macrophages/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
9.
Int J Mol Sci ; 25(10)2024 May 07.
Article En | MEDLINE | ID: mdl-38791110

Vascular co-option is a consequence of the direct interaction between perivascular cells, known as pericytes (PCs), and glioblastoma multiforme (GBM) cells (GBMcs). This process is essential for inducing changes in the pericytes' anti-tumoral and immunoreactive phenotypes. Starting from the initial stages of carcinogenesis in GBM, PCs conditioned by GBMcs undergo proliferation, acquire a pro-tumoral and immunosuppressive phenotype by expressing and secreting immunosuppressive molecules, and significantly hinder the activation of T cells, thereby facilitating tumor growth. Inhibiting the pericyte (PC) conditioning mechanisms in the GBM tumor microenvironment (TME) results in immunological activation and tumor disappearance. This underscores the pivotal role of PCs as a key cell in the TME, responsible for tumor-induced immunosuppression and enabling GBM cells to evade the immune system. Other cells within the TME, such as tumor-associated macrophages (TAMs) and microglia, have also been identified as contributors to this immunomodulation. In this paper, we will review the role of these three cell types in the immunosuppressive properties of the TME. Our conclusion is that the cellular heterogeneity of immunocompetent cells within the TME may lead to the misinterpretation of cellular lineage identification due to different reactive stages and the identification of PCs as TAMs. Consequently, novel therapies could be developed to disrupt GBM-PC interactions and/or PC conditioning through vascular co-option, thereby exposing GBMcs to the immune system.


Brain Neoplasms , Pericytes , Tumor Microenvironment , Pericytes/immunology , Pericytes/pathology , Pericytes/metabolism , Humans , Tumor Microenvironment/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Glioma/immunology , Glioma/pathology , Glioma/metabolism , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/metabolism , Disease Progression , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
10.
Int J Mol Sci ; 25(10)2024 May 12.
Article En | MEDLINE | ID: mdl-38791312

Glioblastomas (GBM) are the most common primary malignant brain tumors, comprising 2% of all cancers in adults. Their location and cellular and molecular heterogeneity, along with their highly infiltrative nature, make their treatment challenging. Recently, our research group reported promising results from a prospective phase II clinical trial involving allogeneic vaccination with dendritic cells (DCs). To date, six out of the thirty-seven reported cases remain alive without tumor recurrence. In this study, we focused on the characterization of infiltrating immune cells observed at the time of surgical resection. An analytical model employing a neural network-based predictive algorithm was used to ascertain the potential prognostic implications of immunological variables on patients' overall survival. Counterintuitively, immune phenotyping of tumor-associated macrophages (TAMs) has revealed the extracellular marker PD-L1 to be a positive predictor of overall survival. In contrast, the elevated expression of CD86 within this cellular subset emerged as a negative prognostic indicator. Fundamentally, the neural network algorithm outlined here allows a prediction of the responsiveness of patients undergoing dendritic cell vaccination in terms of overall survival based on clinical parameters and the profile of infiltrated TAMs observed at the time of tumor excision.


Brain Neoplasms , Dendritic Cells , Glioblastoma , Immunotherapy , Humans , Dendritic Cells/immunology , Glioblastoma/therapy , Glioblastoma/immunology , Glioblastoma/mortality , Glioblastoma/pathology , Immunotherapy/methods , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Male , Female , Middle Aged , B7-H1 Antigen/metabolism , Prognosis , Adult , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Aged , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism
11.
Anticancer Res ; 44(6): 2437-2444, 2024 Jun.
Article En | MEDLINE | ID: mdl-38821624

BACKGROUND/AIM: Non-invasive physical plasma (NIPP) has shown promise in the treatment of cancer. However, conflicting results have been reported regarding the effect of NIPP on macrophage polarization. As tumor-associated macrophages (TAMs) are essential in the regulation of cancer development, this study aimed to determine the role of NIPP treatment in macrophage polarization and tumor-microenvironment (TME) remodeling. MATERIALS AND METHODS: A portable NIPP device, Plasma Care (Terraplasma Medical, Garching, Germany), was employed as the source of NIPP. The human monocytic cell line THP-1 was adopted as the cell model for macrophage differentiation and polarization. The effects of NIPP treatment on temperature, pH value, and oxidative stress induction of the culture medium were examined to validate the feasibility of applying the NIPP device in subsequent cell treatment. The changes in morphology, viability, and proliferation of THP-1 cells after NIPP treatment were determined. The expression of M1/M2 macrophage markers was examined by real-time quantitative polymerase chain reaction. RESULTS: No significant changes were observed in temperature and pH value after NIPP treatment, while the formation of hydrogen peroxide was promoted in a time-dependent manner. Cell morphology, viability, and proliferation were not affected by up to 6 minutes of NIPP treatment. In monocytes, 6 minutes of NIPP treatment significantly increased the expression of M1 markers (TNF-α and IL-6) and suppressed the M2 marker (CD206), findings which were consistent in the monocyte-derived macrophages. Furthermore, NIPP treatment also significantly promoted M1 polarization in the monocyte-derived macrophages induced by phorbol 12-myristate 13-acetate. CONCLUSION: NIPP is a safe and robust oxidative stress inducer and showed potential in TAM regulation by promoting M1 macrophage polarization.


Macrophages , Plasma Gases , Tumor Microenvironment , Humans , Plasma Gases/pharmacology , Macrophages/metabolism , Macrophages/immunology , THP-1 Cells , Oxidative Stress , Cell Differentiation , Cell Proliferation , Macrophage Activation , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology
12.
BMC Med Genomics ; 17(1): 145, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802881

BACKGROUND: Emerging investigations have increasingly highlighted the critical role of tumor-associated macrophages (TAMs) and M2 macrophages in cancer development, progression, and metastasis, marking them as potential targets in various cancer types. The main objective of this research is to discover new biomarkers associated with TAM-M2 macrophages in colorectal cancer (CRC) and to dissect the molecular heterogeneity of CRC by combining single-cell RNA sequencing and bulk RNA-seq data. METHODS: By utilizing weighted gene co-expression network analysis (WGCNA), we acquired TAM-M2-associated genes by intersecting TAM marker genes obtained from scRNA-seq data with module genes of M2 macrophages derived from bulk RNA-seq data. We employed least absolute shrinkage and selection operator (LASSO) Cox analysis to select predictive biomarkers from these TAM-M2-related genes. Quantitative polymerase chain reaction (qPCR) was employed to validate the mRNA expression levels of the genes identified in the screening. This led to the development of the TAM-M2-related signature (TAMM2RS). We also conducted functional and immune landscape analyses of different risk groups. RESULTS: The combination of scRNA-seq and bulk RNA-seq analyses yielded 377 TAM-M2-related genes. DAPK1, NAGK, and TRAF1 emerged as key prognostic genes in CRC, which were identified through LASSO Cox analysis. Utilizing these genes, we constructed and validated the TAMM2RS, demonstrating its effectiveness in predicting survival in CRC patients. CONCLUSION: Our research offers a thorough investigation into the molecular mechanisms associated with TAM-M2 macrophages in CRC and unveils potential therapeutic targets, offering new insights for treatment strategies in colorectal cancer.


Colorectal Neoplasms , Tumor-Associated Macrophages , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Biomarkers, Tumor/genetics , Single-Cell Analysis , Male , Female , Gene Expression Regulation, Neoplastic , Prognosis , Middle Aged , Macrophages/metabolism , Gene Expression Profiling
13.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2376-2384, 2024 May.
Article Zh | MEDLINE | ID: mdl-38812138

The abnormal activation of the mammalian target of rapamycin(mTOR) signaling pathway in non-small cell lung cancer(NSCLC) is closely associated with distant metastasis, drug resistance, tumor immune escape, and low overall survival. The present study reported that betulinic acid(BA), a potent inhibitor of mTOR signaling pathway, exhibited an inhibitory activity against NSCLC in vitro and in vivo. CCK-8 and colony formation results demonstrated that BA significantly inhibited the viability and clonogenic ability of H1299, A549, and LLC cells. Additionally, the treatment with BA induced mitochondrion-mediated apoptosis of H1299 and LLC cells. Furthermore, BA inhibited the mobility and invasion of H1299 and LLC cells by down-regulating the expression level of matrix metalloproteinase 2(MMP2) and impairing epithelial-mesenchymal transition. The results demonstrated that the inhibition of mTOR signaling pathway by BA decreased the proportion of M2 phenotype(CD206 positive) cells in total macrophages. Furthermore, a mouse model of subcutaneous tumor was established with LLC cells to evaluate the anti-tumor efficiency of BA in vivo. The results revealed that the administration of BA dramatically retarded the tumor growth and inhibited the proliferation of tumor cells. More importantly, BA increased the ratio of M1/M2 macrophages in the tumor tissue, which implied the enhancement of anti-tumor immunity. In conclusion, BA demonstrated the inhibitory effect on NSCLC by repolarizing tumor-associated macrophages via the mTOR signaling pathway.


Betulinic Acid , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Pentacyclic Triterpenes , Signal Transduction , TOR Serine-Threonine Kinases , Tumor-Associated Macrophages , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Animals , Mice , Signal Transduction/drug effects , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Pentacyclic Triterpenes/pharmacology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Cell Line, Tumor , Triterpenes/pharmacology , Cell Proliferation/drug effects , Apoptosis/drug effects , Epithelial-Mesenchymal Transition/drug effects
14.
BMC Cancer ; 24(1): 664, 2024 May 31.
Article En | MEDLINE | ID: mdl-38822331

Recent studies have shown that blue light-emitting diode (LED) light has anti-tumor effects, suggesting the possibility of using visible light in cancer therapy. However, the effects of blue light irradiation on cells in the tumor microenvironment, including tumor-associated macrophages (TAMs), are unknown. Here, THP-1 cells were cultured in the conditioned medium (CM) of HCT-116 cells to prepare TAMs. TAMs were divided into LED-irradiated and control groups. Then, the effects of blue LED irradiation on TAM activation were examined. Expression levels of M2 macrophage markers CD163 and CD206 expression were significantly decreased in LED-irradiated TAMs compared with the control group. While control TAM-CM could induce HCT-116 cell migration, these effects were not observed in cells cultured in TAM-CM with LED irradiation. Vascular endothelial growth factor (VEGF) secretion was significantly suppressed in LED-exposed TAMs. PD-L1 expression was upregulated in HCT-116 cells cultured with TAM-CM but attenuated in cells cultured with LED-irradiated TAM-CM. In an in vivo model, protein expression levels of F4/80 and CD163, which are TAM markers, were reduced in the LED-exposed group. These results indicate that blue LED light may have an inhibitory effect on TAMs, as well as anti-tumor effects on colon cancer cells.


Colonic Neoplasms , Light , Tumor-Associated Macrophages , Humans , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Tumor-Associated Macrophages/immunology , Light/adverse effects , Animals , HCT116 Cells , Mice , Tumor Microenvironment/radiation effects , Cell Movement/radiation effects , Culture Media, Conditioned/pharmacology , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Vascular Endothelial Growth Factor A/metabolism , Receptors, Cell Surface/metabolism , Macrophages/metabolism , Macrophages/radiation effects , Macrophages/immunology , Phototherapy/methods , Macrophage Activation/radiation effects , Blue Light
15.
Mol Cancer ; 23(1): 92, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715072

Breast cancer, the most frequent female malignancy, is often curable when detected at an early stage. The treatment of metastatic breast cancer is more challenging and may be unresponsive to conventional therapy. Immunotherapy is crucial for treating metastatic breast cancer, but its resistance is a major limitation. The tumor microenvironment (TME) is vital in modulating the immunotherapy response. Various tumor microenvironmental components, such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), are involved in TME modulation to cause immunotherapy resistance. This review highlights the role of stromal cells in modulating the breast tumor microenvironment, including the involvement of CAF-TAM interaction, alteration of tumor metabolism leading to immunotherapy failure, and other latest strategies, including high throughput genomic screening, single-cell and spatial omics techniques for identifying tumor immune genes regulating immunotherapy response. This review emphasizes the therapeutic approach to overcome breast cancer immune resistance through CAF reprogramming, modulation of TAM polarization, tumor metabolism, and genomic alterations.


Breast Neoplasms , Drug Resistance, Neoplasm , Immunotherapy , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Breast Neoplasms/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Female , Immunotherapy/methods , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Animals , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects
16.
Cancer Rep (Hoboken) ; 7(5): e2066, 2024 May.
Article En | MEDLINE | ID: mdl-38703051

BACKGROUND: The tumor microenvironment of solid tumors governs the differentiation of otherwise non-immunosuppressive macrophages and gamma delta (γδ) T cells into strong immunosuppressors while promoting suppressive abilities of known immunosuppressors such as myeloid-derived suppressor cells (MDSCs) upon infiltration into the tumor beds. RECENT FINDINGS: In epithelial malignancies, tumor-associated macrophages (TAMs), precursor monocytic MDSCs (M-MDSCs), and gamma delta (γδ) T cells often acquire strong immunosuppressive abilities that dampen spontaneous immune responses by tumor-infiltrating T cells and B lymphocytes against cancer. Both M-MDSCs and γδ T cells have been associated with worse prognosis for multiple epithelial cancers. CONCLUSION: Here we discuss recent discoveries on how tumor-associated macrophages and precursor M-MDSCs as well as tumor associated-γδ T cells acquire immunosuppressive abilities in the tumor beds, promote cancer metastasis, and perspectives on how possible novel interventions could restore the effective adaptive immune responses in epithelial cancers.


Lymphocytes, Tumor-Infiltrating , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid-Derived Suppressor Cells/immunology , Intraepithelial Lymphocytes/immunology , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Immune Tolerance , Animals , Tumor-Associated Macrophages/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Myeloid Cells/immunology
17.
Cancer Immunol Immunother ; 73(6): 115, 2024 May 02.
Article En | MEDLINE | ID: mdl-38693304

In the malignant progression of tumors, there is deposition and cross-linking of collagen, as well as an increase in hyaluronic acid content, which can lead to an increase in extracellular matrix stiffness. Recent research evidence have shown that the extracellular matrix plays an important role in angiogenesis, cell proliferation, migration, immunosuppression, apoptosis, metabolism, and resistance to chemotherapeutic by the alterations toward both secretion and degradation. The clinical importance of tumor-associated macrophage is increasingly recognized, and macrophage polarization plays a central role in a series of tumor immune processes through internal signal cascade, thus regulating tumor progression. Immunotherapy has gradually become a reliable potential treatment strategy for conventional chemotherapy resistance and advanced cancer patients, but the presence of immune exclusion has become a major obstacle to treatment effectiveness, and the reasons for their resistance to these approaches remain uncertain. Currently, there is a lack of exact mechanism on the regulation of extracellular matrix stiffness and tumor-associated macrophage polarization on immune exclusion. An in-depth understanding of the relationship between extracellular matrix stiffness, tumor-associated macrophage polarization, and immune exclusion will help reveal new therapeutic targets and guide the development of clinical treatment methods for advanced cancer patients. This review summarized the different pathways and potential molecular mechanisms of extracellular matrix stiffness and tumor-associated macrophage polarization involved in immune exclusion and provided available strategies to address immune exclusion.


Extracellular Matrix , Neoplasms , Tumor-Associated Macrophages , Humans , Extracellular Matrix/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/therapy , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Animals , Tumor Microenvironment/immunology , Immunotherapy/methods , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism
18.
Breast Cancer Res ; 26(1): 75, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720366

BACKGROUND: Tumor-associated macrophages (TAMs) are a prominent immune subpopulation in the tumor microenvironment that could potentially serve as therapeutic targets for breast cancer. Thus, it is important to characterize this cell population across different tumor subtypes including patterns of association with demographic and prognostic factors, and breast cancer outcomes. METHODS: We investigated CD163+ macrophages in relation to clinicopathologic variables and breast cancer outcomes in the Women's Circle of Health Study and Women's Circle of Health Follow-up Study populations of predominantly Black women with breast cancer. We evaluated 611 invasive breast tumor samples (507 from Black women, 104 from White women) with immunohistochemical staining of tissue microarray slides followed by digital image analysis. Multivariable Cox proportional hazards models were used to estimate hazard ratios for overall survival (OS) and breast cancer-specific survival (BCSS) for 546 cases with available survival data (median follow-up time 9.68 years (IQR: 7.43-12.33). RESULTS: Women with triple-negative breast cancer showed significantly improved OS in relation to increased levels of tumor-infiltrating CD163+ macrophages in age-adjusted (Q3 vs. Q1: HR = 0.36; 95% CI 0.16-0.83) and fully adjusted models (Q3 vs. Q1: HR = 0.30; 95% CI 0.12-0.73). A similar, but non-statistically significant, association was observed for BCSS. Macrophage infiltration in luminal and HER2+ tumors was not associated with OS or BCSS. In a multivariate regression model that adjusted for age, subtype, grade, and tumor size, there was no significant difference in CD163+ macrophage density between Black and White women (RR = 0.88; 95% CI 0.71-1.10). CONCLUSIONS: In contrast to previous studies, we observed that higher densities of CD163+ macrophages are independently associated with improved OS and BCSS in women with invasive triple-negative breast cancer. Trial registration Not applicable.


Antigens, CD , Antigens, Differentiation, Myelomonocytic , Receptors, Cell Surface , Triple Negative Breast Neoplasms , Tumor Microenvironment , Humans , Female , Tumor Microenvironment/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Middle Aged , Receptors, Cell Surface/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Follow-Up Studies , Prognosis , Adult , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Macrophages/metabolism , Macrophages/immunology , Macrophages/pathology , Aged , Biomarkers, Tumor/metabolism , Proportional Hazards Models
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 472-478, 2024 May.
Article Zh | MEDLINE | ID: mdl-38790105

Tumor-associated macrophages (TAM) can be differentiated into M1-type and M2-type macrophage phenotypes in the tumor microenvironment (TME), with M2-type macrophages playing a crucial role in malignant tumors. In cervical cancer, TAM exacerbates human papilloma virus (HPV) infection, promotes the proliferation, invasion, and metastasis of tumor cells, stimulates angiogenesis, and induces immune tolerance. TAM targeting strategies have emerged as a hot topic in cervical cancer immunotherapy.


Tumor Microenvironment , Tumor-Associated Macrophages , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/virology , Female , Tumor-Associated Macrophages/immunology , Tumor Microenvironment/immunology , Immunotherapy/methods , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Animals , Macrophages/immunology
20.
J Cell Mol Med ; 28(10): e18395, 2024 May.
Article En | MEDLINE | ID: mdl-38774995

Tumour-associated macrophages (TAMs), encompassing M1 and M2 subtypes, exert significant effects on osteosarcoma (OS) progression and immunosuppression. However, the impacts of TAM-derived biomarkers on the progression of OS remains limited. The GSE162454 profile was subjected to single-cell RNA (scRNA) sequencing analysis to identify crucial mediators between TAMs and OS cells. The clinical features, effects and mechanisms of these mediators on OS cells and tumour microenvironment were evaluated via biological function experiments and molecular biology experiments. Phosphodiesterase 4C (PDE4C) was identified as a pivotal mediator in the communication between M2 macrophages and OS cells. Elevated levels of PDE4C were detected in OS tissues, concomitant with M2 macrophage level, unfavourable prognosis and metastasis. The expression of PDE4C was observed to increase during the conversion process of THP-1 cells to M2 macrophages, which transferred the PDE4C mRNA to OS cells through exosome approach. PDE4C increased OS cell proliferation and mobility via upregulating the expression of collagens. Furthermore, a positive correlation was observed between elevated levels of PDE4C and increased TIDE score, decreased response rate following immune checkpoint therapy, reduced TMB and diminished PDL1 expression. Collectively, PDE4C derived from M2 macrophages has the potential to enhance the proliferation and mobility of OS cells by augmenting collagen expression. PDE4C may serve as a valuable biomarker for prognosticating patient outcomes and response rates following immunotherapy.


Bone Neoplasms , Cyclic Nucleotide Phosphodiesterases, Type 4 , Immunotherapy , Macrophages , Osteosarcoma , Tumor Microenvironment , Osteosarcoma/pathology , Osteosarcoma/immunology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/therapy , Humans , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Prognosis , Immunotherapy/methods , Tumor Microenvironment/immunology , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Macrophages/metabolism , Macrophages/immunology , Cell Line, Tumor , Cell Proliferation , Male , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Female , Neoplasm Metastasis , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Cell Movement
...