Itraconazole inhibits the Wnt/β-catenin signaling pathway to induce tumor-associated macrophages polarization to enhance the efficacy of immunotherapy in endometrial cancer.

Front Oncol

Department of Gynecology, Dalian Women and Children's Medical Group, Dalian, Liaoning, China.

Published: July 2025


Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Background: Endometrial cancer (EC) is a common gynecologic malignancy with limited treatment options. This study aimed to evaluate the potential of itraconazole (ITZ), a widely used antifungal drug, as an anti-tumor agent and an adjuvant to immunotherapy for EC.

Methods: The effects of ITZ on Ishikawa cells were assessed using proliferation assays, apoptosis assays, and invasion assays. The combination of ITZ and immune checkpoint inhibitors (ICIs) was evaluated to determine their synergistic effects on tumor invasion. Tumor-associated macrophages (TAMs) polarization and cytokine levels were analyzed by flow cytometry and enzyme linked immunosorbent assay (ELISA). Western blotting and Real-time reverse transcription polymerase chain reaction (RT-PCR) were used to investigate the impact of ITZ on the Wnt/β-catenin signaling pathway. Finally, experiments were conducted using a mice tumor model to validate the anti-tumor effects of ITZ and its combination with ICIs.

Results: ITZ inhibits Ishikawa cells proliferation and invasion through apoptosis induction. When combined with ICIs, ITZ significantly enhanced the inhibition of tumor invasion, an effect associated with TAMs polarization. ITZ increased IFN-γ secretion, reduced IL-10 levels, and promoted TAMs polarization from the M2 to the M1 phenotype. Mechanistically, ITZ downregulated Wnt-3a and β-catenin expression while upregulating Axin-1, thereby suppressing Wnt/β-catenin signaling in TAMs. , ITZ and ICIs synergistically reduced tumor volume and weight, shifted TAMs polarization toward the M1 phenotype, and suppressed Wnt/β-catenin signaling.

Conclusions: ITZ demonstrated robust anti-tumor activity against EC by inhibiting Ishikawa cells proliferation, invasion, and enhancing the efficacy of ICIs. Through its dual role in directly targeting tumor cells and modulating the tumor microenvironment, ITZ shows promise as a multitargeted therapeutic agent and a valuable adjuvant to immunotherapy for EC.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC12279481PMC
http://dx.doi.org/10.3389/fonc.2025.1590095DOI Listing

Publication Analysis

Top Keywords

tams polarization
16
wnt/β-catenin signaling
12
itz
12
ishikawa cells
12
signaling pathway
8
tumor-associated macrophages
8
endometrial cancer
8
adjuvant immunotherapy
8
effects itz
8
tumor invasion
8

Similar Publications

Breast cancer remains the most frequently diagnosed malignancy and a leading cause of cancer-related mortality among women worldwide. Increasing evidence underscores the pivotal yet paradoxical roles of innate immune cells and their associated cytokines in orchestrating the dynamic landscape of the breast tumor immune microenvironment (TIME). Innate immune effectors, including tumor-associated macrophages (TAMs) and natural killer (NK) cells, exert dual functions by either initiating robust antitumor responses or facilitating immune evasion, metastatic dissemination, and therapeutic resistance.

View Article and Find Full Text PDF

Tumor-associated macrophages (TAMs) display remarkable functional heterogeneity, yet the molecular mechanisms driving their diverse phenotypes remain elusive. Using CRISPR screens in primary macrophages, we identified tumor-derived factors, including lactic acid, PGE2, and GM-CSF, as key modulators of TAM polarization. These factors interact synergistically and antagonistically to shape distinct TAM phenotypes that are highly conserved across human cancers.

View Article and Find Full Text PDF

Tumor-associated macrophages (TAMs) are prominent constituents of solid tumors, and their prevalence is often associated with poor clinical outcomes. These highly adaptable immune cells undergo dynamic functional changes within the immunosuppressive tumor microenvironment (TME), engaging in reciprocal interactions with malignant cells. This bidirectional communication facilitates concurrent phenotypic transformation: tumor cells shift toward invasive mesenchymal states, whereas TAMs develop immunosuppressive, pro-tumorigenic traits.

View Article and Find Full Text PDF

Background: Human papillomavirus (HPV) is closely associated with tumor progression and the tumor microenvironment (TME), but its role in breast cancer (BC), which can be affected by HPV, has not been reported.

Methods: Ten independent BC cohorts were included to generate two HPV-related gene-based signatures. The CIBERSORT and ESTIMATE algorithms were used to quantify the immune cell fraction and TME scores, and the correlations between HPV-related gene-based signatures and scores were analyzed.

View Article and Find Full Text PDF

Metastasis remains the leading cause of cancer-related mortality, driven by complex interactions within the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play a pivotal role in metastatic progression, yet their molecular diversity and upstream regulators remain poorly defined. Glycoprotein nonmetastatic melanoma protein B (GPNMB), overexpressed in subsets of tumors including triple-negative breast cancer (TNBC), is implicated in epithelial-mesenchymal transition (EMT) and cancer stemness.

View Article and Find Full Text PDF