Skip to main content
Breast Cancer Research : BCR logoLink to Breast Cancer Research : BCR
. 2025 Jul 9;27:128. doi: 10.1186/s13058-025-02086-7

Erdafitinib inhibits the tumorigenicity of MDA-MB-231 triple-negative breast cancer cells by inducing TRIM25/ubiquitin-dependent degradation of FGFR4

Qing Luo 1,2,#, Li Zhang 2,#, Yue Hao 3, Chunwei Xu 4, Xiaojia Wang 5, Zhen Jia 6, Xiandong Xie 7, Zhihong Huang 8,9, Xiaomin Gao 1,2, Yu Chen 2, Xue Zhu 8,9, Jing Fang 8,9, Ke Wang 8,9,, Yongxiang Yin 1,2,
PMCID: PMC12239495  PMID: 40635078

Abstract

Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer (BC), characterized by limited treatment options and poor clinical outcomes. Aberrant FGFR signaling has been implicated in TNBC; however, the therapeutic potential of targeting FGFRs for TNBC treatment remains unclear. This study investigated the anti-cancer activity of the selective pan-FGFR inhibitor Erdafitinib and its underlying mechanisms using both in vitro and in vivo models. The results demonstrated that Erdafitinib suppressed TNBC tumorigenicity by promoting FGFR1/4 degradation, generating reactive oxygen species (ROS), inducing DNA damage, and ultimately triggering cell death. Mechanistic analyses revealed that Erdafitinib facilitated FGFR1/4 degradation through ubiquitination, enhanced interaction between TRIM25 and FGFR1/4, and subsequent lysosomal degradation. Furthermore, RNA-seq data from the TCGA and GEO databases, along with paired tumor tissues from TNBC patients, indicated that FGFR4 was significantly upregulated in TNBC. Notably, co-knockdown of FGFR1 and FGFR4 induced cytotoxicity in MDA-MB-231 cells, highlighting the therapeutic relevance of FGFR1/4 degradation by Erdafitinib in TNBC. These findings provide novel insights into the mechanisms underlying the anti-cancer efficacy of Erdafitinib, supporting its potential as a promising therapeutic agent for TNBC.

Supplementary Information

The online version contains supplementary material available at 10.1186/s13058-025-02086-7.

Keywords: TNBC, Erdafitinib, FGFR1/4, TRIM25, Ubiquitination, Biomarker

Introduction

Triple-negative breast cancer (TNBC), characterized by its aggressive behavior and poor prognosis, represents approximately 10–15% of all breast cancer (BC) cases [1]. Due to the lack of estrogen receptor alpha (ERα) expression, progesterone receptor (PR) expression, and absence of human epidermal growth factor receptor 2 (HER2) gene amplification or protein overexpression, chemotherapy has remained the primary first-line treatment option for TNBC patients over decades [24]. However, patients with residual disease after chemotherapy demonstrate significantly poorer clinical outcomes, with a six-fold increased risk of tumor recurrence and a twelve-fold higher likelihood of mortality from metastatic progression [5]. Additionally, the observed heterogeneity in treatment responses to neoadjuvant chemotherapy implies the existence of an intrinsic chemo-resistant subpopulation within TNBC [6]. Recent advancements in the molecular characterization of TNBC have paved the way for novel targeted therapies, which have shown promise in improving clinical outcomes for both early-stage and metastatic TNBC [7]. For instance, the PARP inhibitor Olaparib significantly improved overall survival (OS) in patients with HER2-negative breast cancer compared to placebo, demonstrating a notable increase in 3-year invasive disease-free survival (iDFS) (85.9% vs. 77.1%) and distant disease-free survival (DDFS) (87.5% vs. 80.4%) [8]. Similarly, the antibody-drug conjugate sacituzumab govitecan (SG) improved the objective response rate (ORR) (35% vs. 5%), progression-free survival (PFS) (5.6 months vs. 1.7 months), and OS (12.1 months vs. 6.7 months) compared to standard chemotherapy [9]. Notwithstanding these therapeutic advancements, the pronounced molecular heterogeneity, aggressive nature, and propensity for rapid development of treatment resistance in TNBC underscore the urgent need for novel therapeutic targets and treatment strategies.

Fibroblast growth factor receptors (FGFRs) constitute a conserved family of transmembrane receptors characterized by an intracellular tyrosine kinase domains. Functioning in concert with their ligands, fibroblast growth factors (FGFs), FGFRs orchestrate a diverse array of fundamental physiological processes, including embryonic development, cellular proliferation and migration, as well as tissue homeostasis [10]. Ligand-dependent activation of FGFRs through FGF binding initiates receptor dimerization and autophosphorylation, subsequently triggering the transduction of downstream signaling pathways including Ras/Raf-MEK-MAPK, signal transducer and activator of transcription (STAT), and the phosphatidylinositol 3-kinase (PI3K)/AKT pathway [11]. Aberrant activation of FGFR signaling cascades, mediated through genomic alterations including gene amplification, activating mutations, or oncogenic fusions events, has been well-documented across multiple cancer types. These molecular perturbations establish FGFRs as a promising therapeutic target for anti-cancer drug development [12]. Emerging evidence links FGFR dysfunction to an increased incidence and progression of BC [13]. Public datasets reveal FGFR2 amplifications, fusions, or mutations in TNBC and other BC subtypes, while FGFR4 overexpression and amplification are observed across all BC subtypes and are associated with poor prognosis [14]. Consequently, targeted modulation of FGFRs has emerged as a potential strategy for TNBC treatment. Several small-molecule inhibitors targeting FGFRs have been developed, including the pan-FGFR inhibitor Erdafitinib (JNJ-42756493), which is the first FGFR inhibitor approved by the US Food and Drug Administration (FDA) for the treatment of metastatic or unresectable urothelial carcinoma (mUC) [15].

Our previous studies have demonstrated that Erdafitinib exerts anti-tumor activity in human lung adenocarcinoma (LUAD) and uveal melanoma (UM) by inducing S-phase cell cycle arrest and ferroptosis in cancer cells [16, 17]. However, its potential therapeutic effects and molecular mechanisms in breast cancer, particularly in the aggressive triple-negative subtype, remain unexplored. In this study, we aimed to investigate the cytotoxic effects and underlying mechanisms of Erdafitinib in TNBC using in vitro and in vivo models, as well as clinical data from TNBC patients.

Materials and methods

Chemicals and reagents

Erdafitinib (HY-18708), the proteasome inhibitor MG132 (HY-13259), and the lysosome inhibitors chloroquine (CQ) (HY-17589 A) and Bafilomycin A1 (HY-100558) were purchased from MedChemExpress (Shanghai, China). The primary antibodies used in this study included: p-FGFR1 (ab173305), FGFR1 (ab76464), FGFR2 (ab109372), FGFR3 (ab133644), FGFR4 (ab178396), p-mTOR (ab109268), mTOR (ab32028), p-AKT (ab192623), AKT (ab18785), LAMP1 (ab24170), γ-H2AX (ab81299), and GADPH (ab8245), all obtained from Abcam (Cambridge, MA, USA). Additional antibodies, including p-FGFR2 (abs140266), p-FGFR3 (abs140268), and p-FGFR4 (abs139979), were purchased from Absin (Shanghai, China). Erk1/2 (AF1051) and p-Erk1 (Thr202/Thr204)/Erk2 (Thr185/Thr187) (AF1891) were obtained from Beyotime (Nantong, China). Antibodies for ubiquitin (10201-2-AP), TRIM25 (12573-1-AP), HA-tag (51064-2-AP), and Flag-tag (20543-1-AP) were sourced from Proteintech (Wuhan, China). Other materials were obtained from Sangon (Shanghai, China) and Beyotime (Nantong, China).

Cell lines and culture

Human TNBC cell lines MDA-MB-231 and MDA-MB-468 were obtained from the National Cell Bank of China (Shanghai, China). MCF-10 A and HEK293T cell lines were provided by the Shanghai Institute of Biochemistry and Cell Biology (SIBCB) (Shanghai, China). MCF10A cells were cultured in specialized medium, while other cells were maintained in high-glucose DMEM supplemented with 10% (v/v) fetal bovine serum (FBS). All cells were incubated at 37 °C in a humidified atmosphere containing 5% CO2.

Measurement of cell growth and apoptosis

The short-term effects of Erdafitinib on cell growth were evaluated using the MTT assay. Cells (10⁴ cells/well) were seeded into 96-well plates and treated for 24 h. After treatment, cells were incubated with 5 mg/mL MTT (ST1537, Beyotime) for 2.5–4 h, followed by dissolution in DMSO. Absorbance was measured at a wavelength of 490 nm using a fluorescence spectrophotometer (SpectraMax M5, Molecular Devices, San Jose, CA, USA). The long-term effects of Erdafitinib on cell growth were assessed using a colony formation assay. Cells were seeded into 6-well plates and subjected to drug treatment for 24 h. The medium was then replaced with fresh medium, and cells were cultured for 7 days. Colonies were fixed with 4% paraformaldehyde (P0099, Beyotime), stained with crystal violet (C0121, Beyotime), and photographed. Colony numbers were quantified using ImageJ software. Apoptosis was measured using the One Step TUNEL Apoptosis Assay Kit (C1089, Beyotime). Cells (10⁴ cells/well) were cultured in 24-well plates and treated with Erdafitinib at the indicated concentrations for 24 h. After treatment, cells were fixed with 4% paraformaldehyde and incubated with the TdT reaction mixture at 37℃. Nuclear staining was performed using DAPI (4’,6-diamidino-2-phenylindole, P0131, Beyotime). Fluorescence signals were detected using a fluorescence microscope (Olympus IX53, Olympus Corporation, Tokyo, Japan).

Detection of intracellular ROS

Intracellular ROS production was detected using the DCFH-DA kit (S0035, Beyotime). DCFH-DA is a cell-permeable, non-fluorescent dye that reacts with reactive oxygen species (ROS) to produce carboxy dichlorofluorescein, which emits fluorescence. After the indicated treatments, cells were incubated with DCFH-DA (10 µM) for 20 min in the dark at 37℃. Following two washes with D-PBS, fluorescence signals were detected using a fluorescence microscope (Olympus IX53).

Quantitative real-time PCR analysis

Total RNA from the indicated cell types was extracted using Trizol reagents (R0016, Beyotime) following the manufacturer’s instructions. RNA extraction and purification were performed as previously described [18]. First-strand cDNA was synthesized from 2 µg of total RNA using the HiScript II 1st Strand cDNA Synthesis Kit (R211-01, Vazyme, Nanjing, China). Quantification was conducted using the SYBR Premix Ex Taq™ (Q712-02, Vazyme) on an ABI 7500 Fast Real-Time system (Thermo Fisher, Waltham, CA, USA). GAPDH was used as an endogenous control for normalization. The primer sequences used were as follows: FGFR1: Forward: 5′-CCCGTAGCTCCATATTGGACA-3′, Reverse: 5′-TTTGCCATTTTTCAACCAGCG-3′; FGFR2: Forward: 5′-AGCACCATACTGGACCAACAC-3′, Reverse: 5′-GGCAGCGAAACTTGACAGTG-3′; FGFR3: Forward: 5′-TGCGTCGTGGAGAACAAGTTT-3′, Reverse: 5′-GCACGGTAACGTAGGGTGTG-3′; FGFR4: Forward: 5′-GCACTGGAGTCTCGTGATGG-3′, Reverse: 5′-CCACAGCGTTCTCTACCAGG-3′ GAPDH: Forward: 5′-GGAGCGAGATCCCTCCAAAAT-3′, Reverse: 5′-GGCTGTTGTCATACTTCTCATGG-3′.

Western blot analysis

Total protein was extracted using radio immunoprecipitation assay (RIPA) lysis buffer (P0013C, Beyotime). Protein concentrations were measured with a bicinchoninic acid (BCA) protein assay kit (P0009, Beyotime). Proteins were separated using 10–15% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride (PVDF) membranes (FFP22, Beyotime). Membranes were blocked and incubated with primary antibodies overnight at 4℃, followed by secondary antibody incubation at 37℃ for 2 h. Protein bands were visualized using the BeyoECL Star assay kit (P0018AM, Beyotime). GAPDH was used as an endogenous control for normalization of band intensities.

Immunofluorescence and confocal microscopy

Cells (10⁴ cells/well) were seeded into 24-well plates or confocal dishes and allowed to attach for 24 h. After the indicated treatments, cells were fixed with 4% paraformaldehyde at room temperature for 15 min with gentle shaking. Subsequently, cells were permeabilized using immunofluorescence staining buffer at room temperature for 10 min. Blocking was performed with immunofluorescence blocking buffer for 30 min at room temperature. Cells were then incubated with the primary antibody overnight at 4℃. The corresponding fluorescent secondary antibody was applied at room temperature for 1 h. Nuclei were stained with DAPI, and fluorescence signals were detected using a fluorescence microscope (Olympus IX53). Confocal images were captured using a Nikon laser scanning confocal microscope (Nikon, Tokyo, Japan).

Cell transfection

Lentiviruses containing shRNAs targeting human TRIM25 were obtained from GeneChem. Cells were seeded 12 h prior to infection, and to establish stable cell lines, cells were selected using puromycin (2 µg/mL) for at least two weeks. Cells were transfected with pcDNA3.1-Flag-FGFR1, pcDNA3.1-Flag-FGFR4, or HA-Ub plasmid (GenePharma, Shanghai, China) using Lipofectamine® 3000 (L3000015, ThermoFisher) according to the manufacturer’s protocol. For gene silencing experiments, small interfering RNA (siRNA) sequences targeting the desired genes were directly synthesized (GenePharma). The siRNA sequences were as follows:

FGFR1 siRNA constructs: siRNA-FGFR1#1: 5′-GCAGUGACACCACCUACUUTT-3′ (sense) and 5′-AAGUAGGUGGUGUCACUGCTT-3′ (antisense); siRNA-FGFR1#2: 5′-GGAGGUGCUUCACUUAAGATT-3′ (sense) and 5′-UCUUAAGUGAAGCACCUCCTT-3′ (antisense); siRNA-FGFR1#3: 5′-GCAGGAUGGUCCCUUGUAUTT-3′ (sense) and 5′-AUACAAGGGACCAUCCUGCTT-3′ (antisense); siRNA-FGFR1#4: 5′-GCACCAACGAGCUGUACAUTT-3′ (sense) and 5′-AUGUACAGCUCGUUGGUGCTT-3′ (antisense).

FGFR4 siRNA constructs: siRNA-FGFR4#1: 5′-GCAGAAUCUCACCUUGAUUTT-3′ (sense) and 5′-AAUCAAGGUGAGAUUCUGCTT-3′ (antisense); siRNA-FGFR4#2: 5′-GGCUGAAGCACAUCGUCAUTT-3′ (sense) and 5′-AUGACGAUGUGCUUCAGCCTT-3′ (antisense); siRNA-FGFR4#3: 5′-CCAGGUAUACGGACAUCAUTT-3′ (sense) and 5′-AUGAUGUCCGUAUACCUGGTT-3′ (antisense); siRNA-FGFR4#4: 5′-CCUCCAGCGAUUCUGUCUUTT-3′ (sense) and 5′-AAGACAGAAUCGCUGGAGGTT-3′ (antisense).

TRIM25 siRNA constructs (Sanggon Biotech): siRNA-TRIM25#1: 5′-GGUGGAGCAGCUACAACA-3′ (sense) and 5′-UUGUUGUAGCUGCUCCAC-3′ (antisense); siRNA-TRIM25#2: 5′-CCUCGACAAGGAAGAUAA-3′ (sense) and 5′-UUUAUCUUCCUUGUCGAG-3′ (antisense); siRNA-TRIM25#3: 5′-GCAAGUUUGACACCAUUU-3′ (sense) and 5′-UAAAUGGUGUACAACUUG-3′ (antisense).

Cells were transfected with siRNA using Lipofectamine® 3000 (L3000015, ThermoFisher). The effectiveness of gene silencing or overexpression was evaluated 48 h post-transfection by assessing target protein expression through immunoblotting assays.

Docking and molecular dynamics simulations

AutoDockTools 1.5.6 was used for receptor protein and small molecule ligand preparation, including the addition of polar hydrogens, charge calculations, and setting up rotatable bonds. Docking of receptor proteins with small molecule ligands was performed using AutoDock Vina, and the results were visualized with PyMOL software. For protein-protein docking, FGFR1 (PDB 1AGW) and FGFR4 (PDB 4TYE) were docked with TRIM25 (PDB 5FER) using the ZDOCK server (http://zdock.umassmed.edu) [19]. The docking results were visualized using PyMOL software.

Immunoprecipitation and co-immunoprecipitation assays

Cell lysates were prepared using a lysis buffer (P0013B, Beyotime) containing 1× complete protease inhibitor cocktail (P1005, Beyotime). For immunoprecipitation (IP) experiments, 1 mg of lysate was incubated with protein A + G agarose beads (P2055, Beyotime) and the appropriate antibodies, including anti-FGFR4, anti-FGFR1, and anti-Flag antibodies, at 4℃ for 24 h. Beads were washed three times with TBS (ST661, Beyotime), and bound proteins were eluted with 2× SDS-PAGE sample buffer. The samples were analyzed by western blot.

LC-MS/MS analysis

To identify E3 ubiquitin ligase candidates, MDA-MB-231 cells were treated with Erdafitinib for 24 h and lysed using RIPA buffer (P0013, Beyotime). Immunoprecipitation was performed using anti-FGFR1 or anti-FGFR4 antibodies, and the precipitates were bound to agarose beads. The bound beads were sent to Shanghai Applied Protein Technology (Shanghai, China) for LC-MS/MS analysis.

Nude mice tumorigenesis assay

BALB/c female nude mice (∼ 5 weeks old) were purchased from Changzhou Cavens (Changzhou, China). The animals were housed in a pathogen-free environment and provided food and water ad libitum. MDA-MB-231 cells (8 × 10⁶) were mixed with Matrigel (2:1) (M8370, Solarbio, Beijing, China) and injected subcutaneously into the mice. When tumor volumes reached approximately 100 mm³, the mice were randomly divided into three groups (n = 4 per group): vehicle control, Erdafitinib (20 mg/kg), and shTRIM25 + Erdafitinib group. Body weight and tumor volumes were measured every other day. After the experiment, tumors were excised, weighed, and photographed. The study was approved by the Laboratory Animal Ethics Committee of Jiangsu Institute of Nuclear Medicine (JSINM-2022-007).

Immunohistochemistry

Tumor tissues were embedded in paraffin and sectioned into 4 μm-thick slices. Hematoxylin and eosin Hematoxylin and eosin (H&E) staining, Ki67 immunohistochemical staining, and TUNEL assay were performed to assess tissue morphology, proliferation, and apoptosis, respectively. The expression of FGFR1/4 was evaluated via immunohistochemical staining using a DAB kit for visualization. Images were recorded with a light microscope (Olympus IX53).

Bioinformatic analysis

RNA-seq data for BC were downloaded as log2(TPM + 1), where TPM represents transcripts per million mapped reads, from the TCGA and GEO databases. Statistical analysis was conducted using R software, and data visualization was performed using the “ggplot2” package (version 3.4.2). The Kaplan–Meier Plotter (https://kmplot.com/analysis) was used to analyze distant metastasis-free survival (DMFS) for FGFR1/4 in TNBC. Tumor immune microenvironment analysis, including tumor purity estimates for TNBC, was conducted using the “estimate” package (version 1.0.13) in R software.

Statistical analysis

All experiments were conducted in triplicate. Data were presented as means ± standard deviation (SD). Differences between groups were analyzed using Student’s t-test or ANOVA, and a P-value < 0.05 was considered statistically significant.

Results

Erdafitinib induces cytotoxicity, cell apoptosis and DNA damage in TNBC cells

The chemical structure of Erdafitinib is shown in Fig. 1A. Its cytotoxic effects on TNBC cell viability were initially evaluated using the MTT assay. As shown in Fig. 1B, Erdafitinib significantly inhibited the growth of TNBC cells in a dose-dependent manner, with half-maximal inhibitory concentrations (IC5₀) of 14.8 µM for MDA-MB-231 cells, 16.82 µM for MDA-MB-468 cells, and 16.11 µM for MDA-MB-453 cells. Importantly, Erdafitinib exhibited minimal cytotoxicity in normal breast epithelial cells, with an IC5₀ of 114.2 µM for MCF-10 A cells. Additionally, the colony formation assay revealed a substantial reduction in TNBC cell growth following Erdafitinib treatment, as evidenced by a marked decrease in the number of colonies (Fig. 1C and Figure S1). MDA-MB-231 cells were subsequently selected for further experiments. The TUNEL assay demonstrated that Erdafitinib induced cytotoxicity by triggering apoptosis in MDA-MB-231 cells in a dose-dependent manner (Fig. 1D). ROS play a critical role in regulating apoptosis and modulating cancer cell survival. As shown in Fig. 1E, Erdafitinib treatment resulted in a dose-dependent increase in intracellular ROS levels. Pre-treatment with the antioxidant N-acetylcysteine (NAC, 10 µM) significantly reduced ROS generation induced by Erdafitinib. ROS, as short-lived and highly reactive oxygen-containing molecules, are known to induce DNA damage and affect the DNA damage response (DDR). To determine whether Erdafitinib promoted DNA damage in MDA-MB-231 cells, γ-H2AX expression was assessed. Western blot analysis revealed a significant increase in γ-H2AX expression in Erdafitinib-treated MDA-MB-231 cells (Fig. 1F). This finding was further supported by immunofluorescence staining, which showed elevated γ-H2AX levels in Erdafitinib-treated cells; however, pre-treatment with NAC (10 µM) significantly attenuated γ-H2AX expression. This suggests that Erdafitinib induces DNA damage in a ROS-dependent manner (Fig. 1G).

Fig. 1.

Fig. 1

Erdafitinib inhibited cell growth, induced ROS production, and led to DNA damage in TNBC cells. (A) Chemical structure of Erdafitinib; (B) MTT assay showing cell viability of MCF-10 A, MDA-MB-231, and MDA-MB-468 cells treated with Erdafitinib for 24 h at the indicated concentrations; (C) Colony formation assay demonstrating the effects of Erdafitinib (0, 7.5, and 15 µM) on the growth of MDA-MB-231 and MDA-MB-468 cells over 7 days; (D) TUNEL staining analysis showing apoptosis in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 24 h; (E) DCFH-DA fluorescent probe analysis of intracellular ROS levels in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 6 h, with or without pre-treatment with NAC (10 µM) for 4 h; (F) Western blot analysis of γ-H2AX expression in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 24 h; (G) Immunofluorescence analysis of γ-H2AX in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 24 h, with or without pre-treatment with NAC (10 µM) for 4 h. *p < 0.05, **p < 0.01, ***p < 0.001 vs. control; ###p < 0.01 vs. Erdafitinib (15 µM)

Erdafitinib induces cytotoxicity by dephosphorylation and downregulation of FGFR1/4 in MDA-MB-231 cells

Erdafitinib is a selective and potent pan-FGFR1-4 tyrosine kinase inhibitor. Based on RNA-Seq data, the mRNA expression levels of FGFR1-4 in MDA-MB-231 cells were unaffected by Erdafitinib treatment (Fig. 2A). Consistent with the RNA-Seq results, Erdafitinib did not alter the mRNA expression of FGFR1-4 in MDA-MB-231 cells (Fig. 2B). Next, the effects of Erdafitinib on the protein expression and phosphorylation status of FGFR1-4 in MDA-MB-231 cells were examined. As shown in Fig. 2C, Erdafitinib treatment for 24 h significantly reduced the protein levels of FGFR1 and FGFR4, along with their phosphorylation, in a dose-dependent manner. Additionally, semi-flexible docking analysis revealed that Erdafitinib had strong binding affinity to FGFR1 and FGFR4, with binding energies of -8.3 and − 8.4 kcal/mol, respectively (Fig. 2D), while its binding affinities for FGFR2 and FGFR3 were − 8.2 and − 7.5 kcal/mol (Figure S2). This suggests that Erdafitinib induces degradation of FGFR1/4 by binding to these proteins. Finally, the effects of Erdafitinib on the downstream signaling pathways of FGFRs in MDA-MB-231 cells were investigated. The results showed that Erdafitinib treatment led to significant suppression of key signaling molecules, including phosphorylated p-AKT, p-STAT3, and p-ERK (Fig. 2E). These data suggest that Erdafitinib exerts its cytotoxic effects in TNBC cells through coordinated inhibition of multiple oncogenic pathways, particularly the AKT, STAT3, and ERK signaling cascades.

Fig. 2.

Fig. 2

Erdafitinib dephosphorylated and downregulated FGFR1/4 and inhibited their downstream signaling in MDA-MB-231 cells. (A) RNA-Seq analysis of FGFR1-4 expression in MDA-MB-231 cells treated with Erdafitinib (0, 15 µM) for 24 h; (B) qRT-PCR analysis of FGFR1-4 expression in MDA-MB-231 cells treated with Erdafitinib (0, 15 µM) for 24 h; (C) Western blot analysis of FGFR1-4 and p-FGFR1-4 expression in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 12 h; (D) Docking model of Erdafitinib binding to human FGFR1/4 proteins. Silver: Receptor FGFR1/FGFR4; Green: Ligand Erdafitinib; Blue: Amino acids on the receptor interacting with the ligand; Yellow: Hydrophobic interactions of molecular bonds; Red: Intermolecular hydrogen bonds; (E) Western blot analysis of FGFRs downstream protein expression in MDA-MB-231 cells treated with Erdafitinib (0, 7.5, and 15 µM) for 12 h. *p < 0.05, **p < 0.01, ***p < 0.001 vs. control

Erdafitinib degrades FGFR1/4 through the ubiquitin/lysosome system in MDA-MB-231 cells

To investigate the mechanism by which Erdafitinib degrades FGFR1/4 proteins, an in-depth analysis of the signaling pathways associated with the ubiquitin system was conducted. As shown in Fig. 3A & B, Erdafitinib-treated cells exhibited a strong ubiquitin signal in the FGFR1/4 immunoprecipitants, along with enhanced endocytosis of FGFR1/4. These findings suggest that Erdafitinib promotes the degradation of FGFR1/4 via ubiquitination and endocytosis of these proteins. Proteasomal and lysosomal degradation systems are the two main intracellular protein degradation pathways. As shown in Fig. 3C & D, treatment with 3-MA and CQ (lysosome inhibitors), but not MG132 (proteasome inhibitor), significantly attenuated the protein expression of FGFR1/4 upon Erdafitinib treatment. This suggests that Erdafitinib likely degrades FGFR1/4 via the lysosomal system. To further confirm this, the effects of Erdafitinib on lysosome-related biomarkers were examined. The results showed that Erdafitinib upregulated LC3-II and LAMP1, downregulated p62, and enhanced the co-localization of FGFR1/4 with LAMP1 in MDA-MB-231 cells (Fig. 3E & F). These findings collectively indicate that Erdafitinib induces the degradation of FGFR1/4 through the ubiquitin/lysosome system in MDA-MB-231 cells.

Fig. 3.

Fig. 3

The ubiquitination and intracellular degradation of FGFR1/4 were induced by Erdafitinib in MDA-MB-231 cells. (A) Western blot analysis of FGFR1/4 ubiquitination in MDA-MB-231 cells treated with Erdafitinib (15 µM) for 24 h; (B) Immunofluorescence analysis of FGFR1/4 endocytosis in MDA-MB-231 cells treated with Erdafitinib (15 µM) for 24 h. Red: FGFR1/4, Blue: DAPI; (C) Western blot analysis of FGFR1/4 protein expression in MDA-MB-231 cells co-treated with Erdafitinib (15 µM) and MG132 (10 µM) for 24 h; (D) Western blot analysis of FGFR1/4 expression in MDA-MB-231 cells co-treated with Erdafitinib (15 µM) and CQ (10 µM) or BafA1 (10 nM) for 24 h; (E) Western blot analysis of LC3-II, p62, and LAMP1 in MDA-MB-231 cells treated with Erdafitinib (15 µM) for 24 h; (F) Immunofluorescence analysis of co-localization of FGFR1/4 with LAMP1 in MDA-MB-231 cells treated with Erdafitinib (15 µM) for 24 h. Green: FGFR1/4, Blue: DAPI, Red: LAMP1. *p < 0.05, **p < 0.01, ***p < 0.001 vs. control, ##p < 0.01 vs. Erdafitinib (15 µM), & p < 0.01 vs. Erdafitinib (15 µM)

Erdafitinib ubiquitinates FGFR1/4 through the E3 ubiquitin ligase TRIM25 in MDA-MB-231 cells

To identify the potential E3 ubiquitin ligase involved in the degradation of FGFR1/4, IP-LC-MS/MS was performed. Mass spectrometry results indicated an interaction between TRIM25 and FGFR1/4 (Fig. 4A). To confirm these findings, reciprocal immunoprecipitation was conducted, and it was found that Erdafitinib induced a marked interaction between TRIM25 and FGFR1/4 (Fig. 4B). Additionally, TRIM25 knockdown using siRNA transfection led to a significant increase in FGFR1/4 protein levels in MDA-MB-231 cells (Fig. 4C). To further confirm the direct involvement of the E3 ubiquitin ligase TRIM25 in the ubiquitination of FGFR1/4, HK293T cells were co-transfected with HA-Ub and Flag-FGFR1/4 plasmids. As shown in Fig. 4D, FGFR1/4 ubiquitination was enhanced in HK293T cells upon co-transfection with FGFR1/4 and Ub. However, TRIM25 knockdown attenuated this process, suggesting that TRIM25 directly interacted with FGFR1/4 and promoted its ubiquitination. Furthermore, ZDOCK docking analysis revealed that TRIM25 exhibited a strong binding affinity for FGFR1/4, characterized by multiple hydrogen bonds (Fig. 4E). Finally, it was investigated whether TRIM25 knockdown affected the cell viability of MDA-MB-231 cells upon Erdafitinib treatment. As shown in Figs. 4F & G, Erdafitinib’s inhibitory effect was significantly rescued by TRIM25 knockdown, as demonstrated by MTT and colony formation assays in MDA-MB-231 cells.

Fig. 4.

Fig. 4

Erdafitinib ubiquitinated FGFR1/4 through the E3 ubiquitin ligase TRIM25 in MDA-MB-231 cells. (A) IP-LC-MS/MS analysis of E3 ubiquitin ligase interacting with FGFR1/4 ubiquitination in MDA-MB-231 cells treated with Erdafitinib (15 µM) for 24 h; (B) Co-IP analysis of the interaction between the E3 ubiquitin ligase TRIM25 and FGFR1/4; (C) Western blot analysis of FGFR1/4 protein levels in cells transfected with siTRIM25; (D) Effect of siTRIM25#3 transfection on FGFR1/4 ubiquitination in HK293T cells co-transfected with Flag-FGFR1/4 and HA-Ub plasmids; (E) Docking model of TRIM25 RING binding to H. sapiens FGFR1/4 protein. Silver: Receptor FGFR1/4; Blue: Ligand TRIM25; Green: Amino acids of receptor at contact interface; Yellow: Amino acids of ligand at contact interface; Red: Intermolecular hydrogen bonds; (F) Cell growth analysis of cells with TRIM25 knockdown (si-TRIM25#3) followed by Erdafitinib (15 µM, 24 h) based on MTT assay; (G) Cell growth analysis of cells with TRIM25 knockdown (si-TRIM25#3) followed by Erdafitinib (15 µM, 24 h) based on colony formation assay. *p < 0.05, **p < 0.01, ***p < 0.001

Erdafitinib inhibits tumor growth in an MDA-MB-231 xenograft mice model

To evaluate the inhibitory effects of Erdafitinib on TNBC in vivo and explore the functional role of TRIM25, we established an MDA-MB-231 xenograft mouse model. As illustrated in Fig. 5A, treatment with Erdafitinib resulted in a marked reduction in tumor volume compared to the control group. Intriguingly, this anti-tumor effect was significantly attenuated in the group receiving combined shTRIM25 and Erdafitinib treatment. Histopathological analysis of resected tumor tissues included H&E staining, as well as immunohistochemical assessment of Ki67, TUNEL, FGFR1, and FGFR4 (Fig. 5C). H&E staining of Erdafitinib-treated tumors revealed distinct morphological changes indicative of cell death, accompanied by inflammatory cell infiltration. Moreover, the Erdafitinib-treated group exhibited reduced Ki67 and FGFR1/4 expression, along with increased TUNEL-positive cells, compared to controls. Notably, the shTRIM25 + Erdafitinib combination group showed restored FGFR1 and FGFR4 protein levels, suggesting TRIM25’s critical role in mediating Erdafitinib-induced receptor degradation. To further corroborate these observations, we performed FGFR1/4 overexpression experiments in conjunction with Erdafitinib treatment. Both MTT and colony formation assays demonstrated that FGFR1/4 overexpression partially counteracted Erdafitinib’s cytotoxic effects, with FGFR4 overexpression exhibiting the most pronounced rescue (Figure S3). These findings strongly implicate FGFR4 degradation as the primary mechanism underlying Erdafitinib’s anti-TNBC activity. Importantly, Erdafitinib treatment did not significantly affect mouse body weight (Fig. 5B) or induce pathological alterations in major organs (Fig. 5D), confirming its favorable safety profile at the administered dose. In summary, our in vivo and in vitro data collectively demonstrate that Erdafitinib effectively inhibits TNBC tumor growth in the MDA-MB-231 xenograft model, with minimal toxicity. The anti-tumor effects are predominantly mediated through TRIM25-dependent degradation of FGFR1/4, with FGFR4 playing a particularly crucial role in this process.

Fig. 5.

Fig. 5

Erdafitinib inhibited tumor growth in an MDA-MB-231 xenograft mice model. (A) Tumor volumes of MDA-MB-231 xenograft mice were measured every three days, and tumor growth curves are displayed; (B) Body weight of MDA-MB-231 xenograft mice was measured every three days (n = 4); (C) H&E staining of Ki67, TUNEL, and FGFR1/4 in tumor sections from MDA-MB-231 xenograft mice. Representative images are shown; (D) H&E staining of the heart, liver, spleen, lung, and kidney from tumor-bearing mice. *p < 0.05, **p < 0.01, ***p < 0.001

FGFR1/4 are over-expressed in TNBC patients and associated with poor clinical outcomes

There are relatively few reports on the role of FGFR1/4 in the tumorigenicity of TNBC to date. To further explore the correlation between FGFR1/4 expression and TNBC phenotypes, RNA-seq data and clinicopathological parameters from the TCGA database were retrieved. As shown in Fig. 6A&B, FGFR4 was significantly upregulated in TNBC tissues, while FGFR1 was highly expressed in the paracancerous tissues. These findings were further confirmed by analysis of GEO datasets (GSE53752). Moreover, TNBC patients with high FGFR4 expression exhibited poor prognosis and higher tumor purity, while FGFR1 was not identified as a significant biomarker (Fig. 6C&D). Subsequently, 50 paired tumor tissues and corresponding adjacent tissues from TNBC patients were collected, and FGFR1/4 expression was detected using immunohistochemistry (IHC) staining. The results showed that FGFR4 was significantly upregulated in TNBC tissues, but FGFR1 was not (Fig. 6E). To further investigate the function of FGFR1/4 in TNBC cells, MDA-MB-231 cells were transfected with siFGFR1, siFGFR4, and siFGFR1 + 4, with knockdown efficiency confirmed by immunoblotting (Fig. 7A-B). As shown in Fig. 7C&D, siFGFR1 (siFGFR1#2), siFGFR4 (siFGFR4#2), and siFGFR1 + 4 significantly suppressed cell viability in MTT assays, which was further confirmed by colony formation assays. In addition, co-knockdown of FGFR1 and FGFR4 resulted in greater cytotoxicity in MDA-MB-231 cells. These findings suggest that FGFR1/4 degradation by Erdafitinib is an effective treatment approach in TNBC.

Fig. 6.

Fig. 6

The expressions of FGFR1/4 in TNBC from database and clinical tissues. (A) TCGA database analysis of FGFR1/4 expression among normal, non-TNBC, and TNBC breast tissues. Log2 (TPM + 1) was applied for the log scale; (B) Public microarray dataset (GSE53752) analysis of FGFR1/4 expression between normal and TNBC breast tissues; (C) Kaplan-Meier survival analysis of the correlation between FGFR1/4 expression and DMFS of TNBC patients based on TCGA database; (D) The correlation of FGFR1/4 expression with tumor purity score in the tumor microenvironment (TME) based on TCGA database; (E) Immunohistochemistry analysis of FGFR1/4 expression in tumor tissues and corresponding adjacent tissues from TNBC patients. *p < 0.05, **p < 0.01, ***p < 0.001

Fig. 7.

Fig. 7

Knockdown of FGFR1/4 inhibited tumor growth in MDA-MB-231 cells. (A&B) Western blot analysis of FGFR1/4 knockdown efficiency; (C) MTT analysis of cell growth in MDA-MB-231 cells with knockdown of FGFR1, FGFR4, and FGFR1 + 4; (D) Colony formation analysis of cell growth in MDA-MB-231 cells with knockdown of FGFR1, FGFR4, and FGFR1 + 4. *p < 0.05, **p < 0.01, ***p < 0.001 vs. control; #p<0.05, ##p < 0.01 vs. FGFR1/4#2 group

Discussion

Receptor tyrosine kinases (RTKs), characterized by their intrinsic cytoplasmic tyrosine kinase domains, serve as crucial regulators in cancer development and are frequently overexpressed during carcinogenesis [20]. The comprehensive understanding of RTK signaling pathways and their modulation by inhibitors is fundamental for developing targeted therapies that improve clinical outcomes [21]. In recent years, small-molecule tyrosine kinase inhibitors (TKIs) targeting the ATP-binding pocket of RTKs have emerged as promising therapeutic agents, demonstrating improved treatment efficacy in various cancers [22]. Among these, Erdafitinib represents a significant advancement as the first FDA-approved oral pan-FGFR TKI, exhibiting selective inhibition across all four FGFR isoforms (FGFR1-4) to attenuate cellular signaling and induce apoptosis [23]. For our investigation, we selected the MDA-MB-231 cell line as the primary model system due to its well-characterized properties in TNBC tumorigenesis research and established utility in studying FGFR-targeted therapeutic mechanisms [2426]. Our previous studies have reported the anti-cancer activities of Erdafitinib in LUAD and UM, and this study further explores its effect on TNBC, providing deeper mechanistic insights. The results revealed that Erdafitinib inhibited the tumorigenicity of TNBC both in vitro and in vivo by inducing FGFR1/4 degradation, ROS generation, DNA damage, and subsequent cell death. Further mechanistic studies showed that Erdafitinib degraded FGFR1/4 by inducing their ubiquitination, interacting with TRIM25, and promoting lysosomal degradation. Therefore, our findings offer novel insights into the anti-cancer mechanism of Erdafitinib in TNBC and identify TRIM25 as a specific E3 ubiquitin ligase targeting FGFR1/4.

An increasingly important concept in TKI functionality is the membrane trafficking, internalization (endocytosis), and proteasomal or lysosomal degradation of targeted RTKs, which inhibit subsequent downstream reactions in carcinogenesis [27]. In this study, it was found that Erdafitinib induced the ubiquitination, endocytosis, and lysosomal degradation of FGFR1/4 in TNBC cells. Ubiquitination is one of the most important post-translational modifications, where a protein is tagged with ubiquitin for proteasomal degradation [28]. This is one of the most versatile cellular regulatory mechanisms for controlling both physiological and pathological events [29]. Ubiquitination is achieved through the covalent attachment of ubiquitin to a target protein via an enzymatic cascade involving three classes of enzymes: E1 (activation), E2 (conjugation), and E3 (ligation) [30]. Among these, E3 ubiquitin ligases play the most prominent role [31]. In this study, IP-LC-MS/MS was performed to identify the potential E3 ubiquitin ligase involved in the degradation of FGFR1/4. Mass spectrometry suggested an interaction between TRIM25 and FGFR1/4. Reciprocal immunoprecipitation in TNBC cells and transfection experiments using HEK293T cells confirmed that Erdafitinib ubiquitinates FGFR1/4 through the E3 ubiquitin ligase TRIM25. TRIM25 is a 17-beta-estradiol and type I interferon-inducible E3 ligase encoded by the TRIM25 gene [32, 33]. It is characterized by the presence of three conserved N-terminal domains: a RING domain, one or two B-Boxes (B1/B2), and a coiled-coil (CC) domain [34]. TRIM25 participates in various cellular processes, including tumor progression [35]. Previous studies have reported its role as a transcriptional regulator of BC metastasis networks and its ability to promote breast cancer progression by enhancing estrogen receptor signaling [36, 37]. Notably, most studies report that TRIM25 primarily functions through its E3 ubiquitin ligase activity to regulate cellular processes, often leading to suppression of cancer cell death - a finding consistent with our observations [3841]. Our study is the first to show that Erdafitinib inhibits the growth of TNBC cells through a TRIM25-dependent FGFR1/4 degradation mechanism, highlighting the important role of TRIM25 in Erdafitinib’s anti-cancer activity in TNBC. However, the specific ubiquitinated lysine (K) residues of FGFR1/4 (such as K6, K11, K27, K29, K33, K48, and K63) by TRIM25 upon Erdafitinib treatment in TNBC require further investigation. Proteasomal and lysosomal degradation are the two main pathways for protein degradation via ubiquitination [42]. In addition, it was found that lysosomal inhibitors 3-MA and CQ, but not the proteasome inhibitor MG132, significantly attenuated the protein levels of FGFR1/4 upon Erdafitinib treatment. This suggests that Erdafitinib may degrade FGFR1/4 via the lysosomal pathway.

To explore the correlation between FGFR1/4 expression and TNBC phenotypes in clinical settings, RNA-seq data from the TCGA/GEO databases and paired tumor tissues from TNBC patients were analyzed. The results showed that FGFR4 was significantly upregulated in TNBC tissues, while FGFR1 was highly expressed in paracancerous tissues. Notably, accumulating evidence indicates that FGFR1 overexpression significantly contributes to TNBC pathogenesis by promoting tumorigenesis, growth and metastasis [43, 44]. Most importantly, our functional rescue experiments in MDA-MB-231 cells showed that overexpression of either FGFR1 or FGFR4 partially reversed the cytotoxic effects of erdafitinib, but overexpression of FGFR4 produced a greater rescue effect. These findings strongly indicate that Erdafitinib’s primary anti-TNBC activity is mediated through FGFR4 degradation, despite its ability to target both receptors. Additionally, siFGFR1 (siFGFR1#2), siFGFR4 (siFGFR4#2), and siFGFR1 + 4 significantly suppressed cell growth, as shown by MTT and colony formation assays. Co-knockdown of FGFR1 and FGFR4 demonstrated enhanced cytotoxicity in MDA-MB-231 cells, suggesting that FGFR1/4 degradation by Erdafitinib is an effective treatment approach in TNBC.

Conclusion

In conclusion, this study reveals the anti-cancer activity of Erdafitinib in TNBC by inducing TRIM25/ubiquitin-dependent degradation of FGFR1/4 and ROS-dependent DNA damage (Fig. 8). These findings provide novel insights into the mechanism of action of Erdafitinib’s anti-cancer efficacy, supporting its potential as a therapeutic drug for TNBC.

Fig. 8.

Fig. 8

The proposed molecular mechanism of Erdafitinib on TNBC

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1 (1.4MB, docx)
Supplementary Material 2 (28.9MB, docx)

Acknowledgements

Not applicable.

Author contributions

Conception and design: KW and XYY. Acquisition of data: LZ, CXW and XJW. Analysis and interpretation of data: YH, ZHH, XDX. Write, review, and/or revision if manuscript: QL, KW, YXY. Administrative, technical, or material support: XMG, YC, XZ and JF.

Funding

This work was supported by Major Project of Wuxi Municipal Health Commission (Z202303), Project of Jiangsu Commission of Health (H2023150), Project of Jiangsu Administration of Traditional Chinese Medicine (MS2022145, MS2023166), Projects of Wuxi Science and Technology Bureau (K20241012), and Key Projects of Wuxi Health Commission (Z202408).

Data availability

No datasets were generated or analysed during the current study.

Declarations

Ethics approval and consent to participate

The study was approved by the Medical Ethics Committee of Wuxi Maternal and Child Health Hospital (approval no. 2023-01-0421-06; approval date, April 21, 2023) and was conducted in accordance with The Declaration of Helsinki. The participants provided written informed consent prior to taking part in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Qing Luo and Li Zhang contributed equally to this work.

Contributor Information

Ke Wang, Email: wangke@jsinm.org.

Yongxiang Yin, Email: yinyrh@jiangnan.edu.cn.

References

  • 1.Xue Y, Gao S, Gou J, et al. Platinum-based chemotherapy in combination with PD-1/PD-L1 inhibitors: preclinical and clinical studies and mechanism of action. Expert Opin Drug Deliv Feb. 2021;18(2):187–203. 10.1080/17425247.2021.1825376 [DOI] [PubMed] [Google Scholar]
  • 2.Won KA, Spruck C. Triple–negative breast cancer therapy: current and future perspectives (Review). Int J Oncol Dec. 2020;57(6):1245–61. 10.3892/ijo.2020.5135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zagami P, Carey LA. Triple negative breast cancer: pitfalls and progress. NPJ Breast Cancer Aug. 2022;20(1):95. 10.1038/s41523-022-00468-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Anurag M, Jaehnig EJ, Krug K, et al. Proteogenomic markers of chemotherapy resistance and response in Triple-Negative breast Cancer. Cancer Discov Nov. 2022;2(11):2586–605. 10.1158/2159-8290.Cd-22-0200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Leon-Ferre RA, Goetz MP. Advances in systemic therapies for triple negative breast cancer. Bmj May 30. 2023;381:e071674. 10.1136/bmj-2022-071674 [DOI] [PubMed] [Google Scholar]
  • 6.Pinilla K, Drewett LM, Lucey R, Abraham JE. Precision breast Cancer medicine: early stage triple negative breast Cancer-A review of molecular characterisation, therapeutic targets and future trends. Front Oncol. 2022;12:866889. 10.3389/fonc.2022.866889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ensenyat-Mendez M, Llinàs-Arias P, Orozco JIJ, et al. Current Triple-Negative breast Cancer subtypes: dissecting the most aggressive form of breast Cancer. Front Oncol. 2021;11:681476. 10.3389/fonc.2021.681476 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhu S, Wu Y, Song B, et al. Recent advances in targeted strategies for triple-negative breast cancer. J Hematol Oncol Aug. 2023;28(1):100. 10.1186/s13045-023-01497-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nader-Marta G, Molinelli C, Debien V, et al. Antibody-drug conjugates: the evolving field of targeted chemotherapy for breast cancer treatment. Ther Adv Med Oncol. 2023;15:17588359231183679. 10.1177/17588359231183679 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Giacomini A, Grillo E, Rezzola S, et al. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev Apr. 2021;1(2):569–610. 10.1152/physrev.00005.2020 [DOI] [PubMed] [Google Scholar]
  • 11.Hallinan N, Finn S, Cuffe S, Rafee S, O’Byrne K, Gately K. Targeting the fibroblast growth factor receptor family in cancer. Cancer Treat Rev May. 2016;46:51–62. 10.1016/j.ctrv.2016.03.015 [DOI] [PubMed] [Google Scholar]
  • 12.Tiseo M, Gelsomino F, Alfieri R, et al. FGFR as potential target in the treatment of squamous Non small cell lung cancer. Cancer Treat Rev Jun. 2015;41(6):527–39. 10.1016/j.ctrv.2015.04.011 [DOI] [PubMed] [Google Scholar]
  • 13.Sankofi BM, Valencia-Rincón E, Sekhri M, Ponton-Almodovar AL, Bernard JJ, Wellberg EA. The impact of poor metabolic health on aggressive breast cancer: adipose tissue and tumor metabolism. Front Endocrinol (Lausanne). 2023;14:1217875. 10.3389/fendo.2023.1217875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chew NJ, Lim Kam Sian TCC, Nguyen EV, et al. Evaluation of FGFR targeting in breast cancer through interrogation of patient-derived models. Breast Cancer Res Aug. 2021;3(1):82. 10.1186/s13058-021-01461-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hosni S, Kilian V, Klümper N, et al. Adipocyte Precursor-Derived NRG1 promotes resistance to FGFR Inhibition in urothelial carcinoma. Cancer Res Mar. 2024;4(5):725–40. 10.1158/0008-5472.Can-23-1398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhu X, Wang L, Wang K, Yao Y, Zhou F. Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med Sep. 2024;222:552–68. 10.1016/j.freeradbiomed.2024.07.002 [DOI] [PubMed] [Google Scholar]
  • 17.Meng X, Zhu X, Ji J, et al. Erdafitinib inhibits tumorigenesis of human lung adenocarcinoma A549 by inducing S-Phase Cell-Cycle arrest as a CDK2 inhibitor. Molecules Oct. 2022;9(19). 10.3390/molecules27196733 [DOI] [PMC free article] [PubMed]
  • 18.Fiedler SD, Carletti MZ, Christenson LK. Quantitative RT-PCR methods for mature MicroRNA expression analysis. Methods Mol Biol. 2010;630:49–64. 10.1007/978-1-60761-629-0_4 [DOI] [PubMed] [Google Scholar]
  • 19.Pierce BG, Wiehe K, Hwang H, Kim BH, Vreven T, Weng Z. ZDOCK server: interactive Docking prediction of protein-protein complexes and symmetric multimers. Bioinformatics Jun. 2014;15(12):1771–3. 10.1093/bioinformatics/btu097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Saraon P, Pathmanathan S, Snider J, Lyakisheva A, Wong V, Stagljar I. Receptor tyrosine kinases and cancer: oncogenic mechanisms and therapeutic approaches. Oncogene Jun. 2021;40(24):4079–93. 10.1038/s41388-021-01841-2 [DOI] [PubMed] [Google Scholar]
  • 21.Park H, Park H, Baek J, Moon H, Ro SW. Target therapy for hepatocellular carcinoma: beyond receptor tyrosine kinase inhibitors and immune checkpoint inhibitors. Biology (Basel). Apr. 2022;12(4). 10.3390/biology11040585 [DOI] [PMC free article] [PubMed]
  • 22.Wang H, Du X, Liu W, et al. Combination of betulinic acid and EGFR-TKIs exerts synergistic anti-tumor effects against wild-type EGFR NSCLC by inducing autophagy-related cell death via EGFR signaling pathway. Respir Res May. 2024;20(1):215. 10.1186/s12931-024-02844-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Roskoski R. Jr. The role of fibroblast growth factor receptor (FGFR) protein-tyrosine kinase inhibitors in the treatment of cancers including those of the urinary bladder. Pharmacol Res Jan. 2020;151:104567. 10.1016/j.phrs.2019.104567 [DOI] [PubMed] [Google Scholar]
  • 24.Liu Z, Zhang S, Wang T, Shao H, Gao J, Wang Y, Ge Y. Neferine inhibits MDA-MB-231 cells growth and metastasis by regulating miR-374a/FGFR-2. Chem Biol Interact Aug. 2019;25:309:108716. 10.1016/j.cbi.2019.06.029 [DOI] [PubMed] [Google Scholar]
  • 25.Wang Y, Pan Y, Lv Z, Gou S. Discovery of N-(4-((6-(3,5- Dimethoxyphenyl)-9H-purine derivatives as irreversible covalent FGFR inhibitors. Eur J Med Chem May. 2024;5:271:116415. 10.1016/j.ejmech.2024.116415 [DOI] [PubMed] [Google Scholar]
  • 26.Chen Z, Tong LJ, Tang BY, Liu HY, Wang X, Zhang T, Cao XW, Chen Y, Li HL, Qian XH, Xu YF, Xie H, Ding J. C11, a novel fibroblast growth factor receptor 1 (FGFR1) inhibitor, suppresses breast cancer metastasis and angiogenesis. Acta Pharmacol Sin Jun. 2019;40(6):823–32. 10.1038/s41401-018-0191-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Liu X, Rothe K, Yen R, et al. A novel AHI-1-BCR-ABL-DNM2 complex regulates leukemic properties of primitive CML cells through enhanced cellular endocytosis and ROS-mediated autophagy. Leukemia Nov. 2017;31(11):2376–87. 10.1038/leu.2017.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Swatek KN, Komander D. Ubiquitin modifications. Cell Res Apr. 2016;26(4):399–422. 10.1038/cr.2016.39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Martínez-Férriz A, Ferrando A, Fathinajafabadi A, Farràs R. Ubiquitin-mediated mechanisms of translational control. Semin Cell Dev Biol Dec. 2022;132:146–54. 10.1016/j.semcdb.2021.12.009 [DOI] [PubMed] [Google Scholar]
  • 30.Morreale FE, Walden H. Types of ubiquitin ligases. Cell Mar 24. 2016;165(1):248–e2481. 10.1016/j.cell.2016.03.003 [DOI] [PubMed] [Google Scholar]
  • 31.Han S, Wang R, Zhang Y, et al. The role of ubiquitination and deubiquitination in tumor invasion and metastasis. Int J Biol Sci. 2022;18(6):2292–303. 10.7150/ijbs.69411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Marcos-Villar L, Nistal-Villan E, Zamarreño N, Garaigorta U, Gastaminza P, Nieto A. Interferon-β stimulation elicited by the influenza virus is regulated by the histone Methylase Dot1L through the RIG-I-TRIM25 signaling Axis. Cells Mar. 2020;16(3). 10.3390/cells9030732 [DOI] [PMC free article] [PubMed]
  • 33.Orimo A, Inoue S, Minowa O, et al. Underdeveloped uterus and reduced Estrogen responsiveness in mice with disruption of the Estrogen-responsive finger protein gene, which is a direct target of Estrogen receptor alpha. Proc Natl Acad Sci U S A Oct. 1999;12(21):12027–32. 10.1073/pnas.96.21.12027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kong HJ, Lee YJ, Shin J, et al. Molecular characterization of tripartite motif protein 25 (TRIM25) involved in ERα-mediated transcription in the Korean Rose Bitterling Rhodeus uyekii. Comp Biochem Physiol B Biochem Mol Biol Sep. 2012;163(1):147–53. 10.1016/j.cbpb.2012.05.015 [DOI] [PubMed] [Google Scholar]
  • 35.Rahimi-Tesiye M, Zaersabet M, Salehiyeh S, Jafari SZ. The role of TRIM25 in the occurrence and development of cancers and inflammatory diseases. Biochim Biophys Acta Rev Cancer Sep. 2023;1878(5):188954. 10.1016/j.bbcan.2023.188954 [DOI] [PubMed] [Google Scholar]
  • 36.Walsh LA, Alvarez MJ, Sabio EY, et al. An integrated systems biology approach identifies TRIM25 as a key determinant of breast Cancer metastasis. Cell Rep Aug. 2017;15(7):1623–40. 10.1016/j.celrep.2017.07.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Azuma K, Inoue S. Efp/TRIM25 and its related protein, TRIM47, in Hormone-Dependent cancers. Cells Aug. 2022;8(15):2464. 10.3390/cells11152464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jian Y, Kong L, Xu H, et al. Protein phosphatase 1 regulatory inhibitor subunit 14 C promotes triple-negative breast cancer progression via sustaining inactive glycogen synthase kinase 3 beta. Clin Transl Med Jan. 2022;12(1):e725. 10.1002/ctm2.725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wong D, Sogerer L, Lee SS, Wong V, Lum A, Levine AB, Marra MA, Yip S. TRIM25 promotes Capicua degradation independently of ERK in the absence of ATXN1L. BMC Biol Oct. 2020;28(1):154. 10.1186/s12915-020-00895-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yun H, Jeong HR, Kim DY, You JE, Lee JU, Kang DH, Koh DI, Ryu YS, Bae S, Jin DH. Degradation of AZGP1 suppresses apoptosis and facilitates cholangiocarcinoma tumorigenesis via TRIM25. J Cell Mol Med Feb. 2024;28(3):e18104. 10.1111/jcmm.18104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang K, He Q, Jiang X, et al. Targeting UBE2T suppresses breast cancer stemness through CBX6-mediated transcriptional repression of SOX2 and NANOG. Cancer Lett Dec. 2024;21:611:217409. 10.1016/j.canlet.2024.217409 [DOI] [PubMed] [Google Scholar]
  • 42.Dikic I. Proteasomal and autophagic degradation systems. Annu Rev Biochem Jun. 2017;20:86:193–224. 10.1146/annurev-biochem-061516-044908 [DOI] [PubMed] [Google Scholar]
  • 43.Liu Q, Huang J, Yan W, et al. FGFR families: biological functions and therapeutic interventions in tumors. MedComm (2020). Sep. 2023;23(5):e367. 10.1002/mco2.367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang Y, Shen Z, Chen R, et al. Discovery and characterization of novel FGFR1 inhibitors in triple-negative breast cancer via hybrid virtual screening and molecular dynamics simulations. Bioorg Chem Sep. 2024;150:107553. 10.1016/j.bioorg.2024.107553 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Material 1 (1.4MB, docx)
Supplementary Material 2 (28.9MB, docx)

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Breast Cancer Research : BCR are provided here courtesy of BMC

RESOURCES