Open Access

Doxazosin inhibits vasculogenic mimicry in human non‑small cell lung cancer through inhibition of the VEGF‑A/VE‑cadherin/mTOR/MMP pathway

  • Authors:
    • Jui-Ling Hsu
    • Wohn-Jenn Leu
    • Lih-Ching Hsu
    • Chia-Hsun Hsieh
    • Jih-Hwa Guh
  • View Affiliations

  • Published online on: February 22, 2024     https://doi.org/10.3892/ol.2024.14303
  • Article Number: 170
  • Copyright: © Hsu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Lung cancer is the leading cause of cancer‑related death worldwide, and ~85% of lung cancers are non‑small cell lung cancer (NSCLC), which has a low 5‑year overall survival rate and high mortality. Several therapeutic strategies have been developed, such as targeted therapy, immuno-oncotherapy and combination therapy. However, the low survival rate indicates the urgent need for new NSCLC treatments. Vasculogenic mimicry (VM) is an endothelial cell‑free tumor blood supply system of aggressive and metastatic tumor cells present during tumor neovascularization. VM is clinically responsible for tumor metastasis and resistance, and is correlated with poor prognosis in NSCLC, making it a potential therapeutic target. In the present study, A549 cells formed glycoprotein‑rich lined tubular structures, and transcript levels of VM‑related genes were markedly upregulated in VM‑forming cells. Based on a drug repurposing strategy, it was demonstrated that doxazosin (an antihypertensive drug) displayed inhibitory activity on VM formation at non‑cytotoxic concentrations. Doxazosin significantly reduced the levels of vascular endothelial growth factor A (VEGF‑A) and matrix metalloproteinase‑2 (MMP‑2) in the cell media during VM formation. Further experiments revealed that the protein expression levels of VEGF‑A and vascular endothelial‑cadherin (VE‑cadherin), which contribute to tumor aggressiveness and VM formation, were downregulated following doxazosin treatment. Moreover, the downstream signaling Ephrin type‑A receptor 2 (EphA2)/AKT/mTOR/MMP/Laminin‑5γ2 network was inhibited in response to doxazosin treatment. In conclusion, the present study demonstrated that doxazosin displayed anti‑VM activity in an NSCLC cell model through the downregulation of VEGF‑A and VE‑cadherin levels, and the suppression of signaling pathways related to the receptor tyrosine kinase, EphA2, protein kinases, AKT and mTOR, and proteases, MMP‑2 and MMP‑9. These results support the add‑on anti‑VM effect of doxazosin as a potential agent against NSCLC.

Introduction

Lung cancer is the leading cause of cancer-related death worldwide. Non-small cell lung cancer (NSCLC), including adenocarcinoma, squamous cell carcinoma and large cell carcinoma, comprises ~85% of all lung cancer cases (1,2). Advances in recent decades have led to the identification of numerous oncogenic factors in NSCLC, such as gene mutations in EGFR, BRAF, ROS proto-oncogene 1, p16INK4a and human epidermal growth factor receptor 2, and rearrangements in anaplastic lymphoma kinase and RET (36) Although target therapies and immunotherapies have enhanced the overall survival time of patients, the occurrence of metastasis and resistance (primary or acquired) may cause difficulties for effective second-line treatment options. This has resulted in an urgent unmet need for NSCLC treatment (7).

Vasculogenic mimicry (VM), a new blood supply network with abundant extracellular matrix (ECM), is the formation of microvascular channels by aggressive and metastatic tumor cells (8). VM is suggested to be responsible for tumor metastasis and poor prognosis in patients with cancer, such as lung cancer, colorectal cancer, liver cancer, sarcoma and melanoma (810). VM offers functional perfusion pathways for rapidly growing tumors by transferring fluid from leaky vessels and connecting with vasculature, representing a non-angiogenic pathway. Aggressive tumors possess VM structures, which are inaccessible to anti-angiogenic therapies, resulting in cancer resistance (11,12). Analysis of NSCLC tissue samples has indicated that the levels of VM, slug and vimentin [two key regulators in epithelial-mesenchymal transition (EMT)] are higher in NSCLC tissues compared with normal lung tissues (13,14). Furthermore, CDK5 kinase induces focal adhesion kinase (FAK)/AKT signaling and subsequent VM formation. Blockade of CDK5 by inhibitors or siRNA inhibits VM formation and tumor growth in an NSCLC A549 cell line and animal models (15).

Drug repurposing has emerged as an attractive approach in combating malignant tumors (1619). Doxazosin, an α1-adrenergic blocker for the treatment of hypertension and the symptoms of benign prostatic hyperplasia, has been reported to display anticancer activity through the inhibition of cell proliferation, migration and metastasis, and the induction of autophagy and apoptosis of cancer cells (2023). Furthermore, doxazosin is reported to inhibit vascular endothelial growth factor (VEGF), suppressing the migration and invasion of endothelial cells (24). However, the impact of doxazosin on VM formation has not yet, to the best of our knowledge, been reported. In the present study, a VM model containing hollow lumens was established using an NSCLC cell model and the inhibitory activity of doxazosin was studied. To the best of our knowledge, the present study is the first report to elucidate the anti-VM effect of doxazosin.

Materials and methods

Materials

The human NSCLC cell line, A549, was purchased from the American Type Culture Collection. RPMI 1640 medium, fetal bovine serum (FBS), penicillin and streptomycin were purchased from Gibco (Thermo Fisher Scientific, Inc.). The GAPDH (cat. no. 32233) and fibronectin (cat. no. 18825) antibodies were purchased from Santa Cruz Biotechnology, Inc. Vascular endothelial (VE)-cadherin (cat. no. 2500), Ephrin type-A receptor 2 (EpHA2; cat. no. 6997), phosphorylated (p-)EpHA2Ser897 (cat. no. 6347), 3-phosphoinositide-dependent kinase 1 (PDK1; cat. no. 3062), p-PDK1Ser241 (cat. no. 3061), AKT (cat. no. 9272), p-AKTSer473 (cat. no. 4060), mTOR (cat. no. 2972), mTORSer2448 (cat. no. 2971), P70s6k (cat. no. 9202), p-P70s6kThr389 (cat. no. 9234), ERK (cat. no. 9102), p-ERKThr202/Tyr204 (cat. no. 9101) and vimentin (cat. no. 5741) antibodies were purchased from Cell Signaling Technologies, Inc. VEGF-A (cat. no. ab1316) antibodies were purchased from Abcam. Matrix metalloproteinase (MMP)-2 (cat. no. AB19167), MMP-9 (cat. no. AB19016) and Laminin 5γ2 (cat. no. MAB19562) antibodies were purchased from Millipore. Matrigel was purchased from BD Biosciences. Anti-mouse (cat. no. 115-035-062) and anti-rabbit (cat. no. 111-035-045) IgGs were purchased from Jackson ImmunoResearch Laboratories, Inc. Doxazosin (cat. no. D9815), thiazolyl blue tetrazolium blue (MTT; cat. no. M2128) and sulforhodamine B (SRB; cat. no. S9012) were purchased from Sigma-Aldrich.

Cell culture

A549 cells were cultured in RPMI 1640 medium supplemented with 10% (v/v) FBS and 1% (v/v) penicillin-streptomycin-amphotericin B solution. Cell cultures were maintained in a 37°C incubator with 5% CO2. Adherent cell cultures were passaged using 0.05% trypsin-EDTA after reaching ~80% confluence.

MTT assay

After a 24-h treatment with the indicated concentrations of doxazosin, the cells were incubated with MTT (final concentration 0.5 mg/ml) for 2 h. Then, the medium was removed and replaced with 100 µl DMSO to dissolve the formed purple formazan. An ELISA reader (570 nm) was used to assess the absorbance values (25).

SRB assay

Firstly, cells were seeded into 96-well plates. After overnight incubation, cells in partial wells were fixed with 10% trichloroacetic acid (TCA) for 10 min at room temperature and washed with ddH2O. Cells at this stage represented the cell population at the time of drug addition (T0). The other cells were treated with (Tx) or without [control (C) in 0.1% DMSO] the indicated concentrations of doxazosin for an additional 48 h. Then, the cells (T0 and Tx) were fixed with 10% TCA for 10 min at room temperature and washed with ddH2O. All cells were stained with 0.4% (w/v) SRB in 1% acetic acid for 10 min at room temperature, and then washed with 1% acetic acid to remove unbound dye. SRB bound cells were solubilized with 10 mM trizma base. Using the absorbance (515 nm) measurements for T0, C and Tx, the percentage of doxazosin effect was calculated as follows: [1-(Tx-T0)/(C-T0)] ×100% (26,27).

Flow cytometry with propidium iodide (PI) staining

Cells were harvested by trypsinization, fixed with 70% (v/v) ethanol for 30 min at 4°C and washed with PBS. The cells were then centrifuged at 500 × g for 10 min at room temperature and re-suspended with 0.3 ml PI solution containing Triton X-100 (0.1%, v/v), RNase (100 µg/ml) and PI (80 µg/ml). The cellular DNA content was analyzed using an FACScan flow cytometer and CellQuest software (V6.0.4; Becton, Dickinson and Company) (28).

VM formation assays

VM formation assays were performed in 48-well culture plates coated with 100 µl Matrigel (9.5 mg/ml). Following Matrigel polymerization at 37°C for 30 min, the cells were seeded at 3.2×105 cells/ml in serum-free RPMI medium onto the Matrigel. After cell adhesion to the Matrigel for 4 h (basal condition, representing non-VM forming condition), 0 or 25 µM doxazosin was added to the serum-free medium and a medium change was performed every 2 days at 37°C for the indicated time, before western blotting and microscopic examination.

Periodic acid Schiff (PAS) staining and confocal microscopy

Cells were cultured on 18×18 mm glass coverslips coated with Matrigel at 37°C for 30 min. After 72 h treatment with different concentration of doxazosin, 3D culture cells were fixed with 4% paraformaldehyde in PBS for 15 min at room temperature, then quickly washed with PBS. Vascular channels were stained using a PAS kit (cat. no. SI-395B; Sigma) for 10 min at room temperature, washed with PBS for 10 min and treated with Schiff reagent (cat. no. SI-395B; Sigma-Aldrich) for 20 min at room temperature. The stained cells were washed with PBS for 15 min and vasculogenic morphogenesis was visualized using fluorescence microscopy (Zeiss Axio Imager, M1). Vascular channels were quantified using MetaMorph software (V7.8.0; Molecular Devices, LLC) (29). For 3D reconstruction, the 3D culture cells were stained with PAS and observed using a ZEISS Cell Observer SD Confocal Microscope (Zeiss GmbH) and ZEN software (V2.3; Zeiss GmbH) (30). The results were determined as follows: Total tube length=total length of tube (excluding nodes); mean tube length=(total tube length)/(number of segments); total tube area=total tube area (excluding nodes); mean tube area=(total tube area)/(number of segments); segments=total number of tube segments connecting branch points and/or ends. IC50 is the half maximal inhibitory concentration.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR were performed to assess mRNA expression levels. Corning Cell Recovery Solution (Corning, Inc.) was used to recover cells from 3D Matrigel cultures according to the manufacturer's instructions. Total RNA was extracted from the cells using an RNAspin Mini Kit (Cytivia), then cDNA was reverse transcribed from the total RNA (0.7 µg) using an iScript cDNA synthesis kit (Bio-Rad Laboratories, Inc.). The reverse transcription was performed at 37°C for 60 min, and then at 85°C for 5 min, according to the manufacturer's protocol. qPCR was performed using iTag Universal SYBR Green Supermix (Bio-Rad Laboratories, Inc.) and primer sequences as follows: Human VEGF-A forward (F), 5′-CTACCTCCACCATGCCAAGT-3′ and reverse (R), 5′-GCAGTAGCTGCGCTGATAGA-3′; human EPHA2 F, 5′-CCTCTAGTGCCTTCTTTAG-3′ and R, 5′-GAATGTTTGACACCCTCT-3′; human VE-cadherin F, 5′-CGTGTTCGCCATTGAGAG-3′ and R, 5′-TTCGCCAGTGTCCTTGTC-3′; human N-cadherin F, 5′-AGTACAGAAGCACTGGGATT-3′ and R, 5′-AAGCGTGTTGAAGCATATCAT-3′; human vimentin F, 5′-AGTCCACTGAGTACCGGAGAC-3′ and R, 5′-CATTTCACGCATCTGGCGTTC-3′; human FAK F, 5′-GTAGCGTGGCGTAAGTTA-3′ and R, 5′-TTCCTTGACAAGTGAATTATGC-3′; and human GAPDH F, 5′-CAGGGCTGCTTTTAACTCTGGT-3′ and R, 5′-GATTTTGGAGGGATCTCGCT-3′. DNA was amplified with an initial denaturation at 95°C for 5 min, followed by 40 cycles of 95°C for 5 sec, and 60°C for 30 sec. GAPDH was chosen as the internal reference. The data were expressed as relative mRNA levels by Cq values and then subsequently converted to fold change (31).

Western blotting

Corning Cell Recovery Solution (Corning, Inc.) was used to recover cells from 3D Matrigel cultures according to the manufacturer's instructions. The cells were harvested, centrifuged at 500 × g for 10 min at 4°C and lysed in 80 µl ice-cold lysis buffer (150 mM NaCl, 1% Triton X-100, 20 mM Tris-HCl pH 7.4, 1 mM EDTA, 1 mM EGTA, 1 mM PMSF, 10 µg/ml leupeptin, 1 mM Na3VO4, 1 mM NaF and 1 mM dithiothreitol) for 30 min. The concentration of the total protein was quantified using the Bradford method. Total protein was mixed with sample buffer and heated at 95°C for 10 min. An equal amount of protein (30 µg) per lane was separated by 8 or 12% SDS-PAGE, transferred to PVDF membranes. The membrane was then blocked with 5% skimmed milk for 1 h at room temperature. Protein expression levels were detected with specific antibodies (1:1,000 dilution for the primary antibodies at 4°C overnight and 1:7,000 dilution for the secondary antibodies at room temperature for 2 h). The membranes were washed three times with PBS-T (0.1% Tween 20) for 10 min after incubations with the primary and secondary antibodies. The immunoreactive proteins were detected with an enhanced chemiluminescence detection kit (LumiFlash™ Prime Chemiluminescent Substrate; cat. no. LF01-500; Visual Protein; Energenesis Biomedical Co., Ltd.) and the images were captured using a ChemiDoc™ MP System (Bio-Rad Laboratories, Inc.). LabTM Software (V6.0; Bio-Rad Laboratories, Inc.) was used to semi-quantify the data from western blotting.

VEGF-A secretion quantification

ELISA was applied to assess VEGF-A secretion (32). 3D culture cells were treated with 25 µM doxazocin for 96 h. Then, the concentration of VEGF-A in the supernatant was determined using a Human VEGF Quantikine ELISA kit (cat. no. DEV00; R&D Systems, Inc.) according to the manufacturer's instructions.

MMP-2 secretion and quantification

3D culture cells were treated with 25 µM doxazocin for 96 h. Then, the concentration of MMP-2 in the supernatant was determined using a Human MMP-2 Quantikine ELISA kit (cat. no. MMP200; R&D Systems, Inc.) according to the manufacturer's instructions.

Statistical analysis

Data are presented as the mean ± SEM. Student's unpaired t-test was performed for the statistical analysis of data comparing two groups. One-way ANOVA followed by the Bonferroni's post hoc test was used to perform the statistical analysis of multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

3D tubular structures form in A549 cells

A Matrigel-based VM formation assay was performed to examine whether the NSCLC cell line, A549, generated channels in 3D cultures. The results demonstrated that the cells formed vessel-like structures when cultured on Matrigel (Fig. 1A). PAS staining was performed to identify the glycoprotein-rich inner area of VM vessels, and lumen-containing tubular structures were subsequently observed in cultures using confocal microscopy and ZEN software (Fig. 1A and Video S1) (30). VM-related gene expression levels were also determined. The results revealed that VEGF-A and VE-cadherin expression levels were markedly upregulated in VM-forming cells (Fig. 1B). N-cadherin and vimentin (EMT markers) levels were also increased in 3D cultures. In addition, the levels of MMP-9, which is responsible for cell motility and ECM remodeling (33), were raised. MMP-2, which is involved in degrading basement membrane components for cancer metastasis along with MMP-9 (33), expression levels showed an increased trend, albeit this was not significant. FAK expression was also increased, although not significantly These data demonstrated that glycoprotein-rich lined tubular structures and VM-related molecules were present in the constructed in vitro VM model. Therefore, this model was utilized to evaluate the anti-VM effect of doxazosin.

Effect of doxazosin on VM formation

Quinazoline-based α1-adrenergic receptor blockers are widely used therapeutic drugs for treating hypertension (34), one of which was examined in the present study using the constructed VM cell model. Vasculogenic morphogenesis on a Matrigel surface was visualized in the control group using fluorescence microscopy following PAS staining (Figs. 1A and 2A). Doxazosin inhibited capillary-like tube formation and caused a dispersed morphology in A549 cells (Fig. 2A). The total tube length, mean tube length, total tube area and mean tube area of mimetic vessels (quantified using MetaMorph software) were also reduced by doxazosin, with IC50 values of 36.07±1.38, 28.38±2.10, 31.27±0.32 and 24.17±2.35 µM, respectively (Fig. 2B). The results therefore indicated that doxazosin treatment of the 3D Matrigel cell culture model significantly reduced VM channel formation.

Effect of doxazosin on cytotoxicity

The results of the SRB and MTT assays demonstrated that doxazosin also induced cytotoxic effects in A549 cells (Fig. 3). Doxazosin was more effective in inducing toxicities (6.3 µM, 13.1±4.6%; 12.5 µM, 20.8±5.6%; 25.0 µM, 45.1±3.5%; 37.5 µM, 98.2±9.1%) when using the SRB assay than when using MTT assay (cell survival: 37.5 µM, 72.6±4.4%; 50.0 µM, 47.8±1.8%; 100.0 µM, 14.5±0.1%) (Fig. 3A and B).

The effect of doxazosin on cell cycle progression was also examined, which revealed an increase in the apoptotic sub-G1 phase population with an initial apoptotic effect at 37.5 µM, compared with the 0 µM doxazosin control group (Figs. 3C and S1), which was similar to the aforementioned cytotoxicity assessment (Fig. 3B). The results therefore indicated that doxazosin was more effective in inducing anti-VM activities than in inducing cytotoxic effects.

Effect of doxazosin on VEGF-A and MMP-2 levels in 3D culture media

To further examine the doxazosin-mediated VM-blocking effect on underlying signaling pathways, the levels of two key mediators were determined in the medium during VM formation. The results revealed that the levels of both VEGF-A and MMP-2 in the control treatment group (0 µM doxazosin) were significantly increased compared with basal condition group, supporting VM formation (Fig. 4). Treatment with doxazosin significantly decreased these secretion levels.

Effect of doxazosin on the expression of pro-VM formation regulators

VE-cadherin is required for proper vascular development and is typically examined as an indicator of VM formation (3539). VE-cadherin expression was significantly increased following a 96-h incubation of the 3D Matrigel cultures (Fig. S2). The VM-related protein expression levels were then examined following treatment with doxazosin. For this, A549 cells were seeded in serum-free RPMI medium onto Matrigel in the absence or presence of 25 µM doxazosin for 96 h. Then, Corning Cell Recovery Solution was used to recover cells from the 3D Matrigel cultures. The protein expression of several signaling pathways, including VEGF-A/VE-cadherin/p-EphA2/p-PDK1/p-AKT/p-mTOR/p-p70S6k/p-ERK, MMP-2/MMP-9/laminin 5γ2 and vimentin/fibronectin were determined by western blotting. Doxazosin markedly decreased the cellular protein expression of VEGF-A monomer and dimer (Fig. 5A). Furthermore, doxazosin significantly decreased the protein expression of VE-cadherin and EphA2, and significantly reduced the phosphorylation levels of PDK1, AKT, mTOR, P70S6K and ERK in the 3D Matrigel cell culture model (Fig. 5A). The aforementioned results revealed the doxazosin-mediated suppression of VM formation. MMP-2 and MMP-9 are reported to degrade collagen in the basement membrane, supporting remodeling of the ECM and the regulation of VM formation (40). Although the laminin 5γ2 chain is mainly cleaved by activated MMP-2 (not MMP-9) to produce the 5γ2′ and 5γ2× cleaved fragments (which subsequently trigger the migration and invasion of tumor cells), both MMP-2 and MMP-9 colocalize with VM networks to assist VM formation (41). In the present study, doxazosin significantly reduced the protein expression levels of MMP-2 and MMP-9 and downregulated the protein expression levels of the cleaved forms of laminin, 5γ2′ and 5γ2× (Fig. 5B). EMT is reported to be implicated in VM formation and is associated with the tumor invasion-metastasis cascade (42). Doxazosin also significantly downregulated the protein expression levels of the two EMT markers, vimentin and fibronectin, in the 3D Matrigel-cultured A549 model (Fig. 5C). Collectively, these results confirmed that doxazosin displayed anti-VM activity in the NSCLC cell model.

Discussion

When tumors metastasize and grow in size, neovasculature is required to achieve sufficient nutrition through diffusion. VM serves as a mode of vascularization to mimic the vasculogenesis of endothelial cells (11,12,43). The vascular structure of VM has been reported to be built by cancer cells on rich ECMs, including proteoglycans and glycoproteins, which are positive for PAS staining. To identify VMs in human tumor biopsies and experimental settings, positive PAS staining in the absence of endothelial cell markers, such as CD31 or CD34, is generally used to assess vessel-like structures (30,43). However, certain studies raise an issue doubting the validity of concluding the presence of VM based solely on positive PAS staining without a definitive proof of a lumen in patterned vessel-like structures (44). In the present study, in accordance with the definition of VM formation, confocal microscopy examination was performed to detect structures with a lumen. The images showed lumen-like tubular structures after a 6-day growth of A549 cells in a Matrigel-cultured model. Furthermore, high PAS+ staining of VM vessels was observed. Notably, the gene expression levels of VEGF-A, VE-cadherin, N-cadherin, vimentin and MMP-9, which are crucial molecules in driving VM formation (9,44), were also markedly increased. Collectively, these data indicated VM formation in the constructed 3D Matrigel-cultured A549 model.

Several anticancer mechanisms of doxazosin have been reported, including activation of DNA damage (45,46), the TGF-β pathway (47) and autophagy (20), and the inhibition of angiogenesis through regulating the VEGF/Akt/mTOR signaling (23). In the present study, the anti-VM effect of doxazosin was explored using the constructed NSCLC A549 model. Doxazosin displayed inhibitory activity, as demonstrated by the reduced PAS+ staining of VM vessels and the quantified tube length and tube area. Notably, doxazosin was more efficient in blocking VM formation than inducing cytotoxicity by MTT assay and apoptosis. Although doxazosin induced similar anti-VM efficacy and cytotoxicity by SRB assay, the underlying mechanisms could be distinguished in which a broad panel of markers were used for anti-VM identification, including the protein expression of VEGF-A, MMP-2, MMP-9, VE-cadherin, EpHA2, vimentin and fibronectin, and laminin 5γ2 cleavage.

VE-cadherin is responsible for homotypic cell-cell interactions and vasculogenic events (48), and is exclusively expressed in highly aggressive tumors (49). Downregulation of VE-cadherin expression in aggressive melanoma cells abolished the ability to generate vasculogenic networks (49). Several studies have examined VE-cadherin as the only indicator for VM formation due to it having a prominent role in the acquisition of vascular-like structures (3638). Recent research and review articles have also addressed the key role of VE-cadherin in VM formation (38,39). Hepatocellular carcinoma cells, cultured on plates coated with a fusion protein comprising a human VE-cadherin extracellular domain and an immunoglobulin G Fc region (hVE-cad-Fc), markedly formed patterned tubular structures and exhibited increased levels or activity of EphA2, MMP-2, MMP-9 and EMT makers (50). p-EphA2 has been reported to trigger PI3K and increase membrane type 1 matrix metalloproteinase/MMP/MMP-14 expression and MMP-2 activation through the FAK and ERK1/2 pathways (8). Subsequently, the laminin 5γ2-chain is cleaved into γ2′ and γ2× fragments that subsequently trigger migration, invasion and VM formation in melanoma (8,41). Apatinib and its combination with melatonin are reported to decrease the expression of VE-cadherin and EphA2, and inhibit the phosphorylation of PI3K and AKT, leading to the inhibition of VM formation, survival and invasion of breast cancer stem cells (51). In the present study, similar inhibitory effects of doxazosin were observed on VM formation in the A549 model through the downregulation of VE-cadherin and the EphA2/PI3K/PDK-1/AKT/mTOR pathway. The downregulation of both MMP-2 and MMP-9 also contributed to the anti-VM mechanism. Furthermore, doxazosin significantly inhibited the generation of γ2′ and γ2× fragments, further supporting its anti-VM capability.

VEGF-A is another key mediator in VM formation. Secreted as a dimer, VEGF-A binds to VEGFR-1, which is highly expressed in malignant tumor cells with the capacity to induce VM formation (52). VEGF-A exposure triggers VM formation in several types of cancer cells, such as ovarian carcinoma (53) and melanoma (54), through increased expression of VE-cadherin, EphA2, MMP-2 and MMP-9, indicating that VEGF-A can stimulate tumor cell plasticity (53). VEGF gene silencing was reported to reduce VM formation and impair the expression levels of MMP-2 and MMP-9 via the PI3K/AKT-dependent pathway (55). Moreover, the blockade of EphA2 expression or activity inhibits VEGF expression and related angiogenesis in animal models, suggesting a complex regulation of these key regulators (56,57). In the present study, it was demonstrated that the gene expression and medium content of VEGF-A were upregulated in VM-forming cells, which was consistent with the aforementioned previous studies. Following doxazosin treatment, the reduction in VEGF-A expression might partly contribute to the inhibition of downstream VM signaling. EMT-related regulatory proteins are upregulated in VM-forming cells, suggesting a positive association between EMT and VM (42). MMPs, secreted by tumor cells lining VM networks, serve an important role in modifying cell-to-cell junctions and cell-ECM interactions, in which the ECM facilitates tumor invasion and metastasis (40). Due to the downregulation of MMP-2, MMP-9, vimentin and fibronectin protein expression observed in the present study following doxazosin treatment, doxazosin was hypothesized to inhibit EMT pathways, although this requires further validation.

It is noteworthy that several small molecules including natural products, synthetic compounds and known therapeutic drugs display anticancer potential through the suppression of VM formation (58). Furthermore, certain VM formation signaling pathways are suppressed by these agents (59,60). For example, triptonide potently inhibits VM by reducing the expression of VE-cadherin (59). Combretastatin A-4 and the synthetic compound, DHPAC, inhibit AKT phosphorylation and decrease the expression levels of VEGF, MMP2, MMP9 and Laminin 5 in NSCLC cell models (60). Following validation of the EphA2 receptor as a new target for treating tumors dependent on angiogenesis and VM (61), a novel synthetic compound, UniPR505, has been developed as an antagonist of the EphA2 receptor (62). Similarly, the present study demonstrated that doxazosin reduced the levels of VEGF-A, MMP-2, MMP-9 and VE-cadherin and inhibited the EphA2/AKT/mTOR/Laminin-5γ2 signaling network. Collectively, these studies together with the results of the present study support that the doxazosin-mediated suppression of VM formation may be an addition to the anti-NSCLC activities. However, the limitation of the present study is that it is a mechanistic study using 3D cancer cell culture without the consideration of interactions between other cell types and environments, as with in real tissues.

In conclusion, the results of the present study suggested that doxazosin displayed anti-VM activity in a 3D A549 model through the downregulation of VEGF-A and VE-cadherin levels, and the suppression of signaling pathways in which the receptor tyrosine kinase, EphA2, protein kinases, AKT and mTOR, and proteases, MMP-2 and MMP-9, are involved. These data support the add-on anti-VM effect of doxazosin as a potential agent against NSCLC.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Ms. Hwa-Man Hsu (Imaging Core, First Core Labs, National Taiwan University College of Medicine, Taipei, Taiwan), for technical assistance in image acquisition and analysis.

Funding

This work was supported by grants from the National Science and Technology Council (grant no. NSTC 112-2320-B-255-014) and the Chang Gung University of Science and Technology (grant no. ZURPF3N0091).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

JLH, LCH, CHH and JHG contributed to the conception and design of the experiments. JLH performed the experiments. JLH and WJL analyzed the data. JLH, WJL and JHG confirm the authenticity of all the raw data. JLH and JHG wrote the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Li S, de Camargo Correia GS, Wang J, Manochakian R, Zhao Y and Lou Y: Emerging targeted therapies in advanced non-small-cell lung cancer. Cancers (Basel). 15:28992023. View Article : Google Scholar : PubMed/NCBI

3 

Tan AC and Tan DSW: Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. J Clin Oncol. 40:611–625. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Alexander M, Kim SY and Cheng H: Update 2020: Management of non-small cell lung cancer. Lung. 198:897–907. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Jurišić V, Obradovic J, Pavlović S and Djordjevic N: Epidermal growth factor receptor gene in non-small-cell lung cancer: The importance of promoter polymorphism investigation. Anal Cell Pathol (Amst). 2018:61921872018.PubMed/NCBI

6 

Jurisic V, Obradovic J, Nikolic N, Javorac J, Perin B and Milasin J: Analyses of P16INK4a gene promoter methylation relative to molecular, demographic and clinical parameters characteristics in non-small cell lung cancer patients: A pilot study. Mol Biol Rep. 50:971–979. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Sun R, Hou Z, Zhang Y and Jiang B: Drug resistance mechanisms and progress in the treatment of EGFR-mutated lung adenocarcinoma. Oncol Lett. 24:4082022. View Article : Google Scholar : PubMed/NCBI

8 

Kirschmann DA, Seftor EA, Hardy KM, Seftor RE and Hendrix MJ: Molecular pathways: Vasculogenic mimicry in tumor cells: Diagnostic and therapeutic implications. Clin Cancer Res. 18:2726–2732. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Cao Z, Bao M, Miele L, Sarkar FH, Wang Z and Zhou Q: Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: A systemic review and meta-analysis. Eur J Cancer. 49:3914–3923. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Hendrix MJ, Seftor EA, Hess AR and Seftor RE: Vasculogenic mimicry and tumour-cell plasticity: Lessons from melanoma. Nat Rev Cancer. 3:411–421. 2003. View Article : Google Scholar : PubMed/NCBI

11 

van der Schaft DW, Seftor RE, Seftor EA, Hess AR, Gruman LM, Kirschmann DA, Yokoyama Y, Griffioen AW and Hendrix MJ: Effects of angiogenesis inhibitors on vascular network formation by human endothelial and melanoma cells. J Natl Cancer Inst. 96:1473–1477. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Angara K, Rashid MH, Shankar A, Ara R, Iskander A, Borin TF, Jain M, Achyut BR and Arbab AS: Vascular mimicry in glioblastoma following anti-angiogenic and anti-20-HETE therapies. Histol Histopathol. 32:917–928. 2017.PubMed/NCBI

13 

Song H, Ci H, Xu J, Xu Z, Zhang Y, Wang Y, Wu S and Tao Y: Vasculogenic mimicry and expression of slug and vimentin correlate with metastasis and prognosis in non-small cell lung cancer. Int J Clin Exp Pathol. 11:2749–2758. 2018.PubMed/NCBI

14 

Ci H, Xu Z, Xu J, Wang Y and Wu S: Expressions of KAI1 and E-cadherin in nonsmall cell lung cancer and their correlation with vasculogenic mimicry. Medicine (Baltimore). 97:e122932018. View Article : Google Scholar : PubMed/NCBI

15 

Zhou X, Gu R, Han X, Wu G and Liu J: Cyclin-dependent kinase 5 controls vasculogenic mimicry formation in non-small cell lung cancer via the FAK-AKT signaling pathway. Biochem Biophys Res Commun. 492:447–452. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Naeem A, Dakshanamurthy S, Walthieu H, Parasido E, Avantaggiati M, Tricoli L, Kumar D, Lee RJ, Feldman A, Noon MS, et al: Predicting new drug indications for prostate cancer: The integration of an in silico proteochemometric network pharmacology platform with patient-derived primary prostate cells. Prostate. 80:1233–1243. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Dalwadi SM, Hunt A, Bonnen MD and Ghebre YT: Computational approaches for drug repurposing in oncology: Untapped opportunity for high value innovation. Front Oncol. 13:11982842023. View Article : Google Scholar : PubMed/NCBI

18 

Wu H, Huang D, Zhou H, Sima X, Wu Z, Sun Y, Wang L, Ruan Y, Wu Q, Wu F, et al: Metformin: A promising drug for human cancers. Oncol Lett. 24:2042022. View Article : Google Scholar : PubMed/NCBI

19 

Pantziarka P, Pirmohamed M and Mirza N: New uses for old drugs. BMJ. 361:k27012018. View Article : Google Scholar : PubMed/NCBI

20 

Batty M, Pugh R, Rathinam I, Simmonds J, Walker E, Forbes A, Anoopkumar-Dukie S, McDermott CM, Spencer B, Christie D and Chess-Williams R: The role of α1-Adrenoceptor antagonists in the treatment of prostate and other cancers. Int J Mol Sci. 17:13392016. View Article : Google Scholar : PubMed/NCBI

21 

Forbes A, Anoopkumar-Dukie S, Chess-Williams R and McDermott C: Relative cytotoxic potencies and cell death mechanisms of α1-adrenoceptor antagonists in prostate cancer cell lines. Prostate. 76:757–766. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Bilbro J, Mart M and Kyprianou N: Therapeutic value of quinazoline-based compounds in prostate cancer. Anticancer Res. 33:4695–4700. 2013.PubMed/NCBI

23 

Keledjian K, Garrison JB and Kyprianou N: Doxazosin inhibits human vascular endothelial cell adhesion, migration, and invasion. J Cell Biochem. 94:374–388. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Park MS, Kim BR, Dong SM, Lee SH, Kim DY and Rho SB: The antihypertension drug doxazosin inhibits tumor growth and angiogenesis by decreasing VEGFR-2/Akt/mTOR signaling and VEGF and HIF-1α expression. Oncotarget. 5:4935–4944. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Scherbakov AM, Vorontsova SK, Khamidullina AI, Mrdjanovic J, Andreeva OE, Bogdanov FB, Salnikova DI, Jurisic V, Zavarzin IV and Shirinian VZ: Novel pentacyclic derivatives and benzylidenes of the progesterone series cause anti-estrogenic and antiproliferative effects and induce apoptosis in breast cancer cells. Invest New Drugs. 41:142–152. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Jurisic V, Bogdanovic G, Kojic V, Jakimov D and Srdic T: Effect of TNF-alpha on Raji cells at different cellular levels estimated by various methods. Ann Hematol. 85:86–94. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, Warren JT, Bokesch H, Kenney S and Boyd MR: New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Cancer Inst. 82:1107–1112. 1990. View Article : Google Scholar : PubMed/NCBI

28 

Jurisic V, Srdic-Rajic T, Konjevic G, Bogdanovic G and Colic M: TNF-α induced apoptosis is accompanied with rapid CD30 and slower CD45 shedding from K-562 cells. J Membr Biol. 239:115–122. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Shaifer CA, Huang J and Lin PC: Glioblastoma cells incorporate into tumor vasculature and contribute to vascular radioresistance. Int J Cancer. 127:2063–2075. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Racordon D, Valdivia A, Mingo G, Erices R, Aravena R, Santoro F, Bravo ML, Ramirez C, Gonzalez P, Sandoval A, et al: Structural and functional identification of vasculogenic mimicry in vitro. Sci Rep. 7:69852017. View Article : Google Scholar : PubMed/NCBI

31 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Jurisic V: Multiomic analysis of cytokines in immuno-oncology. Expert Rev Proteomics. 17:663–674. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N and Martinez-Fierro ML: The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci. 21:97392020. View Article : Google Scholar : PubMed/NCBI

34 

Vincent J, Elliott HL, Meredith PA and Reid JL: Doxazosin, an alpha 1-adrenoceptor antagonist: Pharmacokinetics and concentration-effect relationships in man. Br J Clin Pharmacol. 15:719–725. 1983. View Article : Google Scholar : PubMed/NCBI

35 

Franco P, Camerino I, Merlino F, D'Angelo M, Cimmino A, Carotenuto A, Colucci-D'Amato L and Stoppelli MP: αV–Integrin-Dependent inhibition of glioblastoma cell migration, invasion and vasculogenic mimicry by the uPAcyclin decapeptide. Cancers (Basel). 15:47752023. View Article : Google Scholar : PubMed/NCBI

36 

Qin Y, Zhao W, Cai Z, Wang Q, Gao J, Ci H, Feng Z and Ma L: The biomarker like the correlation between vasculogenic mimicry, vascular endothelial cadherin, sex-determiningregion on Y-Box transcription factor 17, and cyclin D1 in oesophageal squamous cell carcinoma. J Oncol. 2022:89155032022. View Article : Google Scholar : PubMed/NCBI

37 

Zhang Y, Tan Y, Liu S, Yin H, Duan J, Fan L, Zhao X and Jiang B: Implications of Withaferin A for the metastatic potential and drug resistance in hepatocellular carcinoma cells via Nrf2-mediated EMT and ferroptosis. Toxicol Mech Methods. 33:47–55. 2023. View Article : Google Scholar : PubMed/NCBI

38 

Delgado-Bellido D, Garcia-Diaz A and Oliver FJ: Co-immunoprecipitation of protein complexes from different subcellular compartments in vasculogenic mimicry studies. Methods Mol Biol. 2514:61–72. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Delgado-Bellido D, Oliver FJ, Vargas Padilla MV, Lobo-Selma L, Chacón-Barrado A, Díaz-Martin J and de Álava E: VE-Cadherin in cancer-associated angiogenesis: A deceptive strategy of blood vessel formation. Int J Mol Sci. 24:93432023. View Article : Google Scholar : PubMed/NCBI

40 

Winkler J, Abisoye-Ogunniyan A, Metcalf KJ and Werb Z: Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 11:51202020. View Article : Google Scholar : PubMed/NCBI

41 

Seftor RE, Seftor EA, Koshikawa N, Meltzer PS, Gardner LM, Bilban M, Stetler-Stevenson WG, Quaranta V and Hendrix MJ: Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res. 61:6322–6327. 2001.PubMed/NCBI

42 

Liu Q, Qiao L, Liang N, Xie J and Zhang J, Deng G, Luo H and Zhang J: The relationship between vasculogenic mimicry and epithelial-mesenchymal transitions. J Cell Mol Med. 20:1761–1769. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, Trent JM, Meltzer PS and Hendrix MJ: Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am J Pathol. 155:739–752. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Valdivia A, Mingo G, Aldana V, Pinto MP, Ramirez M, Retamal C, Gonzalez A, Nualart F, Corvalan AH and Owen GI: Fact or fiction, it is time for a verdict on vasculogenic mimicry? Front Oncol. 9:6802019. View Article : Google Scholar : PubMed/NCBI

45 

Lin SC, Chueh SC, Hsiao CJ, Li TK, Chen TH, Liao CH, Lyu PC and Guh JH: Prazosin displays anticancer activity against human prostate cancers: Targeting DNA and cell cycle. Neoplasia. 9:830–839. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Arencibia JM, Del Rio M, Bonnin A, Lopes R, Lemoine NR and López-Barahona M: Doxazosin induces apoptosis in LNCaP prostate cancer cell line through DNA binding and DNA-dependent protein kinase down-regulation. Int J Oncol. 27:1617–1623. 2005.PubMed/NCBI

47 

Partin JV, Anglin IE and Kyprianou N: Quinazoline-based alpha 1-adrenoceptor antagonists induce prostate cancer cell apoptosis via TGF-beta signalling and I kappa B alpha induction. Br J Cancer. 88:1615–1621. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Treps L, Le Guelte A and Gavard J: Emerging roles of Semaphorins in the regulation of epithelial and endothelial junctions. Tissue Barriers. 1:e232722013. View Article : Google Scholar : PubMed/NCBI

49 

Hendrix MJ, Seftor EA, Meltzer PS, Gardner LM, Hess AR, Kirschmann DA, Schatteman GC and Seftor RE: Expression and functional significance of VE-cadherin in aggressive human melanoma cells: Role in vasculogenic mimicry. Proc Natl Acad Sci USA. 98:8018–8023. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Shuai Q, Cao L, Qin Z, Zhang Y, Gu Z and Yang J: VE-cadherin fusion protein substrate enhanced the vasculogenic mimicry capability of hepatocellular carcinoma cells. J Mater Chem B. 8:1699–1712. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Maroufi NF, Rashidi M, Vahedian V, Jahanbazi R, Mostafaei S, Akbarzadeh M, Kazemzadeh H, Nejabati HR, Isazadeh A, Rashidi MR and Nouri M: Effect of Apatinib plus melatonin on vasculogenic mimicry formation by cancer stem cells from breast cancer cell line. Breast Cancer. 29:260–273. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Frank NY, Schatton T, Kim S, Zhan Q, Wilson BJ, Ma J, Saab KR, Osherov V, Widlund HR, Gasser M, et al: VEGFR-1 expressed by malignant melanoma-initiating cells is required for tumor growth. Cancer Res. 71:1474–1485. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Wang JY, Sun T, Zhao XL, Zhang SW, Zhang DF, Gu Q, Wang XH, Zhao N, Qie S and Sun BC: Functional significance of VEGF-a in human ovarian carcinoma: Role in vasculogenic mimicry. Cancer Biol Ther. 7:758–766. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Vartanian A, Stepanova E, Grigorieva I, Solomko E, Baryshnikov A and Lichinitser M: VEGFR1 and PKCα signaling control melanoma vasculogenic mimicry in a VEGFR2 kinase-independent manner. Melanoma Res. 21:91–98. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Xu X, Zong Y, Gao Y, Sun X, Zhao H, Luo W and Jia S: VEGF induce vasculogenic mimicry of choroidal melanoma through the PI3k signal pathway. Biomed Res Int. 2019:39091022019. View Article : Google Scholar : PubMed/NCBI

56 

Cheng N, Brantley D, Fang WB, Liu H, Fanslow W, Cerretti DP, Bussell KN, Reith A, Jackson D and Chen J: Inhibition of VEGF-dependent multistage carcinogenesis by soluble EphA receptors. Neoplasia. 5:445–456. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Brantley-Sieders DM, Fang WB, Hwang Y, Hicks D and Chen J: Ephrin-A1 facilitates mammary tumor metastasis through an angiogenesis-dependent mechanism mediated by EphA receptor and vascular endothelial growth factor in mice. Cancer Res. 66:10315–10324. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Guan YY, Luan X, Lu Q, Liu YR, Sun P, Zhao M, Chen HZ and Fang C: Natural products with antiangiogenic and antivasculogenic mimicry activity. Mini Rev Med Chem. 16:1290–1302. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Han H, Du L, Cao Z, Zhang B and Zhou Q: Triptonide potently suppresses pancreatic cancer cell-mediated vasculogenic mimicry by inhibiting expression of VE-cadherin and chemokine ligand 2 genes. Eur J Pharmacol. 818:593–603. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Gong FL, Wang L, Yu LG, Dang YF, Jiang XN, Zhao L and Guo XL: DHPAC, a novel microtubule depolymerizing agent, suppresses angiogenesis and vasculogenic mimicry formation of human non-small cell lung cancer. J Cell Biochem. 121:4756–4771. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Margaryan NV, Strizzi L, Abbott DE, Seftor EA, Rao MS, Hendrix MJ and Hess AR: EphA2 as a promoter of melanoma tumorigenicity. Cancer Biol Ther. 8:279–288. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Incerti M, Russo S, Corrado M, Giorgio C, Ballabeni V, Chiodelli P, Rusnati M, Scalvini L, Callegari D, Castelli R, et al: Optimization of EphA2 antagonists based on a lithocholic acid core led to the identification of UniPR505, a new 3α-carbamoyloxy derivative with antiangiogenetic properties. Eur J Med Chem. 189:1120832020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hsu J, Leu W, Hsu L, Hsieh C and Guh J: Doxazosin inhibits vasculogenic mimicry in human non‑small cell lung cancer through inhibition of the VEGF‑A/VE‑cadherin/mTOR/MMP pathway. Oncol Lett 27: 170, 2024
APA
Hsu, J., Leu, W., Hsu, L., Hsieh, C., & Guh, J. (2024). Doxazosin inhibits vasculogenic mimicry in human non‑small cell lung cancer through inhibition of the VEGF‑A/VE‑cadherin/mTOR/MMP pathway. Oncology Letters, 27, 170. https://doi.org/10.3892/ol.2024.14303
MLA
Hsu, J., Leu, W., Hsu, L., Hsieh, C., Guh, J."Doxazosin inhibits vasculogenic mimicry in human non‑small cell lung cancer through inhibition of the VEGF‑A/VE‑cadherin/mTOR/MMP pathway". Oncology Letters 27.4 (2024): 170.
Chicago
Hsu, J., Leu, W., Hsu, L., Hsieh, C., Guh, J."Doxazosin inhibits vasculogenic mimicry in human non‑small cell lung cancer through inhibition of the VEGF‑A/VE‑cadherin/mTOR/MMP pathway". Oncology Letters 27, no. 4 (2024): 170. https://doi.org/10.3892/ol.2024.14303