Open Access

Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer

  • Authors:
    • Yan Li
    • Chunxia Liu
    • Ke Tang
    • Yan Chen
    • Kang Tian
    • Zhiqiang Feng
    • Jindong Chen
  • View Affiliations

  • Published online on: November 28, 2017     https://doi.org/10.3892/mmr.2017.8179
  • Pages: 2373-2383
  • Copyright: © Li 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

Activation of kinase-associated signaling pathways is one of the leading causes of various malignant phenotypes in breast tumors. Strategies of drug discovery and development have investigated approaches to target the inhibition of protein kinase signaling. In the current study, the anti‑tumor activities of a novel multi‑kinase inhibitor, T03 were evaluated in breast cancer. T03 inhibited Taxol‑resistant breast cancer cell proliferation and induced cell cycle arrest and apoptosis in vitro and in vivo. The current results demonstrated that T03 downregulated c‑Raf, platelet‑derived growth factor receptor‑β and other kinases, thus inhibited Raf/mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase and Akt/mechanistic target of rapamycin survival pathways in MCF‑7 and MCF‑7/Taxol xenograft tumors. At a dose of 100 mg/kg, T03 inhibited tumor growth by 62.90 and 59.98% in tumor weight in MX‑1 and MX‑1/T xenograft models, respectively and by 62.60 and 60.22% in MCF‑7 and MCF‑7/T tumors, respectively. These data indicate that the novel multi‑kinase inhibitor, T03, may present as a potential compound to develop novel treatments against breast cancer and Taxol‑resistant breast tumors.

Introduction

Breast cancer is the most common type of invasive cancer in women. Approximately 1.7 million women are diagnosed with breast cancer annually, and >500,000 succumb to it worldwide (1). While surgery, traditional chemotherapy and radiotherapy are commonly used to treat breast cancer; targeted therapy has drawn the attention of clinicians and researchers in the past decade for its improved therapeutic effect in metastatic cancer, as compared with traditional chemotherapy. Thus, numerous multi-kinase inhibitors have been adopted in the targeted therapy of metastatic breast cancer (24). However, the resistance of breast cancer to these inhibitors and drugs remains challenging in chemotherapy and targeted therapy. Therefore, continuing to develop novel anti-cancer drugs is necessary in cancer therapy.

In tumorigenesis of breast and other types of tissue, protein kinases are important in the regulation of proliferation, apoptosis and migration (5). For example, platelet-derived growth factor receptor-β (PDGFRβ), a member of the tyrosine kinase receptors type III family, is associated with the malignancy of breast carcinoma (68). In response to survival signals, PDGFRβ activates Akt by upregulating phosphoinositide 3-kinase (PI3K) and phosphoinositide-dependent protein kinase-1 (PDK1) (9). Thus, inhibition of PDGFRβ activity by TKI inhibitor(s) may suppress breast tumor growth (10).

In addition, the Ras/Raf/mitogen-activated protein kinases (MAPK) signaling pathway is critical in breast tumorigenesis (11). Studies have demonstrated that constitutive activation of the MAPK signaling pathway is associated with the progression of breast cancer via the induction of chemoresistance and distant metastases (1216). Thus, the MAPK signaling pathway may be a potent target for breast cancer chemotherapy (17).

Currently, certain drugs, such as Trastuzumab, Lapatinib, Bevacizumab and Taxol have been identified for breast cancer targeted therapy. Of them, Taxol is the commonly administered drug for the treatment of breast cancer. However, continuous use of Taxol results in acquired drug-resistance of breast cancer (18). Therefore, development of novel drugs is essential to improve targeted therapy of breast cancer and Taxol-resistant breast cancer. In the current study, a novel multi-kinase inhibitor, T03 is reported. T03 is a novel multi-kinase inhibitor against PDGFRβ and c-Raf, and inhibition of PDGFRβ and c-Raf by T03 may downregulate the Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and PDGFR/Akt/mechanistic target of rapamycin (mTOR) survival pathway. In the present study, the anti-tumor activity and underlying mechanism of T03 in regular and Taxol-resistant breast cancer were investigated in vitro and in vivo.

Materials and methods

Cell culture

Breast cancer cell line MCF-7 was obtained from the cell center of Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC; Beijing, China). Cell lines MX-1 and MX-1/T (Taxol-resistant) A549, A549/T (Taxol-resistant) were obtained from the Professor Yongkui Jing (Mount Sinai School of Medicine, New York, NY, USA). The MCF-7/T (Taxol-resistant) cell was established in our laboratory (Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing China) (19). MCF-7/ADM (Adriamycin-resistant) was obtained from the Assistant Professor Hongbo Wang (Yantai University, Shandong, China). The cells were maintained in Dulbecco's modified Eagle's medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 100 IU/ml penicillin and 100 µg/ml streptomycin in a humidified incubator containing 5% CO2 at 37°C. In the present study, four specific cells (MX-1/T, MCF-7/T, MCF-7/ADM, A549/T) were used.

Drugs

T03, a small molecule compound containing 2-picolinylhydrazide moiety (Chinese patent application no. 201110129115.7) was synthesized by the Department of Pharmacochemistry at the Institute of Materia Medica, CAMS and PUMC (Purity >97%; high-performance liquid chromatography). For in vitro experiments, T03 and Taxol (Beijing Union Pharmaceutical Factory, Beijing, China) were dissolved in dimethyl sulfoxide (DMSO) and stored at 4°C until use. DMSO served as a vehicle control in all experiments at a final concentration of 0.1%. For the in vivo experiments, T03 was dissolved in a solution of Cremophor EL (Aladdin Industrial Corporation, Shanghai, China; cat no. C107105)/ethanol/water (12.5:12.5:75) (20).

Cell viability assay

MX-1, MX-1/T, MCF-7 and MCF-7/T cells (2,500 cells per well) were seeded in a 96-well plate. After 24 h of incubation at 37°C, cells were treated with various different concentrations of Taxol and T03. After 72 h of incubation at 37°C, the Cell Counting kit-8 (CCK-8; cat. no. C0037; Beyotime Institute of Biotechnology, Shanghai, China) assay was performed to evaluate cell viability. Absorbance values which was measured using an ELISA reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA) at 450 nm were normalized to the values obtained for vehicle-treated cells to determine the percentage of surviving cells. The median inhibitory concentration (IC50) was defined as the drug concentration at which cell growth was inhibited by 50%. Each assay was performed in triplicate.

Colony formation assay

MCF-7 and MCF-7/T cells were trypsinized to single-cell suspensions, and resuspended in DMEM culture medium containing 10% FBS. Approximately 500 cells were plated in 6-well tissue culture plates. After a 24-h incubation at 37°C, the cells were treated with either T03, 0.1% DMSO, or nothing. Cells were incubated in 5% CO2 at 37°C for 14 days, and the colonies were washed, fixed and stained with 0.005% crystal violet in methanol. The number of colonies was manually counted without a microscope, and experiments were performed in triplicate and repeated three times.

Apoptosis analysis

MX-1, MX-1/T, MCF-7 and MCF-7/T cells were treated with either T03 or 0.1% DMSO for 72 h. Apoptotic cells were measured using Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection kit (cat. no. 556570; BD Biosciences, Franklin Lakes, NJ, USA). Briefly, cells were trypsinized and washed with PBS following treatment and stained with Annexin V-FITC according to the manufacturer's protocol, then analyzed by ACCURI C6 flow cytometry with BD Accuri C6 software (BD Biosciences).

Cell-cycle analysis

MX-1, MX-1/T, MCF-7 and MCF-7/T cells were incubated at 37°C with either T03 or 0.1% DMSO for 72 h. Cells were washed in PBS and fixed with 4°C ice-cold 70% ethanol overnight. The cells were then suspended in PBS containing RNase A (100 µg/ml; cat. no. R1030; Beijing Solarbio Science and Technology Co., Ltd., Beijing, China), propidium iodide (50 µg/ml; cat. no. P8080; Beijing Solarbio Science and Technology Co., Ltd.), Triton X-100 (0.1%), and incubated on the ice in the dark for at least 1 h (21). The cell cycle profiles were determined by flow cytometric analysis.

Tumor implantation and growth in MX-1, MX-1/T, MCF-7 and MCF-7/T xenografts

All animal studies were performed in compliance with the policies of the Institute of Materia Medica Animal Care and Use Committee. Six-week-old, female BALB/c/nu nude mice were used in the present study (all had 20 mice/experiment). The body weight was 15–16 g for MX-1 and MX-1/T xenograft model, and 16–22 g for MCF-7 and MCF-7/T xenograft model. They were purchased from Vital River Laboratory Animal Technology Co., Ltd., (Beijing, China) and housed in the controlled environment at 25°C on a 12-h light/dark cycle (5 mice per group). When tumors grew to an average volume of 100–250 mm3, tumor-bearing mice were randomly separated into four groups of five animals. A total of one group received per os Cremophor EL/ethanol/water and served as a vehicle control; the other groups received injections of 5 mg/kg Taxol (twice per week), or received an oral dose of 50 or 100 mg/kg T03 six times per week for 13 days (MX-1 and MX-1/T) and 33 days (MCF-7, MCF-7/T). Mice were euthanized at the end of the treatment period. Tumors were removed and weighed, and samples of all of the sections were stored at −80°C for western blot analysis.

Kinase inhibition assay

Inhibition of kinase activity against target kinases was measured using Caliper and Glo-ATP assays (ADP-Glo assay buffer: 25 mM HEPES, 10 mM MgCl2, 0.01% Triton X-100, 100 µg/ml BSA, 2.5 mM DTT, (pH 7.4); Caliper assay buffer: 100 mM HEPES, 10 mM MgCl2, 100 µl/l Brij35 (30%), 1 mM DTT, (pH 7.4); Other reagents: ATP (cat. no. A7699 Sigma-Aldrich; Merck KGaA, Darmstadt, Germany); ADP Gloreagent (cat. no. V9102; Promega Corporation, Madison, WI, USA). The Biochemical assay was performed according to the manufacturer's protocol. The assay was performed by HD Biosciences (China) Co., Ltd. (Shanghai, China).

Western blot analysis

Lysates (portions of two or three randomly selected tumors from MCF-7 and MCF-7/T xenograft mice) were prepared as previously described. The protein extraction buffer was a radioimmunoprecipitation buffer (1 mM phenylmethylsulfonyl fluoride) (21). Protein concentration was determined using the bicinchoninic acid method. Total proteins (50 µg) were separated by 10.0% SDS-PAGE and transferred to a nitrocellulose membrane by semi-wet electrophoresis were incubated with primary antibodies overnight at 4°C following blocking with TBS containing 1% Tween-20 and 5% skimmed dry milk for 1 h at room temperature. The antibodies were as follows: Rabbit anti-phosphorylated (p)-c-Raf (Ser259; cat. no. 9421), c-Raf (cat. no. 9422), p-MEK (cat. no. 9127), MEK (cat. no. 9903), p-ERK (cat. no. 4370), ERK (cat. no. 9101), p-PDGFRβ (cat. no. 3170), PDGFRβ (cat. no. 3169), p-PDK (cat. no. 3061), PDK (cat. no. 3062), p-Akt (Thr 308; cat. no. 9275), Akt (cat. no. 4691), p-mTOR (cat. no. 2971), mTOR (cat. no. 2983), p-AuroraA (cat. no. 2914), AuroraA (cat. no. 14475) (all from Cell Signaling Technology, Inc., Danvers, MA, USA) and mouse anti-actin (cat. no. 3700; CST Biological Reagents Co., Ltd., Shanghai China). All the primary antibodies were used at 1:1,000. The samples were detected with peroxidase-conjugated anti-rabbit or anti-mouse IgG (1:5,000, cat. no. sc-2004, Santa Cruz Biotechnology, Inc., Dallas, TX, USA) for 1 h at room temperature and developed using an enjhanced chemiluminescence system western blot detection and analysis system (Applygen Technologies, Inc., Beijing, China). The membranes were assessed for equal loading by probing for β-actin.

Statistical analysis

Data were expressed as means ± standard deviation. Statistical analysis of the results was performed using one-way analysis of variance followed by a Bonferroni post-hoc test. P<0.05 was considered to indicate a statistically significant difference. Statistical software used was Microsoft Excel 2010 (Microsoft Corporation, Redmond, WA, USA) and SPSS software (version 19.0; SPSS, Inc., Chicago, IL, USA).

Results

T03 inhibits the proliferation of breast cancer cells

To evaluate the suppressive efficacy of T03 on breast cancer cells in vitro, the regular (MX-1, MCF-7) and Taxol-resistant (MX-1/T, MCF-7/T) breast cancer cells were treated with various concentrations of T03 (0.8–25.0 µmol/l) for 72 h. The CCK-8 assay results showed that T03 inhibited the growth of the breast cancer cells in dose-dependent manner (Fig. 1A). The T03 IC50 S values were 8.46±0.28 (MX-1), 11.65±2.19 (MX-1/T), 6.39±1.15 (MCF-7), and 10.95±0.49 µmol/l (MCF-7/T; Table I). These results indicated that T03 effectively inhibited the growth of breast cancer cells, as well as the Taxol-resistant breast cancer cells. To establish whether T03 inhibits other drug-resistant cells, similar experiments on Doxorubicin-resistant breast cancer cells (MCF-7/ADM) were conducted. Similarly, T03 inhibited Adriamycin-resistant cells in a dose-dependent manner (Table II).

Table I.

Effect of T03 on proliferation in breast cancer cells.

Table I.

Effect of T03 on proliferation in breast cancer cells.

Median inhibitory concentration, mol/l

TreatmentMX-1MX-1/TMCF-7MCF-7/T
T03 8.46±0.28×10-6 11.65±2.19×10-6 6.39±1.15×10-6 10.95±0.49×10-6
Taxol 4.44±0.30×10-8 1.52±0.50×10-6 1.83±0.36×10-9 7.66±2.06×10-7

Table II.

Effect of T03 on proliferation in resistant cells.

Table II.

Effect of T03 on proliferation in resistant cells.

Median inhibitory concentration, mol/l

TreatmentMCF-7MCF-7/ADMA549A549/T
T037.91×10-61.83×10-59.83×10-62.76×10-5
Taxol5.97×10-94.37×10-75.09×10-92.30×10-7
T03 suppresses colony formation in breast cancer cells

To confirm the suppressive ability of T03 on tumor cell proliferation, a colony formation assay was conducted in MCF-7 and MCF-7/T cells. The data revealed that T03 effectively inhibited the colony formation in MCF-7 and MCF-7/T colony cells in a dose-dependent manner (Fig. 1B and C). T03 inhibited colony formation by ~50.0% (IC50) at a concentration of 7.61 µmol/l in MCF-7 and 7.45 µmol/l in MCF-7/T cells. The data were consistent with the results of the CCK-8 assay.

T03 treatment led to cell cycle arrest in breast cancer cells

T03 was observed to cause cell cycle arrest in breast cancer cells. T03 treatment led to elevated numbers of G2-phase cells and decreased G1-phase cells in a dose-dependent manner in MX-1 and MX-1/T cells (Fig. 2A and B). With a treatment of 5.0 µmol/l T03, the percentage of G2-phase cells increased to 15.23±0.83% (MX-1) and 29.30±1.37% (MX-1/T), while it was 11.37±0.90 and 11.47±1.26% in the untreated controls. Concomitantly, the percentage of G1 phase cells reduced to 60.37±1.01% (MX-1) and 40.60±1.06% (MX-1/T) compared with 68.10±1.65 and 56.33±0.86% in the untreated controls. Furthermore, while T03 treatment induced cell accumulation at the G2 and G1 phases, it caused the decrease of the S phase cells in MCF-7 and MCF-7/T (Fig. 2C and D). The percentage of G2 phase cells was 25.50±0.92 and 26.80±1.51% in MCF-7 and MCF-7/T cells, respectively, when they were treated with 5.0 µmol/l T03. By contrast, the percentages were 20.20±1.25% (MCF-7) and 21.73±1.66% (MCF-7/T) in the controls. The percentages of S phase cells decreased from 33.43±1.45 and 30.77±0.46% to 16.90±1.49 and 18.53±2.10% (following treatment with 5.0 µmol/l T03), respectively (Fig. 2C and D).

T03 induced apoptosis in breast cancer cells

T03 treatment was demonstrated to induce apoptosis in MX-1, MX-1/T, MCF-7, and MCF-7/T breast cancer cells. Upon treatment with 10.0 µmol/l T03, the early apoptosis increased by 4.16- and 2.90-fold, and late apoptosis increased by 67.25- and 3.23-fold in MX-1 and MX-1/T cells (Fig. 2E and F). In MCF-7 and MCF-7/T cells, early apoptosis increased from 1.28 and 4.51% (control) to 24.75 and 14.70%, respectively, while late apoptosis increased from 2.67 and 4.73% (control) to 24.05 and 18.00%, respectively (Fig. 2G and H).

T03 inhibited kinases in breast cancer cells

To investigate which kinases T03 inhibited, Caliper and ADP-Glo assays were performed on a panel of kinases in T03-treated breast cancer cells. The results indicated that various oncogenic kinases were susceptible to T03 inhibition. Of these kinases, c-Raf, PDGFRβ and RET proto-oncogene may be suppressed by T03, and the IC50s are 0.78, 0.23 and 0.71 µmol/l, respectively. In addition, T03 may inhibit the activity of PDGFRα, fms related tyrosine kinase 1 (FLT1), kinase insert domain receptor, FLT3, and c-kit with IC50 between 0.05 and 1.0 µmol/l. Furthermore, it was found that T03 downregulates FGFR1, FGFR2 and b-Raf at the micromole level. By contrast, T03 exerted little effect on other tested kinases, such as Aurora A, insulin like growth factor 1 receptor, ERK1, ERK2, Src, PI3K, erb-b2 receptor tyrosine kinase 2 and AMPK (Table III).

Table III.

Inhibitory activity of T03 against different kinases (biochemical assay).

Table III.

Inhibitory activity of T03 against different kinases (biochemical assay).

A, Median inhibitory concentration of T03 against different kinases

Kinase targetIn vitro IC50 value, µmol/lSource
c-Raf0.78ADP-Glo
PDGFRβ   0.230Caliper
Ret proto-oncogene0.71Caliper
PDGFRα0.37Caliper
FLT10.93Caliper
Kinase insert domain receptor   0.504Caliper
FLT3   0.046Caliper
c-kit0.16Caliper
FGFR1   3.929Caliper
FGFR2   2.962Caliper
b-Raf   4.063ADP-Glo

B, The inhibition rate of T03 against different kinases

Kinase targetInhibition, 1.0 µmo/la (%)Source

Aurora A9.92Caliper assay
IGFR12.57ADP-Glo
ERK19.73Caliper
ERK20.79Caliper
SRCADP-Glo
PI3Kα2.01ADP-Glo
PI3Kγ19.33Caliper
ERBB28.91Caliper
AMPK (A1/B1/G1)14.65Caliper
AMPK (A2/B1/G1)28.76ADP-Glo

[i] aInhibition at 1.0 µmol/l. Kinase selectivity profiling was performed at HD Biosciences (China) Co., Ltd. (Shanghai, China) usng ADP-Glo and Capliper assays. PDGFR, platelet derived growth factor receptor; FLT, Fms related tyrosine kinase; FGFR, fibroblast growth factor receptor; ERK, extracellular signal regulated kinase; PI3K, phosphoinositide 3-kinase; AMPK, AMP-activated protein kinase; IGFR1, insulin like growth factor 1 receptor; ERBB2, Erb-b2 receptor tyrosine kinase.

T03 inhibited xenograft tumor growth of breast cancer cells

Based on the above results obtained in vitro, further experiments were performed to determine whether T03 inhibits xenograft tumor growth from the breast cancer cells. To obtain xenograft tumors, the MX-1, MX-1/T, MCF-7, and MCF-7/T human breast cells were inoculated into BALB/c/nu nude mice. While T03 was used to treat the xenograft tumors, Taxol was adopted as a reference compound. When 5 mg/kg Taxol was applied to MX-1 and MX-1/T xenografts, the treated/control (T/C) ratio were 55.56 and 95.13%, respectively, according to the relative tumor volume (RTV). Furthermore, the inhibition ratios were 41.56% in MX-1 and 0% in MX-1/T based on the relative tumor weight, indicating that MX-1/T was less sensitive to Taxol at a dose of 5 mg/kg. For T03, the T/C ratio was 46.99% (50 mg/kg) and 34.68% (100 mg/kg) according to RTV, and the inhibition ratio of tumor weight reached 50.00 and 62.90% in MX-1 xenografts (Table IV and Fig. 3A). Furthermore, in MX-1/T xenografts, the T/C ratio of RTV was 45.06% (50 mg/kg) and 31.47% (100 mg/kg), and the inhibition ratio of the tumor weight was 51.02 and 59.98%, respectively (Table V and Fig. 3B), indicating that T03 exerted more effective inhibition on the MX-1 and MX-1/T xenografts.

Table IV.

Antitumor activity of T03 on the breast cancer MX-1 xenograft model.

Table IV.

Antitumor activity of T03 on the breast cancer MX-1 xenograft model.

Body weight, gTumor volume, mm3Relative tumor volumeTumor weight




CompoundDose, mg/kgAnimals, nInitialFinalInitialFinalx±SDTreated/control ratio, %x±SD, gInhibition, %
Control 5/5 16.0±1.0 21.8±1.9 122.6±4.2 2,709.5±633.4 22.09±5.12 1.61±0.45
Taxol     55/5 16.0±0.7 20.8±2.2 140.2±13.3 1,714.4±308.2a 12.27±2.33b55.56 0.94±0.20a41.56
T03  505/5 15.0±1.0 19.2±1.3 121.2±24.9 1,318.2±849.5a 10.38±5.48b46.99 0.81±0.49a50.00
1005/5 15.2±0.5 18.9±1.4 123.7±6.7 945.7±35.8c 7.66±0.48c34.68 0.60±0.03b62.90

a P<0.05

b P<0.01 and P<0.001 vs. control. Means ± standard deviation.

Table V.

Antitumor activity of T03 on the breast cancer MX-1/T xenograft model.

Table V.

Antitumor activity of T03 on the breast cancer MX-1/T xenograft model.

Body weight, gTumor volume, mm3Relative tumor volumeTumor weight




CompoundDose, mg/kgAnimals, nInitialFinalInitialFinalx±SDTreated/control ratio, %x±SD, gInhibition, %
Control 5/5 16.0±1.0 20.8±2.9 91.7±19.7 2,131.3±920.4 23.87±10.22 1.57±0.47
Taxol     55/5 15.6±0.6 20.4±0.9 95.7±17.1 2,183.1±1,091.0 22.71±11.0995.13 1.58±0.79
T03  505/5 15.2±0.5 19.0±2.5 98.5±9.8 1,079.1±887.6 10.76±8.6645.06 0.77±0.6251.02
1005/5 15.8±0.8 18.8±2.2 96.7±6.4 728.6±78.2a 7.51±0.86a31.47 0.63±0.06b59.98

a P<0.05

b P<0.01 vs. control. x±SD, Means ± standard deviation.

In addition, it was observed that T03 presented even higher inhibitory ability to MCF-7 and MCF-7/T xenografts compared with MX-1 or MX-1/T models. While Taxol was applied to the MCF-7 xenograft, the T/C ratio of the RTV was 6.75% (5 mg/kg) and the inhibition rate of the tumor weight was 96.36%. By contrast, Taxol exerted almost no inhibitory effect on the MCF-7/T xenograft model. In contrast to this, when T03 was applied to MCF-7 xenografts, the RTV T/C ratio was 45.63% (50 mg/kg) and 32.55% (100 mg/kg), and the inhibition rate of the tumor weight reached 57.09 and 62.60% (Table VI and Fig. 3C). Furthermore, in MCF-7/T xenografts, the RTV T/C ratios were 41.21% (50 mg/kg) and 25.52% (100 mg/kg) while the inhibition rate of the tumor weight attained 44.06 and 60.22%, respectively (Table VII and Fig. 3D). These data indicated that T03 inhibits Taxol-sensitive and -resistant breast cancer tumors.

Table VI.

Antitumor activity of compounds on the breast cancer MCF-7 xenograft model.

Table VI.

Antitumor activity of compounds on the breast cancer MCF-7 xenograft model.

Body weight, gTumor volume, mm3Relative tumor volumeTumor weight




CompoundDose, mg/kgAnimals, nInitialFinalInitialFinalx±SDTreated/control ratio, %x±SD, gInhibition, %
Control 5/5 21.8±0.8 26.0±1.2 237.5±32.6 1,163.8±503.2 5.06±2.46 1.02±0.46
Taxol     55/5 21.0±1.6 22.8±2.3 227.4±74.6 68.3±32.5a 0.34±0.22a6.75 0.04±0.02a96.36
T03  505/5 20.6±1.1 26.0±4.6 230.6±65.3 521.2±658.1 2.31±2.5045.63 0.44±0.27b57.09
1005/5 21.0±2.2 23.2±3.1 240.9±38.6 419.3±413.6b 1.65±1.55b32.55 0.38±0.4962.60

a P<0.01

b P<0.05 vs. control. x± SD, Means ± standard deviation.

Table VII.

Antitumor activity of compounds on the breast cancer MCF-7/T xenograft model.

Table VII.

Antitumor activity of compounds on the breast cancer MCF-7/T xenograft model.

Body weight, gTumor volume, mm3Relative tumor volumeTumor weight




CompoundDose, mg/kgAnimals, nInitialFinalInitialFinalx±SDTreated/control ratio, %x±SD, gInhibition, %
Control 5/5 18.0±1.4 18.6±2.4 112.0±5.8 1,373.5±363.1 12.19±2.78 1.26±0.24
Taxol     55/4 16.8±1.9 17.5±2.1 132.5±45.3 1,871.0±981.5 12.59±3.17 1.36±0.48
T03  505/5 17.4±1.2 17.4±2.5 142.4±38.7 621.2±243.3a 5.02±2.11a41.21 0.71±0.18a44.06
1005/5 17.3±1.1 15.6±2.7 148.1±37.1 442.6±266.0a 3.11±2.19a25.52 0.50±0.21b60.22

a P<0.01

b P<0.001 vs. control. x±SD, Means ± standard deviation.

T03 downregulated the Raf/MEK/ERK and PDGFRβ/Akt signaling pathway in MCF-7 and MCF-7/T xenograft nude model mice

Subsequently, whether T03 inactivated the above-mentioned kinase-associated signaling pathways was evaluated. As expected, the phosphorylated levels of PDGFRβ decreased in T03-treated tumors of MCF-7 and MCF-7/T xenografts (Fig. 4A and B). As PDK is an important downstream target of PDGFRβ/PI3K signaling and a key upstream kinase of the AKT/mTOR signaling pathway, its activation promotes proliferation and inhibits apoptosis in numerous human cancer types (9,22,23). The current study observed that the activity of PDK, AKT, and mTOR decreased in MCF-7 and MCF-7/T treated with T03 compared with the controls (Fig. 4A and B), indicating that T03 may downregulate PDGFRβ and PDK/AKT/mTOR and in MCF-7 and MCF-7/T xenograft models.

As T03 inhibited c-Raf in the current study, whether T03 downregulates the Ras/Raf/ERK signaling pathway in MCF-7 and MCF-7/T xenografts was examined. Fig. 4C and D demonstrate that T03 treatment significantly and dose-dependently decreased p-c-Raf, p-MEK, and p-ERK in MCF-7 and MCF-7/T tumors, while the basal levels of c-Raf, MEK and ERK were only reduced in the group treated with 100 mg/kg T03.

Furthermore, T03 downregulated Aurora A, a downstream effecter of the Ras/Raf/MEK/ERK signaling pathway (24), in MCF-7 and MCF-7/T xenografts. Aurora A and p-Aurora A were markedly downregulated upon T03 treatment, particularly in MCF-7 tumors, which were consistent with the previous study (24) that Aurora A and p-Aurora A were frequently activated by the Ras/Raf signaling pathway (Fig. 4E and F).

Discussion

Despite improvements in prevention, early detection, and treatment, breast cancer remains one of the most common malignant tumors affecting women in western countries (25). Drug-resistance of breast cancer to Taxol has limited its effect and application in clinical treatment. In the present study, the anti-tumor activity of the novel multi-kinases inhibitor, T03 was investigated, as well as its potential in breast cancer treatment.

As with Taxol, T03 displayed similar antitumor effects on MX-1 and MCF-7 breast cancer cells. Furthermore, T03 inhibited the growth of Taxol-resistant MX-1/T and MCF-7/T breast tumors in vitro and in vivo. It caused G2/M-phase cell accumulation and induced apoptosis, thus resulting in cell growth inhibition. In addition, T03 resulted in tumor regression in MX-1- and MCF-7-derived xenografts, as well as in Taxol-resistant MX-1/T and MCF-7/T tumors, indicating that T03 may exert effects on Taxol-sensitive and -resistant breast cancer cells. In addition, T03 was demonstrated to exert efficient effects on other types of drug-resistant breast cancer cells, such as Doxorubicin (Adriamycin)-resistant cells, which indicated that T03 may be used for treatment of other types of drug-resistant breast cancer.

Previous studies revealed that PDGFRβ was overexpressed in breast cancer (26,27). Highly activated PDGFRβ promoted tumor cell proliferation via PI3K/Akt and Ras/MEK/ERK signaling pathways (28,29), and resulted in distant metastasis and insensitivity to chemotherapy (30). The current study demonstrated that T03 may downregulate the Akt/mTOR and Ras/MEK/ERK signaling pathways, as well as PDGFRβ in MCF-7 and MCF-7/T in vivo. In MCF-7 and MCF-7/T xenograft tumors, T03 significantly reduced p-PDGFRβ, which was further confirmed by performance of the biochemical assay. Furthermore, T03 downregulated PDK, AKT and mTOR. As PDK-1, Akt and mTOR are the downstream components of PDGFRβ, and are involved in cell growth and apoptosis, T03 may cause cell growth inhibition and apoptosis via downregulation of the PDGFRβ/Akt signaling pathway. PDGFRβ activation is known to induce Taxol-resistance in breast cancer (31,32). Therefore, T03 may be used for inhibiting Taxol-resistant breast cancer.

Raf kinase is an upstream member of the Raf/MEK/ERK signaling cascade (33). Activation of the Raf/MEK/ERK signaling pathway has been associated with chemoresistance of breast cancer (3436). The present data indicated that T03 inhibited p-c-Raf, and consequently resulted in downregulation of p-MEK and p-ERK. These results indicate that T03 may inhibit the cell cycle and induce apoptosis via downregulation of the Raf/MEK/ERK signaling pathway in MCF-7 and MCF-7/T breast cancer.

Previous studies observed that activation of c-Raf signaling led to stabilization and accumulation of Aurora A mitotic kinase in breast cancer cells, and deduced that c-Raf may regulate the expression levels of Aurora A (24,37). The current study found that T03 treatment led to inhibition of c-Raf and decreased Aurora A in MCF-7 and MCF-7/T xenografts.

Previous studies have demonstrated that over-expressed Aurora A inhibits apoptosis, promotes cell cycle progression and metastasis, and mediates Taxol-resistance in breast cancer and other types of cancer (38,39). Inhibition of Aurora A by T03 may cause G2/M cell accumulation, apoptosis and sensitivity to Taxol in breast cancer. Based on the current results, the therapeutic efficacy of T03 on breast cancer may be partially attributed to the inhibition of c-Raf, PDGFRβ and the associated signaling pathways (40,41).

Although T03 presented similar anti-tumor activity in Taxol-sensitive and -resistant breast cancer, and Doxorubicin-resistant breast cancer as well, there are certain efficacy differences, which merit further investigation. In addition, based on our existing data, whether the anti-tumor effects are transient or permanent could not be determined.

In conclusion, the current study demonstrated that T03 induces cell cycle arrest and apoptosis, and inhibits cell proliferation in MX-1, MCF-7, MX-1/T, and MCF-7/T breast cancer in vitro and in vivo. These results demonstrate that T03 inhibits breast cancer growth by downregulating PDGFRβ/Akt and Ras/Raf/ERK signaling pathways, which are regulators of apoptosis, proliferation and chemoresistance. These findings indicate that T03 may be a potential candidate for effective chemotherapy of breast cancer, particularly for Taxol-resistant breast cancer.

Acknowledgements

The present study was financially supported by The National Natural Science Foundation of China (grant no. 81102025) and National S&T Major Special Project on ‘12·5 Major New Drug Innovation’ (grant no. 2012ZX09103-101-019).

References

1 

Reese JM, Suman VJ, Subramaniam M, Wu X, Negron V, Gingery A, Pitel KS, Shah SS, Cunliffe HE, McCullough AE, et al: ERβ1: Characterization, prognosis, and evaluation of treatment strategies in ERα-positive and -negative breast cancer. BMC Cancer. 14:7492014. View Article : Google Scholar : PubMed/NCBI

2 

Nahta R, Hortobágyi GN and Esteva FJ: Growth factor receptors in breast cancer: Potential for therapeutic intervention. Oncologist. 8:5–17. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Vlahovic G and Crawford J: Activation of tyrosine kinases in cancer. Oncologist. 8:531–538. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Niehoff P, Mundhenke C, Kimmig B and Maas N: Breast irradiation with brachytherapy: Approved techniques and new concepts. Minerva Ginecol. 59:377–386. 2007.PubMed/NCBI

5 

Hubbard SR: Juxtamembrane autoinhibition in receptor tyrosine kinases. Nat Rev Mol Cell Biol. 5:464–471. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Shchemelinin I, Sefc L and Necas E: Protein kinase inhibitors. Folia Biol (Praha). 52:137–148. 2006.PubMed/NCBI

7 

Weigel MT, Meinhold-Heerlein I, Bauerschlag DO, Schem C, Bauer M, Jonat W, Maass N and Mundhenke C: Combination of imatinib and vinorelbine enhances cell growth inhibition in breast cancer cells via PDGFR beta signalling. Cancer Lett. 273:70–79. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Weigel MT, Banerjee S, Arnedos M, Salter J, A'Hern R, Dowsett M and Martin LA: Enhanced expression of the PDGFR/Abl signaling pathway in aromatase inhibitor-resistant breast cancer. Ann Oncol. 24:126–133. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Lin HJ, Hsieh FC, Song H and Lin J: Elevated phosphorylation and activation of PDK-1/AKT pathway in human breast cancer. Br J Cancer. 93:1372–1381. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Weigel MT, Dahmke L, Schem C, Bauerschlag DO, Weber K, Niehoff P, Bauer M, Strauss A, Jonat W, Maass N and Mundhenke C: In vitro effects of imatinib mesylate on radiosensitivity and chemosensitivity of breast cancer cells. BMC Cancer. 10:4122010. View Article : Google Scholar : PubMed/NCBI

11 

Sridhar SS, Hedley D and Siu LL: Raf kinase as a target for anticancer therapeutics. Mol Cancer Ther. 4:677–685. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Li Z, Min W and Gou J: Knockdown of cyclophilin A reverses paclitaxel resistance in human endometrial cancer cells via suppression of MAPK kinase pathways. Cancer Chemother Pharmacol. 72:1001–1011. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Gong Y, He H, Liu H, Zhang C, Zhao W and Shao RG: Phosphorylation of myofibrillogenesis regulator-1 activates the MAPK signaling pathway and induces proliferation and migration in human breast cancer MCF7 cells. FEBS Lett. 588:2903–2910. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Hashimoto K, Tsuda H, Koizumi F, Shimizu C, Yonemori K, Ando M, Kodaira M, Yunokawa M, Fujiwara Y and Tamura K: Activated PI3K/AKT and MAPK pathways are potential good prognostic markers in node-positive, triple-negative breast cancer. Ann Oncol. 25:1973–1979. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Heckler MM, Thakor H, Schafer CC and Riggins RB: ERK/MAPK regulates ERRγ expression, transcriptional activity and receptor-mediated tamoxifen resistance in ER+ breast cancer. FEBS J. 281:2431–2442. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Im NK, Jang WJ, Jeong CH and Jeong GS: Delphinidin suppresses PMA-induced MMP-9 expression by blocking the NF-κB activation through MAPK signaling pathways in MCF-7 human breast carcinoma cells. J Med Food. 17:855–861. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, Dancey JE and Piccart-Gebhart MJ: Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 39:935–946. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Lu J, Tan M, Huang WC, Li P, Guo H, Tseng LM, Su XH, Yang WT, Treekitkarnmongkol W, Andreeff M, et al: Mitotic deregulation by survivin in ErbB2-overexpressing breast cancer cells contributes to Taxol resistance. Clin Cancer Res. 15:1326–3134. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Li Y, Tang K, Zhang H, Zhang Y, Zhou W and Chen X: Function of Aurora kinase A in Taxol-resistant breast cancer and its correlation with P-gp. Mol Med Rep. 4:739–746. 2011.PubMed/NCBI

20 

Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, et al: BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 64:7099–7109. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Li Y, Zhang ZF, Chen J, Huang D, Ding Y, Tan MH, Qian CN, Resau JH, Kim H and Teh BT: VX680/MK-0457, a potent and selective Aurora kinase inhibitor, targets both tumor and endothelial cells in clear cell renal cell carcinoma. Am J Transl Res. 2:296–308. 2010.PubMed/NCBI

22 

Reynolds TH IV, Bodine SC and Lawrence JC Jr: Control of Ser2448 phosphorylation in the mammalian target of rapamycin by insulin and skeletal muscle load. J Biol Chem. 277:17657–17662. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Salmond RJ, Emery J, Okkenhaug K and Zamoyska R: MAPK, phosphatidylinositol 3-kinase, and mammalian target of rapamycin pathways converge at the level of ribosomal protein S6 phosphorylation to control metabolic signaling in CD8 T cells. J Immunol. 183:7388–7397. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Biran A, Brownstein M, Haklai R and Kloog Y: Downregulation of survivin and aurora A by histone deacetylase and RAS inhibitors: A new drug combination for cancer therapy. Int J Cancer. 128:691–701. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Kim BK, Lee JW, Park PJ, Shin YS, Lee WY, Lee KA, Ye S, Hyun H, Kang KN, Yeo D, et al: The multiplex bead array approach to identifying serum biomarkers associated with breast cancer. Breast Cancer Res. 11:R222009. View Article : Google Scholar : PubMed/NCBI

26 

de Jong JS, van Diest PJ, van der Valk P and Baak JP: Expression of growth factors, growth-inhibiting factors, and their receptors in invasive breast cancer. II: Correlations with proliferation and angiogenesis. J Pathol. 184:53–57. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Lev DC, Kim SJ, Onn A, Stone V, Nam DH, Yazici S, Fidler IJ and Price JE: Inhibition of platelet-derived growth factor receptor signaling restricts the growth of human breast cancer in the bone of nude mice. Clin Cancer Res. 11:306–314. 2005.PubMed/NCBI

28 

Klos KS, Wyszomierski SL, Sun M, Tan M, Zhou X, Li P, Yang W, Yin G, Hittelman WN and Yu D: ErbB2 increases vascular endothelial growth factor protein synthesis via activation of mammalian target of rapamycin/p70S6K leading to increased angiogenesis and spontaneous metastasis of human breast cancer cells. Cancer Res. 66:2028–2037. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Wen XF, Yang G, Mao W, Thornton A, Liu J, Bast RC Jr and Le XF: HER2 signaling modulates the equilibrium between pro- and antiangiogenic factors via distinct pathways: Implications for HER2-targeted antibody therapy. Oncogene. 25:6986–6996. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Seymour L and Bezwoda WR: Positive immunostaining for platelet derived growth factor (PDGF) is an adverse prognostic factor in patients with advanced breast cancer. Breast Cancer Res Treat. 32:229–233. 1994. View Article : Google Scholar : PubMed/NCBI

31 

Blagosklonny MV and Fojo T: Molecular effects of paclitaxel: Myths and reality (a critical review). Int J Cancer. 83:151–156. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Langley RR, Fan D, Tsan RZ, Rebhun R, He J, Kim SJ and Fidler IJ: Activation of the platelet-derived growth factor-receptor enhances survival of murine bone endothelial cells. Cancer Res. 64:3727–3730. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Sambade MJ, Camp JT, Kimple RJ, Sartor CI and Shields JM: Mechanism of lapatinib-mediated radiosensitization of breast cancer cells is primarily by inhibition of the Raf>MEK>ERK mitogen-activated protein kinase cascade and radiosensitization of lapatinib-resistant cells restored by direct inhibition of MEK. Radiother Oncol 93: 639–644, 2009. Radiother Oncol 93: 639–644, 2009. 93: 639–644, 2009:639-644, 2009–644, 2009. 2009.

34 

Frogne T, Benjaminsen RV, Sonne-Hansen K, Sorensen BS, Nexo E, Laenkholm AV, Rasmussen LM, Riese DJ II, de Cremoux P, Stenvang J and Lykkesfeldt AE: Activation of ErbB3, EGFR and Erk is essential for growth of human breast cancer cell lines with acquired resistance to fulvestrant. Breast Cancer Res Treat. 114:263–275. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Lian WJ, Liu G, Liu YJ, Zhao ZW, Yi T and Zhou HY: Downregulation of BMP6 enhances cell proliferation and chemoresistance via activation of the ERK signaling pathway in breast cancer. Oncol Rep. 30:193–200. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Sokolosky M, Chappell WH, Stadelman K, Abrams SL, Davis NM, Steelman LS and McCubrey JA: Inhibition of GSK-3β activity can result in drug and hormonal resistance and alter sensitivity to targeted therapy in MCF-7 breast cancer cells. Cell Cycle. 13:820–833. 2014. View Article : Google Scholar : PubMed/NCBI

37 

D'Assoro AB, Liu T, Quatraro C, Amato A, Opyrchal M, Leontovich A, Ikeda Y, Ohmine S, Lingle W, Suman V, et al: The mitotic kinase Aurora-a promotes distant metastases by inducing epithelial-to-mesenchymal transition in ERα(+) breast cancer cells. Oncogene. 33:599–610. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Anand S, Penrhyn-Lowe S and Venkitaraman AR: AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. Cancer Cell. 3:51–62. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Kwiatkowski N, Deng X, Wang J, Tan L, Villa F, Santaguida S, Huang HC, Mitchison T, Musacchio A and Gray N: Selective aurora kinase inhibitors identified using a taxol-induced checkpoint sensitivity screen. ACS Chem Biol. 7:185–196. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Horowitz JC, Lee DY, Waghray M, Keshamouni VG, Thomas PE, Zhang H, Cui Z and Thannickal VJ: Activation of the pro-survival phosphatidylinositol 3-kinase/AKT pathway by transforming growth factor-beta1 in mesenchymal cells is mediated by p38 MAPK-dependent induction of an autocrine growth factor. J Biol Chem. 279:1359–1367. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Arany I, Faisal A, Nagamine Y and Safirstein RL: p66shc inhibits pro-survival epidermal growth factor receptor/ERK signaling during severe oxidative stress in mouse renal proximal tubule cells. J Biol Chem. 283:6110–6117. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Y, Liu C, Tang K, Chen Y, Tian K, Feng Z and Chen J: Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer. Mol Med Rep 17: 2373-2383, 2018
APA
Li, Y., Liu, C., Tang, K., Chen, Y., Tian, K., Feng, Z., & Chen, J. (2018). Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer. Molecular Medicine Reports, 17, 2373-2383. https://doi.org/10.3892/mmr.2017.8179
MLA
Li, Y., Liu, C., Tang, K., Chen, Y., Tian, K., Feng, Z., Chen, J."Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer". Molecular Medicine Reports 17.2 (2018): 2373-2383.
Chicago
Li, Y., Liu, C., Tang, K., Chen, Y., Tian, K., Feng, Z., Chen, J."Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer". Molecular Medicine Reports 17, no. 2 (2018): 2373-2383. https://doi.org/10.3892/mmr.2017.8179