Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells

  • Authors:
    • Yu‑Chang Liu
    • Reng‑Hong Wu
    • Wei‑Shu Wang
  • View Affiliations

  • Published online on: May 9, 2017     https://doi.org/10.3892/ol.2017.6142
  • Pages: 461-467
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of regorafenib on the nuclear factor κ‑light‑chain‑enhancer of activated B cells (NF)‑κB‑modulated expression of angiogenesis‑ and metastasis‑associated proteins and cell invasion in human hepatocellular carcinoma SK‑Hep1 cells. The SK‑Hep1 cells were treated with different concentrations of NF‑κB inhibitor 4‑N‑[2‑(4‑phenoxyphenyl) ethyl] quinazoline‑4,6‑diamine (QNZ) or regorafenib for 24 or 48 h. The effects of QNZ and regorafenib on cell viability, NF‑κB activation, expression and secretion levels of angiogenesis‑ and metastasis‑associated proteins and cell invasion were evaluated with MTT assays, western blotting, ELISA, gelatin zymography and cell invasion assays. The results demonstrated that QNZ and regorafenib significantly reduced the expression and secretion levels of the angiogenesis‑ and metastasis‑associated proteins vascular endothelial growth factor, tumor necrosis factor‑α, interleukin (IL)‑1β, IL‑6, matrix metalloproteinase (MMP)‑2 and MMP‑9, NF‑κB activation and cell invasion. In conclusion, the inhibition of NF‑κB activation induces anti‑angiogenic and antimetastatic effects in SK‑Hep1 cells. Regorafenib reduces the level of expression and secretion of angiogenesis‑ and metastasis‑associated proteins and cell invasion through the suppression of NF‑κB activation in SK‑Hep1 cells.

Introduction

Human hepatocellular carcinoma (HCC) has the highest mortality rate worldwide (1). Angiogenesis, the development of new blood vessels, is a prerequisite for tumor growth and metastasis in HCC (2). Tumor angiogenesis is modulated by angiogenic factors including vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6. The expression of VEGF, a major mediator of angiogenesis, is upregulated by the stimulation of inflammatory cytokines, such as TNF-α, IL-1β and IL-6. The overexpression of angiogenic factors is exhibited in and associated with poor prognoses in HCC (26).

Metastasis is the major cause of mortality in patients with cancer (7). Matrix metalloproteinases-2 (MMP-2) and MMP-9, which represent 72 kD and 92 kD type IV collagenases, respectively, degrade the basement membrane of normal cells and result in tumor invasion and subsequent metastasis. The detection of high expression levels of MMP-2 and MMP-9 in HCC has been suggested as a biomarker for poor prognosis (8,9).

Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), which is assembled via the dimerization of five subunits: RELA p65; RELB; NF-κB 1 p50; NF-κB 2 p52; and c-Rel, serves an important role in immunity, inflammation and tumorigenesis (10). The NF-κB p50/p65 heterodimer is associated with tumorigenesis and is generally sequestered in the cytoplasm by an inhibitor of κBα, IκBα. Activators of NF-κB induce the phosphorylation and degradation of IκBα through IκB kinase and 26S proteasome, respectively, resulting in the NF-κB nuclear translocation and expression of NF-κB target oncogenes (11). A number of angiogenesis- and metastasis-associated proteins are encoded by NF-κB target oncogenes in HCC cells (12). The inhibition of NF-κB activation induces anti-angiogenic and antimetastatic effects via the suppression of the NF-κB signal cascade (13). NF-κB was revealed to be more highly expressed in the nucleoli of HCC cells compared with normal hepatic tissue, which was associated with poor prognoses (14). However, a number of therapeutic agents, and radiation, induce apoptosis signal and trigger NF-κB activity in HCC in vitro and in vivo (13,15,16). Therefore, the development of novel inhibitors of NF-κB signaling may be useful for preventing angiogenesis and metastasis in patients with HCC.

Regorafenib, or Stivarga®, a novel oral multiple kinase inhibitor, is a member of the group of biaryl urea compounds and is similar to sorafenib in chemical structure. The addition of fluorine to the center of the phenyl group means that regorafenib may exhibit greater range of activity against oncogenic receptor tyrosine kinases and intracellular signaling kinases compared with sorafenib (17). Regorafenib has been approved for the treatment of metastatic colorectal cancer and advanced gastrointestinal stromal tumors. A randomized double-blinded phase III study of regorafenib in patients with HCC who have progressed after sorafenib treatment is ongoing (18). In our previous studies, sorafenib was revealed to be an inhibitor of NF-κB signaling and reduced NF-κB-modulated expression of proteins including MMP-9 and VEGF in HCC in vitro and in vivo (13,19). However, whether regorafenib, a sorafenib derivative, induces anti-angiogenic and antimetastatic effects through the suppression of NF-κB activation in HCC cells remains unknown. The aim of the present study was to investigate the effects of regorafenib on NF-κB-modulated expression of angiogenesis- and metastasis-associated proteins and cell invasion in HCC SK-Hep1 cells by using western blotting, ELISA, gelatin zymography and Matrigel invasion assays. The effects of NF-κB inactivation on the expression of angiogenesis- and metastasis-associated proteins and cell invasion in SK-Hep1 cells were also evaluated.

Materials and methods

Chemicals

Regorafenib was provided by Bayer Corporation (Whippany, NJ, USA). Dulbecco's modified Eagle medium (DMEM), fetal bovine serum (FBS), L-glutamine, and penicillin-streptomycin (PS) were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). TNF-α, IL-1β and IL-6 ELISA kits and NF-κB inhibitor QNZ were purchased from eBioscience, Inc. (San Diego, CA, USA) and Apexbio Technology LLC (Houston, TX, USA), respectively. Matrigel and Transwell (8-µm pore size) were obtained from Selleck Chemicals (Houston, TX, USA) and Corning Life Sciences (Tewksbury, MA, USA), respectively. Primary antibodies for β-actin and TNF-α were obtained from Thermo Fischer Scientific, Inc. (Waltham, MA, USA). Primary antibodies for NF-κB p65 and IL-1β were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Primary antibodies for MMP-9 and VEGF were purchased from EMD Millipore (Billerica, MA, USA). Primary antibodies for MMP-2, IL-6, and NF-κB p65 were purchased from OriGene Technologies, Inc., (Rockville, MD, USA), Abbiotec LLC (San Diego, CA, USA), and Santa Cruz Biotechnology (Dallas, TX, USA). Secondary antibodies were bought from Jackson ImmunoResearch Laboratoires, Inc. (West Grove, PA, USA).

Cell culture

Hepatocellular carcinoma SK-Hep1 cells were used in the present study. The SK-Hep1 cells were provided by Professor Jing-Gung Chung from the Department of Biological Science and Technology, China Medical University, Taichung, Taiwan. The cells were cultured in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/ml penicillin, and 100 µg/ml streptomycin and maintained at 37°C and 5% CO2 in a humidified incubator (1).

MTT assay

The SK-Hep1 cells were seeded into 96-well plates with 2×104 cells/well and incubated at room temperature overnight, then treated with regorafenib (0, 5, 10, 15, 20 and 25 µM) in 0.1% DMSO or QNZ (0, 0.05, 0.1, 0.2 and 0.4 µM) in 0.1% dimethyl sulphoxide for 12, 24, and 48 h. The effects of regorafenib and QNZ on cell viability were analyzed by MTT assay as described previously (16).

Western blotting

A total of 2×106 SK-Hep1 cells were incubated at room temperature in a 10-cm diameter dish overnight, then treated with 20 µM regorafenib or 0.1 µM QNZ for 12 or 24 h. The total cellular proteins in all treatment groups were extracted using 1 mM phenylmethanesulfonyl fluoride, 0.5% NP-40, 120 mM NaCl, and 50 mM Tris-HCl pH 8.0 lysis buffer. The protein expressions of VEGF, MMP-2, MMP-9, IL-1β, IL-6, TNF-α, pNF-κB, and NF-κB were evaluated with western blotting assays as described by Wang et al (20). The protein expression was quantified by densitometry using ImageJ software version 1.50 (National Institutes of Health, Bethesda, MD, USA) in order to determine the density of regions of interest. β-actin was used as the internal control.

ELISA

The SK-Hep1 cells were seeded into 12-well plates with 1×105 cells/well and incubated overnight at room temperature, then treated with 20 µM regorafenib or 0.1 µM QNZ for 12 h. ELISA kits were used to measure the levels of IL-1β, IL-6 and TNF-α secreted by the cells in media. The procedure followed the protocols provided by manufacturer.

Gelatin zymography assay

The SK-Hep1 cells were cultured in 6-well plates with 1×106 cells/well and incubated overnight at room temperature, then treated with 20 µM regorafenib or 0.1 µM QNZ for 12 h. The secretion of active MMP-2 and MMP-9 from the cells in medium was analyzed with gelatin zymography assays as described previously (19). ImageJ software version 1.50 was used to quantify the bands of MMP-2 and MMP-9 activity.

Cell invasion assay

Matrigel-coated Transwell chambers were used to measure cell invasion. The 8-µm pore sized Transwell chambers were inserted into 96-well plates and coated with 50 ml Matrigel, then incubated in 5% CO2 at 37°C for 1 h. The SK-hep1 cells in serum-free DMEM were treated with 20 µM regorafenib or 0.1 µM QNZ and 2×105 cells/well were added in the apical chamber of the Transwell, followed by the addition of DMEM supplemented with 10% FBS to the basolateral chamber and subsequent incubation for 12 h. Sterile cotton swabs were used to remove the Matrigel in the Transwell chamber. The invasive cells in the bottom of the Transwell were fixed with a mixture of 3:1 methanol and acetic acid and stained with 0.5% crystal violet for 15 min and then were counted under a light microscope at magnification, ×100 (21).

Statistical analysis

The significance of difference between treatment group and control was verified by Student's t-test. Data were presented as the mean ± standard error. P<0.05 was considered to indicate a statistically significant difference. All experiments were performed three times.

Results

NF-κB inhibitor reduces cell viability and expression of angiogenesis and metastasis associated proteins in SK-Hep1 cells

To confirm the effects of NF-κB inactivation on the expression of angiogenesis- and metastasis-associated proteins, the SK-Hep1 cells were treated with a range of concentrations of QNZ for various time intervals. The effects of QNZ on cell viability, expression of angiogenesis- and metastasis-associated proteins, and NF-κB p65 in the SK-Hep1 cells were evaluated with an MTT assay and western blotting. Fig. 1A illustrates that the cell viability significantly reduced as the QNZ concentration and incubation time increased compared with the control. Fig. 1B indicates that the levels of expression of angiogenesis- and metastasis-associated proteins, along with NF-κB p65, were significantly inhibited by QNZ in a time-dependent manner.

Regorafenib induces cytotoxicity and inhibits expression of NF-κB-modulated angiogenesis- and metastasis-associated proteins in SK-Hep1 cells

The present study suggested that regorafenib affects the levels of expression of NF-κB-modulated angiogenesis- and metastasis-associated proteins by treating SK-Hep1 cells with different concentration of regorafenib for various time intervals. The effects of regorafenib on the cell viability and expression levels of the angiogenesis- and metastasis-associated proteins and NF-κB p65 in SK-Hep1 cells were evaluated with MTT assay and western blotting. Fig. 2A indicates that cell viability was significantly decreased by regorafenib in a dose- and time-dependent manner. Regorafenib significantly reduced expression of angiogenesis- and metastasis-associated proteins along with NF-κB p65 in a time dependent manner by 20–80% compared with the control (Fig. 2B).

Regorafenib and QNZ reduce secretion of angiogenesis associated cytokines in SK-Hep1 cells

ELISA was used to evaluate the effects of regorafenib and QNZ on the secretion of angiogenesis-associated cytokines TNF-α, IL-1β and IL-6 in the SK-Hep1 cells. TNF-α, IL-1β, and IL-6 secretion were significantly inhibited by 5–55% compared with the control, as demonstrated in Fig. 3A-C with unchanged cell viability, as illustrated in Figs. 1A and 2A, when the cells were treated with 0.1 µM QNZ or 20 µM regorafenib for 12 h.

Regorafenib and QNZ inhibit secretion of metastasis-associated proteins and cell invasion in SK-Hep1 cells

The effect of regorafenib and QNZ on the secretion of active MMP-2 and MMP-9 and cell invasion in the SK-Hep1 cells was investigated using gelatin zymography and cell invasion assays. Regorafenib and QNZ reduced the protein levels of MMP-2 and MMP-9, as demonstrated in Figs. 1B and 2B, and inhibited the secretion of active MMP-2 and MMP-9, as illustrated in Fig. 4A. It was also revealed that regorafenib and QNZ significantly decreased the number of cells that invaded the Matrigel by 50–70% compared with the control, as demonstrated in Fig. 4B.

Discussion

Regorafenib, a novel sorafenib derivative, has been approved by the Food and Drug Administration (FDA) for the treatment of metastatic colorectal cancer and advanced gastrointestinal stromal tumors (18). Our previous studies indicated that sorafenib, as an NF-κB signal inhibitor, inhibits the vorinostat- and 12-O-tetradecanoylphorbol-13-acetate-induced expression of angiogenesis- and metastasis-associated proteins through the inhibition of NF-κB activation in HCC cells in vitro and in vivo (13,19). However, whether regorafenib triggers anti-angiogenic and antimetastatic effects via the inhibition of NF-κB activation in HCC cells remains unknown. Therefore, in the present study, the effects of regorafenib on the levels of expression of NF-κB-modulated angiogenesis- and metastasis-associated proteins and cell invasion in HCC cells were investigated in vitro. It was revealed that QNZ, a specific NF-κB inhibitor, reduces the cell viability and expression levels of angiogenesis- and metastasis-associated proteins VEGF, TNF-α, IL-1β, IL-6, MMP-2, MMP-9 and NF-κB p65. Additionally, it was identified that regorafenib induces cytotoxicity and inhibits the expression of angiogenesis- and metastasis-associated proteins VEGF, TNF-α, IL-1β, IL-6, MMP-2, MMP-9 and NF-κB p65. Regorafenib and QNZ significantly decrease the levels of cytokines TNF-α, IL-1β and IL-6 secreted by the cells in media. Finally, regorafenib and QNZ also significantly decrease the secretion of active MMP-2 and MMP-9 and the number of invading cells.

Pikarsky et al (22) suggested that NF-κB activation is essential for promoting inflammation-associated HCC. NF-κB signaling has been identified as a biomarker for cancer that is indicative of a self-sufficiency in proliferative growth signals, an insensitivity to growth inhibitory signals, an ability to evade apoptosis, an unlimited replicative potential and the induction of angiogenesis and metastasis by modulating the levels of expression of effector proteins (6,23,24). The inhibition of NF-κB signaling reduces the expression of angiogenesis- and metastasis-associated proteins and results in the suppression of tumor progression (13,23,24). Our previous studies revealed that the transfection of inhibitor of nuclear factor κBβ mutant vector, a super repressor of NF-κB activation, decreased the protein expression of MMP-9 and VEGF in HCC and oral cancer cells (19,25). Huang et al (26) demonstrated that the inhibition of NF-κB activation is associated with the suppression of angiogenesis, invasion, and metastasis in prostate cancer cells. In the present study, QNZ, a specific inhibitor of NF-κB activation, was used to investigate the effects of NF-κB inactivation on the levels of angiogenesis- and metastasis associated-proteins and cell invasion in SK-Hep1 cells. It was demonstrated that QNZ significantly decreases the expression levels and secretion of angiogenesis- and metastasis-associated proteins and cell invasion in the SK-Hep1 cells. Baud and Karin (23) suggested that NF-κB signaling may be a potential molecular target for cancer therapy and the development of the NF-κB pathway inhibitors may offer an effective treatment approach for certain types of cancer.

Constitutive NF-κB activation was demonstrated in HCC and used as a biomarker for poor prognosis. Jin et al (14) used immunohistochemistry (IHC) to evaluate the expression and location of NF-κB proteins in HCC and surrounding normal hepatic tissues. It was identified that NF-κB nuclear translocation in HCC was significantly increased compared with adjacent normal liver tissue. In addition, NF-κB nuclear translocation was significantly higher in patients with HCC exhibiting adverse clinical features including advanced staging, portal vein tumor thrombus, and the involvement of regional lymph nodes compared with other patients with early-stage disease. Chemotherapeutic agents and radiation are able to trigger the NF-κB-modulated expression of angiogenesis- and metastasis-associated proteins in HCC cells via the activation of NF-κB signaling (15,16). Sorafenib, a multiple kinase inhibitor, has been approved by the FDA for the treatment of advanced HCC (13). Previous studies have demonstrated that sorafenib inhibits endogenous NF-κB activity and reverses the anticancer drug and carcinogen-induced NF-κB activity with the inhibition of MMP-9 and VEGF protein expression in HCC in vitro and in vivo (13,19). Carr et al (27) suggested that regorafenib, or fluoro-sorafenib, inhibits tumor growth via the induction of caspase-dependent apoptosis in HCC cells. However, the effect of regorafenib on NF-κB-modulated angiogenesis and metastasis in HCC cells remains unknown.

The main purpose of the present study was to investigate if regorafenib affects the NF-κB-modulated expression of angiogenesis- and metastasis-associated proteins and cell invasion in SK-Hep1 cells. The role of NF-κB inactivation on the anti-angiogenic and antimetastatic effects was investigated using QNZ. Based on the data from previous studies, it was suggested that the blockage of NF-κB activation triggers anti-angiogenic and antimetastatic effects in HCC cells. Then, regorafenib was used and revealed similar findings to the experiments with QNZ. Regorafenib significantly decreased the level of NF-κB activation and the levels of expression of angiogenesis- and metastasis-associated proteins. The secretion of inflammatory cytokines TNF-α, L-1β and IL-6 were significantly decreased by regorafenib treatment. Regorafenib suppressed the secretion of active MMP-2 and MMP-9 and the number of cells invaded in SK-Hep1 cells. The effects of regorafenib on NF-κB activation in HCC may be different in other types of cancer cells. Chen et al (28) revealed that regorafenib inhibits colorectal tumor growth through the activation of the NF-κB-modulated protein expression of p53 upregulated modulator of apoptosis. Regorafenib is hypothesized to induce anti-angiogenic and antimetastatic effects through the suppression of NF-κB activation in SK-Hep 1 cells in vitro.

In conclusion, the present study demonstrated that regorafenib reduced the expression and secretion of the angiogenesis- and metastasis-associated proteins VEGF, TNF-α, IL-1β, IL-6, MMP-2 and MMP-9, and cell invasion through the inhibition of NF-κB activation. Therefore, regorafenib may exhibit a potential clinical application as an NF-κB signal inhibitor to limit angiogenesis and metastasis in HCC.

Acknowledgements

The present study was supported by grants from National Yang-Ming University Hospital, Yilan, Taiwan (grant no. RD2016-020) and Chi Mei Medical Center, Tainan, Taiwan (grant no. CTU102-CM-002). The present authors thank Professor Song-Shei Lin of Department of Radiological Technology, Central Taiwan University of Science and Technology, Taichung, for excellent technical assistance.

References

1 

Ma CY, Ji WT, Chueh FS, Yang JS, Chen PY, Yu CC and Chung JG: Butein inhibits the migration and invasion of SK-HEP-1 human hepatocarcinoma cells through suppressing the ERK, JNK, p38 and uPA signaling multiple pathways. J Agric Food Chem. 59:9032–9038. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Zhan P, Qian Q and Yu LK: Serum VEGF level is associated with the outcome of patients with hepatocellular carcinoma: A meta-analysis. Hepatobiliary Surg Nutr. 2:209–215. 2013.PubMed/NCBI

3 

Nakazaki H: Preoperative and postoperative cytokines in patients with cancer. Cancer. 70:709–713. 1992. View Article : Google Scholar : PubMed/NCBI

4 

Okamoto K, Ishida C, Ikebuchi Y, Mandai M, Mimura K, Murawaki Y and Yuasa I: The genotypes of IL-1 beta and MMP-3 are associated with the prognosis of HCV-related hepatocellular carcinoma. Intern Med. 49:887–895. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Jang JW, Oh BS, Kwon JH, You CR, Chung KW, Kay CS and Jung HS: Serum interleukin-6 and C-reactive protein as a prognostic indicator in hepatocellular carcinoma. Cytokine. 60:686–693. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Naugler WE and Karin M: NF-kappaB and cancer-identifying targets and mechanisms. Curr Opin Genet Dev. 18:19–26. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Mehlen P and Puisieux A: Metastasis: A question of life or death. Nat Rev Cancer. 6:449–458. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Daniele A, Divella R, Quaranta M, Mattioli V, Casamassima P, Paradiso A, Garrisi VM, Gadaleta CD, Gadaleta-Caldarola G, Savino E, et al: Clinical and prognostic role of circulating MMP-2 and its inhibitor TIMP-2 in HCC patients prior to and after transhepatic arterial chemo-embolization. Clin Biochem. 47:184–190. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Chen R, Cui J, Xu C, Xue T, Guo K, Gao D, Liu Y, Ye S and Ren Z: The significance of MMP-9 over MMP-2 in HCC invasiveness and recurrence of hepatocellular carcinoma after curative resection. Ann Surg Oncol. 19 Suppl 3:S375–S384. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Liu YC, Chiang IT, Hsu FT and Hwang JJ: Using NF-κB as a molecular target for theranostics in radiation oncology research. Expert Rev Mol Diagn. 12:139–146. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Chen YJ, Yeh MH, Yu MC, Wei YL, Chen WS, Chen JY, Shih CY, Tu CY, Chen CH, Hsia TC, et al: Lapatinib-induced NF-kappaB activation sensitizes triple-negative breast cancer cells to proteasome inhibitors. Breast Cancer Res. 15:R1082013. View Article : Google Scholar : PubMed/NCBI

12 

Wu JM, Sheng H, Saxena R, Skill NJ, Bhat-Nakshatri P, Yu M, Nakshatri H and Maluccio MA: NF-kappaB inhibition in human hepatocellular carcinoma and its potential as adjunct to sorafenib based therapy. Cancer Lett. 278:145–155. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Hsu FT, Liu YC, Chiang IT, Liu RS, Wang HE, Lin WJ and Hwang JJ: Sorafenib increases efficacy of vorinostat against human hepatocellular carcinoma through transduction inhibition of vorinostat-induced ERK/NF-κB signaling. Int J Oncol. 45:177–188. 2014.PubMed/NCBI

14 

Jin Y, Chen J, Feng Z, Fan W, Wang Y, Li J and Tong D: The expression of Survivin and NF-κB associated with prognostically worse clinicopathologic variables in hepatocellular carcinoma. Tumour Biol. 35:9905–9910. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Tietze MK, Wuestefeld T, Paul Y, Zender L, Trautwein C, Manns MP and Kubicka S: IkappaBalpha gene therapy in tumor necrosis factor-alpha- and chemotherapy-mediated apoptosis of hepatocellular carcinomas. Cancer Gene Ther. 7:1315–1323. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Hsu FT, Liu YC, Liu TT and Hwang JJ: Curcumin sensitizes hepatocellular carcinoma cells to radiation via suppression of radiation-induced NF-κB activity. Biomed Res Int. 2015:363–671. 2015. View Article : Google Scholar

17 

Ravi S and Singal AK: Regorafenib: An evidence-based review of its potential in patients with advanced liver cancer. Core Evid. 17:81–87. 2014.

18 

Tai WT, Chu PY, Shiau CW, Chen YL, Li YS, Hung MH, Chen LJ, Chen PL, Su JC, Lin PY, et al: STAT3 mediates regorafenib-induced apoptosis in hepatocellular carcinoma. Clin Cancer Res. 20:5768–5776. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Chiang IT, Liu YC, Wang WH, Hsu FT, Chen HW, Lin WJ, Chang WY and Hwang JJ: Sorafenib inhibits TPA-induced MMP-9 and VEGF expression via suppression of ERK/NF-kB pathway in hepatocellular carcinoma cells. In Vivo. 26:671–681. 2012.PubMed/NCBI

20 

Wang WH, Chiang IT, Ding K, Chung JG, Lin WJ, Lin SS and Hwang JJ: Curcumin-induced apoptosis in human hepatocellular carcinoma j5 cells: Critical role of ca(+2)-dependent pathway. Evid Based Complement Alternat Med. 2012:5129072012.PubMed/NCBI

21 

Liao CL, Lai KC, Huang AC, Yang JS, Lin JJ, Wu SH, Wood W Gibson, Lin JG and Chung JG: Gallic acid inhibits migration and invasion in human osteosarcoma U-2 OS cells through suppressing the matrix metalloproteinase-2/−9, protein kinase B (PKB) and PKC signaling pathways. Food Chem Toxicol. 50:1734–1740. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E and Ben-Neriah Y: NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature. 431:461–466. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Baud V and Karin M: Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov. 8:33–40. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Pacifico F and Leonardi A: NF-kappaB in solid tumors. Biochem Pharmacol. 72:1142–1152. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Chiang IT, Liu YC, Hsu FT, Chien YC, Kao CH, Lin WJ, Chung JG and Hwang JJ: Curcumin synergistically enhances the radiosensitivity of human oral squamous cell carcinoma via suppression of radiation-induced NF-κB activity. Oncol Rep. 4:1729–1737. 2014.

26 

Huang S, Pettaway CA, Uehara H, Bucana CD and Fidler IJ: Blockade of NF-kappaB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene. 20:4188–4197. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Carr BI, Cavallini A, Lippolis C, D'Alessandro R, Messa C, Refolo MG and Tafaro A: Fluoro-Sorafenib (Regorafenib) effects on hepatoma cells: Growth inhibition, quiescence, and recovery. J Cell Physiol. 228:292–297. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Chen D, Wei L, Yu J and Zhang L: Regorafenib inhibits colorectal tumor growth through PUMA-mediated apoptosis. Clin Cancer Res. 20:3472–3484. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 14 Issue 1

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
Liu YC, Wu RH and Wang WS: Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells. Oncol Lett 14: 461-467, 2017
APA
Liu, Y., Wu, R., & Wang, W. (2017). Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells. Oncology Letters, 14, 461-467. https://doi.org/10.3892/ol.2017.6142
MLA
Liu, Y., Wu, R., Wang, W."Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells". Oncology Letters 14.1 (2017): 461-467.
Chicago
Liu, Y., Wu, R., Wang, W."Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells". Oncology Letters 14, no. 1 (2017): 461-467. https://doi.org/10.3892/ol.2017.6142