Open Access

Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line

  • Authors:
    • Yongxi Tong
    • Mingshan Wang
    • Haijun Huang
    • Jiajie Zhang
    • Yicheng Huang
    • Yingjun Chen
    • Hongying Pan
  • View Affiliations

  • Published online on: September 19, 2019     https://doi.org/10.3892/etm.2019.8027
  • Pages: 3793-3800
  • Copyright: © Tong 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

Combination therapy is an important method for treating advanced hepatocellular carcinoma (HCC). Gefitinib is an epidermal growth factor receptor (EGFR) inhibitor, which has profound effects on HCC. The purpose of the present study was to investigate the effects of genistein in combination with gefitinib on the proliferation and apoptosis of HCC cells and the associated mechanism. Cell counting kit‑8 assay was performed to calculate the IC50 values and cytotoxicity, whilst flow cytometry was used to assess cell apoptosis. Protein expression was detected using western blot analysis. The IC50 of genistein and gefitinib on Hep3B cells were calculated to be 128.078 and 13.657 µM, respectively. Genistein in combination with gefitinib significantly inhibited cell viability, promoted apoptosis and reduced EGFR, vascular endothelial growth factor receptor and platelet‑derived growth factor receptor phosphorylation. Genistein in combination with gefitinib promoted the expression of cleaved caspase‑3 and cleaved poly ADP‑ribose polymerase. In addition, combined treatment of genistein and gefitinib strongly inhibited the activation of the Akt/Erk/mTOR signaling pathway. In conclusion, findings from the present study suggest that genistein in combination with gefitinib inhibit HCC cell proliferation and promote apoptosis by inhibiting the Akt/Erk/mTOR pathway.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common malignancies (1). HCC has the fifth highest incidence and the third highest mortality of all malignancies worldwide (1,2), leading to >600,000 deaths annually (3). However, effective treatment for HCC remains elusive (4).

Liver resection and transplantation are currently the primary methods for treating HCC (5). However, due to unobservable symptoms in the early stages of HCC pathogenesis, most patients are diagnosed with HCC in the advanced stages (4). Surgical intervention for advanced HCC is not effective and chemotherapy is the only therapeutic option (6). However, traditional chemotherapeutic agents, including cisplatin, doxorubicin and fluorouracil, produce unsatisfactory outcomes, adverse side effects and drug resistance (6). With the emergence of targeted drugs such as small molecule kinase inhibitors, considerable progress has been made in the treatment of HCC (7). For patients with HCC, where surgery is not recommended, palliative treatment using sorafenib is currently the only clinical solution (8). However, since some patients with HCC also exhibit insensitivity or resistance to sorafenib, other chemotherapeutic options are required (8).

Gefitinib is a small molecule inhibitor of epidermal growth factor receptor (EGFR) (9). As a transmembrane protein, EGFR can induce cell proliferation by phosphorylating mitogen-activated protein kinase (MAPK), Akt and JNK (1012). Previous studies have confirmed that increased expression of EGFR is an important feature to the occurrence and development of HCC. In particular, Schiffer et al (13) has found that gefitinib inhibited hepatoma cell proliferation, however, any further mechanism remains unclear.

Genistein, also known as 5,7,4′-trihydroxyisoflavone, is an isoflavone originally found in Glycine max (14), which has demonstrated the potential to reduce the risk of developing liver, lung and breast cancer (15). In hepatoma cells, a previous study has found that genistein treatment inhibited proliferation whilst promoting apoptosis (16) and inhibited EGFR activation (17). However, to the best of our knowledge, no study on the combined effects of gefitinib and genistein on liver cancer has been conducted.

Therefore, the purpose of the present study was to investigate the effect of gefitinib and genistein on the physiology of hepatocellular carcinoma cells and to investigate the mechanism by testing the Akt/Erk/mTOR pathway.

Materials and methods

Cell culture

The HCC cell line Hep3B was purchased from American Type Culture Collection (cat. no. ATCC® HB-8064). The cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) and 100 U/ml of penicillin-streptomycin maintained in a humidified atmosphere at 37°C under 5% CO2. RPMI 1640, FBS and penicillin-streptomycin were purchased from Gibco (Thermo Fisher Scientific, Inc.). Hep3B cells (5×103 cells/well) were cultured with different concentrations of genistein (10, 20, 40, 80 and 160 µM; cat. no. G0272; Tokyo Chemical Industry Development Co., Ltd.) or gefitinib (1, 2.5, 5, 10 and 20 µM; cat. no. S1025, Selleck Chemicals) for 48 h at 37°C, in order to calculate the IC50 values. Hep3B cells were cultured in the presence of PBS (control group), 120 µM genistein (genistein group), 12 µM gefitinib (gefitinib group) and 127.6 µM genistein + 9.8 µM gefitinib (combination group) at 37°C, with the inhibition of cell growth analyzed at 0, 12, 24, 36, 48, 60 and 72 h for each group.

Cell cytotoxicity assay

Cell viability (5×103 cells/well) treated with genistein (10, 20, 40, 80 and 160 µM) or gefitinib (1, 2.5, 5, 10 and 20 µM) for 48 h at 37°C was detected by using cell counting kit-8 (CCK-8) assay (Beyotime Institute of Biotechnology). Diluted CCK-8 reagent was added and cultured at 37°C in a humidified atmosphere under 5% CO2 for 4 h. Optical density in each well at 450 nm was subsequently measured using a microplate reader (ELx800™; Omega Bio-Tek, Inc.). The cellular proliferation inhibition rates were calculated as: (1-OD/OD0 µM) ×100%.

Flow cytometry assay

After the treatment of 120 µM genistein (genistein group), 12 µM gefitinib (gefitinib group) or 127.6 µM genistein + 9.8 µM gefitinib (combination group) and culturing for 72 h (5×103 cells/well), cell apoptosis was measured using BD Pharmingen™ PE Annexin V-FITC/PI Apoptosis Detection Kit I (BD Biosciences) according to manufacturer's protocol. The samples were incubated at room temperature in the dark for 10 min. BD FACSCalibur™ Flow Cytometer and BD FACStation™ Software v6.1 × (BD Biosciences) was used to analyze cell apoptosis.

Western blot analysis

Cells were lysed using NP40 lysis buffer (Beyotime Institute of Biotechnology). The supernatant was collected by centrifuging the cell lysate at 10,000 × g at 4°C for 15 min. Bicinchoninic acid assay was used to determine the protein concentration. The proteins (20 µg/lane) were separated using 10% SDS-PAGE followed by transfer onto PVDF membranes. The membranes were blocked with 5% fat-free milk, diluted in PBS, at room temperature for 2 h prior to incubation with primary antibodies against anti-pro-caspase-3, (1:800, cat. no. ab13847), anti-cleaved-caspase-3 (1:600; cat. no. ab49822), anti-cleaved-poly ADP ribose polymerase (cleaved-PARP; 1:700; cat. no. ab32064), anti-EGFR (1:2,000; cat. no. ab32562), anti-platelet-derived growth factor α (PDGF; 1:500; cat. no. ab38562), anti-pan-Akt (Akt; 1:800; cat. no. ab8805), anti-phospho-pan-Akt (phospho T308; p-Akt; 1:800, cat. no. ab38449), anti-Erk1/2 (1:800, cat. no. ab54230), anti-p-Erk1/2 (1:600; cat. no. ab201015), anti-mTOR (1:800; cat. no. ab2732) or anti-p-mTOR (1:600; cat. no. ab109268) at 4°C overnight. All primary antibodies were purchased from Abcam. The membranes were subsequently incubated with goat anti-mouse IgG (1:8,000; cat. no. ab6785; Abcam), rabbit anti-mouse IgG, (1:9,000; cat. no. ab99697; Abcam), mouse anti-rabbit IgG (1:7,000; cat. no. BA1034; Invitrogen; Thermo Fisher Scientific, Inc.) and donkey anti-rabbit IgG (1:5,000; cat. no. NL004; R&D Systems, Inc.) secondary antibodies at room temperature for 1.5 h. Protein bands were visualized using BeyoECLMoon ECL reagent (Beyotime Institute of Biotechnology) and Quantity One v4.6.2 (Bio-Rad Laboratories, Inc.) quantitative analysis software.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted using TRIzol (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol and quantified using a NanoDrop spectrometer (Thermo Fisher Scientific, Inc.). RNA was reverse transcribed into cDNA at 42°C for 30 min and 85°C for 5 min, using the iScript™ cDNA Synthesis kit (Bio-Rad Laboratories, Inc.) according to the manufacturer's protocol. qPCR was then performed using FastStart Universal SYBR® Green Master kit (Roche Diagnostics) in the ABI StepOne system (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to manufacturers' protocols. Each reaction system was prepared as follows: cDNA template, 2.5 µl; forward primers (10 µM), 1 µl; reverse primers (10 µM), 1 µl; 2X SYBR® Green master mix, 10 µl and ddH2O, 5.5 µl. The following thermocycling conditions were used for the qPCR: Initial denaturation for 2 min at 95°C; 40 cycles of 15 sec at 95°C, 25 sec at 60°C and 60 sec at 72°C. Expression levels were quantified using the 2−ΔΔCq method with GAPDH as the internal reference (18). The list of primers used for this study is included in Table I.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

Primer sequence (5′-3′)

Primer nameForwardReverse
EGFR AACACCCTGGTCTGGAAGTACG TCGTTGGACAGCCTTCAAGACC
PDGF GAGGAAGCCGAGATGCCCC TGCTGTGGATCTGACTTCGAG
GAPDH AGTATGACTCCACTCACGGC CACCAGTAGACTCCACGACA

[i] EGFR, epidermal growth factor receptor; PDGF, platelet-derived growth factor.

Statistical analysis

Data are presented as the mean ± standard deviation. Statistical analysis was performed using SPSS 20 software (IBM Corp.). One-way ANOVA followed by Tukey's multiple comparisons post hoc test was performed to analyze differences between experimental groups. P<0.05 was considered to indicate a statistically significant difference.

Results

IC50 determination of genistein and gefitinib

By measuring Hep3B cell cytotoxicity under different concentrations of genistein and gefitinib after 48 h, the IC50 values of genistein and gefitinib were calculated to be 127.603 and 9.818 µM, respectively (Fig. 1A and B). These concentrations were therefore used for subsequent experiments.

Apoptosis of Hep3B cells induced by genistein and gefitinib

Comparing the cytotoxicity (killing the cells) of the control, genistein, gefitinib and combination groups over a 72-h time course, it was found that the cytotoxicity in the combination group was the highest (Fig. 2A). Flow cytometry was applied to detect the apoptosis of the control, genistein, gefitinib, and combination groups, after cells were cultured for 72 h. The apoptosis in the genistein alone and gefitinib alone groups were significantly higher compared with that in the control group, whereas that of the combination group was the highest compared with the other three groups (Fig. 2B). This suggests that genistein in combination with gefitinib enhanced hep3b cell apoptosis.

Effects of genistein and gefitinib on the activity of EGFR, PDGF, and the expression of proteins associated with apoptosis

Expression levels of pro-caspase-3, cleaved-caspase-3, cleaved-PARP, EGFR and PDGF were detected using western blot analysis. The expression levels of pro-caspase-3 protein was significantly reduced in the genistein alone and gefitinib alone groups, whilst the protein expression levels of cleaved-caspase-3 and cleaved-PARP were significantly increased compared with the control group (Fig. 3A-D). When genistein and gefitinib were combined together, these observed effects on the expression of proteins associated with apoptosis were significantly enhanced compared with when either drug was used alone (Fig. 3A-D).

RT-qPCR and western blotting results revealed that either genistein or gefitinib treatment alone significantly downregulated the expression of EGFR and PDGF mRNA and protein, which was significantly enhanced by the combined addition of genistein and gefitinib (Fig. 3E-I). This suggest that that the enhanced effects of genistein in combination with gefitinib were achieved by inhibiting EGFR and PDGF expression and promoting the expression of pro-apoptotic proteins.

Effects of genistein and gefitinib on the Akt/Erk/mTOR pathway

Treatment with genistein alone, gefitinib alone or both in combination did not have statistically significant effects on the expression of total Akt, Erk and mTOR proteins (Fig. 4A-D). However, Akt, Erk and mTOR phosphorylation levels were significantly lower in genistein or gefitimib alone group, compared with control group. In addition, Akt, Erk and mTOR phosphorylation levels were significantly lower in the combination group compared with control, genistein or gefitinib alone, while Akt, Erk and Mtor phosphorylation levels were significantly lower in genistein or gefitinib groups compared with control group (Fig. 4A-D). These results suggest that genistein in combination with gefitinib can significantly reduce the Akt/Erk/mTOR pathway activity.

Discussion

Gefitinib is a small-molecule antitumor drug that can selectively inhibit EGFR and inhibit tumor cell proliferation to promote apoptosis (19). Indeed, gefitinib has demonstrated a high efficacy in treating lung and gastric cancer (20,21). A recent study has shown that gefitinib exhibited inhibitory effects on HCC (22). By contrast, genistein is an isoflavone compound and an inhibitor of tyrosine protein kinase (TPK) (14). Studies have shown that inhibiting TPK activity using genistein prevented EGFR-mediated receptor autophosphorylation and mitotic signal transduction (23,24). Genistein has been studied in various malignancies, including breast, lung and prostate cancer (2527). However, only a limited number of studies have been conducted on the treatment of genistein on HCC.

The occurrence and development of HCC is closely associated with the abnormal expression of a number of proteins. In particular, one study has confirmed that the overexpression of EGFR was associated with the occurrence and development of liver cancer (28), where higher levels of EGFR expression were associated with poorer prognosis and higher recurrence rates in poorly differentiated HCC (29). Combination therapy is the most important method for treating advanced HCC (30), and to the best of our knowledge, there is currently no study investigating the effects of genistein in combination with gefitinib on HCC. Therefore, in the present study the effect of genistein combined with gefitinib on the proliferation and apoptosis of the HCC cell line Hep3B was investigated. Both drugs significantly inhibited Hep3B cell viability and promoted apoptosis, and these effects were enhanced when these drugs were used in combination. These results suggest that combined genistein and gefitinib treatment exerted increased anti-proliferative and pro-apoptotic effects, compared with genistein or gefitinib alone. In addition, a previous study has also shown that combined genistein and gefitinib treatment enhanced the effect of growth inhibition and apoptosis on non-small cell lung cancer, where the strongest synergistic effect was observed at low concentrations (31).

To explore the role of genistein in combination with gefitinib in cell proliferation and apoptosis further the expression levels of proteins associated with apoptosis, EGFR and PDGF, were determined. The caspase protein family serves important roles in apoptosis. Caspase-3 is the activator of apoptosis where it can enzymatically cleave PARP (32). Downstream, PARP is a multifunctional post-translational modification enzyme that recognizes structurally damaged DNA fragments. PARP cleavage by caspase-3 increases cell instability and promotes apoptosis (32,33). The results of the present study revealed that genistein in combination with gefitinib displayed the strongest effect on the activation of caspase-3 and PARP compared with the other three groups tested, which caused PARP to lose its enzymatic activity and promote apoptosis. EGFR and PDGF have been demonstrated to promote the division of epithelial cells and proliferation of liver cancer cells (3436). The present study demonstrated that the expression levels of EGFR and PDGF in the combination group were significantly lower compared with genistein and gefitinib groups alone. Previous studies have found that gefitinib inhibited proliferation by suppressing EGFR (37) whereas genistein has also been demonstrated to inhibit EGFR and PDGF in tumor cells (38,39). These results suggest that genistein promoted the effects of gefitinib by inhibiting the expression of EGFR and PDGF.

The downstream mechanism by which genistein in combination with gefitinib inhibited HCC Hep3B cell growth was subsequently explored by assessing the Akt/Erk/mTOR pathway. Gefitinib and genistein have been previously observed to act on EGFR, where the PI3K/Akt/mTOR and MAPK signaling pathways were the most important signal transduction pathways downstream of EGFR (40,41). Therefore, the effects of genistein and gefitinib on the Akt/Erk/mTOR signaling were investigated in the current study. Total Akt, Erk and mTOR protein levels remained relatively stable in all treatment groups; however, their phosphorylation levels decreased significantly. The levels of p-Akt, p-Erk, and p-mTOR in the combination group were significantly lower compared with genistein or gefitinib alone. Akt, alternatively known as protein kinase B, is an important downstream molecule of PI3K and serves an important role in the regulation of cell growth, proliferation, survival and glucose metabolism. Akt can modulate mTOR either directly or through Erk (42,43). mTOR is a type of serine/threonine kinase and activated mTOR promotes the phosphorylation of substrates S6 kinase (S6K) and 4E binding protein 1 (42). As both substrates are key regulators of protein translation, their phosphorylation leads to the initiation and increase in ribosomal protein synthesis. When mTOR activation is inhibited, cells undergo cell cycle arrest at the G1 phase and apoptosis (44). Previous studies have shown that genistein and gefitinib inhibited cell proliferation by reducing mTOR phosphorylation in cervical cancer cells and breast cancer cells (45,46). The present study suggested that the combination of genistein and gefitinib synergistically exerted anti-proliferative and pro-apoptotic effects by inhibiting the activation of the Akt/Erk/mTOR pathway. Indeed, the Akt-mTOR-p70 S6K pathway has been reported to be overactivated and may serve as a potential target in HCC therapy (47). Therefore, p70S6K activity may also have been inhibited in this study. In addition, a previous study has shown that the Akt/Erk/mTOR pathway was associated with cell autophagy in cancer cells (12). Therefore, it would be beneficial to investigate the effect of genistein and gefitinib on HCC cell autophagy in the future. Genistein has been reported to exhibit multi-targeted biological and molecular effects on cancer cells (15), and the specific inhibition of tyrosine-specific protein kinases is one of the best documented (14). Therefore, it would be of interest to validate the mechanism in which genistein acts to enhance the antitumor effects of gefitinib.

In conclusion, genistein in combination with gefitinib could synergistically inhibit HCC cell proliferation and promote apoptosis, in a more powerful manner compared with either applied alone. Such a phenomenon may be associated with the inhibition of the Akt/Erk/mTOR pathway. The present study provides support for the clinical application of genistein-gefitinib combination treatment on HCC.

Acknowledgements

Not applicable.

Funding

The present study was supported by Zhejiang Science and Technology Department Public Welfare Project (grant no. 2016C37149).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YT and MW performed western blot analysis and RT-qPCR. HH and JZ performed flow cytometry assay. YH detected cell cytotoxicity. YC and HP analyzed data and wrote the manuscript. All authors read and approved the final manuscript.

Ethical 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 

Hartke J, Johnson M and Ghabril M: The diagnosis and treatment of hepatocellular carcinoma. Semin Diagn Pathol. 34:153–159. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Costentin C: Hepatocellular carcinoma surveillance. Presse Med. 46:381–385. 2017.(In French). View Article : Google Scholar : PubMed/NCBI

3 

Jiang JF, Lao YC, Yuan BH, Yin J, Liu X, Chen L and Zhong JH: Treatment of hepatocellular carcinoma with portal vein tumor thrombus: Advances and challenges. Oncotarget. 8:33911–33921. 2017.PubMed/NCBI

4 

Bruix J, Gores GJ and Mazzaferro V: Hepatocellular carcinoma: Clinical frontiers and perspectives. Gut. 63:844–855. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Hsueh KC, Lee TY, Kor CT, Chen TM, Chang TM, Yang SF and Hsieh CB: The role of liver transplantation or resection for patients with early hepatocellular carcinoma. Tumour Biol. 37:4193–4201. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Hollebecque A, Malka D, Ferte C, Ducreux M and Boige V: Systemic treatment of advanced hepatocellular carcinoma: From disillusions to new horizons. Eur J Cancer. 51:327–339. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Qi XS, Guo XZ, Han GH, Li HY and Chen J: MET inhibitors for treatment of advanced hepatocellular carcinoma: A review. World J Gastroenterol. 21:5445–5453. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Abdel-Rahman O and Fouad M: Sorafenib-based combination as a first line treatment for advanced hepatocellular carcinoma: A systematic review of the literature. Crit Rev Oncol Hematol. 91:1–8. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, et al: EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science. 304:1497–1500. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Liu BN, Yan HQ, Wu X, Pan ZH, Zhu Y, Meng ZW, Zhou QH and Xu K: Apoptosis induced by benzyl isothiocyanate in gefitinib-resistant lung cancer cells is associated with Akt/MAPK pathways and generation of reactive oxygen species. Cell Biochem Biophys. 66:81–92. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Zhao ZQ, Yu ZY, Li J and Ouyang XN: Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway. Oncol Lett. 12:63–68. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Lu PH, Kuo TC, Chang KC, Chang CH and Chu CY: Gefitinib-induced epidermal growth factor receptor-independent keratinocyte apoptosis is mediated by the JNK activation pathway. Br J Dermatol. 164:38–46. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Schiffer E, Housset C, Cacheux W, Wendum D, Desbois-Mouthon C, Rey C, Clergue F, Poupon R, Barbu V and Rosmorduc O: Gefitinib, an EGFR inhibitor, prevents hepatocellular carcinoma development in the rat liver with cirrhosis. Hepatology. 41:307–314. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, Shibuya M and Fukami Y: Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem. 262:5592–5595. 1987.PubMed/NCBI

15 

Banerjee S, Li Y, Wang Z and Sarkar FH: Multi-targeted therapy of cancer by genistein. Cancer Lett. 269:226–242. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Li S, Li J, Dai W, Zhang Q, Feng J, Wu L, Liu T, Yu Q, Xu S, Wang W, et al: Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br J Cancer. 117:1518–1528. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Gruca A, Krawczyk Z, Szeja W, Grynkiewicz G and Rusin A: Synthetic genistein glycosides inhibiting EGFR phosphorylation enhance the effect of radiation in HCT 116 colon cancer cells. Molecules. 19:18558–18573. 2014. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Wu JF, Ji J, Dong SY, Li BB, Yu ML, Wu DD, Tao L and Tong XH: Gefitinib enhances oxaliplatin-induced apoptosis mediated by Src and PKC-modulated gap junction function. Oncol Rep. 36:3251–3258. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, Gemma A, Harada M, Yoshizawa H, Kinoshita I, et al: Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 362:2380–2388. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Kishida O, Miyazaki Y, Murayama Y, Ogasa M, Miyazaki T, Yamamoto T, Watabe K, Tsutsui S, Kiyohara T, Shimomura I and Shinomura Y: Gefitinib (‘Iressa’, ZD1839) inhibits SN38-triggered EGF signals and IL-8 production in gastric cancer cells. Cancer Chemother Pharmacol. 55:584–594. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Yang T, Gu J, Liu T, Ma H, Ma X, Tao J, Jin Y and Liang X: Expression of acetaldehyde dehydrogenase in gefitinib-resistant human lung adenocarcinoma HCC-827/GR cells. Zhongguo Fei Ai Za Zhi. 21:431–436. 2018.(In Chinese). PubMed/NCBI

23 

Nakashima S, Koike T and Nozawa Y: Genistein, a protein tyrosine kinase inhibitor, inhibits thromboxane A2-mediated human platelet responses. Mol Pharmacol. 39:475–480. 1991.PubMed/NCBI

24 

Papazisis KT, Zambouli D, Kimoundri OT, Papadakis ES, Vala V, Geromichalos GD, Voyatzi S, Markala D, Destouni E, Boutis L and Kortsaris AH: Protein tyrosine kinase inhibitor, genistein, enhances apoptosis and cell cycle arrest in K562 cells treated with gamma-irradiation. Cancer Lett. 160:107–113. 2000. View Article : Google Scholar : PubMed/NCBI

25 

van Duursen MB, Nijmeijer SM, de Morree ES, de Jong PC and van den Berg M: Genistein induces breast cancer-associated aromatase and stimulates estrogen-dependent tumor cell growth in in vitro breast cancer model. Toxicology. 289:67–73. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Geller J, Sionit L, Partido C, Li L, Tan X, Youngkin T, Nachtsheim D and Hoffman RM: Genistein inhibits the growth of human-patient BPH and prostate cancer in histoculture. Prostate. 34:75–79. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Tian T, Li J, Li B, Wang Y, Li M, Ma D and Wang X: Genistein exhibits anti-cancer effects via down-regulating FoxM1 in H446 small-cell lung cancer cells. Tumour Biol. 35:4137–4145. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Han C, Michalopoulos GK and Wu T: Prostaglandin E2 receptor EP1 transactivates EGFR/MET receptor tyrosine kinases and enhances invasiveness in human hepatocellular carcinoma cells. J Cell Physiol. 207:261–270. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Wang W, Ma XP, Shi Z, Zhang P, Ding DL, Huang HX, Saiyin HG, Chen TY, Lu PX, Wang NJ, et al: Epidermal growth factor receptor pathway polymorphisms and the prognosis of hepatocellular carcinoma. Am J Cancer Res. 5:396–410. 2014.PubMed/NCBI

30 

Lin J, Wu L, Bai X, Xie Y, Wang A, Zhang H, Yang X, Wan X, Lu X, Sang X and Zhao H: Combination treatment including targeted therapy for advanced hepatocellular carcinoma. Oncotarget. 7:71036–71051. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Zhu H, Cheng H, Ren Y, Liu ZG, Zhang YF and De Luo B: Synergistic inhibitory effects by the combination of gefitinib and genistein on NSCLC with acquired drug-resistance in vitro and in vivo. Mol Biol Rep. 39:4971–4979. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Boulares AH, Yakovlev AG, Ivanova V, Stoica BA, Wang G, Iyer S and Smulson M: Role of poly(ADP-ribose) polymerase (PARP) cleavage in apoptosis. Caspase 3-resistant PARP mutant increases rates of apoptosis in transfected cells. J Biol Chem. 274:22932–22940. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Luo H, Liang H, Chen J, Xu Y, Chen Y, Xu L, Yun L, Liu J, Yang H, Liu L, et al: Hydroquinone induces TK6 cell growth arrest and apoptosis through PARP-1/p53 regulatory pathway. Environ Toxicol. 32:2163–2171. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Xu J, Zhang X, Wang H, Ge S, Gao T, Song L, Wang X, Li H, Qin Y and Zhang Z: HCRP1 downregulation promotes hepatocellular carcinoma cell migration and invasion through the induction of EGFR activation and epithelial-mesenchymal transition. Biomed Pharmacother. 88:421–429. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Lv X, Fang C, Yin R, Qiao B, Shang R, Wang J, Song W, He Y and Chen Y: Agrin para-secreted by PDGF-activated human hepatic stellate cells promotes hepatocarcinogenesis in vitro and in vivo. Oncotarget. 8:105340–105355. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Wang R, Li Y, Hou Y, Yang Q, Chen S, Wang X, Wang Z, Yang Y, Chen C, Wang Z and Wu Q: The PDGF-D/miR-106a/Twist1 pathway orchestrates epithelial-mesenchymal transition in gemcitabine resistance hepatoma cells. Oncotarget. 6:7000–7010. 2015.PubMed/NCBI

37 

Ni J, Zhou LL, Ding L, Zhao X, Cao H, Fan F, Li H, Lou R, Du Y, Dong S, et al: PPARγ agonist efatutazone and gefitinib synergistically inhibit the proliferation of EGFR-TKI-resistant lung adenocarcinoma cells via the PPARgamma/PTEN/Akt pathway. Exp Cell Res. 361:246–256. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Nakamura H and Wang Y, Kurita T, Adomat H, Cunha GR and Wang Y: Genistein increases epidermal growth factor receptor signaling and promotes tumor progression in advanced human prostate cancer. PLoS One. 6:e200342011. View Article : Google Scholar : PubMed/NCBI

39 

Little PJ, Getachew R, Rezaei HB, Sanchez-Guerrero E, Khachigian LM, Wang H, Liao S, Zheng W, Ballinger ML and Osman N: Genistein inhibits PDGF-stimulated proteoglycan synthesis in vascular smooth muscle without blocking PDGFβ receptor phosphorylation. Arch Biochem Biophys. 525:25–31. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Ono M and Kuwano M: Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs. Clin Cancer Res. 12:7242–7251. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Tanjak P, Thiantanawat A, Watcharasit P and Satayavivad J: Genistein reduces the activation of AKT and EGFR, and the production of IL6 in cholangiocarcinoma cells involving estrogen and estrogen receptors. Int J Oncol. 53:177–188. 2018.PubMed/NCBI

42 

Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ and Yancopoulos GD: Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 3:1014–1019. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, Piemonti L, Capurso G, Di Florio A, delle Fave G, et al: Pancreatic endocrine tumors: Expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 28:245–255. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Zoncu R, Efeyan A and Sabatini DM: mTOR: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 12:21–35. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Sahin K, Tuzcu M, Basak N, Caglayan B, Kilic U, Sahin F and Kucuk O: Sensitization of cervical cancer cells to cisplatin by genistein: The role of NFκB and Akt/mTOR signaling pathways. J Oncol. 2012:4615622012. View Article : Google Scholar : PubMed/NCBI

46 

Block M, Gründker C, Fister S, Kubin J, Wilkens L, Mueller MD, Hemmerlein B, Emons G and Günthert AR: Inhibition of the AKT/mTOR and erbB pathways by gefitinib, perifosine and analogs of gonadotropin-releasing hormone I and II to overcome tamoxifen resistance in breast cancer cells. Int J Oncol. 41:1845–1854. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Li W, Tan D, Zhang Z, Liang JJ and Brown RE: Activation of Akt-mTOR-p70S6K pathway in angiogenesis in hepatocellular carcinoma. Oncol Rep. 20:713–719. 2008.PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tong Y, Wang M, Huang H, Zhang J, Huang Y, Chen Y and Pan H: Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line. Exp Ther Med 18: 3793-3800, 2019
APA
Tong, Y., Wang, M., Huang, H., Zhang, J., Huang, Y., Chen, Y., & Pan, H. (2019). Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line. Experimental and Therapeutic Medicine, 18, 3793-3800. https://doi.org/10.3892/etm.2019.8027
MLA
Tong, Y., Wang, M., Huang, H., Zhang, J., Huang, Y., Chen, Y., Pan, H."Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line". Experimental and Therapeutic Medicine 18.5 (2019): 3793-3800.
Chicago
Tong, Y., Wang, M., Huang, H., Zhang, J., Huang, Y., Chen, Y., Pan, H."Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line". Experimental and Therapeutic Medicine 18, no. 5 (2019): 3793-3800. https://doi.org/10.3892/etm.2019.8027