Skip to main content
Top
Published in: Cancer Cell International 1/2019

Open Access 01-12-2019 | Thyroid Cancer | Primary research

Epigallocatechin-3-gallate inhibits the growth and increases the apoptosis of human thyroid carcinoma cells through suppression of EGFR/RAS/RAF/MEK/ERK signaling pathway

Authors: Dongdong Wu, Zhengguo Liu, Jianmei Li, Qianqian Zhang, Peiyu Zhong, Tieshan Teng, Mingliang Chen, Zhongwen Xie, Ailing Ji, Yanzhang Li

Published in: Cancer Cell International | Issue 1/2019

Login to get access

Abstract

Background

Thyroid cancer is the most common type of endocrine malignancy and the incidence rate is rapidly increasing worldwide. Epigallocatechin-3-gallate (EGCG) could suppress cancer growth and induce apoptosis in many types of cancer cells. However, the mechanism of action of EGCG on the growth of human thyroid carcinoma cells has not been fully illuminated.

Methods

Cell proliferation and viability were detected by EdU and MTS assays. Cell cycle distribution was measured by flow cytometry. Migration and invasion were evaluated by scratch and transwell assays. Apoptotic levels were detected by TUNEL staining and western blotting. The protein levels of EGFR/RAS/RAF/MEK/ERK signaling pathway were detected by western blotting. The in vivo results were determined by tumor xenografts in nude mice. The in vivo proliferation, tumor microvessel density, and apoptosis were detected by immunohistochemistry.

Results

EGCG inhibited the proliferation, viability, and cell cycle progression in human thyroid carcinoma cells. EGCG decreased the migration and invasion, but increased the apoptosis of human thyroid carcinoma cells. EGCG reduced the protein levels of phospho (p)-epidermal growth factor receptor (EGFR), H-RAS, p-RAF, p-MEK1/2, and p-extracellular signal-regulated protein kinase 1/2 (ERK1/2) in human thyroid carcinoma cells. EGCG inhibited the growth of human thyroid carcinoma xenografts by inducing apoptosis and down-regulating angiogenesis.

Conclusions

EGCG could reduce the growth and increase the apoptosis of human thyroid carcinoma cells through suppressing the EGFR/RAS/RAF/MEK/ERK signaling pathway. EGCG can be developed as an effective therapeutic agent for the treatment of thyroid cancer.
Literature
2.
go back to reference Kitahara CM, Sosa JA. The changing incidence of thyroid cancer. Nat Rev Endocrinol. 2016;12:646–53.CrossRefPubMed Kitahara CM, Sosa JA. The changing incidence of thyroid cancer. Nat Rev Endocrinol. 2016;12:646–53.CrossRefPubMed
4.
go back to reference Schlumberger M, Elisei R, Müller S, et al. Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann Oncol. 2017;28:2813–9.CrossRefPubMedCentralPubMed Schlumberger M, Elisei R, Müller S, et al. Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann Oncol. 2017;28:2813–9.CrossRefPubMedCentralPubMed
5.
go back to reference Molinaro E, Romei C, Biagini A, et al. Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol. 2017;13:644–60.CrossRefPubMed Molinaro E, Romei C, Biagini A, et al. Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol. 2017;13:644–60.CrossRefPubMed
7.
go back to reference Liu D, Hou P, Liu Z, et al. Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res. 2009;69:7311–9.CrossRefPubMedCentralPubMed Liu D, Hou P, Liu Z, et al. Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res. 2009;69:7311–9.CrossRefPubMedCentralPubMed
8.
go back to reference Capdevila J, Mayor R, Mancuso FM, et al. Early evolutionary divergence between papillary and anaplastic thyroid cancers. Ann Oncol. 2018;29:1454–60.CrossRefPubMed Capdevila J, Mayor R, Mancuso FM, et al. Early evolutionary divergence between papillary and anaplastic thyroid cancers. Ann Oncol. 2018;29:1454–60.CrossRefPubMed
9.
go back to reference Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384:319–28.CrossRefPubMedCentralPubMed Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384:319–28.CrossRefPubMedCentralPubMed
10.
go back to reference Broecker-Preuss M, Müller S, Britten M, et al. Sorafenib inhibits intracellular signaling pathways and induces cell cycle arrest and cell death in thyroid carcinoma cells irrespective of histological origin or BRAF mutational status. BMC Cancer. 2015;15:184.CrossRefPubMedCentralPubMed Broecker-Preuss M, Müller S, Britten M, et al. Sorafenib inhibits intracellular signaling pathways and induces cell cycle arrest and cell death in thyroid carcinoma cells irrespective of histological origin or BRAF mutational status. BMC Cancer. 2015;15:184.CrossRefPubMedCentralPubMed
11.
go back to reference Wang J, Man GCW, Chan TH, et al. A prodrug of green tea polyphenol (–)-epigallocatechin-3-gallate (Pro-EGCG) serves as a novel angiogenesis inhibitor in endometrial cancer. Cancer Lett. 2018;412:10–20.CrossRefPubMed Wang J, Man GCW, Chan TH, et al. A prodrug of green tea polyphenol (–)-epigallocatechin-3-gallate (Pro-EGCG) serves as a novel angiogenesis inhibitor in endometrial cancer. Cancer Lett. 2018;412:10–20.CrossRefPubMed
12.
go back to reference Torello CO, Shiraishi RN, Della Via FI, et al. Reactive oxygen species production triggers green tea-induced anti-leukaemic effects on acute promyelocytic leukaemia model. Cancer Lett. 2018;414:116–26.CrossRefPubMed Torello CO, Shiraishi RN, Della Via FI, et al. Reactive oxygen species production triggers green tea-induced anti-leukaemic effects on acute promyelocytic leukaemia model. Cancer Lett. 2018;414:116–26.CrossRefPubMed
13.
go back to reference Ugai T, Matsuo K, Sawada N, et al. Coffee and green tea consumption and subsequent risk of acute myeloid leukemia and myelodysplastic syndromes in Japan. Int J Cancer. 2018;142:1130–8.CrossRefPubMed Ugai T, Matsuo K, Sawada N, et al. Coffee and green tea consumption and subsequent risk of acute myeloid leukemia and myelodysplastic syndromes in Japan. Int J Cancer. 2018;142:1130–8.CrossRefPubMed
14.
go back to reference Ko H, So Y, Jeon H, et al. TGF-β1-induced epithelial-mesenchymal transition and acetylation of Smad2 and Smad3 are negatively regulated by EGCG in human A549 lung cancer cells. Cancer Lett. 2013;335:205–13.CrossRefPubMed Ko H, So Y, Jeon H, et al. TGF-β1-induced epithelial-mesenchymal transition and acetylation of Smad2 and Smad3 are negatively regulated by EGCG in human A549 lung cancer cells. Cancer Lett. 2013;335:205–13.CrossRefPubMed
15.
go back to reference Gan RY, Li HB, Sui ZQ, et al. Absorption, metabolism, anti-cancer effect and molecular targets of epigallocatechin gallate (EGCG): an updated review. Crit Rev Food Sci Nutr. 2018;58:924–41.CrossRefPubMed Gan RY, Li HB, Sui ZQ, et al. Absorption, metabolism, anti-cancer effect and molecular targets of epigallocatechin gallate (EGCG): an updated review. Crit Rev Food Sci Nutr. 2018;58:924–41.CrossRefPubMed
16.
go back to reference Afzal M, Safer AM, Menon M. Green tea polyphenols and their potential role in health and disease. Inflammopharmacology. 2015;23:151–61.CrossRefPubMed Afzal M, Safer AM, Menon M. Green tea polyphenols and their potential role in health and disease. Inflammopharmacology. 2015;23:151–61.CrossRefPubMed
17.
go back to reference Oliveira MR, Nabavi SF, Daglia M, et al. Epigallocatechin gallate and mitochondria—a story of life and death. Pharmacol Res. 2016;104:70–85.CrossRefPubMed Oliveira MR, Nabavi SF, Daglia M, et al. Epigallocatechin gallate and mitochondria—a story of life and death. Pharmacol Res. 2016;104:70–85.CrossRefPubMed
18.
go back to reference Modernelli A, Naponelli V, Giovanna Troglio M, et al. EGCG antagonizes bortezomib cytotoxicity in prostate cancer cells by an autophagic mechanism. Sci Rep. 2015;5:15270.CrossRefPubMedCentralPubMed Modernelli A, Naponelli V, Giovanna Troglio M, et al. EGCG antagonizes bortezomib cytotoxicity in prostate cancer cells by an autophagic mechanism. Sci Rep. 2015;5:15270.CrossRefPubMedCentralPubMed
19.
go back to reference Butt MS, Ahmad RS, Sultan MT, et al. Green tea and anticancer perspectives: updates from last decade. Crit Rev Food Sci Nutr. 2015;55:792–805.CrossRefPubMed Butt MS, Ahmad RS, Sultan MT, et al. Green tea and anticancer perspectives: updates from last decade. Crit Rev Food Sci Nutr. 2015;55:792–805.CrossRefPubMed
20.
go back to reference Wang H, Bian S, Yang CS. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1α. Carcinogenesis. 2011;32:1881–9.CrossRefPubMedCentralPubMed Wang H, Bian S, Yang CS. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1α. Carcinogenesis. 2011;32:1881–9.CrossRefPubMedCentralPubMed
21.
go back to reference Singh AK, Sharma N, Ghosh M, et al. Emerging importance of dietary phytochemicals in fight against cancer: role in targeting cancer stem cells. Crit Rev Food Sci Nutr. 2017;57:3449–63.CrossRefPubMed Singh AK, Sharma N, Ghosh M, et al. Emerging importance of dietary phytochemicals in fight against cancer: role in targeting cancer stem cells. Crit Rev Food Sci Nutr. 2017;57:3449–63.CrossRefPubMed
22.
go back to reference Shammas MA, Neri P, Koley H, et al. Specific killing of multiple myeloma cells by (–)-epigallocatechin-3-gallate extracted from green tea: biologic activity and therapeutic implications. Blood. 2006;108:2804–10.CrossRefPubMedCentralPubMed Shammas MA, Neri P, Koley H, et al. Specific killing of multiple myeloma cells by (–)-epigallocatechin-3-gallate extracted from green tea: biologic activity and therapeutic implications. Blood. 2006;108:2804–10.CrossRefPubMedCentralPubMed
23.
go back to reference Chow HH, Cai Y, Hakim IA, et al. Pharmacokinetics and safety of green tea polyphenols after multiple-dose administration of epigallocatechin gallate and polyphenon E in healthy individuals. Clin Cancer Res. 2003;9:3312–9.PubMed Chow HH, Cai Y, Hakim IA, et al. Pharmacokinetics and safety of green tea polyphenols after multiple-dose administration of epigallocatechin gallate and polyphenon E in healthy individuals. Clin Cancer Res. 2003;9:3312–9.PubMed
24.
go back to reference Wu D, Luo N, Wang L, et al. Hydrogen sulfide ameliorates chronic renal failure in rats by inhibiting apoptosis and inflammation through ROS/MAPK and NF-κB signaling pathways. Sci Rep. 2017;7:455.CrossRefPubMedCentralPubMed Wu D, Luo N, Wang L, et al. Hydrogen sulfide ameliorates chronic renal failure in rats by inhibiting apoptosis and inflammation through ROS/MAPK and NF-κB signaling pathways. Sci Rep. 2017;7:455.CrossRefPubMedCentralPubMed
25.
go back to reference Wu D, Li M, Tian W, et al. Hydrogen sulfide acts as a double-edged sword in human hepatocellular carcinoma cells through EGFR/ERK/MMP-2 and PTEN/AKT signaling pathways. Sci Rep. 2017;7:5134.CrossRefPubMedCentralPubMed Wu D, Li M, Tian W, et al. Hydrogen sulfide acts as a double-edged sword in human hepatocellular carcinoma cells through EGFR/ERK/MMP-2 and PTEN/AKT signaling pathways. Sci Rep. 2017;7:5134.CrossRefPubMedCentralPubMed
26.
go back to reference Wu DD, Gao YR, Li T, et al. PEST-containing nuclear protein mediates the proliferation, migration, and invasion of human neuroblastoma cells through MAPK and PI3K/AKT/mTOR signaling pathways. BMC Cancer. 2018;18:499.CrossRefPubMedCentralPubMed Wu DD, Gao YR, Li T, et al. PEST-containing nuclear protein mediates the proliferation, migration, and invasion of human neuroblastoma cells through MAPK and PI3K/AKT/mTOR signaling pathways. BMC Cancer. 2018;18:499.CrossRefPubMedCentralPubMed
27.
go back to reference Heilmann AM, Perera RM, Ecker V, et al. CDK4/6 and IGF1 receptor inhibitors synergize to suppress the growth of p16INK4A-deficient pancreatic cancers. Cancer Res. 2014;74:3947–58.CrossRefPubMedCentralPubMed Heilmann AM, Perera RM, Ecker V, et al. CDK4/6 and IGF1 receptor inhibitors synergize to suppress the growth of p16INK4A-deficient pancreatic cancers. Cancer Res. 2014;74:3947–58.CrossRefPubMedCentralPubMed
28.
go back to reference Determann O, Hoster E, Ott G, et al. Ki-67 predicts outcome in advanced-stage mantle cell lymphoma patients treated with anti-CD20 immunochemotherapy: results from randomized trials of the European MCL network and the German low grade lymphoma study group. Blood. 2008;111:2385–7.CrossRefPubMed Determann O, Hoster E, Ott G, et al. Ki-67 predicts outcome in advanced-stage mantle cell lymphoma patients treated with anti-CD20 immunochemotherapy: results from randomized trials of the European MCL network and the German low grade lymphoma study group. Blood. 2008;111:2385–7.CrossRefPubMed
29.
go back to reference Koukourakis MI, Giatromanolaki A, Thorpe PE, et al. Vascular endothelial growth factor/KDR activated microvessel density versus CD31 standard microvessel density in non-small cell lung cancer. Cancer Res. 2000;60:3088–95.PubMed Koukourakis MI, Giatromanolaki A, Thorpe PE, et al. Vascular endothelial growth factor/KDR activated microvessel density versus CD31 standard microvessel density in non-small cell lung cancer. Cancer Res. 2000;60:3088–95.PubMed
31.
go back to reference Wang SS, Lv Y, Xu XC, et al. Triptonide inhibits human nasopharyngeal carcinoma cell growth via disrupting Lnc-RNA THOR-IGF2BP1 signaling. Cancer Lett. 2019;443:13–24.CrossRefPubMed Wang SS, Lv Y, Xu XC, et al. Triptonide inhibits human nasopharyngeal carcinoma cell growth via disrupting Lnc-RNA THOR-IGF2BP1 signaling. Cancer Lett. 2019;443:13–24.CrossRefPubMed
32.
go back to reference Pitchakarn P, Suzuki S, Ogawa K, et al. Induction of G1 arrest and apoptosis in androgen-dependent human prostate cancer by Kuguacin J, a triterpenoid from Momordica charantia leaf. Cancer Lett. 2011;306:142–50.CrossRefPubMed Pitchakarn P, Suzuki S, Ogawa K, et al. Induction of G1 arrest and apoptosis in androgen-dependent human prostate cancer by Kuguacin J, a triterpenoid from Momordica charantia leaf. Cancer Lett. 2011;306:142–50.CrossRefPubMed
33.
go back to reference Chen M, Wang X, Zha D, et al. Apigenin potentiates TRAIL therapy of non-small cell lung cancer via upregulating DR4/DR5 expression in a p53-dependent manner. Sci Rep. 2016;6:35468.CrossRefPubMedCentralPubMed Chen M, Wang X, Zha D, et al. Apigenin potentiates TRAIL therapy of non-small cell lung cancer via upregulating DR4/DR5 expression in a p53-dependent manner. Sci Rep. 2016;6:35468.CrossRefPubMedCentralPubMed
34.
go back to reference Kitagawa M, Liao PJ, Lee KH, et al. Dual blockade of the lipid kinase PIP4Ks and mitotic pathways leads to cancer-selective lethality. Nat Commun. 2017;8:2200.CrossRefPubMedCentralPubMed Kitagawa M, Liao PJ, Lee KH, et al. Dual blockade of the lipid kinase PIP4Ks and mitotic pathways leads to cancer-selective lethality. Nat Commun. 2017;8:2200.CrossRefPubMedCentralPubMed
35.
go back to reference Knight T, Irving JA. Ras/Raf/MEK/ERK pathway activation in childhood acute lymphoblastic leukemia and its therapeutic targeting. Front Oncol. 2014;4:160.CrossRefPubMedCentralPubMed Knight T, Irving JA. Ras/Raf/MEK/ERK pathway activation in childhood acute lymphoblastic leukemia and its therapeutic targeting. Front Oncol. 2014;4:160.CrossRefPubMedCentralPubMed
36.
go back to reference Poulikakos PI, Rosen N. Mutant BRAF melanomas-dependence and resistance. Cancer Cell. 2011;19:11–5.CrossRefPubMed Poulikakos PI, Rosen N. Mutant BRAF melanomas-dependence and resistance. Cancer Cell. 2011;19:11–5.CrossRefPubMed
37.
go back to reference Peng J, Gassama-Diagne A. Apicobasal polarity and Ras/Raf/MEK/ERK signalling in cancer. Gut. 2017;66:986–7.CrossRefPubMed Peng J, Gassama-Diagne A. Apicobasal polarity and Ras/Raf/MEK/ERK signalling in cancer. Gut. 2017;66:986–7.CrossRefPubMed
39.
go back to reference Troiani T, Napolitano S, Vitagliano D, et al. Primary and acquired resistance of colorectal cancer cells to anti-EGFR antibodies converge on MEK/ERK pathway activation and can be overcome by combined MEK/EGFR inhibition. Clin Cancer Res. 2014;20:3775–86.CrossRefPubMed Troiani T, Napolitano S, Vitagliano D, et al. Primary and acquired resistance of colorectal cancer cells to anti-EGFR antibodies converge on MEK/ERK pathway activation and can be overcome by combined MEK/EGFR inhibition. Clin Cancer Res. 2014;20:3775–86.CrossRefPubMed
40.
go back to reference Bentzien F, Zuzow M, Heald N, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.CrossRefPubMedCentralPubMed Bentzien F, Zuzow M, Heald N, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.CrossRefPubMedCentralPubMed
41.
go back to reference Hong S, Yu S, Li J, et al. MiR-20b displays tumor-suppressor functions in papillary thyroid carcinoma by regulating the MAPK/ERK signaling pathway. Thyroid. 2016;26:1733–43.CrossRefPubMed Hong S, Yu S, Li J, et al. MiR-20b displays tumor-suppressor functions in papillary thyroid carcinoma by regulating the MAPK/ERK signaling pathway. Thyroid. 2016;26:1733–43.CrossRefPubMed
42.
go back to reference So Y, Lee YJ, Lee WW, et al. Determination of the optimal time for radioiodine therapy in anaplastic thyroid carcinoma using the adenovirus-mediated transfer of sodium iodide symporter gene. Oncol Rep. 2013;29:1666–70.CrossRefPubMed So Y, Lee YJ, Lee WW, et al. Determination of the optimal time for radioiodine therapy in anaplastic thyroid carcinoma using the adenovirus-mediated transfer of sodium iodide symporter gene. Oncol Rep. 2013;29:1666–70.CrossRefPubMed
43.
go back to reference De Amicis F, Perri A, Vizza D, et al. Epigallocatechin gallate inhibits growth and epithelial-to-mesenchymal transition in human thyroid carcinoma cell lines. J Cell Physiol. 2013;228:2054–62.CrossRefPubMed De Amicis F, Perri A, Vizza D, et al. Epigallocatechin gallate inhibits growth and epithelial-to-mesenchymal transition in human thyroid carcinoma cell lines. J Cell Physiol. 2013;228:2054–62.CrossRefPubMed
44.
go back to reference Wu D, Si W, Wang M, et al. Hydrogen sulfide in cancer: friend or foe? Nitric Oxide. 2015;50:38–45.CrossRefPubMed Wu D, Si W, Wang M, et al. Hydrogen sulfide in cancer: friend or foe? Nitric Oxide. 2015;50:38–45.CrossRefPubMed
45.
go back to reference Palumbo T, Poultsides GA, Kouraklis G, et al. A functional microRNA library screen reveals miR-410 as a novel anti-apoptotic regulator of cholangiocarcinoma. BMC Cancer. 2016;16:353.CrossRefPubMedCentralPubMed Palumbo T, Poultsides GA, Kouraklis G, et al. A functional microRNA library screen reveals miR-410 as a novel anti-apoptotic regulator of cholangiocarcinoma. BMC Cancer. 2016;16:353.CrossRefPubMedCentralPubMed
46.
go back to reference Wu D, Gao Y, Qi Y, et al. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett. 2014;351:13–22.CrossRefPubMed Wu D, Gao Y, Qi Y, et al. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett. 2014;351:13–22.CrossRefPubMed
47.
go back to reference Ke FFS, Vanyai HK, Cowan AD, et al. Embryogenesis and adult life in the absence of intrinsic apoptosis effectors BAX, BAK, and BOK. Cell. 2018;173:1217–30.CrossRefPubMed Ke FFS, Vanyai HK, Cowan AD, et al. Embryogenesis and adult life in the absence of intrinsic apoptosis effectors BAX, BAK, and BOK. Cell. 2018;173:1217–30.CrossRefPubMed
48.
go back to reference Manohar M, Fatima I, Saxena R, et al. (–)-Epigallocatechin-3-gallate induces apoptosis in human endometrial adenocarcinoma cells via ROS generation and p38 MAP kinase activation. J Nutr Biochem. 2013;24:940–7.CrossRefPubMed Manohar M, Fatima I, Saxena R, et al. (–)-Epigallocatechin-3-gallate induces apoptosis in human endometrial adenocarcinoma cells via ROS generation and p38 MAP kinase activation. J Nutr Biochem. 2013;24:940–7.CrossRefPubMed
49.
go back to reference Hu Q, Chang X, Yan R, et al. (–)-Epigallocatechin-3-gallate induces cancer cell apoptosis via acetylation of amyloid precursor protein. Med Oncol. 2015;32:390.CrossRefPubMed Hu Q, Chang X, Yan R, et al. (–)-Epigallocatechin-3-gallate induces cancer cell apoptosis via acetylation of amyloid precursor protein. Med Oncol. 2015;32:390.CrossRefPubMed
50.
go back to reference Tang GQ, Yan TQ, Guo W, et al. (–)-Epigallocatechin-3-gallate induces apoptosis and suppresses proliferation by inhibiting the human Indian Hedgehog pathway in human chondrosarcoma cells. J Cancer Res Clin Oncol. 2010;136:1179–85.CrossRefPubMed Tang GQ, Yan TQ, Guo W, et al. (–)-Epigallocatechin-3-gallate induces apoptosis and suppresses proliferation by inhibiting the human Indian Hedgehog pathway in human chondrosarcoma cells. J Cancer Res Clin Oncol. 2010;136:1179–85.CrossRefPubMed
51.
go back to reference Wang J, Xie Y, Feng Y, et al. (–)-Epigallocatechingallate induces apoptosis in B lymphoma cells via caspase-dependent pathway and Bcl-2 family protein modulation. Int J Oncol. 2015;46:1507–15.CrossRefPubMedCentralPubMed Wang J, Xie Y, Feng Y, et al. (–)-Epigallocatechingallate induces apoptosis in B lymphoma cells via caspase-dependent pathway and Bcl-2 family protein modulation. Int J Oncol. 2015;46:1507–15.CrossRefPubMedCentralPubMed
52.
go back to reference Alessio N, Del Gaudio S, Capasso S, et al. Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process. Oncotarget. 2015;6:8155–66.CrossRefPubMed Alessio N, Del Gaudio S, Capasso S, et al. Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process. Oncotarget. 2015;6:8155–66.CrossRefPubMed
53.
go back to reference Mandal R, Becker S, Strebhardt K. Stamping out RAF and MEK1/2 to inhibit the ERK1/2 pathway: an emerging threat to anticancer therapy. Oncogene. 2016;35:2547–61.CrossRefPubMed Mandal R, Becker S, Strebhardt K. Stamping out RAF and MEK1/2 to inhibit the ERK1/2 pathway: an emerging threat to anticancer therapy. Oncogene. 2016;35:2547–61.CrossRefPubMed
54.
go back to reference Nichols RJ, Haderk F, Stahlhut C, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20:1064–73.CrossRefPubMedCentralPubMed Nichols RJ, Haderk F, Stahlhut C, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20:1064–73.CrossRefPubMedCentralPubMed
55.
go back to reference Gourlaouen M, Welti JC, Vasudev NS, et al. Essential role for endocytosis in the growth factor-stimulated activation of ERK1/2 in endothelial cells. J Biol Chem. 2013;288:7467–80.CrossRefPubMedCentralPubMed Gourlaouen M, Welti JC, Vasudev NS, et al. Essential role for endocytosis in the growth factor-stimulated activation of ERK1/2 in endothelial cells. J Biol Chem. 2013;288:7467–80.CrossRefPubMedCentralPubMed
56.
go back to reference Roskoski R Jr. Targeting oncogenic Raf protein-serine/threonine kinases in human cancers. Pharmacol Res. 2018;135:239–58.CrossRefPubMed Roskoski R Jr. Targeting oncogenic Raf protein-serine/threonine kinases in human cancers. Pharmacol Res. 2018;135:239–58.CrossRefPubMed
58.
go back to reference Koustas E, Karamouzis MV, Mihailidou C, et al. Co-targeting of EGFR and autophagy signaling is an emerging treatment strategy in metastatic colorectal cancer. Cancer Lett. 2017;396:94–102.CrossRefPubMed Koustas E, Karamouzis MV, Mihailidou C, et al. Co-targeting of EGFR and autophagy signaling is an emerging treatment strategy in metastatic colorectal cancer. Cancer Lett. 2017;396:94–102.CrossRefPubMed
59.
go back to reference Li K, Guo Q, Yang J, et al. FOXD3 is a tumor suppressor of colon cancer by inhibiting EGFR-Ras-Raf-MEK-ERK signal pathway. Oncotarget. 2017;8:5048–56.PubMed Li K, Guo Q, Yang J, et al. FOXD3 is a tumor suppressor of colon cancer by inhibiting EGFR-Ras-Raf-MEK-ERK signal pathway. Oncotarget. 2017;8:5048–56.PubMed
60.
go back to reference Ouyang L, Chen Y, Wang XY, et al. Polygonatum odoratum lectin induces apoptosis and autophagy via targeting EGFR-mediated Ras-Raf-MEK-ERK pathway in human MCF-7 breast cancer cells. Phytomedicine. 2014;21:1658–65.CrossRefPubMed Ouyang L, Chen Y, Wang XY, et al. Polygonatum odoratum lectin induces apoptosis and autophagy via targeting EGFR-mediated Ras-Raf-MEK-ERK pathway in human MCF-7 breast cancer cells. Phytomedicine. 2014;21:1658–65.CrossRefPubMed
61.
go back to reference Angelova M, Mlecnik B, Vasaturo A, et al. Evolution of metastases in space and time under immune selection. Cell. 2018;175:751–65.CrossRefPubMed Angelova M, Mlecnik B, Vasaturo A, et al. Evolution of metastases in space and time under immune selection. Cell. 2018;175:751–65.CrossRefPubMed
62.
go back to reference Liu Y, Jang S, Xie L, et al. Host deficiency in caveolin-2 inhibits lung carcinoma tumor growth by impairing tumor angiogenesis. Cancer Res. 2014;74:6452–62.CrossRefPubMedCentralPubMed Liu Y, Jang S, Xie L, et al. Host deficiency in caveolin-2 inhibits lung carcinoma tumor growth by impairing tumor angiogenesis. Cancer Res. 2014;74:6452–62.CrossRefPubMedCentralPubMed
Metadata
Title
Epigallocatechin-3-gallate inhibits the growth and increases the apoptosis of human thyroid carcinoma cells through suppression of EGFR/RAS/RAF/MEK/ERK signaling pathway
Authors
Dongdong Wu
Zhengguo Liu
Jianmei Li
Qianqian Zhang
Peiyu Zhong
Tieshan Teng
Mingliang Chen
Zhongwen Xie
Ailing Ji
Yanzhang Li
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2019
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-0762-9

Other articles of this Issue 1/2019

Cancer Cell International 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine