Skip to main content
Top
Published in: Cellular Oncology 5/2019

Open Access 01-10-2019 | Ovarian Cancer | Original paper

Apatinib inhibits glycolysis by suppressing the VEGFR2/AKT1/SOX5/GLUT4 signaling pathway in ovarian cancer cells

Authors: Lihua Chen, Xi Cheng, Wenzhi Tu, Zihao Qi, Haoran Li, Fei Liu, Yufei Yang, Zhe Zhang, Ziliang Wang

Published in: Cellular Oncology | Issue 5/2019

Login to get access

Abstract

Background

Apatinib is a tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor-2 (VEGFR2), and has shown encouraging therapeutic effects in various malignant tumors. As yet, however, the role of apatinib in ovarian cancer has remained unknown. Here, we sought to elucidate the role of apatinib in the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as in glucose metabolism in these cells.

Methods

The effects of apatinib on ovarian cancer cell viability and proliferation were assessed using Cell Counting Kit-8 (CCK-8) and colony formation assays, respectively. The expression of VEGFR2/AKT1/SOX5/GLUT4 pathway proteins was assessed using Western blotting, and glucose uptake and lactate production assays were used to detect glycolysis in ovarian cancer cells. SOX5 was exogenously over-expressed and silenced in ovarian cancer cells using expression vector and shRNA-based methods, respectively. RNA expression analyses were performed using RNA-seq and gene-chip-based methods. GLUT4 promoter activity was assessed using a dual-luciferase reporter assay. The expression of p-VEGFR2 (Tyr1175), p-AKT1 (Ser473), p-GSK3β (Ser9), SOX5 and GLUT4 in xenograft tissues was assessed using immunohistochemistry (IHC).

Results

We found that apatinib inhibited the in vitro and in vivo viability and proliferation in Hey and OVCA433 ovarian cancer cells in a dose-dependent and time-dependent manner. We also found that apatinib effectively suppressed glucose uptake and lactate production by blocking the expression of GLUT4 in these cells. In addition, we found that SOX5 predominantly rescued the inhibitory effect of apatinib on GLUT4 expression by activating its promoter. Finally, we found that apatinib regulated the expression of SOX5 by suppressing the VEGFR2/AKT1/GSK3β signaling pathway.

Conclusions

From our results, we conclude that apatinib suppresses the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as glycolysis by inhibiting the VEGFR2/AKT1/GSK3β/SOX5/GLUT4 signaling pathway. Apatinib may serve as a promising drug for the treatment of ovarian cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, D.M. Parkin, D. Forman, F. Bray, Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136, E359–E386 (2015)CrossRefPubMed J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, D.M. Parkin, D. Forman, F. Bray, Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136, E359–E386 (2015)CrossRefPubMed
2.
go back to reference A. Markowska, S. Sajdak, J. Markowska, A. Huczynski, Angiogenesis and cancer stem cells: New perspectives on therapy of ovarian cancer. Eur J Med Chem 142, 87–94 (2017)CrossRefPubMed A. Markowska, S. Sajdak, J. Markowska, A. Huczynski, Angiogenesis and cancer stem cells: New perspectives on therapy of ovarian cancer. Eur J Med Chem 142, 87–94 (2017)CrossRefPubMed
3.
go back to reference L.A. Torre, B. Trabert, C.E. DeSantis, K.D. Miller, G. Samimi, C.D. Runowicz, M.M. Gaudet, A. Jemal, R.L. Siegel, Ovarian cancer statistics, 2018. CA Cancer J Clin 68, 284–296 (2018)CrossRefPubMedPubMedCentral L.A. Torre, B. Trabert, C.E. DeSantis, K.D. Miller, G. Samimi, C.D. Runowicz, M.M. Gaudet, A. Jemal, R.L. Siegel, Ovarian cancer statistics, 2018. CA Cancer J Clin 68, 284–296 (2018)CrossRefPubMedPubMedCentral
4.
go back to reference K. El Bairi, A.H. Kandhro, A. Gouri, W. Mahfoud, N. Louanjli, B. Saadani, S. Afqir, M. Amrani, Emerging diagnostic, prognostic and therapeutic biomarkers for ovarian cancer. Cell Oncol 40, 105–118 (2017)CrossRef K. El Bairi, A.H. Kandhro, A. Gouri, W. Mahfoud, N. Louanjli, B. Saadani, S. Afqir, M. Amrani, Emerging diagnostic, prognostic and therapeutic biomarkers for ovarian cancer. Cell Oncol 40, 105–118 (2017)CrossRef
5.
go back to reference W.A. Spannuth, A.M. Nick, N.B. Jennings, G.N. Armaiz-Pena, L.S. Mangala, C.G. Danes, Y.G. Lin, W.M. Merritt, P.H. Thaker, A.A. Kamat, L.Y. Han, J.R. Tonra, R.L. Coleman, L.M. Ellis, A.K. Sood, Functional significance of VEGFR-2 on ovarian cancer cells. Int J Cancer 124, 1045–1053 (2009)CrossRefPubMedPubMedCentral W.A. Spannuth, A.M. Nick, N.B. Jennings, G.N. Armaiz-Pena, L.S. Mangala, C.G. Danes, Y.G. Lin, W.M. Merritt, P.H. Thaker, A.A. Kamat, L.Y. Han, J.R. Tonra, R.L. Coleman, L.M. Ellis, A.K. Sood, Functional significance of VEGFR-2 on ovarian cancer cells. Int J Cancer 124, 1045–1053 (2009)CrossRefPubMedPubMedCentral
6.
go back to reference D.J. Price, T. Miralem, S. Jiang, R. Steinberg, H. Avraham, Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ 12, 129–136 (2001) D.J. Price, T. Miralem, S. Jiang, R. Steinberg, H. Avraham, Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ 12, 129–136 (2001)
7.
go back to reference B.P. Kumar, S. Rajput, K.K. Dey, A. Parekh, S. Das, A. Mazumdar, M. Mandal, Celecoxib alleviates tamoxifen-instigated angiogenic effects by ROS-dependent VEGF/VEGFR2 autocrine signaling. BMC Cancer 13, 273 (2013)CrossRefPubMedPubMedCentral B.P. Kumar, S. Rajput, K.K. Dey, A. Parekh, S. Das, A. Mazumdar, M. Mandal, Celecoxib alleviates tamoxifen-instigated angiogenic effects by ROS-dependent VEGF/VEGFR2 autocrine signaling. BMC Cancer 13, 273 (2013)CrossRefPubMedPubMedCentral
8.
go back to reference S. Chatterjee, L.C. Heukamp, M. Siobal, J. Schottle, C. Wieczorek, M. Peifer, D. Frasca, M. Koker, K. Konig, L. Meder, D. Rauh, R. Buettner, J. Wolf, R.A. Brekken, B. Neumaier, G. Christofori, R.K. Thomas, R.T. Ullrich, Tumor VEGF:VEGFR2 autocrine feed-forward loop triggers angiogenesis in lung cancer. J Clin Invest 123, 1732–1740 (2013)CrossRefPubMedPubMedCentral S. Chatterjee, L.C. Heukamp, M. Siobal, J. Schottle, C. Wieczorek, M. Peifer, D. Frasca, M. Koker, K. Konig, L. Meder, D. Rauh, R. Buettner, J. Wolf, R.A. Brekken, B. Neumaier, G. Christofori, R.K. Thomas, R.T. Ullrich, Tumor VEGF:VEGFR2 autocrine feed-forward loop triggers angiogenesis in lung cancer. J Clin Invest 123, 1732–1740 (2013)CrossRefPubMedPubMedCentral
9.
go back to reference R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol Cancer Res 6, 1630–1638 (2008)CrossRefPubMed R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol Cancer Res 6, 1630–1638 (2008)CrossRefPubMed
10.
go back to reference F. Shan, R. Miao, K. Xue, Z. Li, Z. Li, Z. Bu, A. Wu, L. Zhang, X. Wu, X. Zong, X. Wang, S. Li, X. Ji, Z. Jia, Z. Li, J. Ji, Controlling angiogenesis in gastric cancer: A systematic review of anti-angiogenic trials. Cancer Lett 380, 598–607 (2016)CrossRefPubMed F. Shan, R. Miao, K. Xue, Z. Li, Z. Li, Z. Bu, A. Wu, L. Zhang, X. Wu, X. Zong, X. Wang, S. Li, X. Ji, Z. Jia, Z. Li, J. Ji, Controlling angiogenesis in gastric cancer: A systematic review of anti-angiogenic trials. Cancer Lett 380, 598–607 (2016)CrossRefPubMed
11.
go back to reference G. Roviello, A. Ravelli, K. Polom, R. Petrioli, L. Marano, D. Marrelli, F. Roviello, D. Generali, Apatinib: A novel receptor tyrosine kinase inhibitor for the treatment of gastric cancer. Cancer Lett 372, 187–191 (2016)CrossRefPubMed G. Roviello, A. Ravelli, K. Polom, R. Petrioli, L. Marano, D. Marrelli, F. Roviello, D. Generali, Apatinib: A novel receptor tyrosine kinase inhibitor for the treatment of gastric cancer. Cancer Lett 372, 187–191 (2016)CrossRefPubMed
12.
go back to reference R. Geng, J. Li, Apatinib for the treatment of gastric cancer. Expert Opin Pharmacother 16, 117–122 (2015)CrossRefPubMed R. Geng, J. Li, Apatinib for the treatment of gastric cancer. Expert Opin Pharmacother 16, 117–122 (2015)CrossRefPubMed
13.
go back to reference J. Li, S. Qin, J. Xu, J. Xiong, C. Wu, Y. Bai, W. Liu, J. Tong, Y. Liu, R. Xu, Z. Wang, Q. Wang, X. Ouyang, Y. Yang, Y. Ba, J. Liang, X. Lin, D. Luo, R. Zheng, X. Wang, G. Sun, L. Wang, L. Zheng, H. Guo, J. Wu, N. Xu, J. Yang, H. Zhang, Y. Cheng, N. Wang, L. Chen, Z. Fan, P. Sun, H. Yu, Randomized, double-blind, placebo-controlled phase III trial of Apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction. J Clin Oncol 34, 1448–1454 (2016)CrossRefPubMed J. Li, S. Qin, J. Xu, J. Xiong, C. Wu, Y. Bai, W. Liu, J. Tong, Y. Liu, R. Xu, Z. Wang, Q. Wang, X. Ouyang, Y. Yang, Y. Ba, J. Liang, X. Lin, D. Luo, R. Zheng, X. Wang, G. Sun, L. Wang, L. Zheng, H. Guo, J. Wu, N. Xu, J. Yang, H. Zhang, Y. Cheng, N. Wang, L. Chen, Z. Fan, P. Sun, H. Yu, Randomized, double-blind, placebo-controlled phase III trial of Apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction. J Clin Oncol 34, 1448–1454 (2016)CrossRefPubMed
14.
go back to reference X. Hu, J. Zhang, B. Xu, Z. Jiang, J. Ragaz, Z. Tong, Q. Zhang, X. Wang, J. Feng, D. Pang, M. Fan, J. Li, B. Wang, Z. Wang, Q. Zhang, S. Sun, C. Liao, Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer 135, 1961–1969 (2014)CrossRefPubMed X. Hu, J. Zhang, B. Xu, Z. Jiang, J. Ragaz, Z. Tong, Q. Zhang, X. Wang, J. Feng, D. Pang, M. Fan, J. Li, B. Wang, Z. Wang, Q. Zhang, S. Sun, C. Liao, Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer 135, 1961–1969 (2014)CrossRefPubMed
15.
go back to reference M. Zhang, Z. Tian, Y. Sun, Successful treatment of ovarian cancer with apatinib combined with chemotherapy: A case report. Medicine 96, e8570 (2017)CrossRefPubMedPubMedCentral M. Zhang, Z. Tian, Y. Sun, Successful treatment of ovarian cancer with apatinib combined with chemotherapy: A case report. Medicine 96, e8570 (2017)CrossRefPubMedPubMedCentral
16.
go back to reference L. Deng, Y. Wang, W. Lu, Q. Liu, J. Wu, J. Jin, Apatinib treatment combined with chemotherapy for advanced epithelial ovarian cancer: A case report. Onco Targets Ther 10, 1521–1525 (2017)CrossRefPubMedPubMedCentral L. Deng, Y. Wang, W. Lu, Q. Liu, J. Wu, J. Jin, Apatinib treatment combined with chemotherapy for advanced epithelial ovarian cancer: A case report. Onco Targets Ther 10, 1521–1525 (2017)CrossRefPubMedPubMedCentral
17.
go back to reference M. Miao, G. Deng, S. Luo, J. Zhou, L. Chen, J. Yang, J. He, J. Li, J. Yao, S. Tan, J. Tang, A phase II study of apatinib in patients with recurrent epithelial ovarian cancer. Gynecol Oncol 148, 286–290 (2018)CrossRefPubMed M. Miao, G. Deng, S. Luo, J. Zhou, L. Chen, J. Yang, J. He, J. Li, J. Yao, S. Tan, J. Tang, A phase II study of apatinib in patients with recurrent epithelial ovarian cancer. Gynecol Oncol 148, 286–290 (2018)CrossRefPubMed
18.
go back to reference C.-Y. Lan, Y. Wang, Y. Xiong, J.-D. Li, J.-X. Shen, Y.-F. Li, M. Zheng, Y.-N. Zhang, Y.-L. Feng, Q. Liu, H.-Q. Huang, X. Huang, Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): A phase 2, single-arm, prospective study. Lancet Oncol 19, 1239–1246 (2018)CrossRefPubMed C.-Y. Lan, Y. Wang, Y. Xiong, J.-D. Li, J.-X. Shen, Y.-F. Li, M. Zheng, Y.-N. Zhang, Y.-L. Feng, Q. Liu, H.-Q. Huang, X. Huang, Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): A phase 2, single-arm, prospective study. Lancet Oncol 19, 1239–1246 (2018)CrossRefPubMed
19.
go back to reference I. Kurtsdotter, D. Topcic, A. Karlen, B. Singla, D.W. Hagey, M. Bergsland, P. Siesjo, M. Nister, J.W. Carlson, V. Lefebvre, O. Persson, J. Holmberg, J. Muhr, SOX5/6/21 prevent oncogene-driven transformation of brain stem cells. Cancer Res 77, 4985–4997 (2017)CrossRefPubMed I. Kurtsdotter, D. Topcic, A. Karlen, B. Singla, D.W. Hagey, M. Bergsland, P. Siesjo, M. Nister, J.W. Carlson, V. Lefebvre, O. Persson, J. Holmberg, J. Muhr, SOX5/6/21 prevent oncogene-driven transformation of brain stem cells. Cancer Res 77, 4985–4997 (2017)CrossRefPubMed
20.
go back to reference E. Tchougounova, Y. Jiang, D. Brasater, N. Lindberg, M. Kastemar, A. Asplund, B. Westermark, L. Uhrbom, Sox5 can suppress platelet-derived growth factor B-induced glioma development in Ink4a-deficient mice through induction of acute cellular senescence. Oncogene. 28, 1537–1548 (2009)CrossRefPubMed E. Tchougounova, Y. Jiang, D. Brasater, N. Lindberg, M. Kastemar, A. Asplund, B. Westermark, L. Uhrbom, Sox5 can suppress platelet-derived growth factor B-induced glioma development in Ink4a-deficient mice through induction of acute cellular senescence. Oncogene. 28, 1537–1548 (2009)CrossRefPubMed
21.
go back to reference H. Zou, S. Wang, S. Wang, H. Wu, J. Yu, Q. Chen, W. Cui, Y. Yuan, X. Wen, J. He, SOX5 interacts with YAP1 to drive malignant potential of non-small cell lung cancer cells. Am J Cancer Res 8, 866 (2018)PubMedPubMedCentral H. Zou, S. Wang, S. Wang, H. Wu, J. Yu, Q. Chen, W. Cui, Y. Yuan, X. Wen, J. He, SOX5 interacts with YAP1 to drive malignant potential of non-small cell lung cancer cells. Am J Cancer Res 8, 866 (2018)PubMedPubMedCentral
22.
go back to reference A.S. Axelsson, T. Mahdi, H.A. Nenonen, T. Singh, S. Hanzelmann, A. Wendt, A. Bagge, T.M. Reinbothe, J. Millstein, X. Yang, B. Zhang, E.G. Gusmao, L. Shu, M. Szabat, Y. Tang, J. Wang, S. Salo, L. Eliasson, I. Artner, M. Fex, J.D. Johnson, C.B. Wollheim, J.M.J. Derry, B. Mecham, P. Spegel, H. Mulder, I.G. Costa, E. Zhang, A.H. Rosengren, Sox5 regulates beta-cell phenotype and is reduced in type 2 diabetes. Nat Commun 8, 15652 (2017)CrossRefPubMedPubMedCentral A.S. Axelsson, T. Mahdi, H.A. Nenonen, T. Singh, S. Hanzelmann, A. Wendt, A. Bagge, T.M. Reinbothe, J. Millstein, X. Yang, B. Zhang, E.G. Gusmao, L. Shu, M. Szabat, Y. Tang, J. Wang, S. Salo, L. Eliasson, I. Artner, M. Fex, J.D. Johnson, C.B. Wollheim, J.M.J. Derry, B. Mecham, P. Spegel, H. Mulder, I.G. Costa, E. Zhang, A.H. Rosengren, Sox5 regulates beta-cell phenotype and is reduced in type 2 diabetes. Nat Commun 8, 15652 (2017)CrossRefPubMedPubMedCentral
23.
go back to reference T.Y. Shi, L. Yang, G. Yang, X.Y. Tu, X. Wu, X. Cheng, Q. Wei, DNA polymerase zeta as a potential biomarker of chemoradiation resistance and poor prognosis for cervical cancer. Med Oncol 30, 500 (2013)CrossRefPubMed T.Y. Shi, L. Yang, G. Yang, X.Y. Tu, X. Wu, X. Cheng, Q. Wei, DNA polymerase zeta as a potential biomarker of chemoradiation resistance and poor prognosis for cervical cancer. Med Oncol 30, 500 (2013)CrossRefPubMed
24.
go back to reference H. Li, Y. Lu, Y. Pang, M. Li, X. Cheng, J. Chen, Propofol enhances the cisplatin-induced apoptosis on cervical cancer cells via EGFR/JAK2/STAT3 pathway. Biomed Pharmacother 86, 324–333 (2017)CrossRefPubMed H. Li, Y. Lu, Y. Pang, M. Li, X. Cheng, J. Chen, Propofol enhances the cisplatin-induced apoptosis on cervical cancer cells via EGFR/JAK2/STAT3 pathway. Biomed Pharmacother 86, 324–333 (2017)CrossRefPubMed
25.
26.
go back to reference G. van Niekerk, A.M. Engelbrecht, Role of PKM2 in directing the metabolic fate of glucose in cancer: A potential therapeutic target. Cell Oncol 41, 343–351 (2018)CrossRef G. van Niekerk, A.M. Engelbrecht, Role of PKM2 in directing the metabolic fate of glucose in cancer: A potential therapeutic target. Cell Oncol 41, 343–351 (2018)CrossRef
27.
go back to reference M.G. Vander Heiden, L.C. Cantley, C.B. Thompson, Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324, 1029–1033 (2009)CrossRefPubMedPubMedCentral M.G. Vander Heiden, L.C. Cantley, C.B. Thompson, Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324, 1029–1033 (2009)CrossRefPubMedPubMedCentral
28.
go back to reference J.G. Zhao, K.M. Ren, J. Tang, Overcoming 5-Fu resistance in human non-small cell lung cancer cells by the combination of 5-Fu and cisplatin through the inhibition of glucose metabolism. Tumour Biol 35, 12305–12315 (2014)CrossRefPubMed J.G. Zhao, K.M. Ren, J. Tang, Overcoming 5-Fu resistance in human non-small cell lung cancer cells by the combination of 5-Fu and cisplatin through the inhibition of glucose metabolism. Tumour Biol 35, 12305–12315 (2014)CrossRefPubMed
29.
go back to reference D.A. Tennant, R.V. Duran, E. Gottlieb, Targeting metabolic transformation for cancer therapy. Nat Rev Cancer 10, 267–277 (2010)CrossRefPubMed D.A. Tennant, R.V. Duran, E. Gottlieb, Targeting metabolic transformation for cancer therapy. Nat Rev Cancer 10, 267–277 (2010)CrossRefPubMed
30.
go back to reference X. Hu, J. Cao, W. Hu, C. Wu, Y. Pan, L. Cai, Z. Tong, S. Wang, J. Li, Z. Wang, Multicenter phase II study of Apatinib in non-triple-negative metastatic breast cancer. BMC Cancer 14, 820 (2014)CrossRefPubMedPubMedCentral X. Hu, J. Cao, W. Hu, C. Wu, Y. Pan, L. Cai, Z. Tong, S. Wang, J. Li, Z. Wang, Multicenter phase II study of Apatinib in non-triple-negative metastatic breast cancer. BMC Cancer 14, 820 (2014)CrossRefPubMedPubMedCentral
31.
go back to reference J. Li, S. Qin, J. Xu, W. Guo, J. Xiong, Y. Bai, G. Sun, Y. Yang, L. Wang, N. Xu, Y. Cheng, Z. Wang, L. Zheng, M. Tao, X. Zhu, D. Ji, X. Liu, H. Yu, Apatinib for chemotherapy-refractory advanced metastatic gastric cancer: Results from a randomized, placebo-controlled, parallel-arm, phase II trial. J Clin Oncol 31, 3219–3225 (2013)CrossRefPubMed J. Li, S. Qin, J. Xu, W. Guo, J. Xiong, Y. Bai, G. Sun, Y. Yang, L. Wang, N. Xu, Y. Cheng, Z. Wang, L. Zheng, M. Tao, X. Zhu, D. Ji, X. Liu, H. Yu, Apatinib for chemotherapy-refractory advanced metastatic gastric cancer: Results from a randomized, placebo-controlled, parallel-arm, phase II trial. J Clin Oncol 31, 3219–3225 (2013)CrossRefPubMed
32.
go back to reference S. Tian, H. Quan, C. Xie, H. Guo, F. Lü, Y. Xu, J. Li, L. Lou, YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci 102, 1374–1380 (2011)CrossRefPubMed S. Tian, H. Quan, C. Xie, H. Guo, F. Lü, Y. Xu, J. Li, L. Lou, YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci 102, 1374–1380 (2011)CrossRefPubMed
33.
go back to reference F. Li, Z. Liao, C. Zhang, J. Zhao, R. Xing, S. Teng, J. Zhang, Y. Yang, J. Yang, Apatinib as targeted therapy for sarcoma. Oncotarget 9, 24548 (2018)PubMedPubMedCentral F. Li, Z. Liao, C. Zhang, J. Zhao, R. Xing, S. Teng, J. Zhang, Y. Yang, J. Yang, Apatinib as targeted therapy for sarcoma. Oncotarget 9, 24548 (2018)PubMedPubMedCentral
34.
go back to reference X.-z. Tong, F. Wang, S. Liang, X. Zhang, J.-h. He, X.-G. Chen, Y.-j. Liang, Y.-j. Mi, K.K.W. To, L.-w. Fu, Apatinib (YN968D1) enhances the efficacy of conventional chemotherapeutical drugs in side population cells and ABCB1-overexpressing leukemia cells. Biochem Pharmacol 83, 586–597 (2012)CrossRefPubMed X.-z. Tong, F. Wang, S. Liang, X. Zhang, J.-h. He, X.-G. Chen, Y.-j. Liang, Y.-j. Mi, K.K.W. To, L.-w. Fu, Apatinib (YN968D1) enhances the efficacy of conventional chemotherapeutical drugs in side population cells and ABCB1-overexpressing leukemia cells. Biochem Pharmacol 83, 586–597 (2012)CrossRefPubMed
35.
go back to reference Y.-j. Mi, Y.-j. Liang, H.-b. Huang, H.-y. Zhao, C.-P. Wu, F. Wang, L.-y. Tao, C.-z. Zhang, C.-L. Dai, A.K. Tiwari, Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux function of multiple ATP-binding cassette transporters. Cancer Res 70, 7981–7991 (2010)CrossRefPubMedPubMedCentral Y.-j. Mi, Y.-j. Liang, H.-b. Huang, H.-y. Zhao, C.-P. Wu, F. Wang, L.-y. Tao, C.-z. Zhang, C.-L. Dai, A.K. Tiwari, Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux function of multiple ATP-binding cassette transporters. Cancer Res 70, 7981–7991 (2010)CrossRefPubMedPubMedCentral
36.
go back to reference B. Bhattacharya, S.H. Low, C. Soh, N. Kamal Mustapa, M. Beloueche-Babari, K.X. Koh, J. Loh, R. Soong, Increased drug resistance is associated with reduced glucose levels and an enhanced glycolysis phenotype. Br J Pharmacol 171, 3255–3267 (2014)CrossRefPubMedPubMedCentral B. Bhattacharya, S.H. Low, C. Soh, N. Kamal Mustapa, M. Beloueche-Babari, K.X. Koh, J. Loh, R. Soong, Increased drug resistance is associated with reduced glucose levels and an enhanced glycolysis phenotype. Br J Pharmacol 171, 3255–3267 (2014)CrossRefPubMedPubMedCentral
37.
go back to reference T. Egawa-Takata, H. Endo, M. Fujita, Y. Ueda, T. Miyatake, H. Okuyama, K. Yoshino, S. Kamiura, T. Enomoto, T. Kimura, M. Inoue, Early reduction of glucose uptake after cisplatin treatment is a marker of cisplatin sensitivity in ovarian cancer. Cancer Sci 101, 2171–2178 (2010)CrossRefPubMed T. Egawa-Takata, H. Endo, M. Fujita, Y. Ueda, T. Miyatake, H. Okuyama, K. Yoshino, S. Kamiura, T. Enomoto, T. Kimura, M. Inoue, Early reduction of glucose uptake after cisplatin treatment is a marker of cisplatin sensitivity in ovarian cancer. Cancer Sci 101, 2171–2178 (2010)CrossRefPubMed
38.
go back to reference K. Liu, T. Ren, Y. Huang, K. Sun, X. Bao, S. Wang, B. Zheng, W. Guo, Apatinib promotes autophagy and apoptosis through VEGFR2/STAT3/BCL-2 signaling in osteosarcoma. Cell Death Dis 8, e3015 (2017)CrossRefPubMedPubMedCentral K. Liu, T. Ren, Y. Huang, K. Sun, X. Bao, S. Wang, B. Zheng, W. Guo, Apatinib promotes autophagy and apoptosis through VEGFR2/STAT3/BCL-2 signaling in osteosarcoma. Cell Death Dis 8, e3015 (2017)CrossRefPubMedPubMedCentral
39.
go back to reference X. Cheng, H. Feng, H. Wu, Z. Jin, X. Shen, J. Kuang, Z. Huo, X. Chen, H. Gao, F. Ye, X. Ji, X. Jing, Y. Zhang, T. Zhang, W. Qiu, R. Zhao, Targeting autophagy enhances apatinib-induced apoptosis via endoplasmic reticulum stress for human colorectal cancer. Cancer Lett 431, 105–114 (2018)CrossRefPubMed X. Cheng, H. Feng, H. Wu, Z. Jin, X. Shen, J. Kuang, Z. Huo, X. Chen, H. Gao, F. Ye, X. Ji, X. Jing, Y. Zhang, T. Zhang, W. Qiu, R. Zhao, Targeting autophagy enhances apatinib-induced apoptosis via endoplasmic reticulum stress for human colorectal cancer. Cancer Lett 431, 105–114 (2018)CrossRefPubMed
40.
go back to reference J. Hu, J. Tian, S. Zhu, L. Sun, J. Yu, H. Tian, Q. Dong, Q. Luo, N. Jiang, Y. Niu, Z. Shang, Sox5 contributes to prostate cancer metastasis and is a master regulator of TGF-beta-induced epithelial mesenchymal transition through controlling Twist1 expression. Br J Cancer 118, 88–97 (2018)CrossRefPubMed J. Hu, J. Tian, S. Zhu, L. Sun, J. Yu, H. Tian, Q. Dong, Q. Luo, N. Jiang, Y. Niu, Z. Shang, Sox5 contributes to prostate cancer metastasis and is a master regulator of TGF-beta-induced epithelial mesenchymal transition through controlling Twist1 expression. Br J Cancer 118, 88–97 (2018)CrossRefPubMed
41.
go back to reference W. Renjie, L. Haiqian, MiR-132, miR-15a and miR-16 synergistically inhibit pituitary tumor cell proliferation, invasion and migration by targeting Sox5. Cancer Lett 356, 568–578 (2015)CrossRefPubMed W. Renjie, L. Haiqian, MiR-132, miR-15a and miR-16 synergistically inhibit pituitary tumor cell proliferation, invasion and migration by targeting Sox5. Cancer Lett 356, 568–578 (2015)CrossRefPubMed
Metadata
Title
Apatinib inhibits glycolysis by suppressing the VEGFR2/AKT1/SOX5/GLUT4 signaling pathway in ovarian cancer cells
Authors
Lihua Chen
Xi Cheng
Wenzhi Tu
Zihao Qi
Haoran Li
Fei Liu
Yufei Yang
Zhe Zhang
Ziliang Wang
Publication date
01-10-2019
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 5/2019
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00455-x

Other articles of this Issue 5/2019

Cellular Oncology 5/2019 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