Skip to main content
Erschienen in: Gastric Cancer 6/2020

02.07.2020 | Original Article

Resistance to FGFR1-targeted therapy leads to autophagy via TAK1/AMPK activation in gastric cancer

verfasst von: Rui Peng, Yan Chen, Liangnian Wei, Gang Li, Dongju Feng, Siru Liu, Runqiu Jiang, Shaojiang Zheng, Yun Chen

Erschienen in: Gastric Cancer | Ausgabe 6/2020

Einloggen, um Zugang zu erhalten

Abstract

Background

Fibroblast growth factor receptor 1 (FGFR1) is frequently dysregulated in various tumors. FGFR inhibitors have shown promising therapeutic value in several preclinical models. However, tumors resistant to FGFR inhibitors have emerged, compromising therapeutic outcomes by demonstrating markedly aggressive metastatic progression; however, the underlying signaling mechanism of resistance remains unknown.

Methods

We established FGFR inhibitor-resistant cell models using two gastric cancer (GC) cell lines, MGC-803 and BGC-823. RNA-seq was performed to determine the continuous cellular transcriptome changes between parental and resistant cells. We explored the mechanism of resistance to FGFR inhibitor, using a subcutaneous tumor model and GC patient-derived tumor organotypic culture.

Results

We observed that FGFR1 was highly expressed in GC and FGFR1 inhibitor-resistant cell lines, demonstrating elevated levels of autophagic activity. These resistant cells were characterized by epithelial-mesenchymal transition (EMT) required to facilitate metastatic outgrowth. In drug-resistant cells, the FGFR1 inhibitor regulated GC cell autophagy via AMPK/mTOR signal activation, which could be blocked using either pharmacological inhibitors or essential gene knockdown. Furthermore, TGF-β-activated kinase 1 (TAK1) amplification and metabolic restrictions led to AMPK pathway activation and autophagy. In vitro and in vivo results demonstrated that the FGFR inhibitor AZD4547 and TAK1 inhibitor NG25 synergistically inhibited proliferation and autophagy in AZD4547-resistant cell lines and patient-derived GC organotypic cultures.

Conclusions

We elucidated the molecular mechanisms underlying primary resistance to FGFR1 inhibitors in GC, and revealed that the inhibition of FGFR1 and TAK1 signaling could present a potential novel therapeutic strategy for FGFR1 inhibitor-resistant GC patients.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 2013;4:e838.PubMedPubMedCentralCrossRef Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 2013;4:e838.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Schafer MH, Lingohr P, Strasser A, Lehnen NC, Braun M, Perner S, et al. Fibroblast growth factor receptor 1 gene amplification in gastric adenocarcinoma. Hum Pathol. 2015;46(10):1488–95.PubMedCrossRef Schafer MH, Lingohr P, Strasser A, Lehnen NC, Braun M, Perner S, et al. Fibroblast growth factor receptor 1 gene amplification in gastric adenocarcinoma. Hum Pathol. 2015;46(10):1488–95.PubMedCrossRef
3.
Zurück zum Zitat Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer. 2010;10(2):116–29.PubMedCrossRef Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer. 2010;10(2):116–29.PubMedCrossRef
4.
Zurück zum Zitat Ying S, Du X, Fu W, Yun D, Chen L, Cai Y, et al. Synthesis, biological evaluation, QSAR and molecular dynamics simulation studies of potential fibroblast growth factor receptor 1 inhibitors for the treatment of gastric cancer. Eur J Med Chem. 2017;127:885–99.PubMedCrossRef Ying S, Du X, Fu W, Yun D, Chen L, Cai Y, et al. Synthesis, biological evaluation, QSAR and molecular dynamics simulation studies of potential fibroblast growth factor receptor 1 inhibitors for the treatment of gastric cancer. Eur J Med Chem. 2017;127:885–99.PubMedCrossRef
5.
Zurück zum Zitat Carter EP, Fearon AE, Grose RP. Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction. Trends Cell Biol. 2015;25(4):221–33.PubMedCrossRef Carter EP, Fearon AE, Grose RP. Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction. Trends Cell Biol. 2015;25(4):221–33.PubMedCrossRef
6.
Zurück zum Zitat Liang G, Liu Z, Wu J, Cai Y, Li X. Anticancer molecules targeting fibroblast growth factor receptors. Trends Pharmacol Sci. 2012;33(10):531–41.PubMedCrossRef Liang G, Liu Z, Wu J, Cai Y, Li X. Anticancer molecules targeting fibroblast growth factor receptors. Trends Pharmacol Sci. 2012;33(10):531–41.PubMedCrossRef
7.
Zurück zum Zitat Touat M, Ileana E, Postel-Vinay S, Andre F, Soria JC. Targeting FGFR signaling in cancer. Clin Cancer Res. 2015;21(12):2684–94.PubMedCrossRef Touat M, Ileana E, Postel-Vinay S, Andre F, Soria JC. Targeting FGFR signaling in cancer. Clin Cancer Res. 2015;21(12):2684–94.PubMedCrossRef
8.
Zurück zum Zitat Xue WJ, Li MT, Chen L, Sun LP, Li YY. Recent developments and advances of FGFR as a potential target in cancer. Future Med Chem. 2018;10(17):2109–26.PubMedCrossRef Xue WJ, Li MT, Chen L, Sun LP, Li YY. Recent developments and advances of FGFR as a potential target in cancer. Future Med Chem. 2018;10(17):2109–26.PubMedCrossRef
9.
Zurück zum Zitat Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, et al. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol. 2014;25(3):552–63.PubMedCrossRef Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, et al. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol. 2014;25(3):552–63.PubMedCrossRef
10.
Zurück zum Zitat Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2(62):62–93.CrossRef Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2(62):62–93.CrossRef
11.
Zurück zum Zitat Freier K, Schwaenen C, Sticht C, Flechtenmacher C, Muhling J, Hofele C, et al. Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral Oncol. 2007;43(1):60–6.PubMedCrossRef Freier K, Schwaenen C, Sticht C, Flechtenmacher C, Muhling J, Hofele C, et al. Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral Oncol. 2007;43(1):60–6.PubMedCrossRef
12.
Zurück zum Zitat Elbauomy Elsheikh S, Green AR, Lambros MB, Turner NC, Grainge MJ, Powe D, et al. FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Res. 2007;9(2):R23.PubMedPubMedCentralCrossRef Elbauomy Elsheikh S, Green AR, Lambros MB, Turner NC, Grainge MJ, Powe D, et al. FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Res. 2007;9(2):R23.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Theillet C, Adelaide J, Louason G, Bonnet-Dorion F, Jacquemier J, Adnane J, et al. FGFRI and PLAT genes and DNA amplification at 8p12 in breast and ovarian cancers. Genes Chromosomes Cancer. 1993;7(4):219–26.PubMedCrossRef Theillet C, Adelaide J, Louason G, Bonnet-Dorion F, Jacquemier J, Adnane J, et al. FGFRI and PLAT genes and DNA amplification at 8p12 in breast and ovarian cancers. Genes Chromosomes Cancer. 1993;7(4):219–26.PubMedCrossRef
14.
Zurück zum Zitat Simon R, Richter J, Wagner U, Fijan A, Bruderer J, Schmid U, et al. High-throughput tissue microarray analysis of 3p25 (RAF1) and 8p12 (FGFR1) copy number alterations in urinary bladder cancer. Cancer Res. 2001;61(11):4514–9.PubMed Simon R, Richter J, Wagner U, Fijan A, Bruderer J, Schmid U, et al. High-throughput tissue microarray analysis of 3p25 (RAF1) and 8p12 (FGFR1) copy number alterations in urinary bladder cancer. Cancer Res. 2001;61(11):4514–9.PubMed
15.
Zurück zum Zitat Lehnen NC, von Massenhausen A, Kalthoff H, Zhou H, Glowka T, Schutte U, et al. Fibroblast growth factor receptor 1 gene amplification in pancreatic ductal adenocarcinoma. Histopathology. 2013;63(2):157–66.PubMedCrossRef Lehnen NC, von Massenhausen A, Kalthoff H, Zhou H, Glowka T, Schutte U, et al. Fibroblast growth factor receptor 1 gene amplification in pancreatic ductal adenocarcinoma. Histopathology. 2013;63(2):157–66.PubMedCrossRef
17.
Zurück zum Zitat Yu L, Chen Y, Tooze SA. Autophagy pathway: cellular and molecular mechanisms. Autophagy. 2018;14(2):207–15.PubMedCrossRef Yu L, Chen Y, Tooze SA. Autophagy pathway: cellular and molecular mechanisms. Autophagy. 2018;14(2):207–15.PubMedCrossRef
18.
Zurück zum Zitat Din FV, Valanciute A, Houde VP, Zibrova D, Green KA, Sakamoto K, et al. Aspirin inhibits mTOR signaling, activates AMP-activated protein kinase, and induces autophagy in colorectal cancer cells. Gastroenterology. 2012;142(7):1504–15 (e3).PubMedCrossRef Din FV, Valanciute A, Houde VP, Zibrova D, Green KA, Sakamoto K, et al. Aspirin inhibits mTOR signaling, activates AMP-activated protein kinase, and induces autophagy in colorectal cancer cells. Gastroenterology. 2012;142(7):1504–15 (e3).PubMedCrossRef
19.
Zurück zum Zitat Tsuchihara K, Fujii S, Esumi H. Autophagy and cancer: dynamism of the metabolism of tumor cells and tissues. Cancer Lett. 2009;278(2):130–8.PubMedCrossRef Tsuchihara K, Fujii S, Esumi H. Autophagy and cancer: dynamism of the metabolism of tumor cells and tissues. Cancer Lett. 2009;278(2):130–8.PubMedCrossRef
20.
Zurück zum Zitat Ajibade AA, Wang HY, Wang RF. Cell type-specific function of TAK1 in innate immune signaling. Trends Immunol. 2013;34(7):307–16.PubMedCrossRef Ajibade AA, Wang HY, Wang RF. Cell type-specific function of TAK1 in innate immune signaling. Trends Immunol. 2013;34(7):307–16.PubMedCrossRef
21.
Zurück zum Zitat Sakurai H. Targeting of TAK1 in inflammatory disorders and cancer. Trends Pharmacol Sci. 2012;33(10):522–30.PubMedCrossRef Sakurai H. Targeting of TAK1 in inflammatory disorders and cancer. Trends Pharmacol Sci. 2012;33(10):522–30.PubMedCrossRef
23.
Zurück zum Zitat Herrero-Martin G, Hoyer-Hansen M, Garcia-Garcia C, Fumarola C, Farkas T, Lopez-Rivas A, et al. TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells. EMBO J. 2009;28(6):677–85.PubMedPubMedCentralCrossRef Herrero-Martin G, Hoyer-Hansen M, Garcia-Garcia C, Fumarola C, Farkas T, Lopez-Rivas A, et al. TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells. EMBO J. 2009;28(6):677–85.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Inokuchi-Shimizu S, Park EJ, Roh YS, Yang L, Zhang B, Song J, et al. TAK1-mediated autophagy and fatty acid oxidation prevent hepatosteatosis and tumorigenesis. J Clin Invest. 2014;124(8):3566–78.PubMedPubMedCentralCrossRef Inokuchi-Shimizu S, Park EJ, Roh YS, Yang L, Zhang B, Song J, et al. TAK1-mediated autophagy and fatty acid oxidation prevent hepatosteatosis and tumorigenesis. J Clin Invest. 2014;124(8):3566–78.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Yuan K, Huang C, Fox J, Laturnus D, Carlson E, Zhang B, et al. Autophagy plays an essential role in the clearance of pseudomonas aeruginosa by alveolar macrophages. J Cell Sci. 2012;125(Pt 2):507–15.PubMedPubMedCentralCrossRef Yuan K, Huang C, Fox J, Laturnus D, Carlson E, Zhang B, et al. Autophagy plays an essential role in the clearance of pseudomonas aeruginosa by alveolar macrophages. J Cell Sci. 2012;125(Pt 2):507–15.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Krishan S, Richardson DR, Sahni S. Gene of the month. AMP kinase (PRKAA1). J Clin Pathol. 2014;67(9):758–63.PubMedCrossRef Krishan S, Richardson DR, Sahni S. Gene of the month. AMP kinase (PRKAA1). J Clin Pathol. 2014;67(9):758–63.PubMedCrossRef
27.
Zurück zum Zitat Nakanishi Y, Mizuno H, Sase H, Fujii T, Sakata K, Akiyama N, et al. ERK signal suppression and sensitivity to CH5183284/Debio 1347, a selective FGFR inhibitor. Mol Cancer Ther. 2015;14(12):2831–9.PubMedCrossRef Nakanishi Y, Mizuno H, Sase H, Fujii T, Sakata K, Akiyama N, et al. ERK signal suppression and sensitivity to CH5183284/Debio 1347, a selective FGFR inhibitor. Mol Cancer Ther. 2015;14(12):2831–9.PubMedCrossRef
28.
Zurück zum Zitat Raoof S, Mulford IJ, Frisco-Cabanos H, Nangia V, Timonina D, Labrot E, et al. Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer. Oncogene. 2019;38(37):6399–413.PubMedPubMedCentralCrossRef Raoof S, Mulford IJ, Frisco-Cabanos H, Nangia V, Timonina D, Labrot E, et al. Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer. Oncogene. 2019;38(37):6399–413.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zhao Z, Wang L, Bartom E, Marshall S, Rendleman E, Ryan C, et al. Beta-catenin/Tcf7l2-dependent transcriptional regulation of GLUT1 gene expression by Zic family proteins in colon cancer. Sci Adv. 2019;5(7):eaax0698.PubMedPubMedCentralCrossRef Zhao Z, Wang L, Bartom E, Marshall S, Rendleman E, Ryan C, et al. Beta-catenin/Tcf7l2-dependent transcriptional regulation of GLUT1 gene expression by Zic family proteins in colon cancer. Sci Adv. 2019;5(7):eaax0698.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Liu W, Jiang Y, Sun J, Geng S, Pan Z, Prinz RA, et al. Activation of TGF-beta-activated kinase 1 (TAK1) restricts salmonella typhimurium growth by inducing AMPK activation and autophagy. Cell Death Dis. 2018;9(5):570.PubMedPubMedCentralCrossRef Liu W, Jiang Y, Sun J, Geng S, Pan Z, Prinz RA, et al. Activation of TGF-beta-activated kinase 1 (TAK1) restricts salmonella typhimurium growth by inducing AMPK activation and autophagy. Cell Death Dis. 2018;9(5):570.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Alers S, Loffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32(1):2–11.PubMedPubMedCentralCrossRef Alers S, Loffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32(1):2–11.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Tripathi V, Shin JH, Stuelten CH, Zhang YE. TGF-beta-induced alternative splicing of TAK1 promotes EMT and drug resistance. Oncogene. 2019;38(17):3185–200.PubMedPubMedCentralCrossRef Tripathi V, Shin JH, Stuelten CH, Zhang YE. TGF-beta-induced alternative splicing of TAK1 promotes EMT and drug resistance. Oncogene. 2019;38(17):3185–200.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Wu J, Du X, Li W, Zhou Y, Bai E, Kang Y, et al. A novel non-ATP competitive FGFR1 inhibitor with therapeutic potential on gastric cancer through inhibition of cell proliferation, survival and migration. Apoptosis. 2017;22(6):852–64.PubMedCrossRef Wu J, Du X, Li W, Zhou Y, Bai E, Kang Y, et al. A novel non-ATP competitive FGFR1 inhibitor with therapeutic potential on gastric cancer through inhibition of cell proliferation, survival and migration. Apoptosis. 2017;22(6):852–64.PubMedCrossRef
35.
Zurück zum Zitat Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472–6.PubMedPubMedCentralCrossRef Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472–6.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527(7579):525–30.PubMedPubMedCentralCrossRef Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527(7579):525–30.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Kitai H, Ebi H, Tomida S, Floros KV, Kotani H, Adachi Y, et al. Epithelial-to-mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS-mutant lung cancer. Cancer Discov. 2016;6(7):754–69.PubMedPubMedCentralCrossRef Kitai H, Ebi H, Tomida S, Floros KV, Kotani H, Adachi Y, et al. Epithelial-to-mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS-mutant lung cancer. Cancer Discov. 2016;6(7):754–69.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov. 2016;6(10):1118–33.PubMedPubMedCentralCrossRef Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov. 2016;6(10):1118–33.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117(2):326–36.PubMedPubMedCentralCrossRef Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117(2):326–36.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Liu D, Yang Y, Liu Q, Wang J. Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells. Med Oncol. 2011;28(1):105–11.PubMedCrossRef Liu D, Yang Y, Liu Q, Wang J. Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells. Med Oncol. 2011;28(1):105–11.PubMedCrossRef
41.
Zurück zum Zitat Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68(5):1485–94.PubMedCrossRef Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68(5):1485–94.PubMedCrossRef
42.
Zurück zum Zitat Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, et al. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int J Cancer. 2009;124(5):1060–71.PubMedCrossRef Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, et al. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int J Cancer. 2009;124(5):1060–71.PubMedCrossRef
43.
Zurück zum Zitat Hu YL, Jahangiri A, Delay M, Aghi MK. Tumor cell autophagy as an adaptive response mediating resistance to treatments such as antiangiogenic therapy. Cancer Res. 2012;72(17):4294–9.PubMedPubMedCentralCrossRef Hu YL, Jahangiri A, Delay M, Aghi MK. Tumor cell autophagy as an adaptive response mediating resistance to treatments such as antiangiogenic therapy. Cancer Res. 2012;72(17):4294–9.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Dai L, Aye Thu C, Liu XY, Xi J, Cheung PC. TAK1, more than just innate immunity. IUBMB Life. 2012;64(10):825–34.PubMedCrossRef Dai L, Aye Thu C, Liu XY, Xi J, Cheung PC. TAK1, more than just innate immunity. IUBMB Life. 2012;64(10):825–34.PubMedCrossRef
45.
Zurück zum Zitat Neumann D. Is TAK1 a Direct Upstream Kinase of AMPK? Int J Mol Sci. 2018;19(8). Neumann D. Is TAK1 a Direct Upstream Kinase of AMPK? Int J Mol Sci. 2018;19(8).
46.
Zurück zum Zitat Papa S, Choy PM, Bubici C. The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene. 2019;38(13):2223–40.PubMedCrossRef Papa S, Choy PM, Bubici C. The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene. 2019;38(13):2223–40.PubMedCrossRef
47.
Zurück zum Zitat Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, et al. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 2012;14(12):1295–304.PubMedPubMedCentralCrossRef Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, et al. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 2012;14(12):1295–304.PubMedPubMedCentralCrossRef
48.
Metadaten
Titel
Resistance to FGFR1-targeted therapy leads to autophagy via TAK1/AMPK activation in gastric cancer
verfasst von
Rui Peng
Yan Chen
Liangnian Wei
Gang Li
Dongju Feng
Siru Liu
Runqiu Jiang
Shaojiang Zheng
Yun Chen
Publikationsdatum
02.07.2020
Verlag
Springer Nature Singapore
Erschienen in
Gastric Cancer / Ausgabe 6/2020
Print ISSN: 1436-3291
Elektronische ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-020-01088-y

Weitere Artikel der Ausgabe 6/2020

Gastric Cancer 6/2020 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.