Skip to main content
Top
Published in: Cancer Cell International 1/2018

Open Access 01-12-2018 | Primary Research

Inhibition of the PI3K but not the MEK/ERK pathway sensitizes human glioma cells to alkylating drugs

Authors: Bodo Haas, Veronika Klinger, Christina Keksel, Verena Bonigut, Daniela Kiefer, Julia Caspers, Julia Walther, Maria Wos-Maganga, Sandra Weickhardt, Gabriele Röhn, Marco Timmer, Roland Frötschl, Niels Eckstein

Published in: Cancer Cell International | Issue 1/2018

Login to get access

Abstract

Background

Intrinsic chemoresistance of glioblastoma (GBM) is frequently owed to activation of the PI3K and MEK/ERK pathways. These signaling cascades are tightly interconnected however the quantitative contribution of both to intrinsic resistance is still not clear. Here, we aimed at determining the activation status of these pathways in human GBM biopsies and cells and investigating the quantitative impact of both pathways to chemoresistance.

Methods

Receptor tyrosine kinase (RTK) pathways in temozolomide (TMZ) treatment naive or TMZ resistant human GBM biopsies and GBM cells were investigated by proteome profiling and immunoblotting of a subset of proteins. Resistance to drugs and RTK pathway inhibitors was assessed by MTT assays. Apoptotic rates were determined by Annexin V staining and DNA damage with comet assays and immunoblotting.

Results

We analyzed activation of RTK pathways by proteome profiling of tumor samples of patients which were diagnosed a secondary GBM and underwent surgery and patients which underwent a second surgery after TMZ treatment due to recurrence of the tumor. We observed substantial activation of the PI3K and MEK/ERK pathways in both groups. However, AKT and CREB phosphorylation was reduced in biopsies of resistant tumors while ERK phosphorylation remained unchanged. Subsequent proteome profiling revealed that multiple RTKs and downstream targets are also activated in three GBM cell lines. We then systematically describe a mechanism of resistance of GBM cell lines and human primary GBM cells to the alkylating drugs TMZ and cisplatin. No specific inhibitor of the upstream RTKs sensitized cells to drug treatment. In contrast, we were able to restore sensitivity to TMZ and cisplatin by inhibiting PI3K in all cell lines and in human primary GBM cells. Interestingly, an opposite effect was observed when we inhibited the MEK/ERK signaling cascade with two different inhibitors.

Conclusions

Temozolomide treatment naive and TMZ resistant GBM biopsies show a distinct activation pattern of the MEK/ERK and PI3K signaling cascades indicating a role of these pathways in resistance development. Both pathways are also activated in GBM cell lines, however, only the PI3K pathway seems to play a crucial role in resistance to alkylating agents and might serve as drug target for chemosensitization.
Appendix
Available only for authorised users
Literature
1.
go back to reference Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803–20.CrossRefPubMed Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803–20.CrossRefPubMed
2.
3.
go back to reference Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed
4.
go back to reference Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–66.CrossRefPubMed Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–66.CrossRefPubMed
5.
go back to reference Ostrom QT, Gittleman H, Xu J, Kromer C, Wolinsky Y, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013. Neuro Oncol. 2016;18(suppl_5):v1–75.CrossRefPubMed Ostrom QT, Gittleman H, Xu J, Kromer C, Wolinsky Y, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013. Neuro Oncol. 2016;18(suppl_5):v1–75.CrossRefPubMed
7.
go back to reference Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19(4):764–72.CrossRefPubMed Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19(4):764–72.CrossRefPubMed
8.
go back to reference Molenaar RJ, Verbaan D, Lamba S, Zanon C, Jeuken JW, Boots-Sprenger SH, Wesseling P, Hulsebos TJ, Troost D, van Tilborg AA, et al. The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone. Neuro Oncol. 2014;16(9):1263–73.CrossRefPubMedPubMedCentral Molenaar RJ, Verbaan D, Lamba S, Zanon C, Jeuken JW, Boots-Sprenger SH, Wesseling P, Hulsebos TJ, Troost D, van Tilborg AA, et al. The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone. Neuro Oncol. 2014;16(9):1263–73.CrossRefPubMedPubMedCentral
9.
go back to reference Yang P, Zhang W, Wang Y, Peng X, Chen B, Qiu X, Li G, Li S, Wu C, Yao K, et al. IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry. Oncotarget. 2015;6(38):40896–906.CrossRefPubMedPubMedCentral Yang P, Zhang W, Wang Y, Peng X, Chen B, Qiu X, Li G, Li S, Wu C, Yao K, et al. IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry. Oncotarget. 2015;6(38):40896–906.CrossRefPubMedPubMedCentral
10.
go back to reference Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2(1):48–58.CrossRefPubMed Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2(1):48–58.CrossRefPubMed
12.
go back to reference Ishii N, Maier D, Merlo A, Tada M, Sawamura Y, Diserens AC, Van Meir EG. Frequent co-alterations of TP53, p16/CDKN2A, p14ARF, PTEN tumor suppressor genes in human glioma cell lines. Brain Pathol. 1999;9(3):469–79.CrossRefPubMed Ishii N, Maier D, Merlo A, Tada M, Sawamura Y, Diserens AC, Van Meir EG. Frequent co-alterations of TP53, p16/CDKN2A, p14ARF, PTEN tumor suppressor genes in human glioma cell lines. Brain Pathol. 1999;9(3):469–79.CrossRefPubMed
13.
go back to reference Iwadate Y, Fujimoto S, Tagawa M, Namba H, Sueyoshi K, Hirose M, Sakiyama S. Association of p53 gene mutation with decreased chemosensitivity in human malignant gliomas. Int J Cancer. 1996;69(3):236–40.CrossRefPubMed Iwadate Y, Fujimoto S, Tagawa M, Namba H, Sueyoshi K, Hirose M, Sakiyama S. Association of p53 gene mutation with decreased chemosensitivity in human malignant gliomas. Int J Cancer. 1996;69(3):236–40.CrossRefPubMed
14.
16.
go back to reference Tokunaga E, Oki E, Egashira A, Sadanaga N, Morita M, Kakeji Y, Maehara Y. Deregulation of the Akt pathway in human cancer. Curr Cancer Drug Targets. 2008;8(1):27–36.CrossRefPubMed Tokunaga E, Oki E, Egashira A, Sadanaga N, Morita M, Kakeji Y, Maehara Y. Deregulation of the Akt pathway in human cancer. Curr Cancer Drug Targets. 2008;8(1):27–36.CrossRefPubMed
17.
go back to reference Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2(7):489–501.CrossRefPubMed Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2(7):489–501.CrossRefPubMed
18.
go back to reference Martini M, De Santis MC, Braccini L, Gulluni F, Hirsch E. PI3K/AKT signaling pathway and cancer: an updated review. Ann Med. 2014;46(6):372–83.CrossRefPubMed Martini M, De Santis MC, Braccini L, Gulluni F, Hirsch E. PI3K/AKT signaling pathway and cancer: an updated review. Ann Med. 2014;46(6):372–83.CrossRefPubMed
19.
go back to reference Eyler CE, Foo WC, LaFiura KM, McLendon RE, Hjelmeland AB, Rich JN. Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells. 2008;26(12):3027–36.CrossRefPubMedPubMedCentral Eyler CE, Foo WC, LaFiura KM, McLendon RE, Hjelmeland AB, Rich JN. Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells. 2008;26(12):3027–36.CrossRefPubMedPubMedCentral
20.
go back to reference Hirose Y, Katayama M, Mirzoeva OK, Berger MS, Pieper RO. Akt activation suppresses Chk2-mediated, methylating agent-induced G2 arrest and protects from temozolomide-induced mitotic catastrophe and cellular senescence. Cancer Res. 2005;65(11):4861–9.CrossRefPubMed Hirose Y, Katayama M, Mirzoeva OK, Berger MS, Pieper RO. Akt activation suppresses Chk2-mediated, methylating agent-induced G2 arrest and protects from temozolomide-induced mitotic catastrophe and cellular senescence. Cancer Res. 2005;65(11):4861–9.CrossRefPubMed
21.
go back to reference Molina JR, Hayashi Y, Stephens C, Georgescu MM. Invasive glioblastoma cells acquire stemness and increased Akt activation. Neoplasia. 2010;12(6):453–63.CrossRefPubMedPubMedCentral Molina JR, Hayashi Y, Stephens C, Georgescu MM. Invasive glioblastoma cells acquire stemness and increased Akt activation. Neoplasia. 2010;12(6):453–63.CrossRefPubMedPubMedCentral
22.
go back to reference Haas-Kogan D, Shalev N, Wong M, Mills G, Yount G, Stokoe D. Protein kinase B (PKB/Akt) activity is elevated in glioblastoma cells due to mutation of the tumor suppressor PTEN/MMAC. Curr Biol. 1998;8(21):1195–8.CrossRefPubMed Haas-Kogan D, Shalev N, Wong M, Mills G, Yount G, Stokoe D. Protein kinase B (PKB/Akt) activity is elevated in glioblastoma cells due to mutation of the tumor suppressor PTEN/MMAC. Curr Biol. 1998;8(21):1195–8.CrossRefPubMed
23.
go back to reference Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–77.CrossRefPubMedPubMedCentral Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–77.CrossRefPubMedPubMedCentral
24.
go back to reference Chakravarti A, Zhai G, Suzuki Y, Sarkesh S, Black PM, Muzikansky A, Loeffler JS. The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. J Clin Oncol. 2004;22(10):1926–33.CrossRefPubMed Chakravarti A, Zhai G, Suzuki Y, Sarkesh S, Black PM, Muzikansky A, Loeffler JS. The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. J Clin Oncol. 2004;22(10):1926–33.CrossRefPubMed
26.
go back to reference Carico C, Nuno M, Mukherjee D, Elramsisy A, Dantis J, Hu J, Rudnick J, Yu JS, Black KL, Bannykh SI, et al. Loss of PTEN is not associated with poor survival in newly diagnosed glioblastoma patients of the temozolomide era. PLoS ONE. 2012;7(3):e33684.CrossRefPubMedPubMedCentral Carico C, Nuno M, Mukherjee D, Elramsisy A, Dantis J, Hu J, Rudnick J, Yu JS, Black KL, Bannykh SI, et al. Loss of PTEN is not associated with poor survival in newly diagnosed glioblastoma patients of the temozolomide era. PLoS ONE. 2012;7(3):e33684.CrossRefPubMedPubMedCentral
27.
go back to reference Pelloski CE, Lin E, Zhang L, Yung WK, Colman H, Liu JL, Woo SY, Heimberger AB, Suki D, Prados M, et al. Prognostic associations of activated mitogen-activated protein kinase and Akt pathways in glioblastoma. Clin Cancer Res. 2006;12(13):3935–41.CrossRefPubMed Pelloski CE, Lin E, Zhang L, Yung WK, Colman H, Liu JL, Woo SY, Heimberger AB, Suki D, Prados M, et al. Prognostic associations of activated mitogen-activated protein kinase and Akt pathways in glioblastoma. Clin Cancer Res. 2006;12(13):3935–41.CrossRefPubMed
28.
go back to reference Knobbe CB, Reifenberger J, Reifenberger G. Mutation analysis of the Ras pathway genes NRAS, HRAS, KRAS and BRAF in glioblastomas. Acta Neuropathol. 2004;108(6):467–70.CrossRefPubMed Knobbe CB, Reifenberger J, Reifenberger G. Mutation analysis of the Ras pathway genes NRAS, HRAS, KRAS and BRAF in glioblastomas. Acta Neuropathol. 2004;108(6):467–70.CrossRefPubMed
29.
go back to reference Shi Z, Chen Q, Li C, Wang L, Qian X, Jiang C, Liu X, Wang X, Li H, Kang C, et al. MiR-124 governs glioma growth and angiogenesis and enhances chemosensitivity by targeting R-Ras and N-Ras. Neuro Oncol. 2014;16(10):1341–53.CrossRefPubMedPubMedCentral Shi Z, Chen Q, Li C, Wang L, Qian X, Jiang C, Liu X, Wang X, Li H, Kang C, et al. MiR-124 governs glioma growth and angiogenesis and enhances chemosensitivity by targeting R-Ras and N-Ras. Neuro Oncol. 2014;16(10):1341–53.CrossRefPubMedPubMedCentral
30.
go back to reference Wang L, Shi ZM, Jiang CF, Liu X, Chen QD, Qian X, Li DM, Ge X, Wang XF, Liu LZ, et al. MiR-143 acts as a tumor suppressor by targeting N-RAS and enhances temozolomide-induced apoptosis in glioma. Oncotarget. 2014;5(14):5416–27.PubMedPubMedCentral Wang L, Shi ZM, Jiang CF, Liu X, Chen QD, Qian X, Li DM, Ge X, Wang XF, Liu LZ, et al. MiR-143 acts as a tumor suppressor by targeting N-RAS and enhances temozolomide-induced apoptosis in glioma. Oncotarget. 2014;5(14):5416–27.PubMedPubMedCentral
31.
go back to reference See WL, Tan IL, Mukherjee J, Nicolaides T, Pieper RO. Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency. Cancer Res. 2012;72(13):3350–9.CrossRefPubMedPubMedCentral See WL, Tan IL, Mukherjee J, Nicolaides T, Pieper RO. Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency. Cancer Res. 2012;72(13):3350–9.CrossRefPubMedPubMedCentral
32.
go back to reference Riedel M, Struve N, Muller-Goebel J, Kocher S, Petersen C, Dikomey E, Rothkamm K, Kriegs M. Sorafenib inhibits cell growth but fails to enhance radio- and chemosensitivity of glioblastoma cell lines. Oncotarget. 2016;7(38):61988–95.CrossRefPubMedPubMedCentral Riedel M, Struve N, Muller-Goebel J, Kocher S, Petersen C, Dikomey E, Rothkamm K, Kriegs M. Sorafenib inhibits cell growth but fails to enhance radio- and chemosensitivity of glioblastoma cell lines. Oncotarget. 2016;7(38):61988–95.CrossRefPubMedPubMedCentral
33.
go back to reference Jia J, Zhu F, Ma X, Cao Z, Li Y, Chen YZ. Mechanisms of drug combinations: interaction and network perspectives. Nat Rev Drug Discov. 2009;8(2):111–28.CrossRefPubMed Jia J, Zhu F, Ma X, Cao Z, Li Y, Chen YZ. Mechanisms of drug combinations: interaction and network perspectives. Nat Rev Drug Discov. 2009;8(2):111–28.CrossRefPubMed
34.
go back to reference McDowell KA, Riggins GJ, Gallia GL. Targeting the AKT pathway in glioblastoma. Curr Pharm Des. 2011;17(23):2411–20.CrossRefPubMed McDowell KA, Riggins GJ, Gallia GL. Targeting the AKT pathway in glioblastoma. Curr Pharm Des. 2011;17(23):2411–20.CrossRefPubMed
35.
go back to reference Rodon J, Dienstmann R, Serra V, Tabernero J. Development of PI3K inhibitors: lessons learned from early clinical trials. Nat Rev Clin Oncol. 2013;10(3):143–53.CrossRefPubMed Rodon J, Dienstmann R, Serra V, Tabernero J. Development of PI3K inhibitors: lessons learned from early clinical trials. Nat Rev Clin Oncol. 2013;10(3):143–53.CrossRefPubMed
37.
go back to reference Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9(8):550–62.CrossRefPubMed Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9(8):550–62.CrossRefPubMed
39.
go back to reference Wen PY, Lee EQ, Reardon DA, Ligon KL, Alfred Yung WK. Current clinical development of PI3K pathway inhibitors in glioblastoma. Neuro Oncol. 2012;14(7):819–29.CrossRefPubMedPubMedCentral Wen PY, Lee EQ, Reardon DA, Ligon KL, Alfred Yung WK. Current clinical development of PI3K pathway inhibitors in glioblastoma. Neuro Oncol. 2012;14(7):819–29.CrossRefPubMedPubMedCentral
40.
go back to reference Chen L, Han L, Shi Z, Zhang K, Liu Y, Zheng Y, Jiang T, Pu P, Jiang C, Kang C. LY294002 enhances cytotoxicity of temozolomide in glioma by down-regulation of the PI3K/Akt pathway. Mol Med Report. 2011;5:575–9. Chen L, Han L, Shi Z, Zhang K, Liu Y, Zheng Y, Jiang T, Pu P, Jiang C, Kang C. LY294002 enhances cytotoxicity of temozolomide in glioma by down-regulation of the PI3K/Akt pathway. Mol Med Report. 2011;5:575–9.
41.
go back to reference Carminati PO, Donaires FS, Marques MM, Donadi EA, Passos GA, Sakamoto-Hojo ET. Cisplatin associated with LY294002 increases cytotoxicity and induces changes in transcript profiles of glioblastoma cells. Mol Biol Rep. 2014;41(1):165–77.CrossRefPubMed Carminati PO, Donaires FS, Marques MM, Donadi EA, Passos GA, Sakamoto-Hojo ET. Cisplatin associated with LY294002 increases cytotoxicity and induces changes in transcript profiles of glioblastoma cells. Mol Biol Rep. 2014;41(1):165–77.CrossRefPubMed
42.
go back to reference Westhoff MA, Kandenwein JA, Karl S, Vellanki SH, Braun V, Eramo A, Antoniadis G, Debatin KM, Fulda S. The pyridinylfuranopyrimidine inhibitor, PI-103, chemosensitizes glioblastoma cells for apoptosis by inhibiting DNA repair. Oncogene. 2009;28(40):3586–96.CrossRefPubMed Westhoff MA, Kandenwein JA, Karl S, Vellanki SH, Braun V, Eramo A, Antoniadis G, Debatin KM, Fulda S. The pyridinylfuranopyrimidine inhibitor, PI-103, chemosensitizes glioblastoma cells for apoptosis by inhibiting DNA repair. Oncogene. 2009;28(40):3586–96.CrossRefPubMed
43.
go back to reference Prasad G, Sottero T, Yang X, Mueller S, James CD, Weiss WA, Polley MY, Ozawa T, Berger MS, Aftab DT, et al. Inhibition of PI3K/mTOR pathways in glioblastoma and implications for combination therapy with temozolomide. Neuro Oncol. 2011;13(4):384–92.CrossRefPubMedPubMedCentral Prasad G, Sottero T, Yang X, Mueller S, James CD, Weiss WA, Polley MY, Ozawa T, Berger MS, Aftab DT, et al. Inhibition of PI3K/mTOR pathways in glioblastoma and implications for combination therapy with temozolomide. Neuro Oncol. 2011;13(4):384–92.CrossRefPubMedPubMedCentral
44.
go back to reference Sinnberg T, Lasithiotakis K, Niessner H, Schittek B, Flaherty KT, Kulms D, Maczey E, Campos M, Gogel J, Garbe C, et al. Inhibition of PI3K-AKT-mTOR signaling sensitizes melanoma cells to cisplatin and temozolomide. J Invest Dermatol. 2009;129(6):1500–15.CrossRefPubMed Sinnberg T, Lasithiotakis K, Niessner H, Schittek B, Flaherty KT, Kulms D, Maczey E, Campos M, Gogel J, Garbe C, et al. Inhibition of PI3K-AKT-mTOR signaling sensitizes melanoma cells to cisplatin and temozolomide. J Invest Dermatol. 2009;129(6):1500–15.CrossRefPubMed
45.
go back to reference Strobele S, Schneider M, Schneele L, Siegelin MD, Nonnenmacher L, Zhou S, Karpel-Massler G, Westhoff MA, Halatsch ME, Debatin KM. A potential role for the inhibition of PI3K signaling in glioblastoma therapy. PLoS ONE. 2015;10(6):e0131670.CrossRefPubMedPubMedCentral Strobele S, Schneider M, Schneele L, Siegelin MD, Nonnenmacher L, Zhou S, Karpel-Massler G, Westhoff MA, Halatsch ME, Debatin KM. A potential role for the inhibition of PI3K signaling in glioblastoma therapy. PLoS ONE. 2015;10(6):e0131670.CrossRefPubMedPubMedCentral
46.
go back to reference Westhoff MA, Karpel-Massler G, Bruhl O, Enzenmuller S, La Ferla-Bruhl K, Siegelin MD, Nonnenmacher L, Debatin KM. A critical evaluation of PI3K inhibition in glioblastoma and neuroblastoma therapy. Mol Cell Ther. 2014;2:32.CrossRefPubMedPubMedCentral Westhoff MA, Karpel-Massler G, Bruhl O, Enzenmuller S, La Ferla-Bruhl K, Siegelin MD, Nonnenmacher L, Debatin KM. A critical evaluation of PI3K inhibition in glioblastoma and neuroblastoma therapy. Mol Cell Ther. 2014;2:32.CrossRefPubMedPubMedCentral
47.
go back to reference Will M, Qin AC, Toy W, Yao Z, Rodrik-Outmezguine V, Schneider C, Huang X, Monian P, Jiang X, de Stanchina E, et al. Rapid induction of apoptosis by PI3K inhibitors is dependent upon their transient inhibition of RAS-ERK signaling. Cancer Discov. 2014;4(3):334–47.CrossRefPubMedPubMedCentral Will M, Qin AC, Toy W, Yao Z, Rodrik-Outmezguine V, Schneider C, Huang X, Monian P, Jiang X, de Stanchina E, et al. Rapid induction of apoptosis by PI3K inhibitors is dependent upon their transient inhibition of RAS-ERK signaling. Cancer Discov. 2014;4(3):334–47.CrossRefPubMedPubMedCentral
48.
go back to reference Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI, Scaltriti M, Moskatel E, Baselga J, Guichard S, Rosen N. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011;1(3):248–59.CrossRefPubMedPubMedCentral Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI, Scaltriti M, Moskatel E, Baselga J, Guichard S, Rosen N. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011;1(3):248–59.CrossRefPubMedPubMedCentral
49.
go back to reference Serra V, Scaltriti M, Prudkin L, Eichhorn PJ, Ibrahim YH, Chandarlapaty S, Markman B, Rodriguez O, Guzman M, Rodriguez S, et al. PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer. Oncogene. 2011;30(22):2547–57.CrossRefPubMedPubMedCentral Serra V, Scaltriti M, Prudkin L, Eichhorn PJ, Ibrahim YH, Chandarlapaty S, Markman B, Rodriguez O, Guzman M, Rodriguez S, et al. PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer. Oncogene. 2011;30(22):2547–57.CrossRefPubMedPubMedCentral
50.
go back to reference Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, Majumder PK, Baselga J, Rosen N. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19(1):58–71.CrossRefPubMedPubMedCentral Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, Majumder PK, Baselga J, Rosen N. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19(1):58–71.CrossRefPubMedPubMedCentral
51.
go back to reference Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.CrossRefPubMedPubMedCentral Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.CrossRefPubMedPubMedCentral
52.
go back to reference Werner JM, Kuhl S, Stavrinou P, Rohn G, Krischek B, Blau T, Goldbrunner R, Timmer M. Expression of FAS-L differs from primary to relapsed low-grade gliomas and predicts progression-free survival. Anticancer Res. 2017;37(12):6639–48.PubMed Werner JM, Kuhl S, Stavrinou P, Rohn G, Krischek B, Blau T, Goldbrunner R, Timmer M. Expression of FAS-L differs from primary to relapsed low-grade gliomas and predicts progression-free survival. Anticancer Res. 2017;37(12):6639–48.PubMed
53.
go back to reference Eckstein N, Servan K, Girard L, Cai D, von Jonquieres G, Jaehde U, Kassack MU, Gazdar AF, Minna JD, Royer HD. Epidermal growth factor receptor pathway analysis identifies amphiregulin as a key factor for cisplatin resistance of human breast cancer cells. J Biol Chem. 2008;283(2):739–50.CrossRefPubMed Eckstein N, Servan K, Girard L, Cai D, von Jonquieres G, Jaehde U, Kassack MU, Gazdar AF, Minna JD, Royer HD. Epidermal growth factor receptor pathway analysis identifies amphiregulin as a key factor for cisplatin resistance of human breast cancer cells. J Biol Chem. 2008;283(2):739–50.CrossRefPubMed
54.
go back to reference Haas B, Mayer P, Jennissen K, Scholz D, Diaz MB, Bloch W, Herzig S, Fassler R, Pfeifer A. Protein kinase G controls brown fat cell differentiation and mitochondrial biogenesis. Sci Signal. 2009;2(99):ra78.CrossRefPubMed Haas B, Mayer P, Jennissen K, Scholz D, Diaz MB, Bloch W, Herzig S, Fassler R, Pfeifer A. Protein kinase G controls brown fat cell differentiation and mitochondrial biogenesis. Sci Signal. 2009;2(99):ra78.CrossRefPubMed
55.
go back to reference Eckstein N, Servan K, Hildebrandt B, Politz A, von Jonquières G, Wolf-Kummeth S, Napierski I, Hamacher A, Kassack MU, Budczies J, et al. Hyperactivation of the insulin-like growth factor receptor I signaling pathway is an essential event for cisplatin resistance of ovarian cancer cells. Cancer Res. 2009;69(7):2996–3003.CrossRefPubMed Eckstein N, Servan K, Hildebrandt B, Politz A, von Jonquières G, Wolf-Kummeth S, Napierski I, Hamacher A, Kassack MU, Budczies J, et al. Hyperactivation of the insulin-like growth factor receptor I signaling pathway is an essential event for cisplatin resistance of ovarian cancer cells. Cancer Res. 2009;69(7):2996–3003.CrossRefPubMed
56.
go back to reference Frotschl R, Weickardt S, Staszewski S, Kaufmann G, Kasper P. Effects of chlorpromazine with and without UV irradiation on gene expression of HepG2 cells. Mutat Res. 2005;575(1–2):47–60.CrossRefPubMed Frotschl R, Weickardt S, Staszewski S, Kaufmann G, Kasper P. Effects of chlorpromazine with and without UV irradiation on gene expression of HepG2 cells. Mutat Res. 2005;575(1–2):47–60.CrossRefPubMed
57.
go back to reference Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef
58.
go back to reference Opel D, Westhoff MA, Bender A, Braun V, Debatin KM, Fulda S. Phosphatidylinositol 3-kinase inhibition broadly sensitizes glioblastoma cells to death receptor- and drug-induced apoptosis. Cancer Res. 2008;68(15):6271–80.CrossRefPubMed Opel D, Westhoff MA, Bender A, Braun V, Debatin KM, Fulda S. Phosphatidylinositol 3-kinase inhibition broadly sensitizes glioblastoma cells to death receptor- and drug-induced apoptosis. Cancer Res. 2008;68(15):6271–80.CrossRefPubMed
59.
go back to reference Tisdale MJ. Antitumour imidazotetrazines–XI: effect of 8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one [CCRG 81045; M and B 39831 NSC 362856] on poly(ADP-ribose) metabolism. Br J Cancer. 1985;52(5):789–92.CrossRefPubMedPubMedCentral Tisdale MJ. Antitumour imidazotetrazines–XI: effect of 8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one [CCRG 81045; M and B 39831 NSC 362856] on poly(ADP-ribose) metabolism. Br J Cancer. 1985;52(5):789–92.CrossRefPubMedPubMedCentral
Metadata
Title
Inhibition of the PI3K but not the MEK/ERK pathway sensitizes human glioma cells to alkylating drugs
Authors
Bodo Haas
Veronika Klinger
Christina Keksel
Verena Bonigut
Daniela Kiefer
Julia Caspers
Julia Walther
Maria Wos-Maganga
Sandra Weickhardt
Gabriele Röhn
Marco Timmer
Roland Frötschl
Niels Eckstein
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2018
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0565-4

Other articles of this Issue 1/2018

Cancer Cell International 1/2018 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