Skip to main content
Top
Published in: Journal of Neuro-Oncology 3/2016

01-02-2016 | Topic Review

How to train glioma cells to die: molecular challenges in cell death

Authors: Jeffrey Wojton, Walter Hans Meisen, Balveen Kaur

Published in: Journal of Neuro-Oncology | Issue 3/2016

Login to get access

Abstract

The five-year survival rate for patients with malignant glioma is less than 10 %. Despite aggressive chemo/radiotherapy these tumors have remained resistant to almost every interventional strategy evaluated in patients. Resistance to these agents is attributed to extrinsic mechanisms such as the tumor microenvironment, poor drug penetration, and tumoral heterogeneity. In addition, genetic and molecular examination of these tumors has revealed defective apoptotic regulation, enhanced pro-survival autophagy signaling, and a propensity for necrosis that aids in the adaptation to environmental stress and resistance to treatment. The combination of extrinsic and intrinsic hallmarks in glioma contributes to the multifaceted resistance to traditional anti-tumor agents. Here we describe the biology of the disease relevant to therapeutic resistance, with a specific focus on molecular deregulation of cell death pathways. Emerging studies investigating the targeting of these pathways including BH3 mimetics and autophagy inhibitors that are being evaluated in both the preclinical and clinical settings are discussed. This review highlights the pathways exploited by glioblastoma cells that drive their hallmark pro-survival predisposition and makes therapy development such a challenge.
Literature
1.
go back to reference Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A et al (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109PubMedPubMedCentralCrossRef Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A et al (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109PubMedPubMedCentralCrossRef
2.
go back to reference Jiang YG, Peng Y, Koussougbo KS (2011) Necroptosis: a novel therapeutic target for glioblastoma. Med Hypotheses 76(3):350–352PubMedCrossRef Jiang YG, Peng Y, Koussougbo KS (2011) Necroptosis: a novel therapeutic target for glioblastoma. Med Hypotheses 76(3):350–352PubMedCrossRef
3.
go back to reference Tait SW, Green DR (2010) Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 11(9):621–632PubMedCrossRef Tait SW, Green DR (2010) Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 11(9):621–632PubMedCrossRef
5.
go back to reference Cartron PF, Loussouarn D, Campone M, Martin SA, Vallette FM (2012) Prognostic impact of the expression/phosphorylation of the BH3-only proteins of the BCL-2 family in glioblastoma multiforme. Cell Death Dis 3:e421PubMedPubMedCentralCrossRef Cartron PF, Loussouarn D, Campone M, Martin SA, Vallette FM (2012) Prognostic impact of the expression/phosphorylation of the BH3-only proteins of the BCL-2 family in glioblastoma multiforme. Cell Death Dis 3:e421PubMedPubMedCentralCrossRef
6.
go back to reference Strik H, Deininger M, Streffer J, Grote E, Wickboldt J, Dichgans J et al (1999) BCL-2 family protein expression in initial and recurrent glioblastomas: modulation by radio-chemotherapy. J Neurol Neurosurg Psychiatry 67(6):763–768PubMedPubMedCentralCrossRef Strik H, Deininger M, Streffer J, Grote E, Wickboldt J, Dichgans J et al (1999) BCL-2 family protein expression in initial and recurrent glioblastomas: modulation by radio-chemotherapy. J Neurol Neurosurg Psychiatry 67(6):763–768PubMedPubMedCentralCrossRef
7.
go back to reference Qiu B, Wang Y, Tao J, Wang Y (2012) Expression and correlation of Bcl-2 with pathological grades in human glioma stem cells. Oncol Rep 28(1):155–160PubMed Qiu B, Wang Y, Tao J, Wang Y (2012) Expression and correlation of Bcl-2 with pathological grades in human glioma stem cells. Oncol Rep 28(1):155–160PubMed
8.
go back to reference Nagane M, Levitzki A, Gazit A, Cavenee WK, Huang HJ (1998) Drug resistance of human glioblastoma cells conferred by a tumor-specific mutant epidermal growth factor receptor through modulation of Bcl-XL and caspase-3-like proteases. Proc Natl Acad Sci USA 95(10):5724–5729PubMedPubMedCentralCrossRef Nagane M, Levitzki A, Gazit A, Cavenee WK, Huang HJ (1998) Drug resistance of human glioblastoma cells conferred by a tumor-specific mutant epidermal growth factor receptor through modulation of Bcl-XL and caspase-3-like proteases. Proc Natl Acad Sci USA 95(10):5724–5729PubMedPubMedCentralCrossRef
9.
go back to reference Zhu H, Cao X, Ali-Osman F, Keir S, Lo HW (2010) EGFR and EGFRvIII interact with PUMA to inhibit mitochondrial trans-localization of PUMA and PUMA-mediated apoptosis independent of EGFR kinase activity. Cancer Lett 294(1):101–110PubMedPubMedCentralCrossRef Zhu H, Cao X, Ali-Osman F, Keir S, Lo HW (2010) EGFR and EGFRvIII interact with PUMA to inhibit mitochondrial trans-localization of PUMA and PUMA-mediated apoptosis independent of EGFR kinase activity. Cancer Lett 294(1):101–110PubMedPubMedCentralCrossRef
10.
go back to reference Stegh AH, Kim H, Bachoo RM, Forloney KL, Zhang J, Schulze H et al (2007) Bcl2L12 inhibits post-mitochondrial apoptosis signaling in glioblastoma. Genes Dev 21(1):98–111PubMedPubMedCentralCrossRef Stegh AH, Kim H, Bachoo RM, Forloney KL, Zhang J, Schulze H et al (2007) Bcl2L12 inhibits post-mitochondrial apoptosis signaling in glioblastoma. Genes Dev 21(1):98–111PubMedPubMedCentralCrossRef
11.
go back to reference Stegh AH, Kesari S, Mahoney JE, Jenq HT, Forloney KL, Protopopov A et al (2008) Bcl2L12-mediated inhibition of effector caspase-3 and caspase-7 via distinct mechanisms in glioblastoma. Proc Natl Acad Sci USA 105(31):10703–10708PubMedPubMedCentralCrossRef Stegh AH, Kesari S, Mahoney JE, Jenq HT, Forloney KL, Protopopov A et al (2008) Bcl2L12-mediated inhibition of effector caspase-3 and caspase-7 via distinct mechanisms in glioblastoma. Proc Natl Acad Sci USA 105(31):10703–10708PubMedPubMedCentralCrossRef
12.
go back to reference Stegh AH, Brennan C, Mahoney JA, Forloney KL, Jenq HT, Luciano JP et al (2010) Glioma oncoprotein Bcl2L12 inhibits the p53 tumor suppressor. Genes Dev 24(19):2194–2204PubMedPubMedCentralCrossRef Stegh AH, Brennan C, Mahoney JA, Forloney KL, Jenq HT, Luciano JP et al (2010) Glioma oncoprotein Bcl2L12 inhibits the p53 tumor suppressor. Genes Dev 24(19):2194–2204PubMedPubMedCentralCrossRef
13.
go back to reference Tagscherer KE, Fassl A, Campos B, Farhadi M, Kraemer A, Bock BC et al (2008) Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins. Oncogene 27(52):6646–6656PubMedCrossRef Tagscherer KE, Fassl A, Campos B, Farhadi M, Kraemer A, Bock BC et al (2008) Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins. Oncogene 27(52):6646–6656PubMedCrossRef
14.
go back to reference Manero F, Gautier F, Gallenne T, Cauquil N, Gree D, Cartron PF et al (2006) The small organic compound HA14-1 prevents Bcl-2 interaction with Bax to sensitize malignant glioma cells to induction of cell death. Cancer Res 66(5):2757–2764PubMedCrossRef Manero F, Gautier F, Gallenne T, Cauquil N, Gree D, Cartron PF et al (2006) The small organic compound HA14-1 prevents Bcl-2 interaction with Bax to sensitize malignant glioma cells to induction of cell death. Cancer Res 66(5):2757–2764PubMedCrossRef
15.
go back to reference Dodou K, Anderson RJ, Small DA, Groundwater PW (2005) Investigations on gossypol: past and present developments. Expert Opin Investig Drugs 14(11):1419–1434PubMedCrossRef Dodou K, Anderson RJ, Small DA, Groundwater PW (2005) Investigations on gossypol: past and present developments. Expert Opin Investig Drugs 14(11):1419–1434PubMedCrossRef
16.
go back to reference Wagenknecht B, Glaser T, Naumann U, Kugler S, Isenmann S, Bahr M et al (1999) Expression and biological activity of X-linked inhibitor of apoptosis (XIAP) in human malignant glioma. Cell Death Differ 6(4):370–376PubMedCrossRef Wagenknecht B, Glaser T, Naumann U, Kugler S, Isenmann S, Bahr M et al (1999) Expression and biological activity of X-linked inhibitor of apoptosis (XIAP) in human malignant glioma. Cell Death Differ 6(4):370–376PubMedCrossRef
17.
go back to reference Chakravarti A, Noll E, Black PM, Finkelstein DF, Finkelstein DM, Dyson NJ et al (2002) Quantitatively determined survivin expression levels are of prognostic value in human gliomas. J Clin Oncol 20(4):1063–1068PubMedCrossRef Chakravarti A, Noll E, Black PM, Finkelstein DF, Finkelstein DM, Dyson NJ et al (2002) Quantitatively determined survivin expression levels are of prognostic value in human gliomas. J Clin Oncol 20(4):1063–1068PubMedCrossRef
18.
go back to reference Ziegler DS, Wright RD, Kesari S, Lemieux ME, Tran MA, Jain M et al (2008) Resistance of human glioblastoma multiforme cells to growth factor inhibitors is overcome by blockade of inhibitor of apoptosis proteins. J Clin Investig 118(9):3109–3122PubMedPubMedCentralCrossRef Ziegler DS, Wright RD, Kesari S, Lemieux ME, Tran MA, Jain M et al (2008) Resistance of human glioblastoma multiforme cells to growth factor inhibitors is overcome by blockade of inhibitor of apoptosis proteins. J Clin Investig 118(9):3109–3122PubMedPubMedCentralCrossRef
19.
go back to reference Chakravarti A, Zhai GG, Zhang M, Malhotra R, Latham DE, Delaney MA et al (2004) Survivin enhances radiation resistance in primary human glioblastoma cells via caspase-independent mechanisms. Oncogene 23(45):7494–7506PubMedCrossRef Chakravarti A, Zhai GG, Zhang M, Malhotra R, Latham DE, Delaney MA et al (2004) Survivin enhances radiation resistance in primary human glioblastoma cells via caspase-independent mechanisms. Oncogene 23(45):7494–7506PubMedCrossRef
20.
go back to reference Fulda S, Wick W, Weller M, Debatin KM (2002) Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat Med 8(8):808–815PubMed Fulda S, Wick W, Weller M, Debatin KM (2002) Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat Med 8(8):808–815PubMed
21.
go back to reference Wagner L, Marschall V, Karl S, Cristofanon S, Zobel K, Deshayes K et al (2013) Smac mimetic sensitizes glioblastoma cells to Temozolomide-induced apoptosis in a RIP1- and NF-kappaB-dependent manner. Oncogene 32(8):988–997PubMedCrossRef Wagner L, Marschall V, Karl S, Cristofanon S, Zobel K, Deshayes K et al (2013) Smac mimetic sensitizes glioblastoma cells to Temozolomide-induced apoptosis in a RIP1- and NF-kappaB-dependent manner. Oncogene 32(8):988–997PubMedCrossRef
22.
go back to reference Begg AC, Stewart FA, Vens C (2011) Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer 11(4):239–253PubMedCrossRef Begg AC, Stewart FA, Vens C (2011) Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer 11(4):239–253PubMedCrossRef
23.
go back to reference Venur VA, Peereboom DM, Ahluwalia MS (2015) Current medical treatment of glioblastoma. Cancer Treat Res 163:103–115PubMedCrossRef Venur VA, Peereboom DM, Ahluwalia MS (2015) Current medical treatment of glioblastoma. Cancer Treat Res 163:103–115PubMedCrossRef
24.
go back to reference Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996PubMedCrossRef Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996PubMedCrossRef
26.
go back to reference Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W et al (2010) MGMT promoter methylation in malignant gliomas: ready for personalized medicine? Nat Rev Neurol 6(1):39–51PubMedCrossRef Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W et al (2010) MGMT promoter methylation in malignant gliomas: ready for personalized medicine? Nat Rev Neurol 6(1):39–51PubMedCrossRef
27.
go back to reference Barazzuol L, Jena R, Burnet NG, Meira LB, Jeynes JC, Kirkby KJ et al (2013) Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma. Radiat Oncol 8:65PubMedPubMedCentralCrossRef Barazzuol L, Jena R, Burnet NG, Meira LB, Jeynes JC, Kirkby KJ et al (2013) Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma. Radiat Oncol 8:65PubMedPubMedCentralCrossRef
28.
go back to reference Lin F, de Gooijer MC, Roig EM, Buil LC, Christner SM, Beumer JH et al (2014) ABCB1, ABCG2, and PTEN determine the response of glioblastoma to temozolomide and ABT-888 therapy. Clin Cancer Res Off J Am Assoc Cancer Res 20(10):2703–2713CrossRef Lin F, de Gooijer MC, Roig EM, Buil LC, Christner SM, Beumer JH et al (2014) ABCB1, ABCG2, and PTEN determine the response of glioblastoma to temozolomide and ABT-888 therapy. Clin Cancer Res Off J Am Assoc Cancer Res 20(10):2703–2713CrossRef
29.
go back to reference Tentori L, Leonetti C, Scarsella M, D’Amati G, Vergati M, Portarena I et al (2003) Systemic administration of GPI 15427, a novel poly(ADP-ribose) polymerase-1 inhibitor, increases the antitumor activity of temozolomide against intracranial melanoma, glioma, lymphoma. Clin Cancer Res Off J Am Assoc Cancer Res 9(14):5370–5379 Tentori L, Leonetti C, Scarsella M, D’Amati G, Vergati M, Portarena I et al (2003) Systemic administration of GPI 15427, a novel poly(ADP-ribose) polymerase-1 inhibitor, increases the antitumor activity of temozolomide against intracranial melanoma, glioma, lymphoma. Clin Cancer Res Off J Am Assoc Cancer Res 9(14):5370–5379
30.
go back to reference Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ et al (2007) ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res Off J Am Assoc Cancer Res 13(9):2728–2737CrossRef Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ et al (2007) ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res Off J Am Assoc Cancer Res 13(9):2728–2737CrossRef
32.
go back to reference Liu EY, Ryan KM (2012) Autophagy and cancer–issues we need to digest. J Cell Sci 125(Pt 10):2349–2358PubMedCrossRef Liu EY, Ryan KM (2012) Autophagy and cancer–issues we need to digest. J Cell Sci 125(Pt 10):2349–2358PubMedCrossRef
34.
go back to reference Fan QW, Cheng C, Hackett C, Feldman M, Houseman BT, Nicolaides T et al (2010) Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal 3(147):ra81PubMedPubMedCentralCrossRef Fan QW, Cheng C, Hackett C, Feldman M, Houseman BT, Nicolaides T et al (2010) Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal 3(147):ra81PubMedPubMedCentralCrossRef
35.
go back to reference Shen J, Zheng H, Ruan J, Fang W, Li A, Tian G et al (2013) Autophagy inhibition induces enhanced proapoptotic effects of ZD6474 in glioblastoma. Br J Cancer 109(1):164–171PubMedPubMedCentralCrossRef Shen J, Zheng H, Ruan J, Fang W, Li A, Tian G et al (2013) Autophagy inhibition induces enhanced proapoptotic effects of ZD6474 in glioblastoma. Br J Cancer 109(1):164–171PubMedPubMedCentralCrossRef
36.
go back to reference Gammoh N, Lam D, Puente C, Ganley I, Marks PA, Jiang X (2012) Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc Natl Acad Sci USA 109(17):6561–6565PubMedPubMedCentralCrossRef Gammoh N, Lam D, Puente C, Ganley I, Marks PA, Jiang X (2012) Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc Natl Acad Sci USA 109(17):6561–6565PubMedPubMedCentralCrossRef
37.
go back to reference Li Y, Zhu H, Zeng X, Fan J, Qian X, Wang S et al (2013) Suppression of autophagy enhanced growth inhibition and apoptosis of interferon-beta in human glioma cells. Mol Neurobiol 47(3):1000–1010PubMedCrossRef Li Y, Zhu H, Zeng X, Fan J, Qian X, Wang S et al (2013) Suppression of autophagy enhanced growth inhibition and apoptosis of interferon-beta in human glioma cells. Mol Neurobiol 47(3):1000–1010PubMedCrossRef
38.
go back to reference Ge PF, Zhang JZ, Wang XF, Meng FK, Li WC, Luan YX et al (2009) Inhibition of autophagy induced by proteasome inhibition increases cell death in human SHG-44 glioma cells. Acta Pharmacol Sin 30(7):1046–1052PubMedPubMedCentralCrossRef Ge PF, Zhang JZ, Wang XF, Meng FK, Li WC, Luan YX et al (2009) Inhibition of autophagy induced by proteasome inhibition increases cell death in human SHG-44 glioma cells. Acta Pharmacol Sin 30(7):1046–1052PubMedPubMedCentralCrossRef
39.
go back to reference Chen Y, Meng D, Wang H, Sun R, Wang D, Wang S, et al (2014) VAMP8 facilitates cellular proliferation and temozolomide resistance in human glioma cells. Neuro Oncol 17(3):407–418 Chen Y, Meng D, Wang H, Sun R, Wang D, Wang S, et al (2014) VAMP8 facilitates cellular proliferation and temozolomide resistance in human glioma cells. Neuro Oncol 17(3):407–418
40.
go back to reference Cho HY, Wang W, Jhaveri N, Lee DJ, Sharma N, Dubeau L et al (2014) NEO212, temozolomide conjugated to perillyl alcohol, is a novel drug for effective treatment of a broad range of temozolomide-resistant gliomas. Mol Cancer Ther 13(8):2004–2017PubMedCrossRef Cho HY, Wang W, Jhaveri N, Lee DJ, Sharma N, Dubeau L et al (2014) NEO212, temozolomide conjugated to perillyl alcohol, is a novel drug for effective treatment of a broad range of temozolomide-resistant gliomas. Mol Cancer Ther 13(8):2004–2017PubMedCrossRef
41.
go back to reference Curry RC, Dahiya S, Alva Venur V, Raizer JJ, Ahluwalia MS (2015) Bevacizumab in high-grade gliomas: past, present, and future. Expert Rev Anticancer Ther 15(4):387–397PubMedCrossRef Curry RC, Dahiya S, Alva Venur V, Raizer JJ, Ahluwalia MS (2015) Bevacizumab in high-grade gliomas: past, present, and future. Expert Rev Anticancer Ther 15(4):387–397PubMedCrossRef
42.
go back to reference Gilbert MR, Sulman EP, Mehta MP (2014) Bevacizumab for newly diagnosed glioblastoma. New Engl J Med 370(21):2048–2049PubMedCrossRef Gilbert MR, Sulman EP, Mehta MP (2014) Bevacizumab for newly diagnosed glioblastoma. New Engl J Med 370(21):2048–2049PubMedCrossRef
43.
go back to reference Chinot OL, Wick W, Cloughesy T (2014) Bevacizumab for newly diagnosed glioblastoma. New Engl J Med 370(21):2049PubMedCrossRef Chinot OL, Wick W, Cloughesy T (2014) Bevacizumab for newly diagnosed glioblastoma. New Engl J Med 370(21):2049PubMedCrossRef
44.
go back to reference Hu YL, DeLay M, Jahangiri A, Molinaro AM, Rose SD, Carbonell WS et al (2012) Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res 72(7):1773–1783PubMedPubMedCentralCrossRef Hu YL, DeLay M, Jahangiri A, Molinaro AM, Rose SD, Carbonell WS et al (2012) Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res 72(7):1773–1783PubMedPubMedCentralCrossRef
45.
go back to reference Golden EB, Cho HY, Jahanian A, Hofman FM, Louie SG, Schonthal AH et al (2014) Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg Focus 37(6):E12PubMedCrossRef Golden EB, Cho HY, Jahanian A, Hofman FM, Louie SG, Schonthal AH et al (2014) Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg Focus 37(6):E12PubMedCrossRef
46.
go back to reference Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S et al (2014) A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 10(8):1359–1368PubMedPubMedCentralCrossRef Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S et al (2014) A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 10(8):1359–1368PubMedPubMedCentralCrossRef
47.
go back to reference Alonso MM, Jiang H, Yokoyama T, Xu J, Bekele NB, Lang FF et al (2008) Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death. Mol Ther 16(3):487–493PubMedCrossRef Alonso MM, Jiang H, Yokoyama T, Xu J, Bekele NB, Lang FF et al (2008) Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death. Mol Ther 16(3):487–493PubMedCrossRef
49.
go back to reference Stegh AH, Chin L, Louis DN, DePinho RA (2008) What drives intense apoptosis resistance and propensity for necrosis in glioblastoma? A role for Bcl2L12 as a multifunctional cell death regulator. Cell Cycle 7(18):2833–2839PubMedCrossRef Stegh AH, Chin L, Louis DN, DePinho RA (2008) What drives intense apoptosis resistance and propensity for necrosis in glioblastoma? A role for Bcl2L12 as a multifunctional cell death regulator. Cell Cycle 7(18):2833–2839PubMedCrossRef
50.
go back to reference Flannery T, McQuaid S, McGoohan C, McConnell RS, McGregor G, Mirakhur M et al (2006) Cathepsin S expression: an independent prognostic factor in glioblastoma tumours—a pilot study. Int J Cancer J Int Du cancer 119(4):854–860CrossRef Flannery T, McQuaid S, McGoohan C, McConnell RS, McGregor G, Mirakhur M et al (2006) Cathepsin S expression: an independent prognostic factor in glioblastoma tumours—a pilot study. Int J Cancer J Int Du cancer 119(4):854–860CrossRef
51.
go back to reference Keerthivasan S, Keerthivasan G, Mittal S, Chauhan SS (2007) Transcriptional upregulation of human cathepsin L by VEGF in glioblastoma cells. Gene 399(2):129–136PubMedCrossRef Keerthivasan S, Keerthivasan G, Mittal S, Chauhan SS (2007) Transcriptional upregulation of human cathepsin L by VEGF in glioblastoma cells. Gene 399(2):129–136PubMedCrossRef
52.
go back to reference Ouyang L, Shi Z, Zhao S, Wang FT, Zhou TT, Liu B et al (2012) Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif 45(6):487–498PubMedCrossRef Ouyang L, Shi Z, Zhao S, Wang FT, Zhou TT, Liu B et al (2012) Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif 45(6):487–498PubMedCrossRef
53.
go back to reference Melo-Lima S, Celeste Lopes M, Mollinedo F (2014) Necroptosis is associated with low procaspase-8 and active RIPK1 and -3 in human glioma cells. Oncoscience 1(10):649–664PubMedPubMedCentralCrossRef Melo-Lima S, Celeste Lopes M, Mollinedo F (2014) Necroptosis is associated with low procaspase-8 and active RIPK1 and -3 in human glioma cells. Oncoscience 1(10):649–664PubMedPubMedCentralCrossRef
54.
go back to reference Kang TB, Yang SH, Toth B, Kovalenko A, Wallach D (2014) Activation of the NLRP3 inflammasome by proteins that signal for necroptosis. Methods Enzymol 545:67–81PubMedCrossRef Kang TB, Yang SH, Toth B, Kovalenko A, Wallach D (2014) Activation of the NLRP3 inflammasome by proteins that signal for necroptosis. Methods Enzymol 545:67–81PubMedCrossRef
55.
go back to reference Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY et al (2013) An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 12(6):597–615PubMedPubMedCentralCrossRef Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY et al (2013) An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 12(6):597–615PubMedPubMedCentralCrossRef
56.
go back to reference Stupp R, Wong ET, Kanner AA, Steinberg D, Engelhard H, Heidecke V et al (2012) NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer 48(14):2192–2202PubMedCrossRef Stupp R, Wong ET, Kanner AA, Steinberg D, Engelhard H, Heidecke V et al (2012) NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer 48(14):2192–2202PubMedCrossRef
58.
go back to reference Blomgran R, Zheng L, Stendahl O (2007) Cathepsin-cleaved Bid promotes apoptosis in human neutrophils via oxidative stress-induced lysosomal membrane permeabilization. J Leukoc Biol 81(5):1213–1223PubMedCrossRef Blomgran R, Zheng L, Stendahl O (2007) Cathepsin-cleaved Bid promotes apoptosis in human neutrophils via oxidative stress-induced lysosomal membrane permeabilization. J Leukoc Biol 81(5):1213–1223PubMedCrossRef
59.
go back to reference Racoma IO, Meisen WH, Wang QE, Kaur B, Wani AA (2013) Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS One 8(9):e72882PubMedPubMedCentralCrossRef Racoma IO, Meisen WH, Wang QE, Kaur B, Wani AA (2013) Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS One 8(9):e72882PubMedPubMedCentralCrossRef
60.
go back to reference Boya P, Kroemer G (2008) Lysosomal membrane permeabilization in cell death. Oncogene 27(50):6434–6451PubMedCrossRef Boya P, Kroemer G (2008) Lysosomal membrane permeabilization in cell death. Oncogene 27(50):6434–6451PubMedCrossRef
Metadata
Title
How to train glioma cells to die: molecular challenges in cell death
Authors
Jeffrey Wojton
Walter Hans Meisen
Balveen Kaur
Publication date
01-02-2016
Publisher
Springer US
Published in
Journal of Neuro-Oncology / Issue 3/2016
Print ISSN: 0167-594X
Electronic ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-015-1980-1

Other articles of this Issue 3/2016

Journal of Neuro-Oncology 3/2016 Go to the issue