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

Open Access 01-12-2018 | Primary research

Levetiracetam enhances the temozolomide effect on glioblastoma stem cell proliferation and apoptosis

Authors: Bianca Maria Scicchitano, Silvia Sorrentino, Gabriella Proietti, Gina Lama, Gabriella Dobrowolny, Angela Catizone, Elena Binda, Luigi Maria Larocca, Gigliola Sica

Published in: Cancer Cell International | Issue 1/2018

Login to get access

Abstract

Background

Glioblastoma multiforme (GBM) is a highly aggressive brain tumor in which cancer cells with stem cell-like features, called cancer stem cells (CSCs), were identified. Two CSC populations have been previously identified in GBM, one derived from the GBM area called enhanced lesion (GCSCs) and the other one from the brain area adjacent to the tumor margin (PCSCs) that greatly differ in their growth properties and tumor-initiating ability. To date the most effective chemotherapy to treat GBM is represented by alkylating agents such as temozolomide (TMZ), whose activity can be regulated by histone deacetylases (HDACs) inhibitors through the modulation of O6-methylguanine-DNA methyltransferase (MGMT) expression. Levetiracetam (LEV), a relatively new antiepileptic drug, modulates HDAC levels ultimately silencing MGMT, thus increasing TMZ effectiveness. However, an improvement in the therapeutic efficacy of TMZ is needed.

Methods

Cell proliferation was investigated by BrdU cell proliferation assay and by Western Blot analysis of PCNA expression. Apoptosis was evaluated by Western Blot and Immunofluorescence analysis of the cleaved Caspase-3 expression. MGMT and HDAC4 expression was analyzed by Western Blotting and Immunofluorescence. Statistical analysis was performed using the Student’s t test and Mann–Whitney test.

Results

Here we evaluated the effect of TMZ on the proliferation rate of the IDH-wildtype GCSCs and PCSCs derived from six patients, in comparison with the effects of other drugs such as etoposide, irinotecan and carboplatin. Our results demonstrated that TMZ was less effective compared to the other agents; hence, we verified the possibility to increase the effect of TMZ by combining it with LEV. Here we show that LEV enhances the effect of TMZ on GCSCs proliferation (being less effective on PCSCs) by decreasing MGMT expression, promoting HDAC4 nuclear translocation and activating apoptotic pathway.

Conclusions

Although further studies are needed to determine the exact mechanism by which LEV makes GBM stem cells more  sensitive to TMZ, these results suggest that the clinical therapeutic efficacy of TMZ in GBM might be enhanced by the combined treatment with LEV.
Appendix
Available only for authorised users
Literature
2.
go back to reference Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.CrossRefPubMed Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.CrossRefPubMed
3.
go back to reference Cheray M, Begaud G, Deluche E, Nivet A, Battu S, Lalloue F, et al. Cancer stem-like cells in glioblastoma. Brisbane (AU): Codon Publication; 2017.CrossRef Cheray M, Begaud G, Deluche E, Nivet A, Battu S, Lalloue F, et al. Cancer stem-like cells in glioblastoma. Brisbane (AU): Codon Publication; 2017.CrossRef
4.
go back to reference Avgeropoulos NG, Batchelor TT. New treatment strategies for malignant gliomas. Oncologist. 1999;4:209–24.PubMed Avgeropoulos NG, Batchelor TT. New treatment strategies for malignant gliomas. Oncologist. 1999;4:209–24.PubMed
5.
go back to reference Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.CrossRefPubMed
6.
go back to reference Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res. 2009;29:5171–84.PubMed Minniti G, Muni R, Lanzetta G, Marchetti P, Enrici RM. Chemotherapy for glioblastoma: current treatment and future perspectives for cytotoxic and targeted agents. Anticancer Res. 2009;29:5171–84.PubMed
7.
go back to reference Mirimanoff RO, Gorlia T, Mason W, Van den Bent MJ, Kortmann RD, Fisher B, et al. Radiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trial. J Clin Oncol. 2006;24:2563–9.CrossRefPubMed Mirimanoff RO, Gorlia T, Mason W, Van den Bent MJ, Kortmann RD, Fisher B, et al. Radiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trial. J Clin Oncol. 2006;24:2563–9.CrossRefPubMed
8.
go back to reference Hart MG, Garside R, Rogers G, Stein K, Grant R. Temozolomide for high grade glioma. Cochrane Database Syst Rev. 2013; 30:CD007415. Hart MG, Garside R, Rogers G, Stein K, Grant R. Temozolomide for high grade glioma. Cochrane Database Syst Rev. 2013; 30:CD007415.
9.
go back to reference Piccirillo SG, Combi R, Cajola L, Patrizi A, Redaelli S, Bentivegna A, Baronchelli S, et al. Distinct pools of cancer stem-like cells coexist within human glioblastomas and display different tumorigenicity and independent genomic evolution. Oncogene. 2009;28:1807–11.CrossRefPubMed Piccirillo SG, Combi R, Cajola L, Patrizi A, Redaelli S, Bentivegna A, Baronchelli S, et al. Distinct pools of cancer stem-like cells coexist within human glioblastomas and display different tumorigenicity and independent genomic evolution. Oncogene. 2009;28:1807–11.CrossRefPubMed
10.
go back to reference Glas M, Rath BH, Simon M, Reinartz R, Schramme A, Trageser D, et al. Residual tumor cells are unique cellular targets in glioblastoma. Ann Neurol. 2010;68:264–9.PubMedPubMedCentral Glas M, Rath BH, Simon M, Reinartz R, Schramme A, Trageser D, et al. Residual tumor cells are unique cellular targets in glioblastoma. Ann Neurol. 2010;68:264–9.PubMedPubMedCentral
11.
go back to reference Piccirillo SG, Dietz S, Madhu B, Griffiths J, Price SJ, Collins VP, Watts C. Fluorescence-guided surgical sampling of glioblastoma identifies phenotypically distinct tumour-initiating cell populations in the tumour mass and margin. Br J Cancer. 2012;107:462–8.CrossRefPubMedPubMedCentral Piccirillo SG, Dietz S, Madhu B, Griffiths J, Price SJ, Collins VP, Watts C. Fluorescence-guided surgical sampling of glioblastoma identifies phenotypically distinct tumour-initiating cell populations in the tumour mass and margin. Br J Cancer. 2012;107:462–8.CrossRefPubMedPubMedCentral
12.
go back to reference Lama G, Mangiola A, Proietti G, Colabianchi A, Angelucci C, D’ Alessio A, et al. Progenitor/Stem Cell Markers in Brain Adjacent to Glioblastoma: GD3 Ganglioside and NG2 Proteoglycan Expression. J Neuropathol Exp Neurol. 2016;75:134–47.CrossRefPubMed Lama G, Mangiola A, Proietti G, Colabianchi A, Angelucci C, D’ Alessio A, et al. Progenitor/Stem Cell Markers in Brain Adjacent to Glioblastoma: GD3 Ganglioside and NG2 Proteoglycan Expression. J Neuropathol Exp Neurol. 2016;75:134–47.CrossRefPubMed
13.
go back to reference Bakshi A, Nag TC, Wadhwa S, Mahapatra AK, Sarkar C. The expression of nitric oxide synthases in human brain tumours and peritumoral areas. J Neurol Sci. 1998;155:196–203.CrossRefPubMed Bakshi A, Nag TC, Wadhwa S, Mahapatra AK, Sarkar C. The expression of nitric oxide synthases in human brain tumours and peritumoral areas. J Neurol Sci. 1998;155:196–203.CrossRefPubMed
14.
go back to reference Roy S, Sarkar C. Ultrastructural study of micro-blood vessels in human brain tumors and peritumoral tissue. J Neurooncol. 1989;7:283–92.CrossRefPubMed Roy S, Sarkar C. Ultrastructural study of micro-blood vessels in human brain tumors and peritumoral tissue. J Neurooncol. 1989;7:283–92.CrossRefPubMed
15.
go back to reference Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia. 2012;60:502–14.CrossRefPubMed Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia. 2012;60:502–14.CrossRefPubMed
17.
go back to reference Fazi B, Felsani A, Grassi L, Moles A, D’Andrea D, Toschi N, et al. The transcriptome and miRNome profiling of glioblastoma tissues and peritumoral regions highlights molecular pathways shared by tumors and surrounding areas and reveals differences between short-term and long-term survivors. Oncotarget. 2015;6:22526–52.CrossRefPubMedPubMedCentral Fazi B, Felsani A, Grassi L, Moles A, D’Andrea D, Toschi N, et al. The transcriptome and miRNome profiling of glioblastoma tissues and peritumoral regions highlights molecular pathways shared by tumors and surrounding areas and reveals differences between short-term and long-term survivors. Oncotarget. 2015;6:22526–52.CrossRefPubMedPubMedCentral
18.
go back to reference Mangiola A, Saulnier N, De Bonis P, Orteschi D, Sica G, Lama G, et al. Gene expression profile of glioblastoma peritumoral tissue: an ex vivo study. PLoS ONE. 2013;8:e57145.CrossRefPubMedPubMedCentral Mangiola A, Saulnier N, De Bonis P, Orteschi D, Sica G, Lama G, et al. Gene expression profile of glioblastoma peritumoral tissue: an ex vivo study. PLoS ONE. 2013;8:e57145.CrossRefPubMedPubMedCentral
19.
go back to reference Park I, Tamai G, Lee MC, Chuang CF, Chang SM, Berger MS, et al. Patterns of recurrence analysis in newly diagnosed glioblastoma multiforme after three-dimensional conformal radiation therapy with respect to pre-radiation therapy magnetic resonance spectroscopic findings. Int J Radiat Oncol Biol Phys. 2007;69:381–9.CrossRefPubMedPubMedCentral Park I, Tamai G, Lee MC, Chuang CF, Chang SM, Berger MS, et al. Patterns of recurrence analysis in newly diagnosed glioblastoma multiforme after three-dimensional conformal radiation therapy with respect to pre-radiation therapy magnetic resonance spectroscopic findings. Int J Radiat Oncol Biol Phys. 2007;69:381–9.CrossRefPubMedPubMedCentral
20.
go back to reference Barciszewska AM, Gurda D, Glodowicz P, Nowak S, Naskret-Barciszewska MZ. A new epigenetic mechanism of temozolomide action in glioma cells. PLoS ONE. 2015;10:e0136669.CrossRefPubMedPubMedCentral Barciszewska AM, Gurda D, Glodowicz P, Nowak S, Naskret-Barciszewska MZ. A new epigenetic mechanism of temozolomide action in glioma cells. PLoS ONE. 2015;10:e0136669.CrossRefPubMedPubMedCentral
21.
go back to reference Alonso MM, Gomez-Manzano C, Bekele BN, Yung WK, Fueyo J. Adenovirus-based strategies overcome temozolomide resistance by silencing the O6-methylguanine-DNA methyltransferase promoter. Cancer Res. 2007;67:11499–504.CrossRefPubMed Alonso MM, Gomez-Manzano C, Bekele BN, Yung WK, Fueyo J. Adenovirus-based strategies overcome temozolomide resistance by silencing the O6-methylguanine-DNA methyltransferase promoter. Cancer Res. 2007;67:11499–504.CrossRefPubMed
22.
23.
go back to reference Nishikawa R. Standard therapy for glioblastoma-a review of where we are. Neurol Med Chir. 2010;50:713–9.CrossRef Nishikawa R. Standard therapy for glioblastoma-a review of where we are. Neurol Med Chir. 2010;50:713–9.CrossRef
25.
go back to reference Marampon F, Megiorni F, Camero S, Crescioli C, McDowell HP, Sferra R, et al. HDAC4 and HDAC6 sustain DNA double strand break repair and stem-like phenotype by promoting radioresistance in glioblastoma cells. Cancer Lett. 2017;397:1–11.CrossRefPubMed Marampon F, Megiorni F, Camero S, Crescioli C, McDowell HP, Sferra R, et al. HDAC4 and HDAC6 sustain DNA double strand break repair and stem-like phenotype by promoting radioresistance in glioblastoma cells. Cancer Lett. 2017;397:1–11.CrossRefPubMed
27.
go back to reference Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: what are the cancer relevant targets? Cancer Lett. 2009;277:8–21.CrossRefPubMed Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: what are the cancer relevant targets? Cancer Lett. 2009;277:8–21.CrossRefPubMed
28.
go back to reference Bobustuc GC, Baker CH, Limaye A, Jenkins WD, Pearl G, Avgeropoulos NG, Konduri SD. Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol. 2010;12:917–27.CrossRefPubMedPubMedCentral Bobustuc GC, Baker CH, Limaye A, Jenkins WD, Pearl G, Avgeropoulos NG, Konduri SD. Levetiracetam enhances p53-mediated MGMT inhibition and sensitizes glioblastoma cells to temozolomide. Neuro Oncol. 2010;12:917–27.CrossRefPubMedPubMedCentral
29.
go back to reference Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803–20.CrossRefPubMed Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803–20.CrossRefPubMed
30.
go back to reference Horbinski C, Kofler J, Kelly LM, Murdoch GH, Nikiforova MN. Diagnostic use of IDH1/2 mutation analysis in routine clinical testing of formalin-fixed, paraffin-embedded glioma tissues. J Neuropathol Exp Neurol. 2009;68:1319–25.CrossRefPubMed Horbinski C, Kofler J, Kelly LM, Murdoch GH, Nikiforova MN. Diagnostic use of IDH1/2 mutation analysis in routine clinical testing of formalin-fixed, paraffin-embedded glioma tissues. J Neuropathol Exp Neurol. 2009;68:1319–25.CrossRefPubMed
32.
go back to reference Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, et al. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res. 2008;14:8205–12.CrossRefPubMed Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, et al. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res. 2008;14:8205–12.CrossRefPubMed
33.
go back to reference Binda E, Visioli A, Giani F, Lamorte G, Copetti M, Pitter KL, et al. The EphA2 receptor drives self-renewal and tumorigenicity in stem-like tumor-propagating cells from human glioblastomas. Cancer Cell. 2012;22:765–80.CrossRefPubMedPubMedCentral Binda E, Visioli A, Giani F, Lamorte G, Copetti M, Pitter KL, et al. The EphA2 receptor drives self-renewal and tumorigenicity in stem-like tumor-propagating cells from human glioblastomas. Cancer Cell. 2012;22:765–80.CrossRefPubMedPubMedCentral
35.
go back to reference Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999;59:793–7.PubMed Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999;59:793–7.PubMed
36.
go back to reference Zeuner A, Pedini F, Signore M, Testa U, Pelosi E, Peschle C, et al. Stem cell factor protects erythroid precursor cells from chemotherapeutic agents via up-regulation of BCL-2 family proteins. Blood. 2003;102:87–93.CrossRefPubMed Zeuner A, Pedini F, Signore M, Testa U, Pelosi E, Peschle C, et al. Stem cell factor protects erythroid precursor cells from chemotherapeutic agents via up-regulation of BCL-2 family proteins. Blood. 2003;102:87–93.CrossRefPubMed
37.
go back to reference Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, Wick W. Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res. 2008;14:2900–8.CrossRefPubMedPubMedCentral Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, Wick W. Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res. 2008;14:2900–8.CrossRefPubMedPubMedCentral
38.
go back to reference Sanai N, Alvarez-Buylla A, Berger MS. Neural stem cells and the origin of gliomas. N Engl J Med. 2005;353:811–22.CrossRefPubMed Sanai N, Alvarez-Buylla A, Berger MS. Neural stem cells and the origin of gliomas. N Engl J Med. 2005;353:811–22.CrossRefPubMed
39.
go back to reference Ignatova TN, Kukekov VG, Laywell ED, Suslov ON, Vrionis FD, Steindler DA. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia. 2002;39:193–206.CrossRefPubMed Ignatova TN, Kukekov VG, Laywell ED, Suslov ON, Vrionis FD, Steindler DA. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia. 2002;39:193–206.CrossRefPubMed
40.
go back to reference Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed
41.
go back to reference Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, Parada LF. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488:522–6.CrossRefPubMedPubMedCentral Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, Parada LF. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488:522–6.CrossRefPubMedPubMedCentral
42.
go back to reference Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.CrossRefPubMed Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.CrossRefPubMed
44.
go back to reference Pandith AA, Qasim I, Zahoor W, Shah P, Bhat AR, Sanadhya D, Shah ZA, Naikoo NA. Concordant association validates MGMT methylation and protein expression as favorable prognostic factors in glioma patients on alkylating chemotherapy (Temozolomide). Sci Rep. 2018;30:6704.CrossRef Pandith AA, Qasim I, Zahoor W, Shah P, Bhat AR, Sanadhya D, Shah ZA, Naikoo NA. Concordant association validates MGMT methylation and protein expression as favorable prognostic factors in glioma patients on alkylating chemotherapy (Temozolomide). Sci Rep. 2018;30:6704.CrossRef
46.
go back to reference Blough MD, Westgate MR, Beauchamp D, Kelly JJ, Stechishin O, Ramirez AL, Weiss S, Cairncross JG. Sensitivity to temozolomide in brain tumor initiating cells. Neuro Oncol. 2010;12:756–60.CrossRefPubMedPubMedCentral Blough MD, Westgate MR, Beauchamp D, Kelly JJ, Stechishin O, Ramirez AL, Weiss S, Cairncross JG. Sensitivity to temozolomide in brain tumor initiating cells. Neuro Oncol. 2010;12:756–60.CrossRefPubMedPubMedCentral
47.
go back to reference van Breemen MS, Wilms EB, Vecht CJ. Epilepsy in patients with brain tumours: epidemiology, mechanisms, and management. Lancet Neurol. 2007;6:421–30.CrossRefPubMed van Breemen MS, Wilms EB, Vecht CJ. Epilepsy in patients with brain tumours: epidemiology, mechanisms, and management. Lancet Neurol. 2007;6:421–30.CrossRefPubMed
49.
go back to reference Peddi P, Ajit NE, Burton GV, El-Osta H. Regression of a glioblastoma multiforme: spontaneous versus a potential antineoplastic effect of dexamethasone and levetiracetam. BMJ Case Rep. 2016;23:2016. Peddi P, Ajit NE, Burton GV, El-Osta H. Regression of a glioblastoma multiforme: spontaneous versus a potential antineoplastic effect of dexamethasone and levetiracetam. BMJ Case Rep. 2016;23:2016.
Metadata
Title
Levetiracetam enhances the temozolomide effect on glioblastoma stem cell proliferation and apoptosis
Authors
Bianca Maria Scicchitano
Silvia Sorrentino
Gabriella Proietti
Gina Lama
Gabriella Dobrowolny
Angela Catizone
Elena Binda
Luigi Maria Larocca
Gigliola Sica
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/s12935-018-0626-8

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