Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Glioblastoma | Research

G-quadruplex ligand RHPS4 radiosensitizes glioblastoma xenograft in vivo through a differential targeting of bulky differentiated- and stem-cancer cells

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Glioblastoma is the most aggressive and most lethal primary brain tumor in the adulthood. Current standard therapies are not curative and novel therapeutic options are urgently required. Present knowledge suggests that the continued glioblastoma growth and recurrence is determined by glioblastoma stem-like cells (GSCs), which display self-renewal, tumorigenic potential, and increased radio- and chemo-resistance. The G-quadruplex ligand RHPS4 displays in vitro radiosensitizing effect in GBM radioresistant cells through the targeting and dysfunctionalization of telomeres but RHPS4 and Ionizing Radiation (IR) combined treatment efficacy in vivo has not been explored so far.

Methods

RHPS4 and IR combined effects were tested in vivo in a heterotopic mice xenograft model and in vitro in stem-like cells derived from U251MG and from four GBM patients. Cell growth assays, cytogenetic analysis, immunoblotting, gene expression and cytofluorimetric analysis were performed in order to characterize the response of differentiated and stem-like cells to RHPS4 and IR in single and combined treatments.

Results

RHPS4 administration and IR exposure is very effective in blocking tumor growth in vivo up to 65 days. The tumor volume reduction and the long-term tumor control suggested the targeting of the stem cell compartment. Interestingly, RHPS4 treatment was able to strongly reduce cell proliferation in GSCs but, unexpectedly, did not synergize with IR. Lack of radiosensitization was supported by the GSCs telomeric-resistance observed as the total absence of telomere-involving chromosomal aberrations. Remarkably, RHPS4 treatment determined a strong reduction of CHK1 and RAD51 proteins and transcript levels suggesting that the inhibition of GSCs growth is determined by the impairment of the replication stress (RS) response and DNA repair.

Conclusions

We propose that the potent antiproliferative effect of RHPS4 in GSCs is not determined by telomeric dysfunction but is achieved by the induction of RS and by the concomitant depletion of CHK1 and RAD51, leading to DNA damage and cell death. These data open to novel therapeutic options for the targeting of GSCs, indicating that the combined inhibition of cell-cycle checkpoints and DNA repair proteins provides the most effective means to overcome resistance of GSC to genotoxic insults.
Appendix
Available only for authorised users
Literature
1.
go back to reference Miller CR, Glioblastoma PA. Morphologic and molecular genetic diversity. Arch Pathol Lab Med. 2007;131:397–406.PubMed Miller CR, Glioblastoma PA. Morphologic and molecular genetic diversity. Arch Pathol Lab Med. 2007;131:397–406.PubMed
2.
go back to reference Smoll NR, Schaller K, Gautschi OP. Long-term survival of patients with glioblastoma multiforme (GBM). J Clin Neurosci Elsevier Ltd. 2013;20:670–5.CrossRef Smoll NR, Schaller K, Gautschi OP. Long-term survival of patients with glioblastoma multiforme (GBM). J Clin Neurosci Elsevier Ltd. 2013;20:670–5.CrossRef
3.
go back to reference Stupp R, Mason WP, Van Den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant Temozolomide for Glioblastoma. N Engl J Med. 2005;352:987–96.CrossRef Stupp R, Mason WP, Van Den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant Temozolomide for Glioblastoma. N Engl J Med. 2005;352:987–96.CrossRef
4.
go back to reference Field KM, Drummond KJ, Yilmaz M, Tacey M, Compston D, Gibbs P, et al. Clinical trial participation and outcome for patients with glioblastoma: multivariate analysis from a comprehensive dataset. J Clin Neurosci. Elsevier Ltd. 2013;20:783–9.CrossRef Field KM, Drummond KJ, Yilmaz M, Tacey M, Compston D, Gibbs P, et al. Clinical trial participation and outcome for patients with glioblastoma: multivariate analysis from a comprehensive dataset. J Clin Neurosci. Elsevier Ltd. 2013;20:783–9.CrossRef
5.
go back to reference Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, et al. 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, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed
6.
go back to reference King HO, Brend T, Payne HL, Wright A, Ward TA, Patel K, et al. RAD51 is a selective DNA repair target to Radiosensitize glioma stem cells. Stem Cell Reports ElsevierCompany. 2017;8:125–39.CrossRef King HO, Brend T, Payne HL, Wright A, Ward TA, Patel K, et al. RAD51 is a selective DNA repair target to Radiosensitize glioma stem cells. Stem Cell Reports ElsevierCompany. 2017;8:125–39.CrossRef
7.
go back to reference Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60.CrossRef Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60.CrossRef
8.
go back to reference Cheng L, Wu Q, Huang Z, Guryanova OA, Huang Q, Shou W, et al. L1CAM regulates DNA damage checkpoint response of glioblastoma stem cells through NBS1. EMBO J Nature Publishing Group. 2011;30:800–13. Cheng L, Wu Q, Huang Z, Guryanova OA, Huang Q, Shou W, et al. L1CAM regulates DNA damage checkpoint response of glioblastoma stem cells through NBS1. EMBO J Nature Publishing Group. 2011;30:800–13.
9.
go back to reference Facchino S, Abdouh M, Chatoo W, Bernier G. BMI1 confers Radioresistance to Normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. J Neurosci. 2010;30:10096–111.CrossRef Facchino S, Abdouh M, Chatoo W, Bernier G. BMI1 confers Radioresistance to Normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. J Neurosci. 2010;30:10096–111.CrossRef
10.
go back to reference Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008;14:123–9.CrossRef Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, et al. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008;14:123–9.CrossRef
11.
go back to reference Lim Y, Roberts T, WD B, Al. E A role for homologous recombination and abnormal cell cycle progression in radioresistance of glioma initiating cells Mol Cancer Ther 2012;11:1863–1872.CrossRef Lim Y, Roberts T, WD B, Al. E A role for homologous recombination and abnormal cell cycle progression in radioresistance of glioma initiating cells Mol Cancer Ther 2012;11:1863–1872.CrossRef
12.
go back to reference Lim YC, Roberts TL, Day BW, Stringer BW, Kozlov S, Fazry S, et al. Increased sensitivity to ionizing radiation by targeting the homologous recombination pathway in glioma initiating cells. Mol Oncol. 2014;8:1603–15.CrossRef Lim YC, Roberts TL, Day BW, Stringer BW, Kozlov S, Fazry S, et al. Increased sensitivity to ionizing radiation by targeting the homologous recombination pathway in glioma initiating cells. Mol Oncol. 2014;8:1603–15.CrossRef
13.
go back to reference Short SC, Giampieri S, Worku M, Alcaide-german M, Sioftanos G, Bourne S, et al. Rad51 inhibition is an effective means of targeting DNA repair in glioma models and CD1331 tumor-derived cells. Neuro-Oncology. 2011;13:487–99.CrossRef Short SC, Giampieri S, Worku M, Alcaide-german M, Sioftanos G, Bourne S, et al. Rad51 inhibition is an effective means of targeting DNA repair in glioma models and CD1331 tumor-derived cells. Neuro-Oncology. 2011;13:487–99.CrossRef
14.
go back to reference Merle P, Evrard B, Petitjean a, Lehn J-M, Teulade-Fichou M-P, Chautard E, et al. Telomere targeting with a new G4 ligand enhances radiation-induced killing of human Glioblastoma cells. Mol Cancer Ther. 2011;10:1784–95.CrossRef Merle P, Evrard B, Petitjean a, Lehn J-M, Teulade-Fichou M-P, Chautard E, et al. Telomere targeting with a new G4 ligand enhances radiation-induced killing of human Glioblastoma cells. Mol Cancer Ther. 2011;10:1784–95.CrossRef
15.
go back to reference Merle P, Gueugneau M, Teulade-Fichou M-P, Müller-Barthélémy M, Amiard S, Chautard E, et al. Highly efficient radiosensitization of human glioblastoma and lung cancer cells by a G-quadruplex DNA binding compound. Sci Rep Nature Publishing Group. 2015;5:16255. Merle P, Gueugneau M, Teulade-Fichou M-P, Müller-Barthélémy M, Amiard S, Chautard E, et al. Highly efficient radiosensitization of human glioblastoma and lung cancer cells by a G-quadruplex DNA binding compound. Sci Rep Nature Publishing Group. 2015;5:16255.
16.
go back to reference Berardinelli F, Siteni S, Tanzarella C, Stevens MF, Sgura A, Antoccia A. The G-quadruplex-stabilising agent RHPS4 induces telomeric dysfunction and enhances radiosensitivity in glioblastoma cells. DNA Repair (Amst) Elsevier BV. 2015;25:104–15.CrossRef Berardinelli F, Siteni S, Tanzarella C, Stevens MF, Sgura A, Antoccia A. The G-quadruplex-stabilising agent RHPS4 induces telomeric dysfunction and enhances radiosensitivity in glioblastoma cells. DNA Repair (Amst) Elsevier BV. 2015;25:104–15.CrossRef
17.
go back to reference Berardinelli F, Sgura A, Facoetti A, Leone S, Vischioni B, Ciocca M, et al. The G-quadruplex-stabilizing ligand RHPS4 enhances sensitivity of U251MG glioblastoma cells to clinical carbon ion beams. FEBS J. 2018;285:1226–36.CrossRef Berardinelli F, Sgura A, Facoetti A, Leone S, Vischioni B, Ciocca M, et al. The G-quadruplex-stabilizing ligand RHPS4 enhances sensitivity of U251MG glioblastoma cells to clinical carbon ion beams. FEBS J. 2018;285:1226–36.CrossRef
18.
19.
go back to reference Bochman ML, Paeschke K, Zakian VA. DNA secondary structures: stability and function of G-quadruplex structures. Nat Rev Genet Nature Publishing Group. 2012;13:770–80.CrossRef Bochman ML, Paeschke K, Zakian VA. DNA secondary structures: stability and function of G-quadruplex structures. Nat Rev Genet Nature Publishing Group. 2012;13:770–80.CrossRef
20.
go back to reference Gowan SM, Heald R, Stevens MF, Kelland LR. Potent inhibition of telomerase by small-molecule pentacyclic acridines capable of interacting with G-quadruplexes. Mol Pharmacol. 2001;60:981–8.CrossRef Gowan SM, Heald R, Stevens MF, Kelland LR. Potent inhibition of telomerase by small-molecule pentacyclic acridines capable of interacting with G-quadruplexes. Mol Pharmacol. 2001;60:981–8.CrossRef
21.
go back to reference Zizza P, Cingolani C, Artuso S, Salvati E, Rizzo A, D’Angelo C, et al. Intragenic G-quadruplex structure formed in the human CD133 and its biological and translational relevance. Nucleic Acids Res. 2016;44:1579–90.CrossRef Zizza P, Cingolani C, Artuso S, Salvati E, Rizzo A, D’Angelo C, et al. Intragenic G-quadruplex structure formed in the human CD133 and its biological and translational relevance. Nucleic Acids Res. 2016;44:1579–90.CrossRef
22.
go back to reference Phatak P, Cookson JC, Dai F, Smith V, Gartenhaus RB, Stevens MFG, et al. Telomere uncapping by the G-quadruplex ligand RHPS4 inhibits clonogenic tumour cell growth in vitro and in vivo consistent with a cancer stem cell targeting mechanism. Br J Cancer. 2007;96:1223–33.CrossRef Phatak P, Cookson JC, Dai F, Smith V, Gartenhaus RB, Stevens MFG, et al. Telomere uncapping by the G-quadruplex ligand RHPS4 inhibits clonogenic tumour cell growth in vitro and in vivo consistent with a cancer stem cell targeting mechanism. Br J Cancer. 2007;96:1223–33.CrossRef
23.
go back to reference Salvati E, Leonetti C, Rizzo A, Scarsella M, Mottolese M, Galati R, et al. Telomere damage induced by the G-quadruplex ligand RHPS4 has an antitumor effect. J Clin Invest. 2007;117:3236–47.CrossRef Salvati E, Leonetti C, Rizzo A, Scarsella M, Mottolese M, Galati R, et al. Telomere damage induced by the G-quadruplex ligand RHPS4 has an antitumor effect. J Clin Invest. 2007;117:3236–47.CrossRef
24.
go back to reference Berardinelli F, Coluzzi E, Sgura A, Antoccia A. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. Mutat Res - Rev Mutat Res Elsevier BV. 2017;773:204–19.CrossRef Berardinelli F, Coluzzi E, Sgura A, Antoccia A. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. Mutat Res - Rev Mutat Res Elsevier BV. 2017;773:204–19.CrossRef
25.
go back to reference Ricci-Vitiani L, Pallini R, Larocca LM, Lombardi DG, Signore M, Pierconti F, et al. Mesenchymal differentiation of glioblastoma stem cells. Cell Death Differ. 2008;15:1491–8.CrossRef Ricci-Vitiani L, Pallini R, Larocca LM, Lombardi DG, Signore M, Pierconti F, et al. Mesenchymal differentiation of glioblastoma stem cells. Cell Death Differ. 2008;15:1491–8.CrossRef
26.
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.CrossRef 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.CrossRef
27.
go back to reference Shachi B, Raul D, Paul AT. Signals and switches in mammalian neural crest cell differentiation. Cold Spring Harb Perspect Biol. 2013;5:1–20. Shachi B, Raul D, Paul AT. Signals and switches in mammalian neural crest cell differentiation. Cold Spring Harb Perspect Biol. 2013;5:1–20.
28.
go back to reference Bai L, Zhou B, Yang CY, Ji J, McEachern D, Przybranowski S, et al. Targeted degradation of BET proteins in triple-negative breast cancer. Cancer Res. 2017;77:2476–87.CrossRef Bai L, Zhou B, Yang CY, Ji J, McEachern D, Przybranowski S, et al. Targeted degradation of BET proteins in triple-negative breast cancer. Cancer Res. 2017;77:2476–87.CrossRef
29.
go back to reference Ricci-Vitiani L, Pedini F, Mollinari C, Condorelli G, Bonci D, Bez A, et al. Absence of caspase 8 and high expression of PED protect primitive neural cells from cell death. J Exp Med. 2004;200:1257–66.CrossRef Ricci-Vitiani L, Pedini F, Mollinari C, Condorelli G, Bonci D, Bez A, et al. Absence of caspase 8 and high expression of PED protect primitive neural cells from cell death. J Exp Med. 2004;200:1257–66.CrossRef
30.
go back to reference Berardinelli F, Antoccia A, Cherubini R, de Nadal V, Gerardi S, Tanzarella C, et al. Telomere alterations and genomic instability in long-term cultures of normal human fibroblasts irradiated with X rays and protons. Radiat Prot Dosim. 2011;143:274–8.CrossRef Berardinelli F, Antoccia A, Cherubini R, de Nadal V, Gerardi S, Tanzarella C, et al. Telomere alterations and genomic instability in long-term cultures of normal human fibroblasts irradiated with X rays and protons. Radiat Prot Dosim. 2011;143:274–8.CrossRef
31.
go back to reference Berardinelli F, Antoccia A, Buonsante R, Gerardi S, Cherubini R, Nadal V. De, et al. the role of telomere length modulation in delayed chromosome instability induced by ionizing radiation in human primary fibroblasts. Environ Mol Mutagen. 2013;54:172–9.CrossRef Berardinelli F, Antoccia A, Buonsante R, Gerardi S, Cherubini R, Nadal V. De, et al. the role of telomere length modulation in delayed chromosome instability induced by ionizing radiation in human primary fibroblasts. Environ Mol Mutagen. 2013;54:172–9.CrossRef
32.
go back to reference Perner S, Bruderlein S, Hasel C, Waibel I, Holdenried A, Ciloglu N, et al. Quantifying telomere lengths of human individual chromosome arms by centromere-calibrated fluorescence in situ hybridization and digital imaging. Am J Pathol. 2003;163:1751–6.CrossRef Perner S, Bruderlein S, Hasel C, Waibel I, Holdenried A, Ciloglu N, et al. Quantifying telomere lengths of human individual chromosome arms by centromere-calibrated fluorescence in situ hybridization and digital imaging. Am J Pathol. 2003;163:1751–6.CrossRef
33.
go back to reference Signore M, Pelacchi F, Di Martino S, Runci D, Biffoni M, Giannetti S, et al. Combined PDK1 and CHK1 inhibition is required to kill glioblastoma stem-like cells in vitro and in vivo. Cell Death Dis Nat Publ Group. 2014;5:e1223–12.CrossRef Signore M, Pelacchi F, Di Martino S, Runci D, Biffoni M, Giannetti S, et al. Combined PDK1 and CHK1 inhibition is required to kill glioblastoma stem-like cells in vitro and in vivo. Cell Death Dis Nat Publ Group. 2014;5:e1223–12.CrossRef
34.
go back to reference Darzynkiewicz Z, Juan G. Analysis of DNA content and BrdU incorporation. Curr Protoc Cytom. 1997:7.7.1–9. Darzynkiewicz Z, Juan G. Analysis of DNA content and BrdU incorporation. Curr Protoc Cytom. 1997:7.7.1–9.
35.
go back to reference Tsurumi C, Esser N, Firat E, Gaedicke S, Follo M, Behe M, et al. Non-invasive in vivo imaging of tumor-associated cd133/prominin. PLoS One. 2010;5.CrossRef Tsurumi C, Esser N, Firat E, Gaedicke S, Follo M, Behe M, et al. Non-invasive in vivo imaging of tumor-associated cd133/prominin. PLoS One. 2010;5.CrossRef
36.
go back to reference Carruthers RD, Ahmed SU, Ramachandran S, Strathdee K, Kurian KM, Hedley A, et al. Replication stress drives constitutive activation of the DNA damage response and radioresistance in glioblastoma stem-like cells. Cancer Res. 2018;78:canres.0569.2018.CrossRef Carruthers RD, Ahmed SU, Ramachandran S, Strathdee K, Kurian KM, Hedley A, et al. Replication stress drives constitutive activation of the DNA damage response and radioresistance in glioblastoma stem-like cells. Cancer Res. 2018;78:canres.0569.2018.CrossRef
37.
go back to reference Manic G, Sistigu A, Corradi F, Musella M, De Maria R, Vitale I. Replication stress response in cancer stem cells as a target for chemotherapy. Semin Cancer Biol Elsevier Ltd. 2018. Manic G, Sistigu A, Corradi F, Musella M, De Maria R, Vitale I. Replication stress response in cancer stem cells as a target for chemotherapy. Semin Cancer Biol Elsevier Ltd. 2018.
38.
go back to reference Zhao X, Zhao YJ, Lin Q, Yu L, Liu Z, Lindsay H, et al. Cytogenetic landscape of paired neurospheres and traditional monolayer cultures in pediatric malignant brain tumors. Neuro-Oncology. 2015;17:965–77.CrossRef Zhao X, Zhao YJ, Lin Q, Yu L, Liu Z, Lindsay H, et al. Cytogenetic landscape of paired neurospheres and traditional monolayer cultures in pediatric malignant brain tumors. Neuro-Oncology. 2015;17:965–77.CrossRef
39.
go back to reference Frosina G. The bright and the dark sides of DNA repair in stem cells. J Biomed Biotechnol. 2010;2010. Frosina G. The bright and the dark sides of DNA repair in stem cells. J Biomed Biotechnol. 2010;2010.
40.
go back to reference Shervington A, Lu C, Patel R, Shervington L. Telomerase downregulation in cancer brain stem cell. Mol Cell Biochem. 2009;331:153–9.CrossRef Shervington A, Lu C, Patel R, Shervington L. Telomerase downregulation in cancer brain stem cell. Mol Cell Biochem. 2009;331:153–9.CrossRef
41.
go back to reference Annovazzi L, Mellai M, Schiffer D. Chemotherapeutic drugs: DNA damage and repair in glioblastoma. Cancers (Basel). 2017;9:1–17. Annovazzi L, Mellai M, Schiffer D. Chemotherapeutic drugs: DNA damage and repair in glioblastoma. Cancers (Basel). 2017;9:1–17.
42.
go back to reference Leavitt JA, Stafford SL, Link MJ, Pollock BE. Long-term evaluation of radiation-induced optic neuropathy after single-fraction stereotactic radiosurgery. Int J Radiat Oncol Biol Phys Elsevier Inc. 2013;87:524–7.CrossRef Leavitt JA, Stafford SL, Link MJ, Pollock BE. Long-term evaluation of radiation-induced optic neuropathy after single-fraction stereotactic radiosurgery. Int J Radiat Oncol Biol Phys Elsevier Inc. 2013;87:524–7.CrossRef
43.
go back to reference Rizzo A, Salvati E, Porru M, D’Angelo C, Stevens MF, D’Incalci M, et al. Stabilization of quadruplex DNA perturbs telomere replication leading to the activation of an ATR-dependent ATM signaling pathway. Nucleic Acids Res. 2009;37:5353–64.CrossRef Rizzo A, Salvati E, Porru M, D’Angelo C, Stevens MF, D’Incalci M, et al. Stabilization of quadruplex DNA perturbs telomere replication leading to the activation of an ATR-dependent ATM signaling pathway. Nucleic Acids Res. 2009;37:5353–64.CrossRef
44.
go back to reference Sfeir A, Kosiyatrakul ST, Hockemeyer D, Macrae SL, Karlseder J, Schildkraut CL. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell Elsevier Ltd. 2009;138:90–103. Sfeir A, Kosiyatrakul ST, Hockemeyer D, Macrae SL, Karlseder J, Schildkraut CL. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell Elsevier Ltd. 2009;138:90–103.
45.
go back to reference Berardinelli F, Antoccia a CR, De Nadal V, Gerardi S, Cirrone G a P, et al. Transient activation of the ALT pathway in human primary fibroblasts exposed to high-LET radiation. Radiat Res. 2010;174:539–49.CrossRef Berardinelli F, Antoccia a CR, De Nadal V, Gerardi S, Cirrone G a P, et al. Transient activation of the ALT pathway in human primary fibroblasts exposed to high-LET radiation. Radiat Res. 2010;174:539–49.CrossRef
46.
go back to reference Muoio D, Berardinelli F, Leone S, Coluzzi E, di Masi A, Doria F, et al. Naphthalene diimide-derivatives G-quadruplex ligands induce cell proliferation inhibition, mild telomeric dysfunction and cell cycle perturbation in U251MG glioma cells. FEBS J. 2018;285:3769–85.CrossRef Muoio D, Berardinelli F, Leone S, Coluzzi E, di Masi A, Doria F, et al. Naphthalene diimide-derivatives G-quadruplex ligands induce cell proliferation inhibition, mild telomeric dysfunction and cell cycle perturbation in U251MG glioma cells. FEBS J. 2018;285:3769–85.CrossRef
47.
go back to reference Chen Y, Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev Elsevier BV. 2012;64:640–65.CrossRef Chen Y, Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev Elsevier BV. 2012;64:640–65.CrossRef
48.
go back to reference D’Alessandris QG, Biffoni M, Martini M, Runci D, Buccarelli M, Cenci T, et al. The clinical value of patient-derived glioblastoma tumorspheres in predicting treatment response. Neuro-Oncology. 2017;19:1097–108.CrossRef D’Alessandris QG, Biffoni M, Martini M, Runci D, Buccarelli M, Cenci T, et al. The clinical value of patient-derived glioblastoma tumorspheres in predicting treatment response. Neuro-Oncology. 2017;19:1097–108.CrossRef
49.
go back to reference Hasegawa D, Okabe S, Okamoto K, Nakano I, Shin-Ya K, Seimiya H. G-quadruplex ligand-induced DNA damage response coupled with telomere dysfunction and replication stress in glioma stem cells. Biochem Biophys Res Commun Elsevier Ltd. 2016;471:75–81.CrossRef Hasegawa D, Okabe S, Okamoto K, Nakano I, Shin-Ya K, Seimiya H. G-quadruplex ligand-induced DNA damage response coupled with telomere dysfunction and replication stress in glioma stem cells. Biochem Biophys Res Commun Elsevier Ltd. 2016;471:75–81.CrossRef
50.
go back to reference Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, Chalmers AJ. Selective inhibition of parallel DNA damage response pathways optimizes radiosensitization of glioblastoma stem-like cells. Cancer Res. 2015;75:4416–28.CrossRef Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, Chalmers AJ. Selective inhibition of parallel DNA damage response pathways optimizes radiosensitization of glioblastoma stem-like cells. Cancer Res. 2015;75:4416–28.CrossRef
51.
go back to reference Iachettini S, Stevens MFG, Frigerio M, Hummersone MG, Hutchinson I, Garner TP, et al. On and off-target effects of telomere uncapping G-quadruplex selective ligands based on pentacyclic acridinium salts. J Exp Clin Cancer Res. 2013;32:1–12.CrossRef Iachettini S, Stevens MFG, Frigerio M, Hummersone MG, Hutchinson I, Garner TP, et al. On and off-target effects of telomere uncapping G-quadruplex selective ligands based on pentacyclic acridinium salts. J Exp Clin Cancer Res. 2013;32:1–12.CrossRef
52.
go back to reference Bartkova J, Hamerlik P, Stockhausen MT, Ehrmann J, Hlobilkova A, Laursen H, et al. Replication stress and oxidative damage contribute to aberrant constitutive activation of DNA damage signalling in human gliomas. Oncogene Nature Publishing Group. 2010;29:5095–102. Bartkova J, Hamerlik P, Stockhausen MT, Ehrmann J, Hlobilkova A, Laursen H, et al. Replication stress and oxidative damage contribute to aberrant constitutive activation of DNA damage signalling in human gliomas. Oncogene Nature Publishing Group. 2010;29:5095–102.
53.
go back to reference Shi W, Feng Z, Zhang J, Gonzalez-Suarez I, Vanderwaal RP, Wu X, et al. The role of RPA2 phosphorylation in homologous recombination in response to replication arrest. Carcinogenesis. 2010;31:994–1002.CrossRef Shi W, Feng Z, Zhang J, Gonzalez-Suarez I, Vanderwaal RP, Wu X, et al. The role of RPA2 phosphorylation in homologous recombination in response to replication arrest. Carcinogenesis. 2010;31:994–1002.CrossRef
54.
go back to reference Osman F, Whitby MC. Exploring the roles of Mus81-Eme1/Mms4 at perturbed replication forks. DNA Repair (Amst). 2007;6:1004–17.CrossRef Osman F, Whitby MC. Exploring the roles of Mus81-Eme1/Mms4 at perturbed replication forks. DNA Repair (Amst). 2007;6:1004–17.CrossRef
55.
go back to reference Tachon G, Cortes U, Guichet P-O, Rivet P, Balbous A, Masliantsev K, et al. Cell cycle changes after Glioblastoma stem cell irradiation: the major role of RAD51. Int J Mol Sci. 2018;19:3018.CrossRef Tachon G, Cortes U, Guichet P-O, Rivet P, Balbous A, Masliantsev K, et al. Cell cycle changes after Glioblastoma stem cell irradiation: the major role of RAD51. Int J Mol Sci. 2018;19:3018.CrossRef
56.
go back to reference Grande S, Palma A, Ricci-Vitiani L, Luciani AM, Buccarelli M, Biffoni M, et al. Metabolic heterogeneity evidenced by MRS among patient-derived glioblastoma multiforme stem-like cells accounts for cell clustering and different responses to drugs. Stem Cells Int. 2018;2018. Grande S, Palma A, Ricci-Vitiani L, Luciani AM, Buccarelli M, Biffoni M, et al. Metabolic heterogeneity evidenced by MRS among patient-derived glioblastoma multiforme stem-like cells accounts for cell clustering and different responses to drugs. Stem Cells Int. 2018;2018.
Metadata
Title
G-quadruplex ligand RHPS4 radiosensitizes glioblastoma xenograft in vivo through a differential targeting of bulky differentiated- and stem-cancer cells
Publication date
01-12-2019
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1293-x

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine