Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Low oxygen tension reverses antineoplastic effect of iron chelator deferasirox in human glioblastoma cells

Authors: Claire Legendre, Sylvie Avril, Catherine Guillet, Emmanuel Garcion

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Overcoming resistance to treatment is an essential issue in many cancers including glioblastoma (GBM), the deadliest primary tumor of the central nervous system. As dependence on iron is a key feature of tumor cells, using chelators to reduce iron represents an opportunity to improve conventional GBM therapies. The aim of the present study was, therefore, to investigate the cytostatic and cytotoxic impact of the new iron chelator deferasirox (DFX) on human GBM cells in well-defined clinical situations represented by radiation therapy and mild-hypoxia.

Results

Under experimental normoxic condition (21 % O2), deferasirox (DFX) used at 10 μM for 3 days reduced proliferation, led cell cycle arrest in S and G2-M phases and induced cytotoxicity and apoptosis in U251 and U87 GBM cells. The abolition of the antineoplastic DFX effects when cells were co-treated with ferric ammonium sulfate supports the hypothesis that its effects result from its ability to chelate iron. As radiotherapy is the main treatment for GBM, the combination of DFX and X-ray beam irradiation was also investigated. Irradiation at a dose of 16 Gy repressed proliferation, cytotoxicity and apoptosis, but only in U251 cells, while no synergy with DFX was observed in either cell line. Importantly, when the same experiment was conducted in mild-hypoxic conditions (3 % O2), the antiproliferative and cytotoxic effects of DFX were abolished, and its ability to deplete iron was also impaired.

Conclusions

Taken together, these in vitro results could raise the question of the benefit of using iron chelators in their native forms under the hypoxic conditions often encountered in solid tumors such as GBM. Developing new chemistry or a new drug delivery system that would keep DFX active in hypoxic cells may be the next step toward their application.
Appendix
Available only for authorised users
Literature
1.
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(10):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(10):987–96.CrossRefPubMed
2.
go back to reference Yu Y, Kovacevic Z, Richardson DR. Tuning cell cycle regulation with an iron key. Cell Cycle. 2007;6(16):1982–94.CrossRefPubMed Yu Y, Kovacevic Z, Richardson DR. Tuning cell cycle regulation with an iron key. Cell Cycle. 2007;6(16):1982–94.CrossRefPubMed
3.
go back to reference Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005;57(4):547–83.CrossRefPubMed Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005;57(4):547–83.CrossRefPubMed
4.
go back to reference Rouault TA. Iron metabolism in the CNS: implications for neurodegenerative diseases. Nat Rev Neurosci. 2013;14(8):551–64.CrossRefPubMed Rouault TA. Iron metabolism in the CNS: implications for neurodegenerative diseases. Nat Rev Neurosci. 2013;14(8):551–64.CrossRefPubMed
5.
go back to reference Neufeld EJ. Oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood. 2006;107(9):3436–41.CrossRefPubMedPubMedCentral Neufeld EJ. Oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood. 2006;107(9):3436–41.CrossRefPubMedPubMedCentral
6.
go back to reference Blatt J, Stitely S. Antineuroblastoma activity of desferoxamine in human cell lines. Cancer Res. 1987;47(7):1749–50.PubMed Blatt J, Stitely S. Antineuroblastoma activity of desferoxamine in human cell lines. Cancer Res. 1987;47(7):1749–50.PubMed
7.
go back to reference Blatt J, Taylor SR, Kontoghiorghes GJ. Comparison of activity of deferoxamine with that of oral iron chelators against human neuroblastoma cell lines. Cancer Res. 1989;49(11):2925–7.PubMed Blatt J, Taylor SR, Kontoghiorghes GJ. Comparison of activity of deferoxamine with that of oral iron chelators against human neuroblastoma cell lines. Cancer Res. 1989;49(11):2925–7.PubMed
8.
go back to reference Blatt J. Deferoxamine in children with recurrent neuroblastoma. Anticancer Res. 1994;14(5B):2109–12.PubMed Blatt J. Deferoxamine in children with recurrent neuroblastoma. Anticancer Res. 1994;14(5B):2109–12.PubMed
9.
go back to reference Donfrancesco A, Deb G, Dominici C, Pileggi D, Castello MA, Helson L. Effects of a single course of deferoxamine in neuroblastoma patients. Cancer Res. 1990;50(16):4929–30.PubMed Donfrancesco A, Deb G, Dominici C, Pileggi D, Castello MA, Helson L. Effects of a single course of deferoxamine in neuroblastoma patients. Cancer Res. 1990;50(16):4929–30.PubMed
10.
go back to reference Selig RA, White L, Gramacho C, Sterling-Levis K, Fraser IW, Naidoo D. Failure of iron chelators to reduce tumor growth in human neuroblastoma xenografts. Cancer Res. 1998;58(3):473–8.PubMed Selig RA, White L, Gramacho C, Sterling-Levis K, Fraser IW, Naidoo D. Failure of iron chelators to reduce tumor growth in human neuroblastoma xenografts. Cancer Res. 1998;58(3):473–8.PubMed
11.
go back to reference Bedford MR, Ford SJ, Horniblow RD, Iqbal TH, Tselepis C. Iron chelation in the treatment of cancer: a new role for deferasirox? J Clin Pharmacol. 2013;53(9):885–91.CrossRefPubMed Bedford MR, Ford SJ, Horniblow RD, Iqbal TH, Tselepis C. Iron chelation in the treatment of cancer: a new role for deferasirox? J Clin Pharmacol. 2013;53(9):885–91.CrossRefPubMed
12.
go back to reference Evans SM, Judy KD, Dunphy I, Jenkins WT, Nelson PT, Collins R, et al. Comparative measurements of hypoxia in human brain tumors using needle electrodes and EF5 binding. Cancer Res. 2004;64(5):1886–92.CrossRefPubMed Evans SM, Judy KD, Dunphy I, Jenkins WT, Nelson PT, Collins R, et al. Comparative measurements of hypoxia in human brain tumors using needle electrodes and EF5 binding. Cancer Res. 2004;64(5):1886–92.CrossRefPubMed
13.
go back to reference Naidu MD, Mason JM, Pica RV, Fung H, Pena LA. Radiation resistance in glioma cells determined by DNA damage repair activity of Ape1/Ref-1. J Radiat Res. 2010;51(4):393–404.CrossRefPubMed Naidu MD, Mason JM, Pica RV, Fung H, Pena LA. Radiation resistance in glioma cells determined by DNA damage repair activity of Ape1/Ref-1. J Radiat Res. 2010;51(4):393–404.CrossRefPubMed
14.
go back to reference Rong Y, Durden DL, Van Meir EG, Brat DJ. ‘Pseudopalisading’ necrosis in glioblastoma: a familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis. J Neuropathol Exp Neurol. 2006;65(6):529–39.CrossRefPubMed Rong Y, Durden DL, Van Meir EG, Brat DJ. ‘Pseudopalisading’ necrosis in glioblastoma: a familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis. J Neuropathol Exp Neurol. 2006;65(6):529–39.CrossRefPubMed
15.
go back to reference Spence AM, Muzi M, Swanson KR, O’Sullivan F, Rockhill JK, Rajendran JG, et al. Regional hypoxia in glioblastoma multiforme quantified with [18F]fluoromisonidazole positron emission tomography before radiotherapy: correlation with time to progression and survival. Clin Cancer Res. 2008;14(9):2623–30.CrossRefPubMedPubMedCentral Spence AM, Muzi M, Swanson KR, O’Sullivan F, Rockhill JK, Rajendran JG, et al. Regional hypoxia in glioblastoma multiforme quantified with [18F]fluoromisonidazole positron emission tomography before radiotherapy: correlation with time to progression and survival. Clin Cancer Res. 2008;14(9):2623–30.CrossRefPubMedPubMedCentral
16.
go back to reference Hoepken HH, Korten T, Robinson SR, Dringen R. Iron accumulation, iron-mediated toxicity and altered levels of ferritin and transferrin receptor in cultured astrocytes during incubation with ferric ammonium citrate. J Neurochem. 2004;88(5):1194–202.CrossRefPubMed Hoepken HH, Korten T, Robinson SR, Dringen R. Iron accumulation, iron-mediated toxicity and altered levels of ferritin and transferrin receptor in cultured astrocytes during incubation with ferric ammonium citrate. J Neurochem. 2004;88(5):1194–202.CrossRefPubMed
17.
18.
go back to reference Yang LP, Keam SJ, Keating GM. Deferasirox : a review of its use in the management of transfusional chronic iron overload. Drugs. 2007;67(15):2211–30.CrossRefPubMed Yang LP, Keam SJ, Keating GM. Deferasirox : a review of its use in the management of transfusional chronic iron overload. Drugs. 2007;67(15):2211–30.CrossRefPubMed
19.
go back to reference Zheng H, Youdim MB, Fridkin M. Selective acetylcholinesterase inhibitor activated by acetylcholinesterase releases an active chelator with neurorescuing and anti-amyloid activities. ACS Chem Neurosci. 2010;1(11):737–46.CrossRefPubMedPubMedCentral Zheng H, Youdim MB, Fridkin M. Selective acetylcholinesterase inhibitor activated by acetylcholinesterase releases an active chelator with neurorescuing and anti-amyloid activities. ACS Chem Neurosci. 2010;1(11):737–46.CrossRefPubMedPubMedCentral
20.
go back to reference Corce V, Renaud S, Cannie I, Julienne K, Gouin SG, Loreal O, et al. Synthesis and biological properties of Quilamines II, new iron chelators with antiproliferative activities. Bioconjug Chem. 2014;25(2):320–34.CrossRefPubMed Corce V, Renaud S, Cannie I, Julienne K, Gouin SG, Loreal O, et al. Synthesis and biological properties of Quilamines II, new iron chelators with antiproliferative activities. Bioconjug Chem. 2014;25(2):320–34.CrossRefPubMed
21.
22.
go back to reference Elstner A, Holtkamp N, von Deimling A. Involvement of Hif-1 in desferrioxamine-induced invasion of glioblastoma cells. Clin Exp Metastasis. 2007;24(1):57–66.CrossRefPubMed Elstner A, Holtkamp N, von Deimling A. Involvement of Hif-1 in desferrioxamine-induced invasion of glioblastoma cells. Clin Exp Metastasis. 2007;24(1):57–66.CrossRefPubMed
23.
go back to reference Weitman SD, Buchanan GR, Kamen BA. Pulmonary toxicity of deferoxamine in children with advanced cancer. J Natl Cancer Inst. 1991;83(24):1834–5.CrossRefPubMed Weitman SD, Buchanan GR, Kamen BA. Pulmonary toxicity of deferoxamine in children with advanced cancer. J Natl Cancer Inst. 1991;83(24):1834–5.CrossRefPubMed
24.
go back to reference Beerepoot LV, Shima DT, Kuroki M, Yeo KT, Voest EE. Up-regulation of vascular endothelial growth factor production by iron chelators. Cancer Res. 1996;56(16):3747–51.PubMed Beerepoot LV, Shima DT, Kuroki M, Yeo KT, Voest EE. Up-regulation of vascular endothelial growth factor production by iron chelators. Cancer Res. 1996;56(16):3747–51.PubMed
25.
go back to reference Schonberg DL, Miller TE, Wu Q, Flavahan WA, Das NK, Hale JS, et al. Preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell. 2015;28(4):441–55.CrossRefPubMedPubMedCentral Schonberg DL, Miller TE, Wu Q, Flavahan WA, Das NK, Hale JS, et al. Preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell. 2015;28(4):441–55.CrossRefPubMedPubMedCentral
27.
go back to reference del Burgo LS, Hernandez RM, Orive G, Pedraz JL. Nanotherapeutic approaches for brain cancer management. Nanomedicine. 2014;10(5):905–19.CrossRef del Burgo LS, Hernandez RM, Orive G, Pedraz JL. Nanotherapeutic approaches for brain cancer management. Nanomedicine. 2014;10(5):905–19.CrossRef
28.
go back to reference Serwer LP, James CD. Challenges in drug delivery to tumors of the central nervous system: an overview of pharmacological and surgical considerations. Adv Drug Deliv Rev. 2012;64(7):590–7.CrossRefPubMed Serwer LP, James CD. Challenges in drug delivery to tumors of the central nervous system: an overview of pharmacological and surgical considerations. Adv Drug Deliv Rev. 2012;64(7):590–7.CrossRefPubMed
29.
go back to reference Kamalinia G, Khodagholi F, Atyabi F, Amini M, Shaerzadeh F, Sharifzadeh M, et al. Enhanced brain delivery of deferasirox-lactoferrin conjugates for iron chelation therapy in neurodegenerative disorders: in vitro and in vivo studies. Mol Pharm. 2013;10(12):4418–31.CrossRefPubMed Kamalinia G, Khodagholi F, Atyabi F, Amini M, Shaerzadeh F, Sharifzadeh M, et al. Enhanced brain delivery of deferasirox-lactoferrin conjugates for iron chelation therapy in neurodegenerative disorders: in vitro and in vivo studies. Mol Pharm. 2013;10(12):4418–31.CrossRefPubMed
30.
go back to reference Liu G, Men P, Harris PL, Rolston RK, Perry G, Smith MA. Nanoparticle iron chelators: a new therapeutic approach in Alzheimer disease and other neurologic disorders associated with trace metal imbalance. Neurosci Lett. 2006;406(3):189–93.CrossRefPubMed Liu G, Men P, Harris PL, Rolston RK, Perry G, Smith MA. Nanoparticle iron chelators: a new therapeutic approach in Alzheimer disease and other neurologic disorders associated with trace metal imbalance. Neurosci Lett. 2006;406(3):189–93.CrossRefPubMed
31.
go back to reference Liu G, Men P, Kudo W, Perry G, Smith MA. Nanoparticle-chelator conjugates as inhibitors of amyloid-beta aggregation and neurotoxicity: a novel therapeutic approach for Alzheimer disease. Neurosci Lett. 2009;455(3):187–90.CrossRefPubMedPubMedCentral Liu G, Men P, Kudo W, Perry G, Smith MA. Nanoparticle-chelator conjugates as inhibitors of amyloid-beta aggregation and neurotoxicity: a novel therapeutic approach for Alzheimer disease. Neurosci Lett. 2009;455(3):187–90.CrossRefPubMedPubMedCentral
33.
go back to reference Pullarkat V, Meng Z, Donohue C, Yamamoto VN, Tomassetti S, Bhatia R, et al. Iron chelators induce autophagic cell death in multiple myeloma cells. Leuk Res. 2014;38(8):988–96.CrossRefPubMed Pullarkat V, Meng Z, Donohue C, Yamamoto VN, Tomassetti S, Bhatia R, et al. Iron chelators induce autophagic cell death in multiple myeloma cells. Leuk Res. 2014;38(8):988–96.CrossRefPubMed
34.
go back to reference Paubelle E, Zylbersztejn F, Alkhaeir S, Suarez F, Callens C, Dussiot M, et al. Deferasirox and vitamin D improves overall survival in elderly patients with acute myeloid leukemia after demethylating agents failure. PLoS One. 2013;8(6):e65998.CrossRefPubMedPubMedCentral Paubelle E, Zylbersztejn F, Alkhaeir S, Suarez F, Callens C, Dussiot M, et al. Deferasirox and vitamin D improves overall survival in elderly patients with acute myeloid leukemia after demethylating agents failure. PLoS One. 2013;8(6):e65998.CrossRefPubMedPubMedCentral
35.
go back to reference Lee DH, Jang PS, Chung NG, Cho B, Jeong DC, Kim HK. Deferasirox shows in vitro and in vivo antileukemic effects on murine leukemic cell lines regardless of iron status. Exp Hematol. 2013;41(6):539–46.CrossRefPubMed Lee DH, Jang PS, Chung NG, Cho B, Jeong DC, Kim HK. Deferasirox shows in vitro and in vivo antileukemic effects on murine leukemic cell lines regardless of iron status. Exp Hematol. 2013;41(6):539–46.CrossRefPubMed
36.
go back to reference Fukushima T, Kawabata H, Nakamura T, Iwao H, Nakajima A, Miki M, et al. Iron chelation therapy with deferasirox induced complete remission in a patient with chemotherapy-resistant acute monocytic leukemia. Anticancer Res. 2011;31(5):1741–4.PubMed Fukushima T, Kawabata H, Nakamura T, Iwao H, Nakajima A, Miki M, et al. Iron chelation therapy with deferasirox induced complete remission in a patient with chemotherapy-resistant acute monocytic leukemia. Anticancer Res. 2011;31(5):1741–4.PubMed
37.
go back to reference Kim JL, Kang HN, Kang MH, Yoo YA, Kim JS, Choi CW. The oral iron chelator deferasirox induces apoptosis in myeloid leukemia cells by targeting caspase. Acta Haematol. 2011;126(4):241–5.CrossRefPubMed Kim JL, Kang HN, Kang MH, Yoo YA, Kim JS, Choi CW. The oral iron chelator deferasirox induces apoptosis in myeloid leukemia cells by targeting caspase. Acta Haematol. 2011;126(4):241–5.CrossRefPubMed
38.
go back to reference Messa E, Carturan S, Maffe C, Pautasso M, Bracco E, Roetto A, et al. Deferasirox is a powerful NF-kappaB inhibitor in myelodysplastic cells and in leukemia cell lines acting independently from cell iron deprivation by chelation and reactive oxygen species scavenging. Haematologica. 2010;95(8):1308–16.CrossRefPubMedPubMedCentral Messa E, Carturan S, Maffe C, Pautasso M, Bracco E, Roetto A, et al. Deferasirox is a powerful NF-kappaB inhibitor in myelodysplastic cells and in leukemia cell lines acting independently from cell iron deprivation by chelation and reactive oxygen species scavenging. Haematologica. 2010;95(8):1308–16.CrossRefPubMedPubMedCentral
39.
go back to reference Ohyashiki JH, Kobayashi C, Hamamura R, Okabe S, Tauchi T, Ohyashiki K. The oral iron chelator deferasirox represses signaling through the mTOR in myeloid leukemia cells by enhancing expression of REDD1. Cancer Sci. 2009;100(5):970–7.CrossRefPubMed Ohyashiki JH, Kobayashi C, Hamamura R, Okabe S, Tauchi T, Ohyashiki K. The oral iron chelator deferasirox represses signaling through the mTOR in myeloid leukemia cells by enhancing expression of REDD1. Cancer Sci. 2009;100(5):970–7.CrossRefPubMed
40.
go back to reference Ford SJ, Bedford MR, Pang W, Wood A, Iqbal T, Tselepis C, et al. A comparative study of the iron status of patients with oesophageal adenocarcinoma to determine suitability for a clinical trial of iron chelation therapy. Ann R Coll Surg Engl. 2014;96(4):275–8.CrossRefPubMedPubMedCentral Ford SJ, Bedford MR, Pang W, Wood A, Iqbal T, Tselepis C, et al. A comparative study of the iron status of patients with oesophageal adenocarcinoma to determine suitability for a clinical trial of iron chelation therapy. Ann R Coll Surg Engl. 2014;96(4):275–8.CrossRefPubMedPubMedCentral
41.
go back to reference Ford SJ, Obeidy P, Lovejoy DB, Bedford M, Nichols L, Chadwick C, et al. Deferasirox (ICL670A) effectively inhibits oesophageal cancer growth in vitro and in vivo. Br J Pharmacol. 2013;168(6):1316–28.CrossRefPubMedPubMedCentral Ford SJ, Obeidy P, Lovejoy DB, Bedford M, Nichols L, Chadwick C, et al. Deferasirox (ICL670A) effectively inhibits oesophageal cancer growth in vitro and in vivo. Br J Pharmacol. 2013;168(6):1316–28.CrossRefPubMedPubMedCentral
42.
go back to reference Vazana-Barad L, Granot G, Mor-Tzuntz R, Levi I, Dreyling M, Nathan I, et al. Mechanism of the antitumoral activity of deferasirox, an iron chelation agent, on mantle cell lymphoma. Leuk Lymphoma. 2013;54(4):851–9.CrossRefPubMed Vazana-Barad L, Granot G, Mor-Tzuntz R, Levi I, Dreyling M, Nathan I, et al. Mechanism of the antitumoral activity of deferasirox, an iron chelation agent, on mantle cell lymphoma. Leuk Lymphoma. 2013;54(4):851–9.CrossRefPubMed
43.
go back to reference Choi JG, Kim JL, Park J, Lee S, Park SJ, Kim JS, et al. Effects of oral iron chelator deferasirox on human malignant lymphoma cells. Korean J Hematol. 2012;47(3):194–201.CrossRefPubMedPubMedCentral Choi JG, Kim JL, Park J, Lee S, Park SJ, Kim JS, et al. Effects of oral iron chelator deferasirox on human malignant lymphoma cells. Korean J Hematol. 2012;47(3):194–201.CrossRefPubMedPubMedCentral
44.
go back to reference Lui GY, Obeidy P, Ford SJ, Tselepis C, Sharp DM, Jansson PJ, et al. The iron chelator, deferasirox, as a novel strategy for cancer treatment: oral activity against human lung tumor xenografts and molecular mechanism of action. Mol Pharmacol. 2013;83(1):179–90.CrossRefPubMed Lui GY, Obeidy P, Ford SJ, Tselepis C, Sharp DM, Jansson PJ, et al. The iron chelator, deferasirox, as a novel strategy for cancer treatment: oral activity against human lung tumor xenografts and molecular mechanism of action. Mol Pharmacol. 2013;83(1):179–90.CrossRefPubMed
45.
go back to reference Song S, Christova T, Perusini S, Alizadeh S, Bao RY, Miller BW, et al. Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 2011;71(24):7628–39.CrossRefPubMed Song S, Christova T, Perusini S, Alizadeh S, Bao RY, Miller BW, et al. Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 2011;71(24):7628–39.CrossRefPubMed
46.
go back to reference Gaboriau F, Leray AM, Ropert M, Gouffier L, Cannie I, Troadec MB, et al. Effects of deferasirox and deferiprone on cellular iron load in the human hepatoma cell line HepaRG. Biometals. 2010;23(2):231–45.CrossRefPubMed Gaboriau F, Leray AM, Ropert M, Gouffier L, Cannie I, Troadec MB, et al. Effects of deferasirox and deferiprone on cellular iron load in the human hepatoma cell line HepaRG. Biometals. 2010;23(2):231–45.CrossRefPubMed
47.
go back to reference Lescoat G, Chantrel-Groussard K, Pasdeloup N, Nick H, Brissot P, Gaboriau F. Antiproliferative and apoptotic effects in rat and human hepatoma cell cultures of the orally active iron chelator ICL670 compared to CP20: a possible relationship with polyamine metabolism. Cell Prolif. 2007;40(5):755–67.CrossRefPubMed Lescoat G, Chantrel-Groussard K, Pasdeloup N, Nick H, Brissot P, Gaboriau F. Antiproliferative and apoptotic effects in rat and human hepatoma cell cultures of the orally active iron chelator ICL670 compared to CP20: a possible relationship with polyamine metabolism. Cell Prolif. 2007;40(5):755–67.CrossRefPubMed
48.
go back to reference Chantrel-Groussard K, Gaboriau F, Pasdeloup N, Havouis R, Nick H, Pierre JL, et al. The new orally active iron chelator ICL670A exhibits a higher antiproliferative effect in human hepatocyte cultures than O-trensox. Eur J Pharmacol. 2006;541(3):129–37.CrossRefPubMed Chantrel-Groussard K, Gaboriau F, Pasdeloup N, Havouis R, Nick H, Pierre JL, et al. The new orally active iron chelator ICL670A exhibits a higher antiproliferative effect in human hepatocyte cultures than O-trensox. Eur J Pharmacol. 2006;541(3):129–37.CrossRefPubMed
Metadata
Title
Low oxygen tension reverses antineoplastic effect of iron chelator deferasirox in human glioblastoma cells
Authors
Claire Legendre
Sylvie Avril
Catherine Guillet
Emmanuel Garcion
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2074-y

Other articles of this Issue 1/2016

BMC Cancer 1/2016 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