Skip to main content
Erschienen in: Journal of Neuro-Oncology 3/2018

21.08.2018 | Laboratory Investigation

Oxaliplatin disrupts pathological features of glioma cells and associated macrophages independent of apoptosis induction

verfasst von: Nathan B. Roberts, Aymen Alqazzaz, Jacqueline R. Hwang, Xiulan Qi, Achsah D. Keegan, Anthony J. Kim, Jeffrey A. Winkles, Graeme F. Woodworth

Erschienen in: Journal of Neuro-Oncology | Ausgabe 3/2018

Einloggen, um Zugang zu erhalten

Abstract

Introduction

Emerging evidence suggests that effective treatment of glioblastoma (GBM), the most common and deadly form of adult primary brain cancer, will likely require concurrent treatment of multiple aspects of tumor pathobiology to overcome tumor heterogeneity and the complex tumor-supporting microenvironment. Recent studies in non-central nervous system (CNS) tumor cells have demonstrated that oxaliplatin (OXA) can induce multi-faceted anti-tumor effects, in particular at drug concentrations below those required to induce apoptosis. These findings motivated re-investigation of OXA for the treatment of GBM.

Methods

The effects of OXA on murine KR158 and GL261 glioma cells including cell growth, cell death, inhibition of signal transducer and activator of transcription (STAT) activity, O-6-methylguanine-DNA methyltransferase (MGMT) expression, and immunogenic cell death (ICD) initiation, were evaluated by cytotoxicity assays, Western blot analysis, STAT3-luciferase reporter assays, qRT-PCR assays, and flow cytometry. Chemical inhibitors of endoplasmic reticulum (ER) stress were used to investigate the contribution of this cell damage response to the observed OXA effects. The effect of OXA on bone marrow-derived macrophages (BMDM) exposed to glioma conditioned media (GCM) was also analyzed by Western blot analysis.

Results

We identified the OXA concentration threshold for induction of apoptosis and from this determined the drug dose and treatment period for sub-cytotoxic treatments of glioma cells. Under these experimental conditions, OXA reduced STAT3 activity, reduced MGMT levels and increased temozolomide sensitivity. In addition, there was evidence of immunogenic cell death (elevated EIF2α phosphorylation and calreticulin exposure) following prolonged OXA treatment. Notably, inhibition of ER stress reversed the OXA-mediated inhibition of STAT3 activity and MGMT expression in the tumor cells. In BMDMs exposed to GCM, OXA also reduced levels of phosphorylated STAT3 and decreased expression of Arginase 1, an enzyme known to contribute to pro-tumor functions in the tumor-immune environment.

Conclusions

OXA can induce notable multi-faceted biological effects in glioma cells and BMDMs at relatively low drug concentrations. These findings may have significant therapeutic relevance against GBM and warrant further investigation.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Rolle CE, Sengupta S, Lesniak MS (2012) Mechanisms of immune evasion by gliomas. Adv Exp Med Biol 746:53–76CrossRefPubMed Rolle CE, Sengupta S, Lesniak MS (2012) Mechanisms of immune evasion by gliomas. Adv Exp Med Biol 746:53–76CrossRefPubMed
2.
Zurück zum Zitat Aum DJ, Kim DH, Beaumont TL, Leuthardt EC, Dunn GP, Kim AH (2014) Molecular and cellular heterogeneity: the hallmark of glioblastoma. Neurosurg Focus 37(6):E11CrossRefPubMed Aum DJ, Kim DH, Beaumont TL, Leuthardt EC, Dunn GP, Kim AH (2014) Molecular and cellular heterogeneity: the hallmark of glioblastoma. Neurosurg Focus 37(6):E11CrossRefPubMed
3.
Zurück zum Zitat Shah AH, Graham R, Bregy A, Thambuswamy M, Komotar RJ (2014) Recognizing and correcting failures in glioblastoma treatment. Cancer Invest 32(6):299–302CrossRefPubMed Shah AH, Graham R, Bregy A, Thambuswamy M, Komotar RJ (2014) Recognizing and correcting failures in glioblastoma treatment. Cancer Invest 32(6):299–302CrossRefPubMed
4.
Zurück zum Zitat Wang H, Xu T, Jiang Y, Xu H, Yan Y, Fu D, Chen J (2015) The challenges and the promise of molecular targeted therapy in malignant gliomas. Neoplasia 17(3):239–255CrossRefPubMedPubMedCentral Wang H, Xu T, Jiang Y, Xu H, Yan Y, Fu D, Chen J (2015) The challenges and the promise of molecular targeted therapy in malignant gliomas. Neoplasia 17(3):239–255CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Cross-Knorr S, Lu S, Perez K, Guevara S, Brilliant K, Pisano C, Quesenberry PJ, Resnick MB, Chatterjee D (2013) RKIP phosphorylation and STAT3 activation is inhibited by oxaliplatin and camptothecin and are associated with poor prognosis in stage II colon cancer patients. BMC Cancer 13:463CrossRefPubMedPubMedCentral Cross-Knorr S, Lu S, Perez K, Guevara S, Brilliant K, Pisano C, Quesenberry PJ, Resnick MB, Chatterjee D (2013) RKIP phosphorylation and STAT3 activation is inhibited by oxaliplatin and camptothecin and are associated with poor prognosis in stage II colon cancer patients. BMC Cancer 13:463CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Lesterhuis WJ, Punt CJ, Hato SV, Eleveld-Trancikova D, Jansen BJ, Nierkens S, Schreibelt G, de Boer A, Van Herpen CM, Kaanders JH, van Krieken JH, Adema GJ, Figdor CG, de Vries IJ (2011) Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice. J Clin Invest 121(8):3100–3108CrossRefPubMedPubMedCentral Lesterhuis WJ, Punt CJ, Hato SV, Eleveld-Trancikova D, Jansen BJ, Nierkens S, Schreibelt G, de Boer A, Van Herpen CM, Kaanders JH, van Krieken JH, Adema GJ, Figdor CG, de Vries IJ (2011) Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice. J Clin Invest 121(8):3100–3108CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Hato SV, Figdor CG, Takahashi S, Pen AE, Halilovic A, Bol KF, Vasaturo A, Inoue Y, de Haas N, Verweij D, Van Herpen CML, Kaanders JH, van Krieken J, Van Laarhoven HWM, Hooijer GKJ, Punt CJA, Asai A, de Vries IJM, Lesterhuis WJ (2017) Direct inhibition of STAT signaling by platinum drugs contributes to their anti-cancer activity. Oncotarget 8(33):54434CrossRefPubMedPubMedCentral Hato SV, Figdor CG, Takahashi S, Pen AE, Halilovic A, Bol KF, Vasaturo A, Inoue Y, de Haas N, Verweij D, Van Herpen CML, Kaanders JH, van Krieken J, Van Laarhoven HWM, Hooijer GKJ, Punt CJA, Asai A, de Vries IJM, Lesterhuis WJ (2017) Direct inhibition of STAT signaling by platinum drugs contributes to their anti-cancer activity. Oncotarget 8(33):54434CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Hato SV, de Vries IJM, Lesterhuis WJ (2012) STATing the importance of immune modulation by platinum chemotherapeutics. Oncoimmunology 1(2):234–236CrossRefPubMedPubMedCentral Hato SV, de Vries IJM, Lesterhuis WJ (2012) STATing the importance of immune modulation by platinum chemotherapeutics. Oncoimmunology 1(2):234–236CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Wang W, Wu L, Zhang J, Wu H, Han E, Guo Q (2017) Chemoimmunotherapy by combining oxaliplatin with immune checkpoint blockades reduced tumor burden in colorectal cancer animal model. Biochem Biophys Res Commun 487(1):1–7CrossRefPubMed Wang W, Wu L, Zhang J, Wu H, Han E, Guo Q (2017) Chemoimmunotherapy by combining oxaliplatin with immune checkpoint blockades reduced tumor burden in colorectal cancer animal model. Biochem Biophys Res Commun 487(1):1–7CrossRefPubMed
10.
Zurück zum Zitat Bruno PM, Liu Y, Park GY, Murai J, Koch CE, Eisen TJ, Pritchard JR, Pommier Y, Lippard SJ, Hemann MT (2017) A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress. Nat Med 23(4):461–471CrossRefPubMedPubMedCentral Bruno PM, Liu Y, Park GY, Murai J, Koch CE, Eisen TJ, Pritchard JR, Pommier Y, Lippard SJ, Hemann MT (2017) A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress. Nat Med 23(4):461–471CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, Aymeric L, Michaud M, Apetoh L, Barault L, Mendiboure J, Pignon JP, Jooste V, van Endert P, Ducreux M, Zitvogel L, Piard F, Kroemer G (2010) Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 29(4):482–491CrossRefPubMed Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, Aymeric L, Michaud M, Apetoh L, Barault L, Mendiboure J, Pignon JP, Jooste V, van Endert P, Ducreux M, Zitvogel L, Piard F, Kroemer G (2010) Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 29(4):482–491CrossRefPubMed
12.
Zurück zum Zitat Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, Yamazaki T, Martins I, Sukkurwala AQ, Michaud M, Senovilla L, Galluzzi L, Kroemer G, Zitvogel L (2013) Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev 24(4):311–318CrossRefPubMed Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, Yamazaki T, Martins I, Sukkurwala AQ, Michaud M, Senovilla L, Galluzzi L, Kroemer G, Zitvogel L (2013) Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev 24(4):311–318CrossRefPubMed
13.
Zurück zum Zitat Hato SV, Khong A, de Vries IJ, Lesterhuis WJ (2014) Molecular pathways: the immunogenic effects of platinum-based chemotherapeutics. Clin Cancer Res 20(11):2831–2837CrossRefPubMed Hato SV, Khong A, de Vries IJ, Lesterhuis WJ (2014) Molecular pathways: the immunogenic effects of platinum-based chemotherapeutics. Clin Cancer Res 20(11):2831–2837CrossRefPubMed
14.
Zurück zum Zitat Roberts NB, Wadajkar AS, Winkles JA, Davila E, Kim AJ, Woodworth GF (2016) Repurposing platinum-based chemotherapies for multi-modal treatment of glioblastoma. Oncoimmunology 5(9):e1208876CrossRefPubMedPubMedCentral Roberts NB, Wadajkar AS, Winkles JA, Davila E, Kim AJ, Woodworth GF (2016) Repurposing platinum-based chemotherapies for multi-modal treatment of glioblastoma. Oncoimmunology 5(9):e1208876CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, Tesniere A, Zitvogel L, Kroemer G (2011) Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene 30(10):1147–1158CrossRefPubMed Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, Tesniere A, Zitvogel L, Kroemer G (2011) Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene 30(10):1147–1158CrossRefPubMed
16.
Zurück zum Zitat Carvalho da Fonseca AC, Badie B (2013) Microglia and macrophages in malignant gliomas: recent discoveries and implications for promising therapies. Clin Dev Immunol 2013:264124CrossRefPubMedCentral Carvalho da Fonseca AC, Badie B (2013) Microglia and macrophages in malignant gliomas: recent discoveries and implications for promising therapies. Clin Dev Immunol 2013:264124CrossRefPubMedCentral
17.
Zurück zum Zitat Ferguson SD, Srinivasan VM, Heimberger AB (2015) The role of STAT3 in tumor-mediated immune suppression. J Neurooncol 123(3):385–394CrossRefPubMed Ferguson SD, Srinivasan VM, Heimberger AB (2015) The role of STAT3 in tumor-mediated immune suppression. J Neurooncol 123(3):385–394CrossRefPubMed
18.
Zurück zum Zitat Morantz RA, Wood GW, Foster M, Clark M, Gollahon K (1979) Macrophages in experimental and human brain tumors. Part 2: studies of the macrophage content of human brain tumors. J Neurosurg 50(3):305–311CrossRefPubMed Morantz RA, Wood GW, Foster M, Clark M, Gollahon K (1979) Macrophages in experimental and human brain tumors. Part 2: studies of the macrophage content of human brain tumors. J Neurosurg 50(3):305–311CrossRefPubMed
19.
Zurück zum Zitat Hersh DS, Peng S, Dancy JG, Galisteo R, Eschbacher JM, Castellani RJ, Heath JE, Legesse T, Kim AJ, Woodworth GF, Tran NL, Winkles JA (2018) Differential expression of the TWEAK receptor Fn14 in IDH1 wild-type and mutant gliomas. J Neurooncol 138(2):241–250CrossRefPubMedPubMedCentral Hersh DS, Peng S, Dancy JG, Galisteo R, Eschbacher JM, Castellani RJ, Heath JE, Legesse T, Kim AJ, Woodworth GF, Tran NL, Winkles JA (2018) Differential expression of the TWEAK receptor Fn14 in IDH1 wild-type and mutant gliomas. J Neurooncol 138(2):241–250CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Ouédraogo ZG, Biau J, Kemeny J-L, Morel L, Verrelle P, Chautard E (2017) Role of STAT3 in genesis and progression of human malignant gliomas. Mol Neurobiol 54(8):5780–5797CrossRefPubMed Ouédraogo ZG, Biau J, Kemeny J-L, Morel L, Verrelle P, Chautard E (2017) Role of STAT3 in genesis and progression of human malignant gliomas. Mol Neurobiol 54(8):5780–5797CrossRefPubMed
21.
Zurück zum Zitat Zhong Z, Wen Z, Darnell JE, Jr (1994) Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 264(5155):95–98CrossRefPubMed Zhong Z, Wen Z, Darnell JE, Jr (1994) Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 264(5155):95–98CrossRefPubMed
22.
Zurück zum Zitat Iwamaru A, Szymanski S, Iwado E, Aoki H, Yokoyama T, Fokt I, Hess K, Conrad C, Madden T, Sawaya R, Kondo S, Priebe W, Kondo Y (2007) A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene 26(17):2435–2444CrossRefPubMed Iwamaru A, Szymanski S, Iwado E, Aoki H, Yokoyama T, Fokt I, Hess K, Conrad C, Madden T, Sawaya R, Kondo S, Priebe W, Kondo Y (2007) A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene 26(17):2435–2444CrossRefPubMed
23.
Zurück zum Zitat Jiang G, Jiang AJ, Xin Y, Li LT, Cheng Q, Zheng JN (2014) Progression of O(6)-methylguanine-DNA methyltransferase and temozolomide resistance in cancer research. Mol Biol Rep 41(10):6659–6665CrossRefPubMed Jiang G, Jiang AJ, Xin Y, Li LT, Cheng Q, Zheng JN (2014) Progression of O(6)-methylguanine-DNA methyltransferase and temozolomide resistance in cancer research. Mol Biol Rep 41(10):6659–6665CrossRefPubMed
24.
Zurück zum Zitat Fan CH, Liu WL, Cao H, Wen C, Chen L, Jiang G (2013) O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas. Cell Death Dis 4:e876CrossRefPubMedPubMedCentral Fan CH, Liu WL, Cao H, Wen C, Chen L, Jiang G (2013) O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas. Cell Death Dis 4:e876CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Shi S, Tan P, Yan B, Gao R, Zhao J, Wang J, Guo J, Li N, Ma Z (2016) ER stress and autophagy are involved in the apoptosis induced by cisplatin in human lung cancer cells. Oncol Rep 35(5):2606–2614CrossRefPubMedPubMedCentral Shi S, Tan P, Yan B, Gao R, Zhao J, Wang J, Guo J, Li N, Ma Z (2016) ER stress and autophagy are involved in the apoptosis induced by cisplatin in human lung cancer cells. Oncol Rep 35(5):2606–2614CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Shi Y, Tang B, Yu P-W, Tang B, Hao Y-X, Lei X, Luo H-X, Zeng D-Z (2012) Autophagy protects against oxaliplatin-induced cell death via ER stress and ROS in Caco-2 cells. PLoS ONE 7(11):e51076CrossRefPubMedPubMedCentral Shi Y, Tang B, Yu P-W, Tang B, Hao Y-X, Lei X, Luo H-X, Zeng D-Z (2012) Autophagy protects against oxaliplatin-induced cell death via ER stress and ROS in Caco-2 cells. PLoS ONE 7(11):e51076CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Boyce M, Bryant KF, Jousse C, Long K, Harding HP, Scheuner D, Kaufman RJ, Ma D, Coen DM, Ron D, Yuan J (2005) A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science 307(5711):935–939CrossRefPubMed Boyce M, Bryant KF, Jousse C, Long K, Harding HP, Scheuner D, Kaufman RJ, Ma D, Coen DM, Ron D, Yuan J (2005) A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science 307(5711):935–939CrossRefPubMed
28.
Zurück zum Zitat Kubota K, Niinuma Y, Kaneko M, Okuma Y, Sugai M, Omura T, Uesugi M, Uehara T, Hosoi T, Nomura Y (2006) Suppressive effects of 4-phenylbutyrate on the aggregation of Pael receptors and endoplasmic reticulum stress. J Neurochem 97(5):1259–1268CrossRefPubMed Kubota K, Niinuma Y, Kaneko M, Okuma Y, Sugai M, Omura T, Uesugi M, Uehara T, Hosoi T, Nomura Y (2006) Suppressive effects of 4-phenylbutyrate on the aggregation of Pael receptors and endoplasmic reticulum stress. J Neurochem 97(5):1259–1268CrossRefPubMed
29.
Zurück zum Zitat Xipell E, Aragon T, Martinez-Velez N, Vera B, Idoate MA, Martinez-Irujo JJ, Garzon AG, Gonzalez-Huarriz M, Acanda AM, Jones C, Lang FF, Fueyo J, Gomez-Manzano C, Alonso MM (2016) Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51. Neuro Oncol 18(8):1109–1119CrossRefPubMedPubMedCentral Xipell E, Aragon T, Martinez-Velez N, Vera B, Idoate MA, Martinez-Irujo JJ, Garzon AG, Gonzalez-Huarriz M, Acanda AM, Jones C, Lang FF, Fueyo J, Gomez-Manzano C, Alonso MM (2016) Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51. Neuro Oncol 18(8):1109–1119CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, Durchschlag M, Joza N, Pierron G, van Endert P, Yuan J, Zitvogel L, Madeo F, Williams DB, Kroemer G (2009) Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. Embo j 28(5):578–590CrossRefPubMedPubMedCentral Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, Durchschlag M, Joza N, Pierron G, van Endert P, Yuan J, Zitvogel L, Madeo F, Williams DB, Kroemer G (2009) Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. Embo j 28(5):578–590CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B (2009) Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia 57(13):1458–1467CrossRefPubMed Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B (2009) Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia 57(13):1458–1467CrossRefPubMed
32.
Zurück zum Zitat Fujiwara Y, Komohara Y, Kudo R, Tsurushima K, Ohnishi K, Ikeda T, Takeya M (2011) Oleanolic acid inhibits macrophage differentiation into the M2 phenotype and glioblastoma cell proliferation by suppressing the activation of STAT3. Oncol Rep 26(6):1533–1537PubMed Fujiwara Y, Komohara Y, Kudo R, Tsurushima K, Ohnishi K, Ikeda T, Takeya M (2011) Oleanolic acid inhibits macrophage differentiation into the M2 phenotype and glioblastoma cell proliferation by suppressing the activation of STAT3. Oncol Rep 26(6):1533–1537PubMed
33.
Zurück zum Zitat Yao Y, Ye H, Qi Z, Mo L, Yue Q, Baral A, Hoon DSB, Vera JC, Heiss JD, Chen CC, Zhang J, Jin K, Wang Y, Zang X, Mao Y, Zhou L (2016) B7-H4(B7x)-mediated cross-talk between glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway lead to poor prognosis in glioma patients. Clin Cancer Res 22(11):2778–2790CrossRefPubMedPubMedCentral Yao Y, Ye H, Qi Z, Mo L, Yue Q, Baral A, Hoon DSB, Vera JC, Heiss JD, Chen CC, Zhang J, Jin K, Wang Y, Zang X, Mao Y, Zhou L (2016) B7-H4(B7x)-mediated cross-talk between glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway lead to poor prognosis in glioma patients. Clin Cancer Res 22(11):2778–2790CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Luwor RB, Stylli SS, Kaye AH (2013) The role of Stat3 in glioblastoma multiforme. J Clin Neurosci 20(7):907–911CrossRefPubMed Luwor RB, Stylli SS, Kaye AH (2013) The role of Stat3 in glioblastoma multiforme. J Clin Neurosci 20(7):907–911CrossRefPubMed
35.
Zurück zum Zitat Kadowaki H, Nishitoh H (2013) Signaling pathways from the endoplasmic reticulum and their roles in disease. Genes (Basel) 4(3):306–333CrossRef Kadowaki H, Nishitoh H (2013) Signaling pathways from the endoplasmic reticulum and their roles in disease. Genes (Basel) 4(3):306–333CrossRef
36.
Zurück zum Zitat Buckner JC, Ballman KV, Michalak JC, Burton GV, Cascino TL, Schomberg PJ, Hawkins RB, Scheithauer BW, Sandler HM, Marks RS, O’Fallon JR (2006) Phase III trial of carmustine and cisplatin compared with carmustine alone and standard radiation therapy or accelerated radiation therapy in patients with glioblastoma multiforme: North Central Cancer Treatment Group 93-72-52 and Southwest Oncology Group 9503 Trials. J Clin Oncol 24(24):3871–3879CrossRefPubMed Buckner JC, Ballman KV, Michalak JC, Burton GV, Cascino TL, Schomberg PJ, Hawkins RB, Scheithauer BW, Sandler HM, Marks RS, O’Fallon JR (2006) Phase III trial of carmustine and cisplatin compared with carmustine alone and standard radiation therapy or accelerated radiation therapy in patients with glioblastoma multiforme: North Central Cancer Treatment Group 93-72-52 and Southwest Oncology Group 9503 Trials. J Clin Oncol 24(24):3871–3879CrossRefPubMed
37.
Zurück zum Zitat Boulikas T (2004) Low toxicity and anticancer activity of a novel liposomal cisplatin (Lipoplatin) in mouse xenografts. Oncol Rep 12(1):3–12PubMed Boulikas T (2004) Low toxicity and anticancer activity of a novel liposomal cisplatin (Lipoplatin) in mouse xenografts. Oncol Rep 12(1):3–12PubMed
38.
Zurück zum Zitat Jeremic B, Grujicic D, Jevremovic S, Stanisavljevic B, Milojevic L, Djuric L, Mijatovic L (1992) Carboplatin and etoposide chemotherapy regimen for recurrent malignant glioma: a phase II study. J Clin Oncol 10(7):1074–1077CrossRefPubMed Jeremic B, Grujicic D, Jevremovic S, Stanisavljevic B, Milojevic L, Djuric L, Mijatovic L (1992) Carboplatin and etoposide chemotherapy regimen for recurrent malignant glioma: a phase II study. J Clin Oncol 10(7):1074–1077CrossRefPubMed
39.
Zurück zum Zitat Peterson K, Harsh Gt, Fisher PG, Adler J, Le Q (2001) Daily low-dose carboplatin as a radiation sensitizer for newly diagnosed malignant glioma. J Neurooncol 53(1):27–32CrossRefPubMed Peterson K, Harsh Gt, Fisher PG, Adler J, Le Q (2001) Daily low-dose carboplatin as a radiation sensitizer for newly diagnosed malignant glioma. J Neurooncol 53(1):27–32CrossRefPubMed
40.
Zurück zum Zitat Brandes AA, Rigon A, Zampieri P, Ermani M, Carollo C, Altavilla G, Turazzi S, Chierichetti F, Florentino MV (1998) Carboplatin and teniposide concurrent with radiotherapy in patients with glioblastoma multiforme: a phase II study. Cancer 82(2):355–361CrossRefPubMed Brandes AA, Rigon A, Zampieri P, Ermani M, Carollo C, Altavilla G, Turazzi S, Chierichetti F, Florentino MV (1998) Carboplatin and teniposide concurrent with radiotherapy in patients with glioblastoma multiforme: a phase II study. Cancer 82(2):355–361CrossRefPubMed
41.
Zurück zum Zitat Jeremic B, Shibamoto Y, Acimovic L, Milicic B, Milisavljevic S, Nikolic N, Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (2001) Hyperfractionated radiation therapy and concurrent low-dose, daily carboplatin/etoposide with or without weekend carboplatin/etoposide chemotherapy in stage III non-small-cell lung cancer: a randomized trial. Int J Radiat Oncol Biol Phys 50(1):19–25CrossRefPubMed Jeremic B, Shibamoto Y, Acimovic L, Milicic B, Milisavljevic S, Nikolic N, Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (2001) Hyperfractionated radiation therapy and concurrent low-dose, daily carboplatin/etoposide with or without weekend carboplatin/etoposide chemotherapy in stage III non-small-cell lung cancer: a randomized trial. Int J Radiat Oncol Biol Phys 50(1):19–25CrossRefPubMed
42.
Zurück zum Zitat Kohsaka S, Wang L, Yachi K, Mahabir R, Narita T, Itoh T, Tanino M, Kimura T, Nishihara H, Tanaka S (2012) STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol Cancer Ther 11(6):1289CrossRefPubMed Kohsaka S, Wang L, Yachi K, Mahabir R, Narita T, Itoh T, Tanino M, Kimura T, Nishihara H, Tanaka S (2012) STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol Cancer Ther 11(6):1289CrossRefPubMed
43.
Zurück zum Zitat Zhang C, Nance EA, Mastorakos P, Chisholm J, Berry S, Eberhart C, Tyler B, Brem H, Suk JS, Hanes J (2017) Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats. J Control Release 263:112–119CrossRefPubMedPubMedCentral Zhang C, Nance EA, Mastorakos P, Chisholm J, Berry S, Eberhart C, Tyler B, Brem H, Suk JS, Hanes J (2017) Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats. J Control Release 263:112–119CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Arshad A, Yang B, Bienemann AS, Barua NU, Wyatt MJ, Woolley M, Johnson DE, Edler KJ, Gill SS (2015) Convection-enhanced delivery of carboplatin PLGA nanoparticles for the treatment of glioblastoma. PLoS ONE 10(7):e0132266CrossRefPubMedPubMedCentral Arshad A, Yang B, Bienemann AS, Barua NU, Wyatt MJ, Woolley M, Johnson DE, Edler KJ, Gill SS (2015) Convection-enhanced delivery of carboplatin PLGA nanoparticles for the treatment of glioblastoma. PLoS ONE 10(7):e0132266CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Shi M, Fortin D, Sanche L, Paquette B (2015) Convection-enhancement delivery of platinum-based drugs and Lipoplatin(TM) to optimize the concomitant effect with radiotherapy in F98 glioma rat model. Invest New Drugs 33(3):555–563CrossRefPubMedPubMedCentral Shi M, Fortin D, Sanche L, Paquette B (2015) Convection-enhancement delivery of platinum-based drugs and Lipoplatin(TM) to optimize the concomitant effect with radiotherapy in F98 glioma rat model. Invest New Drugs 33(3):555–563CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Shi M, Fortin D, Paquette B, Sanche L (2016) Convection-enhancement delivery of liposomal formulation of oxaliplatin shows less toxicity than oxaliplatin yet maintains a similar median survival time in F98 glioma-bearing rat model. Invest New Drugs 34(3):269–276CrossRefPubMedPubMedCentral Shi M, Fortin D, Paquette B, Sanche L (2016) Convection-enhancement delivery of liposomal formulation of oxaliplatin shows less toxicity than oxaliplatin yet maintains a similar median survival time in F98 glioma-bearing rat model. Invest New Drugs 34(3):269–276CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Maes W, Van Gool SW (2011) Experimental immunotherapy for malignant glioma: lessons from two decades of research in the GL261 model. Cancer Immunol Immunother 60(2):153–160CrossRefPubMed Maes W, Van Gool SW (2011) Experimental immunotherapy for malignant glioma: lessons from two decades of research in the GL261 model. Cancer Immunol Immunother 60(2):153–160CrossRefPubMed
48.
Zurück zum Zitat Chen X, Zhang L, Zhang IY, Liang J, Wang H, Ouyang M, Wu S, da Fonseca ACC, Weng L, Yamamoto Y, Yamamoto H, Natarajan R, Badie B (2014) RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res 74(24):7285–7297CrossRefPubMedPubMedCentral Chen X, Zhang L, Zhang IY, Liang J, Wang H, Ouyang M, Wu S, da Fonseca ACC, Weng L, Yamamoto Y, Yamamoto H, Natarajan R, Badie B (2014) RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res 74(24):7285–7297CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Reilly KM, Loisel DA, Bronson RT, McLaughlin ME, Jacks T (2000) Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat Genet 26(1):109–113CrossRefPubMed Reilly KM, Loisel DA, Bronson RT, McLaughlin ME, Jacks T (2000) Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat Genet 26(1):109–113CrossRefPubMed
50.
Zurück zum Zitat Rodrigues JC, Gonzalez GC, Zhang L, Ibrahim G, Kelly JJ, Gustafson MP, Lin Y, Dietz AB, Forsyth PA, Yong VW, Parney IF (2010) Normal human monocytes exposed to glioma cells acquire myeloid-derived suppressor cell-like properties. Neuro Oncol 12(4):351–365CrossRefPubMed Rodrigues JC, Gonzalez GC, Zhang L, Ibrahim G, Kelly JJ, Gustafson MP, Lin Y, Dietz AB, Forsyth PA, Yong VW, Parney IF (2010) Normal human monocytes exposed to glioma cells acquire myeloid-derived suppressor cell-like properties. Neuro Oncol 12(4):351–365CrossRefPubMed
51.
Zurück zum Zitat Bergamin LS, Braganhol E, Figueiro F, Casali EA, Zanin RF, Sevigny J, Battastini AM (2015) Involvement of purinergic system in the release of cytokines by macrophages exposed to glioma-conditioned medium. J Cell Biochem 116(5):721–729CrossRefPubMed Bergamin LS, Braganhol E, Figueiro F, Casali EA, Zanin RF, Sevigny J, Battastini AM (2015) Involvement of purinergic system in the release of cytokines by macrophages exposed to glioma-conditioned medium. J Cell Biochem 116(5):721–729CrossRefPubMed
52.
Zurück zum Zitat Hambardzumyan D, Gutmann DH, Kettenmann H (2016) The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci 19(1):20–27CrossRefPubMedPubMedCentral Hambardzumyan D, Gutmann DH, Kettenmann H (2016) The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci 19(1):20–27CrossRefPubMedPubMedCentral
Metadaten
Titel
Oxaliplatin disrupts pathological features of glioma cells and associated macrophages independent of apoptosis induction
verfasst von
Nathan B. Roberts
Aymen Alqazzaz
Jacqueline R. Hwang
Xiulan Qi
Achsah D. Keegan
Anthony J. Kim
Jeffrey A. Winkles
Graeme F. Woodworth
Publikationsdatum
21.08.2018
Verlag
Springer US
Erschienen in
Journal of Neuro-Oncology / Ausgabe 3/2018
Print ISSN: 0167-594X
Elektronische ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-018-2979-1

Weitere Artikel der Ausgabe 3/2018

Journal of Neuro-Oncology 3/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.