Skip to main content
Top
Published in: Cellular Oncology 4/2019

01-08-2019 | Glioblastoma | Review

The role of caveolin-1 in tumors of the brain - functional and clinical implications

Authors: Pinar Eser Ocak, Umut Ocak, Jiping Tang, John H. Zhang

Published in: Cellular Oncology | Issue 4/2019

Login to get access

Abstract

Background

Caveolin-1 (cav-1) is the major structural protein of caveolae, the flask-shaped invaginations of the plasma membrane mainly involved in cell signaling. Today, cav-1 is believed to play a role in a variety of disease processes including cancer, owing to the variations of its expression in association with tumor progression, invasive behavior, metastasis and therapy resistance. Since first detected in the brain, a number of studies has particularly focused on the role of cav-1 in the various steps of brain tumorigenesis. In this review, we discuss the different roles of cav-1 and its contributions to the molecular mechanisms underlying the pathobiology and natural behavior of brain tumors including glial, non-glial and metastatic subtypes. These contributions could be attributed to its co-localization with important players in tumorigenesis within the lipid-enriched domains of the plasma membrane. In that regard, the ability of cav-1 to interact with various cell signaling molecules as well as the impact of caveolae depletion on important pathways acting in brain tumor pathogenesis are noteworthy. We also discuss conversant causes hampering the treatment of malignant glial tumors such as limited transport of chemotherapeutics across the blood tumor barrier and resistance to chemoradiotherapy, by focusing on the molecular fundamentals involving cav-1 participation.

Conclusions

Cav-1 has the potential to pivot the molecular basis underlying the pathobiology of brain tumors, particularly the malignant glial subtype. In addition, the regulatory effect of cav-1-dependent and caveola-mediated transcellular transport on the permeability of the blood tumor barrier could be of benefit to overcome the restricted transport across brain barriers when applying chemotherapeutics. The association of cav-1 with tumors of the brain other than malignant gliomas deserves to be underlined, as well given the evidence suggesting its potential in predicting tumor grade and recurrence rates together with determining patient prognosis in oligodendrogliomas, ependymomas, meningiomas, vestibular schwannomas and brain metastases.
Literature
1.
go back to reference A.W. Cohen, R. Hnasko, W. Schubert, M.P. Lisanti, Role of caveolae and caveolins in health and disease. Physiol Rev 84, 1341–1379 (2004)CrossRefPubMed A.W. Cohen, R. Hnasko, W. Schubert, M.P. Lisanti, Role of caveolae and caveolins in health and disease. Physiol Rev 84, 1341–1379 (2004)CrossRefPubMed
2.
go back to reference F. Galbiati, D. Volonte, J. Liu, F. Capozza, P.G. Frank, L. Zhu, R.G. Pestell, M.P. Lisanti, Caveolin-1 expression negatively regulates cell cycle progression by inducing G(0)/G(1) arrest via a p53/p21(WAF1/Cip1)-dependent mechanism. Mol Biol Cell 12, 2229–2244 (2001)CrossRefPubMedPubMedCentral F. Galbiati, D. Volonte, J. Liu, F. Capozza, P.G. Frank, L. Zhu, R.G. Pestell, M.P. Lisanti, Caveolin-1 expression negatively regulates cell cycle progression by inducing G(0)/G(1) arrest via a p53/p21(WAF1/Cip1)-dependent mechanism. Mol Biol Cell 12, 2229–2244 (2001)CrossRefPubMedPubMedCentral
3.
go back to reference V.A. Torres, J.C. Tapia, D.A. Rodriguez, M. Parraga, P. Lisboa, M. Montoya, L. Leyton, A.F. Quest, Caveolin-1 controls cell proliferation and cell death by suppressing expression of the inhibitor of apoptosis protein survivin. J Cell Sci 119, 1812–1823 (2006)CrossRefPubMed V.A. Torres, J.C. Tapia, D.A. Rodriguez, M. Parraga, P. Lisboa, M. Montoya, L. Leyton, A.F. Quest, Caveolin-1 controls cell proliferation and cell death by suppressing expression of the inhibitor of apoptosis protein survivin. J Cell Sci 119, 1812–1823 (2006)CrossRefPubMed
4.
go back to reference K.S. Song, P.E. Scherer, Z. Tang, T. Okamoto, S. Li, M. Chafel, C. Chu, D.S. Kohtz, M.P. Lisanti, Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J Biol Chem 271, 15160–15165 (1996)CrossRefPubMed K.S. Song, P.E. Scherer, Z. Tang, T. Okamoto, S. Li, M. Chafel, C. Chu, D.S. Kohtz, M.P. Lisanti, Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J Biol Chem 271, 15160–15165 (1996)CrossRefPubMed
5.
go back to reference M.M. Hill, M. Bastiani, R. Luetterforst, M. Kirkham, A. Kirkham, S.J. Nixon, P. Walser, D. Abankwa, V.M. Oorschot, S. Martin, J.F. Hancock, R.G. Parton, PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function. Cell 132, 113–124 (2008)CrossRefPubMedPubMedCentral M.M. Hill, M. Bastiani, R. Luetterforst, M. Kirkham, A. Kirkham, S.J. Nixon, P. Walser, D. Abankwa, V.M. Oorschot, S. Martin, J.F. Hancock, R.G. Parton, PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function. Cell 132, 113–124 (2008)CrossRefPubMedPubMedCentral
6.
go back to reference A.J. Koleske, D. Baltimore, M.P. Lisanti, Reduction of caveolin and caveolae in oncogenically transformed cells. Proc Natl Acad Sci U S A 92, 1381–1385 (1995)CrossRefPubMedPubMedCentral A.J. Koleske, D. Baltimore, M.P. Lisanti, Reduction of caveolin and caveolae in oncogenically transformed cells. Proc Natl Acad Sci U S A 92, 1381–1385 (1995)CrossRefPubMedPubMedCentral
7.
go back to reference S.W. Lee, C.L. Reimer, P. Oh, D.B. Campbell, J.E. Schnitzer, Tumor cell growth inhibition by caveolin re-expression in human breast cancer cells. Oncogene 16, 1391–1397 (1998)CrossRefPubMed S.W. Lee, C.L. Reimer, P. Oh, D.B. Campbell, J.E. Schnitzer, Tumor cell growth inhibition by caveolin re-expression in human breast cancer cells. Oncogene 16, 1391–1397 (1998)CrossRefPubMed
8.
go back to reference H. Lee, D.S. Park, B. Razani, R.G. Russell, R.G. Pestell, M.P. Lisanti, Caveolin-1 mutations (P132L and null) and the pathogenesis of breast cancer: Caveolin-1 (P132L) behaves in a dominant-negative manner and caveolin-1 (−/−) null mice show mammary epithelial cell hyperplasia. Am J Pathol 161, 1357–1369 (2002)CrossRefPubMedPubMedCentral H. Lee, D.S. Park, B. Razani, R.G. Russell, R.G. Pestell, M.P. Lisanti, Caveolin-1 mutations (P132L and null) and the pathogenesis of breast cancer: Caveolin-1 (P132L) behaves in a dominant-negative manner and caveolin-1 (−/−) null mice show mammary epithelial cell hyperplasia. Am J Pathol 161, 1357–1369 (2002)CrossRefPubMedPubMedCentral
9.
go back to reference Z.D. Nassar, M.M. Hill, R.G. Parton, M.O. Parat, Caveola-forming proteins caveolin-1 and PTRF in prostate cancer. Nat Rev Urol 10, 529–536 (2013)CrossRefPubMed Z.D. Nassar, M.M. Hill, R.G. Parton, M.O. Parat, Caveola-forming proteins caveolin-1 and PTRF in prostate cancer. Nat Rev Urol 10, 529–536 (2013)CrossRefPubMed
10.
go back to reference C. Schoch, T. Haferlach, S. Bursch, D. Gerstner, S. Schnittger, M. Dugas, W. Kern, H. Loffler, W. Hiddemann, Loss of genetic material is more common than gain in acute myeloid leukemia with complex aberrant karyotype: A detailed analysis of 125 cases using conventional chromosome analysis and fluorescence in situ hybridization including 24-color FISH. Genes Chromosomes Cancer 35, 20–29 (2002)CrossRefPubMed C. Schoch, T. Haferlach, S. Bursch, D. Gerstner, S. Schnittger, M. Dugas, W. Kern, H. Loffler, W. Hiddemann, Loss of genetic material is more common than gain in acute myeloid leukemia with complex aberrant karyotype: A detailed analysis of 125 cases using conventional chromosome analysis and fluorescence in situ hybridization including 24-color FISH. Genes Chromosomes Cancer 35, 20–29 (2002)CrossRefPubMed
11.
go back to reference L. Cheng, G.T. MacLennan, S. Zhang, M. Wang, M. Zhou, P.H. Tan, S. Foster, A. Lopez-Beltran, R. Montironi, Evidence for polyclonal origin of multifocal clear cell renal cell carcinoma. Clin Cancer Res 14, 8087–8093 (2008)CrossRefPubMed L. Cheng, G.T. MacLennan, S. Zhang, M. Wang, M. Zhou, P.H. Tan, S. Foster, A. Lopez-Beltran, R. Montironi, Evidence for polyclonal origin of multifocal clear cell renal cell carcinoma. Clin Cancer Res 14, 8087–8093 (2008)CrossRefPubMed
12.
go back to reference J. Xiong, D. Wang, A. Wei, H. Lu, C. Tan, A. Li, J. Tang, Y. Wang, S. He, X. Liu, W. Hu, Deregulated expression of miR-107 inhibits metastasis of PDAC through inhibition PI3K/Akt signaling via caveolin-1 and PTEN. Exp Cell Res 361, 316–323 (2017)CrossRefPubMed J. Xiong, D. Wang, A. Wei, H. Lu, C. Tan, A. Li, J. Tang, Y. Wang, S. He, X. Liu, W. Hu, Deregulated expression of miR-107 inhibits metastasis of PDAC through inhibition PI3K/Akt signaling via caveolin-1 and PTEN. Exp Cell Res 361, 316–323 (2017)CrossRefPubMed
13.
go back to reference J. Jankovic, S. Tatic, V. Bozic, V. Zivaljevic, D. Cvejic, S. Paskas, Inverse expression of caveolin-1 and EGFR in thyroid cancer patients. Hum Pathol 61, 164–172 (2017)CrossRefPubMed J. Jankovic, S. Tatic, V. Bozic, V. Zivaljevic, D. Cvejic, S. Paskas, Inverse expression of caveolin-1 and EGFR in thyroid cancer patients. Hum Pathol 61, 164–172 (2017)CrossRefPubMed
14.
go back to reference B.K. Ryu, M.G. Lee, N.H. Kim, K.Y. Lee, S.J. Oh, J.R. Moon, H.J. Kim, S.G. Chi, Bidirectional alteration of Cav-1 expression is associated with mitogenic conversion of its function in gastric tumor progression. BMC Cancer 17(766) (2017) B.K. Ryu, M.G. Lee, N.H. Kim, K.Y. Lee, S.J. Oh, J.R. Moon, H.J. Kim, S.G. Chi, Bidirectional alteration of Cav-1 expression is associated with mitogenic conversion of its function in gastric tumor progression. BMC Cancer 17(766) (2017)
15.
go back to reference C. Aguirre-Portoles, J. Feliu, G. Reglero, A. Ramirez de Molina, ABCA1 overexpression worsens colorectal cancer prognosis by facilitating tumour growth and caveolin-1-dependent invasiveness, and these effects can be ameliorated using the BET inhibitor apabetalone. Mol Oncol 12, 1735-1752 (2018) C. Aguirre-Portoles, J. Feliu, G. Reglero, A. Ramirez de Molina, ABCA1 overexpression worsens colorectal cancer prognosis by facilitating tumour growth and caveolin-1-dependent invasiveness, and these effects can be ameliorated using the BET inhibitor apabetalone. Mol Oncol 12, 1735-1752 (2018)
16.
go back to reference Y.N. Liang, Y. Liu, L. Wang, G. Yao, X. Li, X. Meng, F. Wang, M. Li, D. Tong, J. Geng, Combined caveolin-1 and epidermal growth factor receptor expression as a prognostic marker for breast cancer. Oncol Lett 15, 9271–9282 (2018)PubMedPubMedCentral Y.N. Liang, Y. Liu, L. Wang, G. Yao, X. Li, X. Meng, F. Wang, M. Li, D. Tong, J. Geng, Combined caveolin-1 and epidermal growth factor receptor expression as a prognostic marker for breast cancer. Oncol Lett 15, 9271–9282 (2018)PubMedPubMedCentral
17.
go back to reference L. Bazzani, S. Donnini, A. Giachetti, G. Christofori, M. Ziche, PGE2 mediates EGFR internalization and nuclear translocation via caveolin endocytosis promoting its transcriptional activity and proliferation in human NSCLC cells. Oncotarget 9, 14939–14958 (2018)CrossRefPubMedPubMedCentral L. Bazzani, S. Donnini, A. Giachetti, G. Christofori, M. Ziche, PGE2 mediates EGFR internalization and nuclear translocation via caveolin endocytosis promoting its transcriptional activity and proliferation in human NSCLC cells. Oncotarget 9, 14939–14958 (2018)CrossRefPubMedPubMedCentral
18.
go back to reference P.L. Cameron, J.W. Ruffin, R. Bollag, H. Rasmussen, R.S. Cameron, Identification of caveolin and caveolin-related proteins in the brain. J Neurosci 17, 9520–9535 (1997)CrossRefPubMedPubMedCentral P.L. Cameron, J.W. Ruffin, R. Bollag, H. Rasmussen, R.S. Cameron, Identification of caveolin and caveolin-related proteins in the brain. J Neurosci 17, 9520–9535 (1997)CrossRefPubMedPubMedCentral
19.
go back to reference F. Galbiati, D. Volonte, O. Gil, G. Zanazzi, J.L. Salzer, M. Sargiacomo, P.E. Scherer, J.A. Engelman, A. Schlegel, M. Parenti, T. Okamoto, M.P. Lisanti, Expression of caveolin-1 and -2 in differentiating PC12 cells and dorsal root ganglion neurons: Caveolin-2 is up-regulated in response to cell injury. Proc Natl Acad Sci U S A 95, 10257–10262 (1998)CrossRefPubMedPubMedCentral F. Galbiati, D. Volonte, O. Gil, G. Zanazzi, J.L. Salzer, M. Sargiacomo, P.E. Scherer, J.A. Engelman, A. Schlegel, M. Parenti, T. Okamoto, M.P. Lisanti, Expression of caveolin-1 and -2 in differentiating PC12 cells and dorsal root ganglion neurons: Caveolin-2 is up-regulated in response to cell injury. Proc Natl Acad Sci U S A 95, 10257–10262 (1998)CrossRefPubMedPubMedCentral
20.
go back to reference T. Ikezu, H. Ueda, B.D. Trapp, K. Nishiyama, J.F. Sha, D. Volonte, F. Galbiati, A.L. Byrd, G. Bassell, H. Serizawa, W.S. Lane, M.P. Lisanti, T. Okamoto, Affinity-purification and characterization of caveolins from the brain: Differential expression of caveolin-1, −2, and −3 in brain endothelial and astroglial cell types. Brain Res 804, 177–192 (1998)CrossRefPubMed T. Ikezu, H. Ueda, B.D. Trapp, K. Nishiyama, J.F. Sha, D. Volonte, F. Galbiati, A.L. Byrd, G. Bassell, H. Serizawa, W.S. Lane, M.P. Lisanti, T. Okamoto, Affinity-purification and characterization of caveolins from the brain: Differential expression of caveolin-1, −2, and −3 in brain endothelial and astroglial cell types. Brain Res 804, 177–192 (1998)CrossRefPubMed
21.
go back to reference P.L. Cameron, C. Liu, D.K. Smart, S.T. Hantus, J.R. Fick, R.S. Cameron, Caveolin-1 expression is maintained in rat and human astroglioma cell lines. Glia 37, 275–290 (2002)CrossRefPubMed P.L. Cameron, C. Liu, D.K. Smart, S.T. Hantus, J.R. Fick, R.S. Cameron, Caveolin-1 expression is maintained in rat and human astroglioma cell lines. Glia 37, 275–290 (2002)CrossRefPubMed
22.
go back to reference C.D. Stiles, Cancer of the central nervous system. Review of an AACR special conference in cancer research with the joint section on tumors of the AANS/CNS (San Diego, CA, June 7-11, 1997). Biochim Biophys Acta 1377, R1–10 (1998)PubMed C.D. Stiles, Cancer of the central nervous system. Review of an AACR special conference in cancer research with the joint section on tumors of the AANS/CNS (San Diego, CA, June 7-11, 1997). Biochim Biophys Acta 1377, R1–10 (1998)PubMed
25.
go back to reference R.S. Faccion, P.S. Bernardo, G.P.F. de Lopes, L.S. Bastos, C.L. Teixeira, J.A. de Oliveira, P.V. Fernandes, L.G. Dubois, L. Chimelli, R.C. Maia, p53 expression and subcellular survivin localization improve the diagnosis and prognosis of patients with diffuse astrocytic tumors. Cell Oncol 41, 141–157 (2018)CrossRef R.S. Faccion, P.S. Bernardo, G.P.F. de Lopes, L.S. Bastos, C.L. Teixeira, J.A. de Oliveira, P.V. Fernandes, L.G. Dubois, L. Chimelli, R.C. Maia, p53 expression and subcellular survivin localization improve the diagnosis and prognosis of patients with diffuse astrocytic tumors. Cell Oncol 41, 141–157 (2018)CrossRef
26.
go back to reference M.O. Taskapilioglu, U. Aktas, P. Eser, S. Tolunay, A. Bekar, Multiple extracranial metastases from secondary glioblastoma: A case report and review of the literature. Turk Neurosurg 23, 824-827 (2013) M.O. Taskapilioglu, U. Aktas, P. Eser, S. Tolunay, A. Bekar, Multiple extracranial metastases from secondary glioblastoma: A case report and review of the literature. Turk Neurosurg 23, 824-827 (2013)
27.
go back to reference R. Stupp, M.E. Hegi, W.P. Mason, M.J. van den Bent, M.J. Taphoorn, R.C. Janzer, S.K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A.A. Brandes, J. Gijtenbeek, C. Marosi, C.J. Vecht, K. Mokhtari, P. Wesseling, S. Villa, E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J.G. Cairncross, R.O. Mirimanoff, R. European organisation for, T. treatment of Cancer brain, G. radiation oncology and G. National Cancer Institute of Canada clinical trials, effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10, 459–466 (2009)CrossRefPubMed R. Stupp, M.E. Hegi, W.P. Mason, M.J. van den Bent, M.J. Taphoorn, R.C. Janzer, S.K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A.A. Brandes, J. Gijtenbeek, C. Marosi, C.J. Vecht, K. Mokhtari, P. Wesseling, S. Villa, E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J.G. Cairncross, R.O. Mirimanoff, R. European organisation for, T. treatment of Cancer brain, G. radiation oncology and G. National Cancer Institute of Canada clinical trials, effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10, 459–466 (2009)CrossRefPubMed
28.
go back to reference R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O. Mirimanoff, R. European organisation for, T. treatment of Cancer brain, G. radiotherapy and G. National Cancer Institute of Canada clinical trials, radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996 (2005)CrossRefPubMed R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O. Mirimanoff, R. European organisation for, T. treatment of Cancer brain, G. radiotherapy and G. National Cancer Institute of Canada clinical trials, radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996 (2005)CrossRefPubMed
29.
go back to reference M.A. Forget, R.R. Desrosiers, M. Del, R. Moumdjian, D. Shedid, F. Berthelet, R. Beliveau, The expression of rho proteins decreases with human brain tumor progression: Potential tumor markers. Clin Exp Metastasis 19, 9–15 (2002)CrossRefPubMed M.A. Forget, R.R. Desrosiers, M. Del, R. Moumdjian, D. Shedid, F. Berthelet, R. Beliveau, The expression of rho proteins decreases with human brain tumor progression: Potential tumor markers. Clin Exp Metastasis 19, 9–15 (2002)CrossRefPubMed
30.
go back to reference A. Abulrob, S. Giuseppin, M.F. Andrade, A. McDermid, M. Moreno, D. Stanimirovic, Interactions of EGFR and caveolin-1 in human glioblastoma cells: Evidence that tyrosine phosphorylation regulates EGFR association with caveolae. Oncogene 23, 6967–6979 (2004)CrossRefPubMed A. Abulrob, S. Giuseppin, M.F. Andrade, A. McDermid, M. Moreno, D. Stanimirovic, Interactions of EGFR and caveolin-1 in human glioblastoma cells: Evidence that tyrosine phosphorylation regulates EGFR association with caveolae. Oncogene 23, 6967–6979 (2004)CrossRefPubMed
31.
go back to reference V. Barresi, F.R. Buttarelli, E.E. Vitarelli, A. Arcella, M. Antonelli, F. Giangaspero, Caveolin-1 expression in diffuse gliomas: Correlation with the proliferation index, epidermal growth factor receptor, p53, and 1p/19q status. Hum Pathol 40, 1738–1746 (2009)CrossRefPubMed V. Barresi, F.R. Buttarelli, E.E. Vitarelli, A. Arcella, M. Antonelli, F. Giangaspero, Caveolin-1 expression in diffuse gliomas: Correlation with the proliferation index, epidermal growth factor receptor, p53, and 1p/19q status. Hum Pathol 40, 1738–1746 (2009)CrossRefPubMed
32.
go back to reference P. Kucharzewska, H.C. Christianson, J.E. Welch, K.J. Svensson, E. Fredlund, M. Ringner, M. Morgelin, E. Bourseau-Guilmain, J. Bengzon, M. Belting, Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A 110, 7312–7317 (2013)CrossRefPubMedPubMedCentral P. Kucharzewska, H.C. Christianson, J.E. Welch, K.J. Svensson, E. Fredlund, M. Ringner, M. Morgelin, E. Bourseau-Guilmain, J. Bengzon, M. Belting, Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A 110, 7312–7317 (2013)CrossRefPubMedPubMedCentral
33.
go back to reference S. Shimato, L.M. Anderson, M. Asslaber, J.N. Bruce, P. Canoll, D.E. Anderson, R.C. Anderson, Inhibition of caveolin-1 restores myeloid cell function in human glioblastoma. PLoS One 8, e77397 (2013)CrossRefPubMedPubMedCentral S. Shimato, L.M. Anderson, M. Asslaber, J.N. Bruce, P. Canoll, D.E. Anderson, R.C. Anderson, Inhibition of caveolin-1 restores myeloid cell function in human glioblastoma. PLoS One 8, e77397 (2013)CrossRefPubMedPubMedCentral
34.
go back to reference P. Cassoni, R. Senetta, I. Castellano, E. Ortolan, M. Bosco, I. Magnani, A. Ducati, Caveolin-1 expression is variably displayed in astroglial-derived tumors and absent in oligodendrogliomas: Concrete premises for a new reliable diagnostic marker in gliomas. Am J Surg Pathol 31, 760–769 (2007)CrossRefPubMed P. Cassoni, R. Senetta, I. Castellano, E. Ortolan, M. Bosco, I. Magnani, A. Ducati, Caveolin-1 expression is variably displayed in astroglial-derived tumors and absent in oligodendrogliomas: Concrete premises for a new reliable diagnostic marker in gliomas. Am J Surg Pathol 31, 760–769 (2007)CrossRefPubMed
35.
go back to reference R. Senetta, E. Trevisan, R. Ruda, E. Maldi, L. Molinaro, F. Lefranc, L. Chiusa, M. Lanotte, R. Soffietti, P. Cassoni, Caveolin 1 expression independently predicts shorter survival in oligodendrogliomas. J Neuropathol Exp Neurol 68, 425–431 (2009)CrossRefPubMed R. Senetta, E. Trevisan, R. Ruda, E. Maldi, L. Molinaro, F. Lefranc, L. Chiusa, M. Lanotte, R. Soffietti, P. Cassoni, Caveolin 1 expression independently predicts shorter survival in oligodendrogliomas. J Neuropathol Exp Neurol 68, 425–431 (2009)CrossRefPubMed
36.
go back to reference R. Senetta, C. Miracco, S. Lanzafame, L. Chiusa, R. Caltabiano, A. Galia, G. Stella, P. Cassoni, Epidermal growth factor receptor and caveolin-1 coexpression identifies adult supratentorial ependymomas with rapid unfavorable outcomes. Neuro-Oncology 13, 176–183 (2011)CrossRefPubMed R. Senetta, C. Miracco, S. Lanzafame, L. Chiusa, R. Caltabiano, A. Galia, G. Stella, P. Cassoni, Epidermal growth factor receptor and caveolin-1 coexpression identifies adult supratentorial ependymomas with rapid unfavorable outcomes. Neuro-Oncology 13, 176–183 (2011)CrossRefPubMed
37.
go back to reference E.C. Cosset, J. Godet, N. Entz-Werle, E. Guerin, D. Guenot, S. Froelich, D. Bonnet, S. Pinel, F. Plenat, P. Chastagner, M. Dontenwill, S. Martin, Involvement of the TGFbeta pathway in the regulation of alpha5 beta1 integrins by caveolin-1 in human glioblastoma. Int J Cancer 131, 601–611 (2012)CrossRefPubMed E.C. Cosset, J. Godet, N. Entz-Werle, E. Guerin, D. Guenot, S. Froelich, D. Bonnet, S. Pinel, F. Plenat, P. Chastagner, M. Dontenwill, S. Martin, Involvement of the TGFbeta pathway in the regulation of alpha5 beta1 integrins by caveolin-1 in human glioblastoma. Int J Cancer 131, 601–611 (2012)CrossRefPubMed
38.
go back to reference C. Chen, L. Chen, Y. Yao, Z. Qin, H. Chen, Nucleolin overexpression is associated with an unfavorable outcome for ependymoma: A multifactorial analysis of 176 patients. J Neuro-Oncol 127, 43–52 (2016)CrossRef C. Chen, L. Chen, Y. Yao, Z. Qin, H. Chen, Nucleolin overexpression is associated with an unfavorable outcome for ependymoma: A multifactorial analysis of 176 patients. J Neuro-Oncol 127, 43–52 (2016)CrossRef
39.
go back to reference P. Duffy, A. Schmandke, A. Schmandke, J. Sigworth, S. Narumiya, W.B. Cafferty, S.M. Strittmatter, Rho-associated kinase II (ROCKII) limits axonal growth after trauma within the adult mouse spinal cord. J Neurosci 29, 15266–15276 (2009)CrossRefPubMedPubMedCentral P. Duffy, A. Schmandke, A. Schmandke, J. Sigworth, S. Narumiya, W.B. Cafferty, S.M. Strittmatter, Rho-associated kinase II (ROCKII) limits axonal growth after trauma within the adult mouse spinal cord. J Neurosci 29, 15266–15276 (2009)CrossRefPubMedPubMedCentral
40.
go back to reference R. Shinohara, D. Thumkeo, H. Kamijo, N. Kaneko, K. Sawamoto, K. Watanabe, H. Takebayashi, H. Kiyonari, T. Ishizaki, T. Furuyashiki, S. Narumiya, A role for mDia, a rho-regulated actin nucleator, in tangential migration of interneuron precursors. Nat Neurosci 15, 373–380, S371–372 (2012)CrossRefPubMed R. Shinohara, D. Thumkeo, H. Kamijo, N. Kaneko, K. Sawamoto, K. Watanabe, H. Takebayashi, H. Kiyonari, T. Ishizaki, T. Furuyashiki, S. Narumiya, A role for mDia, a rho-regulated actin nucleator, in tangential migration of interneuron precursors. Nat Neurosci 15, 373–380, S371–372 (2012)CrossRefPubMed
41.
go back to reference A. Tashiro, A. Minden, R. Yuste, Regulation of dendritic spine morphology by the rho family of small GTPases: Antagonistic roles of Rac and rho. Cereb Cortex 10, 927–938 (2000)CrossRefPubMed A. Tashiro, A. Minden, R. Yuste, Regulation of dendritic spine morphology by the rho family of small GTPases: Antagonistic roles of Rac and rho. Cereb Cortex 10, 927–938 (2000)CrossRefPubMed
42.
go back to reference C.D. Nobes, A. Hall, Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell 81, 53–62 (1995)CrossRefPubMed C.D. Nobes, A. Hall, Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell 81, 53–62 (1995)CrossRefPubMed
43.
go back to reference G. Fritz, I. Just, B. Kaina, Rho GTPases are over-expressed in human tumors. Int J Cancer 81, 682–687 (1999)CrossRefPubMed G. Fritz, I. Just, B. Kaina, Rho GTPases are over-expressed in human tumors. Int J Cancer 81, 682–687 (1999)CrossRefPubMed
44.
go back to reference D. Gingras, F. Gauthier, S. Lamy, R.R. Desrosiers, R. Beliveau, Localization of RhoA GTPase to endothelial caveolae-enriched membrane domains. Biochem Biophys Res Commun 247, 888–893 (1998)CrossRefPubMed D. Gingras, F. Gauthier, S. Lamy, R.R. Desrosiers, R. Beliveau, Localization of RhoA GTPase to endothelial caveolae-enriched membrane domains. Biochem Biophys Res Commun 247, 888–893 (1998)CrossRefPubMed
45.
go back to reference G.J. Pilkington, The paradox of neoplastic glial cell invasion of the brain and apparent metastatic failure. Anticancer Res 17, 4103–4105 (1997)PubMed G.J. Pilkington, The paradox of neoplastic glial cell invasion of the brain and apparent metastatic failure. Anticancer Res 17, 4103–4105 (1997)PubMed
46.
go back to reference N.L. Tran, W.S. McDonough, B.A. Savitch, S.P. Fortin, J.A. Winkles, M. Symons, M. Nakada, H.E. Cunliffe, G. Hostetter, D.B. Hoelzinger, J.L. Rennert, J.S. Michaelson, L.C. Burkly, C.A. Lipinski, J.C. Loftus, L. Mariani, M.E. Berens, Increased fibroblast growth factor-inducible 14 expression levels promote glioma cell invasion via Rac1 and nuclear factor-kappaB and correlate with poor patient outcome. Cancer Res 66, 9535–9542 (2006)CrossRefPubMed N.L. Tran, W.S. McDonough, B.A. Savitch, S.P. Fortin, J.A. Winkles, M. Symons, M. Nakada, H.E. Cunliffe, G. Hostetter, D.B. Hoelzinger, J.L. Rennert, J.S. Michaelson, L.C. Burkly, C.A. Lipinski, J.C. Loftus, L. Mariani, M.E. Berens, Increased fibroblast growth factor-inducible 14 expression levels promote glioma cell invasion via Rac1 and nuclear factor-kappaB and correlate with poor patient outcome. Cancer Res 66, 9535–9542 (2006)CrossRefPubMed
47.
go back to reference S.P. Fortin Ensign, I.T. Mathews, M.H. Symons, M.E. Berens, N.L. Tran, Implications of rho GTPase signaling in Glioma cell invasion and tumor progression. Front Oncol 3(241) (2013) S.P. Fortin Ensign, I.T. Mathews, M.H. Symons, M.E. Berens, N.L. Tran, Implications of rho GTPase signaling in Glioma cell invasion and tumor progression. Front Oncol 3(241) (2013)
48.
go back to reference S. Dauth, T. Grevesse, H. Pantazopoulos, P.H. Campbell, B.M. Maoz, S. Berretta, K.K. Parker, Extracellular matrix protein expression is brain region dependent. J Comp Neurol 524, 1309–1336 (2016)CrossRefPubMedPubMedCentral S. Dauth, T. Grevesse, H. Pantazopoulos, P.H. Campbell, B.M. Maoz, S. Berretta, K.K. Parker, Extracellular matrix protein expression is brain region dependent. J Comp Neurol 524, 1309–1336 (2016)CrossRefPubMedPubMedCentral
49.
go back to reference U. Gunthert, C. Schwarzler, B. Wittig, J. Laman, P. Ruiz, R. Stauder, A. Bloem, F. Smadja-Joffe, M. Zoller, A. Rolink, Functional involvement of CD44, a family of cell adhesion molecules, in immune responses, tumour progression and haematopoiesis. Adv Exp Med Biol 451, 43–49 (1998)CrossRefPubMed U. Gunthert, C. Schwarzler, B. Wittig, J. Laman, P. Ruiz, R. Stauder, A. Bloem, F. Smadja-Joffe, M. Zoller, A. Rolink, Functional involvement of CD44, a family of cell adhesion molecules, in immune responses, tumour progression and haematopoiesis. Adv Exp Med Biol 451, 43–49 (1998)CrossRefPubMed
50.
go back to reference K.N. Sugahara, T. Murai, H. Nishinakamura, H. Kawashima, H. Saya, M. Miyasaka, Hyaluronan oligosaccharides induce CD44 cleavage and promote cell migration in CD44-expressing tumor cells. J Biol Chem 278, 32259–32265 (2003)CrossRefPubMed K.N. Sugahara, T. Murai, H. Nishinakamura, H. Kawashima, H. Saya, M. Miyasaka, Hyaluronan oligosaccharides induce CD44 cleavage and promote cell migration in CD44-expressing tumor cells. J Biol Chem 278, 32259–32265 (2003)CrossRefPubMed
51.
go back to reference Y. Akiyama, S. Jung, B. Salhia, S. Lee, S. Hubbard, M. Taylor, T. Mainprize, K. Akaishi, W. van Furth, J.T. Rutka, Hyaluronate receptors mediating glioma cell migration and proliferation. J Neuro-Oncol 53, 115–127 (2001)CrossRef Y. Akiyama, S. Jung, B. Salhia, S. Lee, S. Hubbard, M. Taylor, T. Mainprize, K. Akaishi, W. van Furth, J.T. Rutka, Hyaluronate receptors mediating glioma cell migration and proliferation. J Neuro-Oncol 53, 115–127 (2001)CrossRef
52.
go back to reference S.M. Ranuncolo, V. Ladeda, S. Specterman, M. Varela, J. Lastiri, A. Morandi, E. Matos, E. Bal de Kier Joffe, L. Puricelli, M.G. Pallotta, CD44 expression in human gliomas. J Surg Oncol 79, 30–35; discussion 35-36 (2002)CrossRefPubMed S.M. Ranuncolo, V. Ladeda, S. Specterman, M. Varela, J. Lastiri, A. Morandi, E. Matos, E. Bal de Kier Joffe, L. Puricelli, M.G. Pallotta, CD44 expression in human gliomas. J Surg Oncol 79, 30–35; discussion 35-36 (2002)CrossRefPubMed
53.
go back to reference S.K. Singh, I.D. Clarke, M. Terasaki, V.E. Bonn, C. Hawkins, J. Squire, P.B. Dirks, Identification of a cancer stem cell in human brain tumors. Cancer Res 63, 5821–5828 (2003)PubMed S.K. Singh, I.D. Clarke, M. Terasaki, V.E. Bonn, C. Hawkins, J. Squire, P.B. Dirks, Identification of a cancer stem cell in human brain tumors. Cancer Res 63, 5821–5828 (2003)PubMed
54.
55.
go back to reference E.M. Ahmed, G. Bandopadhyay, B. Coyle, A. Grabowska, A HIF-independent, CD133-mediated mechanism of cisplatin resistance in glioblastoma cells. Cell Oncol 41, 319–328 (2018)CrossRef E.M. Ahmed, G. Bandopadhyay, B. Coyle, A. Grabowska, A HIF-independent, CD133-mediated mechanism of cisplatin resistance in glioblastoma cells. Cell Oncol 41, 319–328 (2018)CrossRef
56.
go back to reference I. Okamoto, Y. Kawano, H. Tsuiki, J. Sasaki, M. Nakao, M. Matsumoto, M. Suga, M. Ando, M. Nakajima, H. Saya, CD44 cleavage induced by a membrane-associated metalloprotease plays a critical role in tumor cell migration. Oncogene 18, 1435–1446 (1999)CrossRefPubMed I. Okamoto, Y. Kawano, H. Tsuiki, J. Sasaki, M. Nakao, M. Matsumoto, M. Suga, M. Ando, M. Nakajima, H. Saya, CD44 cleavage induced by a membrane-associated metalloprotease plays a critical role in tumor cell migration. Oncogene 18, 1435–1446 (1999)CrossRefPubMed
57.
go back to reference M. Kajita, Y. Itoh, T. Chiba, H. Mori, A. Okada, H. Kinoh, M. Seiki, Membrane-type 1 matrix metalloproteinase cleaves CD44 and promotes cell migration. J Cell Biol 153, 893–904 (2001)CrossRefPubMedPubMedCentral M. Kajita, Y. Itoh, T. Chiba, H. Mori, A. Okada, H. Kinoh, M. Seiki, Membrane-type 1 matrix metalloproteinase cleaves CD44 and promotes cell migration. J Cell Biol 153, 893–904 (2001)CrossRefPubMedPubMedCentral
58.
go back to reference H. Mori, T. Tomari, N. Koshikawa, M. Kajita, Y. Itoh, H. Sato, H. Tojo, I. Yana, M. Seiki, CD44 directs membrane-type 1 matrix metalloproteinase to lamellipodia by associating with its hemopexin-like domain. EMBO J 21, 3949–3959 (2002)CrossRefPubMedPubMedCentral H. Mori, T. Tomari, N. Koshikawa, M. Kajita, Y. Itoh, H. Sato, H. Tojo, I. Yana, M. Seiki, CD44 directs membrane-type 1 matrix metalloproteinase to lamellipodia by associating with its hemopexin-like domain. EMBO J 21, 3949–3959 (2002)CrossRefPubMedPubMedCentral
59.
go back to reference B. Annabi, M. Lachambre, N. Bousquet-Gagnon, M. Page, D. Gingras, R. Beliveau, Localization of membrane-type 1 matrix metalloproteinase in caveolae membrane domains. Biochem J 353, 547–553 (2001)CrossRefPubMedPubMedCentral B. Annabi, M. Lachambre, N. Bousquet-Gagnon, M. Page, D. Gingras, R. Beliveau, Localization of membrane-type 1 matrix metalloproteinase in caveolae membrane domains. Biochem J 353, 547–553 (2001)CrossRefPubMedPubMedCentral
60.
go back to reference B. Annabi, S. Thibeault, R. Moumdjian, R. Beliveau, Hyaluronan cell surface binding is induced by type I collagen and regulated by caveolae in glioma cells. J Biol Chem 279, 21888–21896 (2004)CrossRefPubMed B. Annabi, S. Thibeault, R. Moumdjian, R. Beliveau, Hyaluronan cell surface binding is induced by type I collagen and regulated by caveolae in glioma cells. J Biol Chem 279, 21888–21896 (2004)CrossRefPubMed
61.
go back to reference B. Annabi, M. Bouzeghrane, R. Moumdjian, A. Moghrabi, R. Beliveau, Probing the infiltrating character of brain tumors: Inhibition of RhoA/ROK-mediated CD44 cell surface shedding from glioma cells by the green tea catechin EGCg. J Neurochem 94, 906–916 (2005)CrossRefPubMed B. Annabi, M. Bouzeghrane, R. Moumdjian, A. Moghrabi, R. Beliveau, Probing the infiltrating character of brain tumors: Inhibition of RhoA/ROK-mediated CD44 cell surface shedding from glioma cells by the green tea catechin EGCg. J Neurochem 94, 906–916 (2005)CrossRefPubMed
62.
go back to reference R. Nishikawa, X.D. Ji, R.C. Harmon, C.S. Lazar, G.N. Gill, W.K. Cavenee, H.J. Huang, A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci U S A 91, 7727–7731 (1994)CrossRefPubMedPubMedCentral R. Nishikawa, X.D. Ji, R.C. Harmon, C.S. Lazar, G.N. Gill, W.K. Cavenee, H.J. Huang, A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci U S A 91, 7727–7731 (1994)CrossRefPubMedPubMedCentral
63.
go back to reference C. Lopez-Gines, L. Navarro, L. Munoz-Hidalgo, E. Buso, J.M. Morales, R. Gil-Benso, M. Gregori-Romero, J. Megias, P. Roldan, R. Segura-Sabater, J.M. Almerich-Silla, D. Monleon, M. Cerda-Nicolas, Association between epidermal growth factor receptor amplification and ADP-ribosylation factor 1 methylation in human glioblastoma. Cell Oncol 40, 389–399 (2017)CrossRef C. Lopez-Gines, L. Navarro, L. Munoz-Hidalgo, E. Buso, J.M. Morales, R. Gil-Benso, M. Gregori-Romero, J. Megias, P. Roldan, R. Segura-Sabater, J.M. Almerich-Silla, D. Monleon, M. Cerda-Nicolas, Association between epidermal growth factor receptor amplification and ADP-ribosylation factor 1 methylation in human glioblastoma. Cell Oncol 40, 389–399 (2017)CrossRef
64.
go back to reference B.R. Voldborg, L. Damstrup, M. Spang-Thomsen, H.S. Poulsen, Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol 8, 1197–1206 (1997)CrossRefPubMed B.R. Voldborg, L. Damstrup, M. Spang-Thomsen, H.S. Poulsen, Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol 8, 1197–1206 (1997)CrossRefPubMed
65.
go back to reference M. Nagane, H. Lin, W.K. Cavenee, H.J. Huang, Aberrant receptor signaling in human malignant gliomas: Mechanisms and therapeutic implications. Cancer Lett 162 Suppl, S17–S21 (2001)CrossRefPubMed M. Nagane, H. Lin, W.K. Cavenee, H.J. Huang, Aberrant receptor signaling in human malignant gliomas: Mechanisms and therapeutic implications. Cancer Lett 162 Suppl, S17–S21 (2001)CrossRefPubMed
66.
go back to reference Y. Narita, M. Nagane, K. Mishima, H.J. Huang, F.B. Furnari, W.K. Cavenee, Mutant epidermal growth factor receptor signaling down-regulates p27 through activation of the phosphatidylinositol 3-kinase/Akt pathway in glioblastomas. Cancer Res 62, 6764–6769 (2002)PubMed Y. Narita, M. Nagane, K. Mishima, H.J. Huang, F.B. Furnari, W.K. Cavenee, Mutant epidermal growth factor receptor signaling down-regulates p27 through activation of the phosphatidylinositol 3-kinase/Akt pathway in glioblastomas. Cancer Res 62, 6764–6769 (2002)PubMed
67.
go back to reference J. Couet, M. Sargiacomo, M.P. Lisanti, Interaction of a receptor tyrosine kinase, EGF-R, with caveolins. Caveolin binding negatively regulates tyrosine and serine/threonine kinase activities. J Biol Chem 272, 30429–30438 (1997)CrossRefPubMed J. Couet, M. Sargiacomo, M.P. Lisanti, Interaction of a receptor tyrosine kinase, EGF-R, with caveolins. Caveolin binding negatively regulates tyrosine and serine/threonine kinase activities. J Biol Chem 272, 30429–30438 (1997)CrossRefPubMed
68.
go back to reference C. Mineo, G.N. Gill, R.G. Anderson, Regulated migration of epidermal growth factor receptor from caveolae. J Biol Chem 274, 30636–30643 (1999)CrossRefPubMed C. Mineo, G.N. Gill, R.G. Anderson, Regulated migration of epidermal growth factor receptor from caveolae. J Biol Chem 274, 30636–30643 (1999)CrossRefPubMed
69.
go back to reference T.F. Cloughesy, K. Yoshimoto, P. Nghiemphu, K. Brown, J. Dang, S. Zhu, T. Hsueh, Y. Chen, W. Wang, D. Youngkin, L. Liau, N. Martin, D. Becker, M. Bergsneider, A. Lai, R. Green, T. Oglesby, M. Koleto, J. Trent, S. Horvath, P.S. Mischel, I.K. Mellinghoff, C.L. Sawyers, Antitumor activity of rapamycin in a phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med 5, e8 (2008)CrossRefPubMedPubMedCentral T.F. Cloughesy, K. Yoshimoto, P. Nghiemphu, K. Brown, J. Dang, S. Zhu, T. Hsueh, Y. Chen, W. Wang, D. Youngkin, L. Liau, N. Martin, D. Becker, M. Bergsneider, A. Lai, R. Green, T. Oglesby, M. Koleto, J. Trent, S. Horvath, P.S. Mischel, I.K. Mellinghoff, C.L. Sawyers, Antitumor activity of rapamycin in a phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med 5, e8 (2008)CrossRefPubMedPubMedCentral
70.
go back to reference S.F. Hussain, L.Y. Kong, J. Jordan, C. Conrad, T. Madden, I. Fokt, W. Priebe, A.B. Heimberger, A novel small molecule inhibitor of signal transducers and activators of transcription 3 reverses immune tolerance in malignant glioma patients. Cancer Res 67, 9630–9636 (2007)CrossRefPubMed S.F. Hussain, L.Y. Kong, J. Jordan, C. Conrad, T. Madden, I. Fokt, W. Priebe, A.B. Heimberger, A novel small molecule inhibitor of signal transducers and activators of transcription 3 reverses immune tolerance in malignant glioma patients. Cancer Res 67, 9630–9636 (2007)CrossRefPubMed
71.
go back to reference A. Iwamaru, S. Szymanski, E. Iwado, H. Aoki, T. Yokoyama, I. Fokt, K. Hess, C. Conrad, T. Madden, R. Sawaya, S. Kondo, W. Priebe, Y. Kondo, A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene 26, 2435–2444 (2007)CrossRefPubMed A. Iwamaru, S. Szymanski, E. Iwado, H. Aoki, T. Yokoyama, I. Fokt, K. Hess, C. Conrad, T. Madden, R. Sawaya, S. Kondo, W. Priebe, Y. Kondo, A novel inhibitor of the STAT3 pathway induces apoptosis in malignant glioma cells both in vitro and in vivo. Oncogene 26, 2435–2444 (2007)CrossRefPubMed
72.
go back to reference H.W. Lo, X. Cao, H. Zhu, F. Ali-Osman, Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res 14, 6042–6054 (2008)CrossRefPubMedPubMedCentral H.W. Lo, X. Cao, H. Zhu, F. Ali-Osman, Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res 14, 6042–6054 (2008)CrossRefPubMedPubMedCentral
73.
go back to reference S.O. Rahaman, P.C. Harbor, O. Chernova, G.H. Barnett, M.A. Vogelbaum, S.J. Haque, Inhibition of constitutively active Stat3 suppresses proliferation and induces apoptosis in glioblastoma multiforme cells. Oncogene 21, 8404–8413 (2002)CrossRefPubMed S.O. Rahaman, P.C. Harbor, O. Chernova, G.H. Barnett, M.A. Vogelbaum, S.J. Haque, Inhibition of constitutively active Stat3 suppresses proliferation and induces apoptosis in glioblastoma multiforme cells. Oncogene 21, 8404–8413 (2002)CrossRefPubMed
74.
go back to reference R. Ghildiyal, D. Dixit, E. Sen, EGFR inhibitor BIBU induces apoptosis and defective autophagy in glioma cells. Mol Carcinog 52, 970–982 (2013)CrossRefPubMed R. Ghildiyal, D. Dixit, E. Sen, EGFR inhibitor BIBU induces apoptosis and defective autophagy in glioma cells. Mol Carcinog 52, 970–982 (2013)CrossRefPubMed
75.
go back to reference X.Q. Wang, Q. Yan, P. Sun, J.W. Liu, L. Go, S.M. McDaniel, A.S. Paller, Suppression of epidermal growth factor receptor signaling by protein kinase C-alpha activation requires CD82, caveolin-1, and ganglioside. Cancer Res 67, 9986–9995 (2007)CrossRefPubMed X.Q. Wang, Q. Yan, P. Sun, J.W. Liu, L. Go, S.M. McDaniel, A.S. Paller, Suppression of epidermal growth factor receptor signaling by protein kinase C-alpha activation requires CD82, caveolin-1, and ganglioside. Cancer Res 67, 9986–9995 (2007)CrossRefPubMed
76.
go back to reference M. Jakobisiak, J. Golab, Potential antitumor effects of statins (review). Int J Oncol 23, 1055–1069 (2003)PubMed M. Jakobisiak, J. Golab, Potential antitumor effects of statins (review). Int J Oncol 23, 1055–1069 (2003)PubMed
77.
go back to reference H. Komuro, T. Kumada, Ca2+ transients control CNS neuronal migration. Cell Calcium 37, 387–393 (2005)CrossRefPubMed H. Komuro, T. Kumada, Ca2+ transients control CNS neuronal migration. Cell Calcium 37, 387–393 (2005)CrossRefPubMed
78.
go back to reference A.K. Weaver, V.C. Bomben, H. Sontheimer, Expression and function of calcium-activated potassium channels in human glioma cells. Glia 54, 223–233 (2006)CrossRefPubMedPubMedCentral A.K. Weaver, V.C. Bomben, H. Sontheimer, Expression and function of calcium-activated potassium channels in human glioma cells. Glia 54, 223–233 (2006)CrossRefPubMedPubMedCentral
79.
go back to reference A. Arcangeli, O. Crociani, E. Lastraioli, A. Masi, S. Pillozzi, A. Becchetti, Targeting ion channels in cancer: A novel frontier in antineoplastic therapy. Curr Med Chem 16, 66–93 (2009)CrossRefPubMed A. Arcangeli, O. Crociani, E. Lastraioli, A. Masi, S. Pillozzi, A. Becchetti, Targeting ion channels in cancer: A novel frontier in antineoplastic therapy. Curr Med Chem 16, 66–93 (2009)CrossRefPubMed
80.
go back to reference H. Komuro, P. Rakic, Orchestration of neuronal migration by activity of ion channels, neurotransmitter receptors, and intracellular Ca2+ fluctuations. J Neurobiol 37, 110–130 (1998)CrossRefPubMed H. Komuro, P. Rakic, Orchestration of neuronal migration by activity of ion channels, neurotransmitter receptors, and intracellular Ca2+ fluctuations. J Neurobiol 37, 110–130 (1998)CrossRefPubMed
81.
go back to reference A. Bordey, H. Sontheimer, J. Trouslard, Muscarinic activation of BK channels induces membrane oscillations in glioma cells and leads to inhibition of cell migration. J Membr Biol 176, 31–40 (2000)CrossRefPubMed A. Bordey, H. Sontheimer, J. Trouslard, Muscarinic activation of BK channels induces membrane oscillations in glioma cells and leads to inhibition of cell migration. J Membr Biol 176, 31–40 (2000)CrossRefPubMed
82.
go back to reference A. Fabian, T. Fortmann, P. Dieterich, C. Riethmuller, P. Schon, S. Mally, B. Nilius, A. Schwab, TRPC1 channels regulate directionality of migrating cells. Pflugers Arch 457, 475–484 (2008)CrossRefPubMed A. Fabian, T. Fortmann, P. Dieterich, C. Riethmuller, P. Schon, S. Mally, B. Nilius, A. Schwab, TRPC1 channels regulate directionality of migrating cells. Pflugers Arch 457, 475–484 (2008)CrossRefPubMed
83.
go back to reference V.C. Bomben, H. Sontheimer, Disruption of transient receptor potential canonical channel 1 causes incomplete cytokinesis and slows the growth of human malignant gliomas. Glia 58, 1145–1156 (2010)CrossRefPubMedPubMedCentral V.C. Bomben, H. Sontheimer, Disruption of transient receptor potential canonical channel 1 causes incomplete cytokinesis and slows the growth of human malignant gliomas. Glia 58, 1145–1156 (2010)CrossRefPubMedPubMedCentral
84.
go back to reference V.C. Bomben, H.W. Sontheimer, Inhibition of transient receptor potential canonical channels impairs cytokinesis in human malignant gliomas. Cell Prolif 41, 98–121 (2008)CrossRefPubMedPubMedCentral V.C. Bomben, H.W. Sontheimer, Inhibition of transient receptor potential canonical channels impairs cytokinesis in human malignant gliomas. Cell Prolif 41, 98–121 (2008)CrossRefPubMedPubMedCentral
85.
go back to reference V.C. Bomben, K.L. Turner, T.T. Barclay, H. Sontheimer, Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J Cell Physiol 226, 1879–1888 (2011)CrossRefPubMed V.C. Bomben, K.L. Turner, T.T. Barclay, H. Sontheimer, Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J Cell Physiol 226, 1879–1888 (2011)CrossRefPubMed
86.
go back to reference C.V. Remillard, J.X. Yuan, Transient receptor potential channels and caveolin-1: Good friends in tight spaces. Mol Pharmacol 70, 1151–1154 (2006)CrossRefPubMed C.V. Remillard, J.X. Yuan, Transient receptor potential channels and caveolin-1: Good friends in tight spaces. Mol Pharmacol 70, 1151–1154 (2006)CrossRefPubMed
87.
go back to reference Y. El Hiani, V. Lehen'kyi, H. Ouadid-Ahidouch, A. Ahidouch, Activation of the calcium-sensing receptor by high calcium induced breast cancer cell proliferation and TRPC1 cation channel over-expression potentially through EGFR pathways. Arch Biochem Biophys 486, 58–63 (2009)CrossRefPubMed Y. El Hiani, V. Lehen'kyi, H. Ouadid-Ahidouch, A. Ahidouch, Activation of the calcium-sensing receptor by high calcium induced breast cancer cell proliferation and TRPC1 cation channel over-expression potentially through EGFR pathways. Arch Biochem Biophys 486, 58–63 (2009)CrossRefPubMed
88.
go back to reference S. Feske, Calcium signalling in lymphocyte activation and disease. Nat Rev Immunol 7, 690–702 (2007)CrossRefPubMed S. Feske, Calcium signalling in lymphocyte activation and disease. Nat Rev Immunol 7, 690–702 (2007)CrossRefPubMed
89.
go back to reference P. Parsons-Wingerter, I.M. Kasman, S. Norberg, A. Magnussen, S. Zanivan, A. Rissone, P. Baluk, C.J. Favre, U. Jeffry, R. Murray, D.M. McDonald, Uniform overexpression and rapid accessibility of alpha5beta1 integrin on blood vessels in tumors. Am J Pathol 167, 193–211 (2005)CrossRefPubMedPubMedCentral P. Parsons-Wingerter, I.M. Kasman, S. Norberg, A. Magnussen, S. Zanivan, A. Rissone, P. Baluk, C.J. Favre, U. Jeffry, R. Murray, D.M. McDonald, Uniform overexpression and rapid accessibility of alpha5beta1 integrin on blood vessels in tumors. Am J Pathol 167, 193–211 (2005)CrossRefPubMedPubMedCentral
90.
go back to reference O. Stoeltzing, W. Liu, N. Reinmuth, F. Fan, G.C. Parry, A.A. Parikh, M.F. McCarty, C.D. Bucana, A.P. Mazar, L.M. Ellis, Inhibition of integrin alpha5beta1 function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. Int J Cancer 104, 496–503 (2003)CrossRefPubMed O. Stoeltzing, W. Liu, N. Reinmuth, F. Fan, G.C. Parry, A.A. Parikh, M.F. McCarty, C.D. Bucana, A.P. Mazar, L.M. Ellis, Inhibition of integrin alpha5beta1 function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. Int J Cancer 104, 496–503 (2003)CrossRefPubMed
91.
go back to reference K. Sawada, A.K. Mitra, A.R. Radjabi, V. Bhaskar, E.O. Kistner, M. Tretiakova, S. Jagadeeswaran, A. Montag, A. Becker, H.A. Kenny, M.E. Peter, V. Ramakrishnan, S.D. Yamada, E. Lengyel, Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res 68, 2329–2339 (2008)CrossRefPubMedPubMedCentral K. Sawada, A.K. Mitra, A.R. Radjabi, V. Bhaskar, E.O. Kistner, M. Tretiakova, S. Jagadeeswaran, A. Montag, A. Becker, H.A. Kenny, M.E. Peter, V. Ramakrishnan, S.D. Yamada, E. Lengyel, Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res 68, 2329–2339 (2008)CrossRefPubMedPubMedCentral
92.
go back to reference A. Maglott, P. Bartik, S. Cosgun, P. Klotz, P. Ronde, G. Fuhrmann, K. Takeda, S. Martin, M. Dontenwill, The small alpha5beta1 integrin antagonist, SJ749, reduces proliferation and clonogenicity of human astrocytoma cells. Cancer Res 66, 6002–6007 (2006)CrossRefPubMed A. Maglott, P. Bartik, S. Cosgun, P. Klotz, P. Ronde, G. Fuhrmann, K. Takeda, S. Martin, M. Dontenwill, The small alpha5beta1 integrin antagonist, SJ749, reduces proliferation and clonogenicity of human astrocytoma cells. Cancer Res 66, 6002–6007 (2006)CrossRefPubMed
93.
go back to reference M.A. del Pozo, N. Balasubramanian, N.B. Alderson, W.B. Kiosses, A. Grande-Garcia, R.G. Anderson, M.A. Schwartz, Phospho-caveolin-1 mediates integrin-regulated membrane domain internalization. Nat Cell Biol 7, 901–908 (2005)CrossRefPubMedPubMedCentral M.A. del Pozo, N. Balasubramanian, N.B. Alderson, W.B. Kiosses, A. Grande-Garcia, R.G. Anderson, M.A. Schwartz, Phospho-caveolin-1 mediates integrin-regulated membrane domain internalization. Nat Cell Biol 7, 901–908 (2005)CrossRefPubMedPubMedCentral
94.
go back to reference K.K. Wary, A. Mariotti, C. Zurzolo, F.G. Giancotti, A requirement for caveolin-1 and associated kinase Fyn in integrin signaling and anchorage-dependent cell growth. Cell 94, 625–634 (1998)CrossRefPubMed K.K. Wary, A. Mariotti, C. Zurzolo, F.G. Giancotti, A requirement for caveolin-1 and associated kinase Fyn in integrin signaling and anchorage-dependent cell growth. Cell 94, 625–634 (1998)CrossRefPubMed
95.
go back to reference K.K. Wary, F. Mainiero, S.J. Isakoff, E.E. Marcantonio, F.G. Giancotti, The adaptor protein Shc couples a class of integrins to the control of cell cycle progression. Cell 87, 733–743 (1996)CrossRefPubMed K.K. Wary, F. Mainiero, S.J. Isakoff, E.E. Marcantonio, F.G. Giancotti, The adaptor protein Shc couples a class of integrins to the control of cell cycle progression. Cell 87, 733–743 (1996)CrossRefPubMed
96.
go back to reference S. Martin, E.C. Cosset, J. Terrand, A. Maglott, K. Takeda, M. Dontenwill, Caveolin-1 regulates glioblastoma aggressiveness through the control of alpha(5)beta(1) integrin expression and modulates glioblastoma responsiveness to SJ749, an alpha(5)beta(1) integrin antagonist. Biochim Biophys Acta 1793, 354–367 (2009)CrossRefPubMed S. Martin, E.C. Cosset, J. Terrand, A. Maglott, K. Takeda, M. Dontenwill, Caveolin-1 regulates glioblastoma aggressiveness through the control of alpha(5)beta(1) integrin expression and modulates glioblastoma responsiveness to SJ749, an alpha(5)beta(1) integrin antagonist. Biochim Biophys Acta 1793, 354–367 (2009)CrossRefPubMed
97.
go back to reference A.W. Cohen, D.S. Park, S.E. Woodman, T.M. Williams, M. Chandra, J. Shirani, A. Pereira de Souza, R.N. Kitsis, R.G. Russell, L.M. Weiss, B. Tang, L.A. Jelicks, S.M. Factor, V. Shtutin, H.B. Tanowitz, M.P. Lisanti, Caveolin-1 null mice develop cardiac hypertrophy with hyperactivation of p42/44 MAP kinase in cardiac fibroblasts. Am J Physiol Cell Physiol 284, C457–C474 (2003)CrossRefPubMed A.W. Cohen, D.S. Park, S.E. Woodman, T.M. Williams, M. Chandra, J. Shirani, A. Pereira de Souza, R.N. Kitsis, R.G. Russell, L.M. Weiss, B. Tang, L.A. Jelicks, S.M. Factor, V. Shtutin, H.B. Tanowitz, M.P. Lisanti, Caveolin-1 null mice develop cardiac hypertrophy with hyperactivation of p42/44 MAP kinase in cardiac fibroblasts. Am J Physiol Cell Physiol 284, C457–C474 (2003)CrossRefPubMed
98.
go back to reference A. Wesolowska, A. Kwiatkowska, L. Slomnicki, M. Dembinski, A. Master, M. Sliwa, K. Franciszkiewicz, S. Chouaib, B. Kaminska, Microglia-derived TGF-beta as an important regulator of glioblastoma invasion--an inhibition of TGF-beta-dependent effects by shRNA against human TGF-beta type II receptor. Oncogene 27, 918–930 (2008)CrossRefPubMed A. Wesolowska, A. Kwiatkowska, L. Slomnicki, M. Dembinski, A. Master, M. Sliwa, K. Franciszkiewicz, S. Chouaib, B. Kaminska, Microglia-derived TGF-beta as an important regulator of glioblastoma invasion--an inhibition of TGF-beta-dependent effects by shRNA against human TGF-beta type II receptor. Oncogene 27, 918–930 (2008)CrossRefPubMed
99.
go back to reference A. Bruna, R.S. Darken, F. Rojo, A. Ocana, S. Penuelas, A. Arias, R. Paris, A. Tortosa, J. Mora, J. Baselga, J. Seoane, High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell 11, 147–160 (2007)CrossRefPubMed A. Bruna, R.S. Darken, F. Rojo, A. Ocana, S. Penuelas, A. Arias, R. Paris, A. Tortosa, J. Mora, J. Baselga, J. Seoane, High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell 11, 147–160 (2007)CrossRefPubMed
100.
go back to reference M. Uhl, S. Aulwurm, J. Wischhusen, M. Weiler, J.Y. Ma, R. Almirez, R. Mangadu, Y.W. Liu, M. Platten, U. Herrlinger, A. Murphy, D.H. Wong, W. Wick, L.S. Higgins, M. Weller, SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo. Cancer Res 64, 7954–7961 (2004)CrossRefPubMed M. Uhl, S. Aulwurm, J. Wischhusen, M. Weiler, J.Y. Ma, R. Almirez, R. Mangadu, Y.W. Liu, M. Platten, U. Herrlinger, A. Murphy, D.H. Wong, W. Wick, L.S. Higgins, M. Weller, SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo. Cancer Res 64, 7954–7961 (2004)CrossRefPubMed
101.
go back to reference M.D. Hjelmeland, A.B. Hjelmeland, S. Sathornsumetee, E.D. Reese, M.H. Herbstreith, N.J. Laping, H.S. Friedman, D.D. Bigner, X.F. Wang, J.N. Rich, SB-431542, a small molecule transforming growth factor-beta-receptor antagonist, inhibits human glioma cell line proliferation and motility. Mol Cancer Ther 3, 737–745 (2004)PubMed M.D. Hjelmeland, A.B. Hjelmeland, S. Sathornsumetee, E.D. Reese, M.H. Herbstreith, N.J. Laping, H.S. Friedman, D.D. Bigner, X.F. Wang, J.N. Rich, SB-431542, a small molecule transforming growth factor-beta-receptor antagonist, inhibits human glioma cell line proliferation and motility. Mol Cancer Ther 3, 737–745 (2004)PubMed
102.
go back to reference S. Kim, G. Buchlis, Z.G. Fridlender, J. Sun, V. Kapoor, G. Cheng, A. Haas, H.K. Cheung, X. Zhang, M. Corbley, L.R. Kaiser, L. Ling, S.M. Albelda, Systemic blockade of transforming growth factor-beta signaling augments the efficacy of immunogene therapy. Cancer Res 68, 10247–10256 (2008)CrossRefPubMedPubMedCentral S. Kim, G. Buchlis, Z.G. Fridlender, J. Sun, V. Kapoor, G. Cheng, A. Haas, H.K. Cheung, X. Zhang, M. Corbley, L.R. Kaiser, L. Ling, S.M. Albelda, Systemic blockade of transforming growth factor-beta signaling augments the efficacy of immunogene therapy. Cancer Res 68, 10247–10256 (2008)CrossRefPubMedPubMedCentral
103.
go back to reference P. Hau, P. Jachimczak, R. Schlingensiepen, F. Schulmeyer, T. Jauch, A. Steinbrecher, A. Brawanski, M. Proescholdt, J. Schlaier, J. Buchroithner, J. Pichler, G. Wurm, M. Mehdorn, R. Strege, G. Schuierer, V. Villarrubia, F. Fellner, O. Jansen, T. Straube, V. Nohria, M. Goldbrunner, M. Kunst, S. Schmaus, G. Stauder, U. Bogdahn, K.H. Schlingensiepen, Inhibition of TGF-beta2 with AP 12009 in recurrent malignant gliomas: From preclinical to phase I/II studies. Oligonucleotides 17, 201–212 (2007)CrossRefPubMed P. Hau, P. Jachimczak, R. Schlingensiepen, F. Schulmeyer, T. Jauch, A. Steinbrecher, A. Brawanski, M. Proescholdt, J. Schlaier, J. Buchroithner, J. Pichler, G. Wurm, M. Mehdorn, R. Strege, G. Schuierer, V. Villarrubia, F. Fellner, O. Jansen, T. Straube, V. Nohria, M. Goldbrunner, M. Kunst, S. Schmaus, G. Stauder, U. Bogdahn, K.H. Schlingensiepen, Inhibition of TGF-beta2 with AP 12009 in recurrent malignant gliomas: From preclinical to phase I/II studies. Oligonucleotides 17, 201–212 (2007)CrossRefPubMed
104.
go back to reference U. Bogdahn, P. Hau, G. Stockhammer, N.K. Venkataramana, A.K. Mahapatra, A. Suri, A. Balasubramaniam, S. Nair, V. Oliushine, V. Parfenov, I. Poverennova, M. Zaaroor, P. Jachimczak, S. Ludwig, S. Schmaus, H. Heinrichs, K.H. Schlingensiepen, G. Trabedersen Glioma Study, Targeted therapy for high-grade glioma with the TGF-beta2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study. Neuro-Oncology 13, 132–142 (2011)CrossRefPubMed U. Bogdahn, P. Hau, G. Stockhammer, N.K. Venkataramana, A.K. Mahapatra, A. Suri, A. Balasubramaniam, S. Nair, V. Oliushine, V. Parfenov, I. Poverennova, M. Zaaroor, P. Jachimczak, S. Ludwig, S. Schmaus, H. Heinrichs, K.H. Schlingensiepen, G. Trabedersen Glioma Study, Targeted therapy for high-grade glioma with the TGF-beta2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study. Neuro-Oncology 13, 132–142 (2011)CrossRefPubMed
105.
go back to reference D.A. Goodenough, J.A. Goliger, D.L. Paul, Connexins, connexons, and intercellular communication. Annu Rev Biochem 65, 475–502 (1996)CrossRefPubMed D.A. Goodenough, J.A. Goliger, D.L. Paul, Connexins, connexons, and intercellular communication. Annu Rev Biochem 65, 475–502 (1996)CrossRefPubMed
106.
go back to reference P. Pu, Z. Xia, S. Yu, Q. Huang, Altered expression of Cx43 in astrocytic tumors. Clin Neurol Neurosurg 107, 49–54 (2004)CrossRefPubMed P. Pu, Z. Xia, S. Yu, Q. Huang, Altered expression of Cx43 in astrocytic tumors. Clin Neurol Neurosurg 107, 49–54 (2004)CrossRefPubMed
107.
go back to reference L. Soroceanu, T.J. Manning Jr., H. Sontheimer, Reduced expression of connexin-43 and functional gap junction coupling in human gliomas. Glia 33, 107–117 (2001)CrossRefPubMed L. Soroceanu, T.J. Manning Jr., H. Sontheimer, Reduced expression of connexin-43 and functional gap junction coupling in human gliomas. Glia 33, 107–117 (2001)CrossRefPubMed
108.
go back to reference W. Zhang, C. Nwagwu, D.M. Le, V.W. Yong, H. Song, W.T. Couldwell, Increased invasive capacity of connexin43-overexpressing malignant glioma cells. J Neurosurg 99, 1039–1046 (2003)CrossRefPubMed W. Zhang, C. Nwagwu, D.M. Le, V.W. Yong, H. Song, W.T. Couldwell, Increased invasive capacity of connexin43-overexpressing malignant glioma cells. J Neurosurg 99, 1039–1046 (2003)CrossRefPubMed
109.
go back to reference J.H. Lin, T. Takano, M.L. Cotrina, G. Arcuino, J. Kang, S. Liu, Q. Gao, L. Jiang, F. Li, H. Lichtenberg-Frate, S. Haubrich, K. Willecke, S.A. Goldman, M. Nedergaard, Connexin 43 enhances the adhesivity and mediates the invasion of malignant glioma cells. J Neurosci 22, 4302–4311 (2002)CrossRefPubMedPubMedCentral J.H. Lin, T. Takano, M.L. Cotrina, G. Arcuino, J. Kang, S. Liu, Q. Gao, L. Jiang, F. Li, H. Lichtenberg-Frate, S. Haubrich, K. Willecke, S.A. Goldman, M. Nedergaard, Connexin 43 enhances the adhesivity and mediates the invasion of malignant glioma cells. J Neurosci 22, 4302–4311 (2002)CrossRefPubMedPubMedCentral
110.
go back to reference P.R. Gielen, Q. Aftab, N. Ma, V.C. Chen, X. Hong, S. Lozinsky, C.C. Naus, W.C. Sin, Connexin43 confers Temozolomide resistance in human glioma cells by modulating the mitochondrial apoptosis pathway. Neuropharmacology 75, 539–548 (2013)CrossRefPubMed P.R. Gielen, Q. Aftab, N. Ma, V.C. Chen, X. Hong, S. Lozinsky, C.C. Naus, W.C. Sin, Connexin43 confers Temozolomide resistance in human glioma cells by modulating the mitochondrial apoptosis pathway. Neuropharmacology 75, 539–548 (2013)CrossRefPubMed
111.
go back to reference S.F. Murphy, R.T. Varghese, S. Lamouille, S. Guo, K.J. Pridham, P. Kanabur, A.M. Osimani, S. Sharma, J. Jourdan, C.M. Rodgers, G.R. Simonds, R.G. Gourdie, Z. Sheng, Connexin 43 inhibition sensitizes Chemoresistant glioblastoma cells to Temozolomide. Cancer Res 76, 139–149 (2016)CrossRefPubMed S.F. Murphy, R.T. Varghese, S. Lamouille, S. Guo, K.J. Pridham, P. Kanabur, A.M. Osimani, S. Sharma, J. Jourdan, C.M. Rodgers, G.R. Simonds, R.G. Gourdie, Z. Sheng, Connexin 43 inhibition sensitizes Chemoresistant glioblastoma cells to Temozolomide. Cancer Res 76, 139–149 (2016)CrossRefPubMed
112.
go back to reference P.O. Strale, J. Clarhaut, C. Lamiche, L. Cronier, M. Mesnil, N. Defamie, Down-regulation of Connexin43 expression reveals the involvement of caveolin-1 containing lipid rafts in human U251 glioblastoma cell invasion. Mol Carcinog 51, 845–860 (2012)CrossRefPubMed P.O. Strale, J. Clarhaut, C. Lamiche, L. Cronier, M. Mesnil, N. Defamie, Down-regulation of Connexin43 expression reveals the involvement of caveolin-1 containing lipid rafts in human U251 glioblastoma cell invasion. Mol Carcinog 51, 845–860 (2012)CrossRefPubMed
113.
go back to reference D.W. Laird, The gap junction proteome and its relationship to disease. Trends Cell Biol 20, 92–101 (2010)CrossRefPubMed D.W. Laird, The gap junction proteome and its relationship to disease. Trends Cell Biol 20, 92–101 (2010)CrossRefPubMed
114.
go back to reference M. Mesnil, S. Crespin, J.L. Avanzo, M.L. Zaidan-Dagli, Defective gap junctional intercellular communication in the carcinogenic process. Biochim Biophys Acta 1719, 125–145 (2005)CrossRefPubMed M. Mesnil, S. Crespin, J.L. Avanzo, M.L. Zaidan-Dagli, Defective gap junctional intercellular communication in the carcinogenic process. Biochim Biophys Acta 1719, 125–145 (2005)CrossRefPubMed
115.
go back to reference D.J. Fitzgerald, M. Mesnil, M. Oyamada, H. Tsuda, N. Ito, H. Yamasaki, Changes in gap junction protein (connexin 32) gene expression during rat liver carcinogenesis. J Cell Biochem 41, 97–102 (1989)CrossRefPubMed D.J. Fitzgerald, M. Mesnil, M. Oyamada, H. Tsuda, N. Ito, H. Yamasaki, Changes in gap junction protein (connexin 32) gene expression during rat liver carcinogenesis. J Cell Biochem 41, 97–102 (1989)CrossRefPubMed
116.
go back to reference R. Du, K.V. Lu, C. Petritsch, P. Liu, R. Ganss, E. Passegue, H. Song, S. Vandenberg, R.S. Johnson, Z. Werb, G. Bergers, HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13, 206–220 (2008)CrossRefPubMedPubMedCentral R. Du, K.V. Lu, C. Petritsch, P. Liu, R. Ganss, E. Passegue, H. Song, S. Vandenberg, R.S. Johnson, Z. Werb, G. Bergers, HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13, 206–220 (2008)CrossRefPubMedPubMedCentral
117.
go back to reference B. Raychaudhuri, P. Rayman, J. Ireland, J. Ko, B. Rini, E.C. Borden, J. Garcia, M.A. Vogelbaum, J. Finke, Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro-Oncology 13, 591–599 (2011)CrossRefPubMedPubMedCentral B. Raychaudhuri, P. Rayman, J. Ireland, J. Ko, B. Rini, E.C. Borden, J. Garcia, M.A. Vogelbaum, J. Finke, Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro-Oncology 13, 591–599 (2011)CrossRefPubMedPubMedCentral
118.
go back to reference S.F. Hussain, D. Yang, D. Suki, K. Aldape, E. Grimm, A.B. Heimberger, The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro-Oncology 8, 261–279 (2006)CrossRefPubMedPubMedCentral S.F. Hussain, D. Yang, D. Suki, K. Aldape, E. Grimm, A.B. Heimberger, The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro-Oncology 8, 261–279 (2006)CrossRefPubMedPubMedCentral
119.
go back to reference H. Zhai, F.L. Heppner, S.E. Tsirka, Microglia/macrophages promote glioma progression. Glia 59, 472–485 (2011)CrossRefPubMed H. Zhai, F.L. Heppner, S.E. Tsirka, Microglia/macrophages promote glioma progression. Glia 59, 472–485 (2011)CrossRefPubMed
120.
go back to reference L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell Oncol 40, 471–482 (2017)CrossRef L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell Oncol 40, 471–482 (2017)CrossRef
121.
go back to reference R.A. Morgan, L.A. Johnson, J.L. Davis, Z. Zheng, K.D. Woolard, E.A. Reap, S.A. Feldman, N. Chinnasamy, C.T. Kuan, H. Song, W. Zhang, H.A. Fine, S.A. Rosenberg, Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 23, 1043–1053 (2012)CrossRefPubMedPubMedCentral R.A. Morgan, L.A. Johnson, J.L. Davis, Z. Zheng, K.D. Woolard, E.A. Reap, S.A. Feldman, N. Chinnasamy, C.T. Kuan, H. Song, W. Zhang, H.A. Fine, S.A. Rosenberg, Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 23, 1043–1053 (2012)CrossRefPubMedPubMedCentral
122.
go back to reference H. Gao, I.Y. Zhang, L. Zhang, Y. Song, S. Liu, H. Ren, H. Liu, H. Zhou, Y. Su, Y. Yang, B. Badie, S100B suppression alters polarization of infiltrating myeloid-derived cells in gliomas and inhibits tumor growth. Cancer Lett (2018) H. Gao, I.Y. Zhang, L. Zhang, Y. Song, S. Liu, H. Ren, H. Liu, H. Zhou, Y. Su, Y. Yang, B. Badie, S100B suppression alters polarization of infiltrating myeloid-derived cells in gliomas and inhibits tumor growth. Cancer Lett (2018)
123.
go back to reference J. Harris, D. Werling, M. Koss, P. Monaghan, G. Taylor, C.J. Howard, Expression of caveolin by bovine lymphocytes and antigen-presenting cells. Immunology 105, 190–195 (2002)CrossRefPubMedPubMedCentral J. Harris, D. Werling, M. Koss, P. Monaghan, G. Taylor, C.J. Howard, Expression of caveolin by bovine lymphocytes and antigen-presenting cells. Immunology 105, 190–195 (2002)CrossRefPubMedPubMedCentral
124.
go back to reference R.A. Santizo, H.L. Xu, E. Galea, S. Muyskens, V.L. Baughman, D.A. Pelligrino, Combined endothelial nitric oxide synthase upregulation and caveolin-1 downregulation decrease leukocyte adhesion in pial venules of ovariectomized female rats. Stroke 33, 613–616 (2002)CrossRefPubMed R.A. Santizo, H.L. Xu, E. Galea, S. Muyskens, V.L. Baughman, D.A. Pelligrino, Combined endothelial nitric oxide synthase upregulation and caveolin-1 downregulation decrease leukocyte adhesion in pial venules of ovariectomized female rats. Stroke 33, 613–616 (2002)CrossRefPubMed
125.
go back to reference M. Ryuto, M. Ono, H. Izumi, S. Yoshida, H.A. Weich, K. Kohno, M. Kuwano, Induction of vascular endothelial growth factor by tumor necrosis factor alpha in human glioma cells. Possible roles of SP-1. J Biol Chem 271, 28220–28228 (1996)CrossRefPubMed M. Ryuto, M. Ono, H. Izumi, S. Yoshida, H.A. Weich, K. Kohno, M. Kuwano, Induction of vascular endothelial growth factor by tumor necrosis factor alpha in human glioma cells. Possible roles of SP-1. J Biol Chem 271, 28220–28228 (1996)CrossRefPubMed
126.
go back to reference M. Tsujimoto, Y.K. Yip, J. Vilcek, Tumor necrosis factor: Specific binding and internalization in sensitive and resistant cells. Proc Natl Acad Sci U S A 82, 7626–7630 (1985)CrossRefPubMedPubMedCentral M. Tsujimoto, Y.K. Yip, J. Vilcek, Tumor necrosis factor: Specific binding and internalization in sensitive and resistant cells. Proc Natl Acad Sci U S A 82, 7626–7630 (1985)CrossRefPubMedPubMedCentral
127.
go back to reference A.A. Beg, D. Baltimore, An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 274, 782–784 (1996)CrossRefPubMed A.A. Beg, D. Baltimore, An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 274, 782–784 (1996)CrossRefPubMed
128.
go back to reference D.J. Van Antwerp, S.J. Martin, T. Kafri, D.R. Green, I.M. Verma, Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science 274, 787–789 (1996)CrossRefPubMed D.J. Van Antwerp, S.J. Martin, T. Kafri, D.R. Green, I.M. Verma, Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science 274, 787–789 (1996)CrossRefPubMed
129.
go back to reference Y. Ding, J. Shen, G. Zhang, X. Chen, J. Wu, W. Chen, CD40 controls CXCR5-induced recruitment of myeloid-derived suppressor cells to gastric cancer. Oncotarget 6, 38901–38911 (2015)PubMedPubMedCentral Y. Ding, J. Shen, G. Zhang, X. Chen, J. Wu, W. Chen, CD40 controls CXCR5-induced recruitment of myeloid-derived suppressor cells to gastric cancer. Oncotarget 6, 38901–38911 (2015)PubMedPubMedCentral
130.
go back to reference S.M. Mangsbo, S. Broos, E. Fletcher, N. Veitonmaki, C. Furebring, E. Dahlen, P. Norlen, M. Lindstedt, T.H. Totterman, P. Ellmark, The human agonistic CD40 antibody ADC-1013 eradicates bladder tumors and generates T-cell-dependent tumor immunity. Clin Cancer Res 21, 1115–1126 (2015)CrossRefPubMed S.M. Mangsbo, S. Broos, E. Fletcher, N. Veitonmaki, C. Furebring, E. Dahlen, P. Norlen, M. Lindstedt, T.H. Totterman, P. Ellmark, The human agonistic CD40 antibody ADC-1013 eradicates bladder tumors and generates T-cell-dependent tumor immunity. Clin Cancer Res 21, 1115–1126 (2015)CrossRefPubMed
131.
go back to reference M. Chonan, R. Saito, T. Shoji, I. Shibahara, M. Kanamori, Y. Sonoda, M. Watanabe, T. Kikuchi, N. Ishii, T. Tominaga, CD40/CD40L expression correlates with the survival of patients with glioblastomas and an augmentation in CD40 signaling enhances the efficacy of vaccinations against glioma models. Neuro-Oncology 17, 1453–1462 (2015)CrossRefPubMedPubMedCentral M. Chonan, R. Saito, T. Shoji, I. Shibahara, M. Kanamori, Y. Sonoda, M. Watanabe, T. Kikuchi, N. Ishii, T. Tominaga, CD40/CD40L expression correlates with the survival of patients with glioblastomas and an augmentation in CD40 signaling enhances the efficacy of vaccinations against glioma models. Neuro-Oncology 17, 1453–1462 (2015)CrossRefPubMedPubMedCentral
132.
go back to reference L.V. Pham, A.T. Tamayo, L.C. Yoshimura, P. Lo, N. Terry, P.S. Reid, R.J. Ford, A CD40 signalosome anchored in lipid rafts leads to constitutive activation of NF-kappaB and autonomous cell growth in B cell lymphomas. Immunity 16, 37–50 (2002)CrossRefPubMed L.V. Pham, A.T. Tamayo, L.C. Yoshimura, P. Lo, N. Terry, P.S. Reid, R.J. Ford, A CD40 signalosome anchored in lipid rafts leads to constitutive activation of NF-kappaB and autonomous cell growth in B cell lymphomas. Immunity 16, 37–50 (2002)CrossRefPubMed
133.
go back to reference R. Tewari, S.R. Choudhury, V.S. Mehta, E. Sen, TNFalpha regulates the localization of CD40 in lipid rafts of glioma cells. Mol Biol Rep 39, 8695–8699 (2012)CrossRefPubMed R. Tewari, S.R. Choudhury, V.S. Mehta, E. Sen, TNFalpha regulates the localization of CD40 in lipid rafts of glioma cells. Mol Biol Rep 39, 8695–8699 (2012)CrossRefPubMed
134.
go back to reference H. Li, E.P. Nord, Functional caveolae are a prerequisite for CD40 signaling in human renal proximal tubule cells. Am J Physiol Renal Physiol 286, F711–F719 (2004)CrossRefPubMed H. Li, E.P. Nord, Functional caveolae are a prerequisite for CD40 signaling in human renal proximal tubule cells. Am J Physiol Renal Physiol 286, F711–F719 (2004)CrossRefPubMed
135.
136.
go back to reference J. Pouyssegur, F. Dayan, N.M. Mazure, Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441, 437–443 (2006)CrossRefPubMed J. Pouyssegur, F. Dayan, N.M. Mazure, Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441, 437–443 (2006)CrossRefPubMed
138.
go back to reference B. Kaur, F.W. Khwaja, E.A. Severson, S.L. Matheny, D.J. Brat, E.G. Van Meir, Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro-Oncology 7, 134–153 (2005)CrossRefPubMedPubMedCentral B. Kaur, F.W. Khwaja, E.A. Severson, S.L. Matheny, D.J. Brat, E.G. Van Meir, Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro-Oncology 7, 134–153 (2005)CrossRefPubMedPubMedCentral
139.
140.
141.
go back to reference J. Feng, Y. Zhang, X. She, Y. Sun, L. Fan, X. Ren, H. Fu, C. Liu, P. Li, C. Zhao, Q. Liu, Q. Liu, G. Li, M. Wu, Hypermethylated gene ANKDD1A is a candidate tumor suppressor that interacts with FIH1 and decreases HIF1alpha stability to inhibit cell autophagy in the glioblastoma multiforme hypoxia microenvironment. Oncogene 38, 103-119 (2019) J. Feng, Y. Zhang, X. She, Y. Sun, L. Fan, X. Ren, H. Fu, C. Liu, P. Li, C. Zhao, Q. Liu, Q. Liu, G. Li, M. Wu, Hypermethylated gene ANKDD1A is a candidate tumor suppressor that interacts with FIH1 and decreases HIF1alpha stability to inhibit cell autophagy in the glioblastoma multiforme hypoxia microenvironment. Oncogene 38, 103-119 (2019)
142.
go back to reference J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves, W.T. Curry Jr., B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 10, 1470–1476 (2008)CrossRefPubMedPubMedCentral J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves, W.T. Curry Jr., B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 10, 1470–1476 (2008)CrossRefPubMedPubMedCentral
143.
go back to reference F. Lan, Q. Qing, Q. Pan, M. Hu, H. Yu, X. Yue, Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol 41, 25–33 (2018)CrossRef F. Lan, Q. Qing, Q. Pan, M. Hu, H. Yu, X. Yue, Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol 41, 25–33 (2018)CrossRef
144.
go back to reference H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9, 654–659 (2007)CrossRefPubMed H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J.J. Lee, J.O. Lotvall, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9, 654–659 (2007)CrossRefPubMed
145.
go back to reference H.W. King, M.Z. Michael, J.M. Gleadle, Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 12, 421 (2012) H.W. King, M.Z. Michael, J.M. Gleadle, Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 12, 421 (2012)
146.
go back to reference A. Kuett, C. Rieger, D. Perathoner, T. Herold, M. Wagner, S. Sironi, K. Sotlar, H.P. Horny, C. Deniffel, H. Drolle, M. Fiegl, IL-8 as mediator in the microenvironment-leukaemia network in acute myeloid leukaemia. Sci Rep 5, 18411 (2015) A. Kuett, C. Rieger, D. Perathoner, T. Herold, M. Wagner, S. Sironi, K. Sotlar, H.P. Horny, C. Deniffel, H. Drolle, M. Fiegl, IL-8 as mediator in the microenvironment-leukaemia network in acute myeloid leukaemia. Sci Rep 5, 18411 (2015)
147.
go back to reference A.K. Kozlowska, H.C. Tseng, K. Kaur, P. Topchyan, A. Inagaki, V.T. Bui, N. Kasahara, N. Cacalano, A. Jewett, Resistance to cytotoxicity and sustained release of interleukin-6 and interleukin-8 in the presence of decreased interferon-gamma after differentiation of glioblastoma by human natural killer cells. Cancer Immunol Immunother 65, 1085–1097 (2016)CrossRefPubMedPubMedCentral A.K. Kozlowska, H.C. Tseng, K. Kaur, P. Topchyan, A. Inagaki, V.T. Bui, N. Kasahara, N. Cacalano, A. Jewett, Resistance to cytotoxicity and sustained release of interleukin-6 and interleukin-8 in the presence of decreased interferon-gamma after differentiation of glioblastoma by human natural killer cells. Cancer Immunol Immunother 65, 1085–1097 (2016)CrossRefPubMedPubMedCentral
148.
go back to reference D.J. Brat, A.C. Bellail, E.G. Van Meir, The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncology 7, 122–133 (2005)CrossRefPubMedPubMedCentral D.J. Brat, A.C. Bellail, E.G. Van Meir, The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncology 7, 122–133 (2005)CrossRefPubMedPubMedCentral
149.
go back to reference Y. Wang, O. Roche, C. Xu, E.H. Moriyama, P. Heir, J. Chung, F.C. Roos, Y. Chen, G. Finak, M. Milosevic, B.C. Wilson, B.T. Teh, M. Park, M.S. Irwin, M. Ohh, Hypoxia promotes ligand-independent EGF receptor signaling via hypoxia-inducible factor-mediated upregulation of caveolin-1. Proc Natl Acad Sci U S A 109, 4892–4897 (2012)CrossRefPubMedPubMedCentral Y. Wang, O. Roche, C. Xu, E.H. Moriyama, P. Heir, J. Chung, F.C. Roos, Y. Chen, G. Finak, M. Milosevic, B.C. Wilson, B.T. Teh, M. Park, M.S. Irwin, M. Ohh, Hypoxia promotes ligand-independent EGF receptor signaling via hypoxia-inducible factor-mediated upregulation of caveolin-1. Proc Natl Acad Sci U S A 109, 4892–4897 (2012)CrossRefPubMedPubMedCentral
150.
go back to reference M. Logozzi, A. De Milito, L. Lugini, M. Borghi, L. Calabro, M. Spada, M. Perdicchio, M.L. Marino, C. Federici, E. Iessi, D. Brambilla, G. Venturi, F. Lozupone, M. Santinami, V. Huber, M. Maio, L. Rivoltini, S. Fais, High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One 4, e5219 (2009)CrossRefPubMedPubMedCentral M. Logozzi, A. De Milito, L. Lugini, M. Borghi, L. Calabro, M. Spada, M. Perdicchio, M.L. Marino, C. Federici, E. Iessi, D. Brambilla, G. Venturi, F. Lozupone, M. Santinami, V. Huber, M. Maio, L. Rivoltini, S. Fais, High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One 4, e5219 (2009)CrossRefPubMedPubMedCentral
151.
go back to reference F. Wei, C. Ma, T. Zhou, X. Dong, Q. Luo, L. Geng, L. Ding, Y. Zhang, L. Zhang, N. Li, Y. Li, Y. Liu, Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol Cancer 16, 132 (2017) F. Wei, C. Ma, T. Zhou, X. Dong, Q. Luo, L. Geng, L. Ding, Y. Zhang, L. Zhang, N. Li, Y. Li, Y. Liu, Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol Cancer 16, 132 (2017)
152.
153.
go back to reference M.J. van den Bent, A.A. Brandes, M.J. Taphoorn, J.M. Kros, M.C. Kouwenhoven, J.Y. Delattre, H.J. Bernsen, M. Frenay, C.C. Tijssen, W. Grisold, L. Sipos, R.H. Enting, P.J. French, W.N. Dinjens, C.J. Vecht, A. Allgeier, D. Lacombe, T. Gorlia, K. Hoang-Xuan, Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: Long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 31, 344–350 (2013)CrossRefPubMed M.J. van den Bent, A.A. Brandes, M.J. Taphoorn, J.M. Kros, M.C. Kouwenhoven, J.Y. Delattre, H.J. Bernsen, M. Frenay, C.C. Tijssen, W. Grisold, L. Sipos, R.H. Enting, P.J. French, W.N. Dinjens, C.J. Vecht, A. Allgeier, D. Lacombe, T. Gorlia, K. Hoang-Xuan, Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: Long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 31, 344–350 (2013)CrossRefPubMed
154.
go back to reference G. Cairncross, M. Wang, E. Shaw, R. Jenkins, D. Brachman, J. Buckner, K. Fink, L. Souhami, N. Laperriere, W. Curran, M. Mehta, Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: Long-term results of RTOG 9402. J Clin Oncol 31, 337–343 (2013)CrossRefPubMed G. Cairncross, M. Wang, E. Shaw, R. Jenkins, D. Brachman, J. Buckner, K. Fink, L. Souhami, N. Laperriere, W. Curran, M. Mehta, Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: Long-term results of RTOG 9402. J Clin Oncol 31, 337–343 (2013)CrossRefPubMed
155.
go back to reference M. Kujas, J. Lejeune, A. Benouaich-Amiel, E. Criniere, F. Laigle-Donadey, Y. Marie, K. Mokhtari, M. Polivka, M. Bernier, F. Chretien, A. Couvelard, L. Capelle, H. Duffau, P. Cornu, P. Broet, J. Thillet, A.F. Carpentier, M. Sanson, K. Hoang-Xuan, J.Y. Delattre, Chromosome 1p loss: A favorable prognostic factor in low-grade gliomas. Ann Neurol 58, 322–326 (2005)CrossRefPubMed M. Kujas, J. Lejeune, A. Benouaich-Amiel, E. Criniere, F. Laigle-Donadey, Y. Marie, K. Mokhtari, M. Polivka, M. Bernier, F. Chretien, A. Couvelard, L. Capelle, H. Duffau, P. Cornu, P. Broet, J. Thillet, A.F. Carpentier, M. Sanson, K. Hoang-Xuan, J.Y. Delattre, Chromosome 1p loss: A favorable prognostic factor in low-grade gliomas. Ann Neurol 58, 322–326 (2005)CrossRefPubMed
156.
go back to reference L. Mariani, G. Deiana, E. Vassella, A.R. Fathi, C. Murtin, M. Arnold, I. Vajtai, J. Weis, P. Siegenthaler, M. Schobesberger, M.M. Reinert, Loss of heterozygosity 1p36 and 19q13 is a prognostic factor for overall survival in patients with diffuse WHO grade 2 gliomas treated without chemotherapy. J Clin Oncol 24, 4758–4763 (2006)CrossRefPubMed L. Mariani, G. Deiana, E. Vassella, A.R. Fathi, C. Murtin, M. Arnold, I. Vajtai, J. Weis, P. Siegenthaler, M. Schobesberger, M.M. Reinert, Loss of heterozygosity 1p36 and 19q13 is a prognostic factor for overall survival in patients with diffuse WHO grade 2 gliomas treated without chemotherapy. J Clin Oncol 24, 4758–4763 (2006)CrossRefPubMed
157.
go back to reference D.N. Louis and International Agency for Research on Cancer, WHO classification of tumours of the central nervous system, Revised 4th edition. Edn. (international agency for research on Cancer, Lyon, 2016) D.N. Louis and International Agency for Research on Cancer, WHO classification of tumours of the central nervous system, Revised 4th edition. Edn. (international agency for research on Cancer, Lyon, 2016)
158.
go back to reference T.S. Armstrong, E. Vera-Bolanos, B.N. Bekele, K. Aldape, M.R. Gilbert, Adult ependymal tumors: Prognosis and the M. D. Anderson Cancer center experience. Neuro-Oncology 12, 862–870 (2010)CrossRefPubMedPubMedCentral T.S. Armstrong, E. Vera-Bolanos, B.N. Bekele, K. Aldape, M.R. Gilbert, Adult ependymal tumors: Prognosis and the M. D. Anderson Cancer center experience. Neuro-Oncology 12, 862–870 (2010)CrossRefPubMedPubMedCentral
159.
160.
go back to reference E. Vera-Bolanos, K. Aldape, Y. Yuan, J. Wu, K. Wani, M.J. Necesito-Reyes, H. Colman, G. Dhall, F.S. Lieberman, P. Metellus, T. Mikkelsen, A. Omuro, S. Partap, M. Prados, H.I. Robins, R. Soffietti, J. Wu, M.R. Gilbert, T.S. Armstrong, C. Foundation, Clinical course and progression-free survival of adult intracranial and spinal ependymoma patients. Neuro-Oncology 17, 440–447 (2015)CrossRefPubMed E. Vera-Bolanos, K. Aldape, Y. Yuan, J. Wu, K. Wani, M.J. Necesito-Reyes, H. Colman, G. Dhall, F.S. Lieberman, P. Metellus, T. Mikkelsen, A. Omuro, S. Partap, M. Prados, H.I. Robins, R. Soffietti, J. Wu, M.R. Gilbert, T.S. Armstrong, C. Foundation, Clinical course and progression-free survival of adult intracranial and spinal ependymoma patients. Neuro-Oncology 17, 440–447 (2015)CrossRefPubMed
161.
go back to reference F. Mendrzyk, A. Korshunov, A. Benner, G. Toedt, S. Pfister, B. Radlwimmer, P. Lichter, Identification of gains on 1q and epidermal growth factor receptor overexpression as independent prognostic markers in intracranial ependymoma. Clin Cancer Res 12, 2070–2079 (2006)CrossRefPubMed F. Mendrzyk, A. Korshunov, A. Benner, G. Toedt, S. Pfister, B. Radlwimmer, P. Lichter, Identification of gains on 1q and epidermal growth factor receptor overexpression as independent prognostic markers in intracranial ependymoma. Clin Cancer Res 12, 2070–2079 (2006)CrossRefPubMed
162.
go back to reference R. Ferraldeschi, A. Latif, R.B. Clarke, K. Spence, G. Ashton, J. O'Sullivan, D.G. Evans, A. Howell, W.G. Newman, Lack of caveolin-1 (P132L) somatic mutations in breast cancer. Breast Cancer Res Treat 132, 1185–1186 (2012)CrossRefPubMed R. Ferraldeschi, A. Latif, R.B. Clarke, K. Spence, G. Ashton, J. O'Sullivan, D.G. Evans, A. Howell, W.G. Newman, Lack of caveolin-1 (P132L) somatic mutations in breast cancer. Breast Cancer Res Treat 132, 1185–1186 (2012)CrossRefPubMed
163.
go back to reference S. Koike, Y. Kodera, A. Nakao, H. Iwata, Y. Yatabe, Absence of the caveolin-1 P132L mutation in cancers of the breast and other organs. J Mol Diagn 12, 712–717 (2010)CrossRefPubMedPubMedCentral S. Koike, Y. Kodera, A. Nakao, H. Iwata, Y. Yatabe, Absence of the caveolin-1 P132L mutation in cancers of the breast and other organs. J Mol Diagn 12, 712–717 (2010)CrossRefPubMedPubMedCentral
164.
go back to reference G. Bonuccelli, M.C. Casimiro, F. Sotgia, C. Wang, M. Liu, S. Katiyar, J. Zhou, E. Dew, F. Capozza, K.M. Daumer, C. Minetti, J.N. Milliman, F. Alpy, M.C. Rio, C. Tomasetto, I. Mercier, N. Flomenberg, P.G. Frank, R.G. Pestell, M.P. Lisanti, Caveolin-1 (P132L), a common breast cancer mutation, confers mammary cell invasiveness and defines a novel stem cell/metastasis-associated gene signature. Am J Pathol 174, 1650–1662 (2009)CrossRefPubMedPubMedCentral G. Bonuccelli, M.C. Casimiro, F. Sotgia, C. Wang, M. Liu, S. Katiyar, J. Zhou, E. Dew, F. Capozza, K.M. Daumer, C. Minetti, J.N. Milliman, F. Alpy, M.C. Rio, C. Tomasetto, I. Mercier, N. Flomenberg, P.G. Frank, R.G. Pestell, M.P. Lisanti, Caveolin-1 (P132L), a common breast cancer mutation, confers mammary cell invasiveness and defines a novel stem cell/metastasis-associated gene signature. Am J Pathol 174, 1650–1662 (2009)CrossRefPubMedPubMedCentral
165.
go back to reference J.N. Sarkaria, G.J. Kitange, C.D. James, R. Plummer, H. Calvert, M. Weller, W. Wick, Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res 14, 2900–2908 (2008)CrossRefPubMedPubMedCentral J.N. Sarkaria, G.J. Kitange, C.D. James, R. Plummer, H. Calvert, M. Weller, W. Wick, Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res 14, 2900–2908 (2008)CrossRefPubMedPubMedCentral
166.
go back to reference D. Matias, J. Balca-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosario, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, V. Moura-Neto, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell Oncol 40, 247–261 (2017)CrossRef D. Matias, J. Balca-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosario, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, V. Moura-Neto, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell Oncol 40, 247–261 (2017)CrossRef
167.
go back to reference K.E. Warren, Beyond the blood:Brain barrier: The importance of central nervous system (CNS) pharmacokinetics for the treatment of CNS tumors, including diffuse intrinsic pontine Glioma. Front Oncol 8, 239 (2018) K.E. Warren, Beyond the blood:Brain barrier: The importance of central nervous system (CNS) pharmacokinetics for the treatment of CNS tumors, including diffuse intrinsic pontine Glioma. Front Oncol 8, 239 (2018)
168.
go back to reference N.J. Abbott, A.A. Patabendige, D.E. Dolman, S.R. Yusof, D.J. Begley, Structure and function of the blood-brain barrier. Neurobiol Dis 37, 13–25 (2010)CrossRefPubMed N.J. Abbott, A.A. Patabendige, D.E. Dolman, S.R. Yusof, D.J. Begley, Structure and function of the blood-brain barrier. Neurobiol Dis 37, 13–25 (2010)CrossRefPubMed
169.
go back to reference D. Knowland, A. Arac, K.J. Sekiguchi, M. Hsu, S.E. Lutz, J. Perrino, G.K. Steinberg, B.A. Barres, A. Nimmerjahn, D. Agalliu, Stepwise recruitment of transcellular and paracellular pathways underlies blood-brain barrier breakdown in stroke. Neuron 82, 603–617 (2014)CrossRefPubMedPubMedCentral D. Knowland, A. Arac, K.J. Sekiguchi, M. Hsu, S.E. Lutz, J. Perrino, G.K. Steinberg, B.A. Barres, A. Nimmerjahn, D. Agalliu, Stepwise recruitment of transcellular and paracellular pathways underlies blood-brain barrier breakdown in stroke. Neuron 82, 603–617 (2014)CrossRefPubMedPubMedCentral
170.
go back to reference J.A. Siegenthaler, F. Sohet, R. Daneman, Sealing off the CNS': Cellular and molecular regulation of blood-brain barriergenesis. Curr Opin Neurobiol 23, 1057–1064 (2013)CrossRefPubMedPubMedCentral J.A. Siegenthaler, F. Sohet, R. Daneman, Sealing off the CNS': Cellular and molecular regulation of blood-brain barriergenesis. Curr Opin Neurobiol 23, 1057–1064 (2013)CrossRefPubMedPubMedCentral
171.
go back to reference A. Idbaih, F. Ducray, M. Sierra Del Rio, K. Hoang-Xuan, J.Y. Delattre, Therapeutic application of noncytotoxic molecular targeted therapy in gliomas: Growth factor receptors and angiogenesis inhibitors. Oncologist 13, 978–992 (2008)CrossRefPubMed A. Idbaih, F. Ducray, M. Sierra Del Rio, K. Hoang-Xuan, J.Y. Delattre, Therapeutic application of noncytotoxic molecular targeted therapy in gliomas: Growth factor receptors and angiogenesis inhibitors. Oncologist 13, 978–992 (2008)CrossRefPubMed
172.
go back to reference V. Laquintana, A. Trapani, N. Denora, F. Wang, J.M. Gallo, G. Trapani, New strategies to deliver anticancer drugs to brain tumors. Expert Opin Drug Deliv 6, 1017–1032 (2009)CrossRefPubMedPubMedCentral V. Laquintana, A. Trapani, N. Denora, F. Wang, J.M. Gallo, G. Trapani, New strategies to deliver anticancer drugs to brain tumors. Expert Opin Drug Deliv 6, 1017–1032 (2009)CrossRefPubMedPubMedCentral
173.
go back to reference A. Armulik, G. Genove, M. Mae, M.H. Nisancioglu, E. Wallgard, C. Niaudet, L. He, J. Norlin, P. Lindblom, K. Strittmatter, B.R. Johansson, C. Betsholtz, Pericytes regulate the blood-brain barrier. Nature 468, 557–561 (2010)CrossRefPubMed A. Armulik, G. Genove, M. Mae, M.H. Nisancioglu, E. Wallgard, C. Niaudet, L. He, J. Norlin, P. Lindblom, K. Strittmatter, B.R. Johansson, C. Betsholtz, Pericytes regulate the blood-brain barrier. Nature 468, 557–561 (2010)CrossRefPubMed
174.
go back to reference A. Ben-Zvi, B. Lacoste, E. Kur, B.J. Andreone, Y. Mayshar, H. Yan, C. Gu, Mfsd2a is critical for the formation and function of the blood-brain barrier. Nature 509, 507–511 (2014)CrossRefPubMedPubMedCentral A. Ben-Zvi, B. Lacoste, E. Kur, B.J. Andreone, Y. Mayshar, H. Yan, C. Gu, Mfsd2a is critical for the formation and function of the blood-brain barrier. Nature 509, 507–511 (2014)CrossRefPubMedPubMedCentral
175.
go back to reference P. Eser Ocak, U. Ocak, P. Sherchan, J.H. Zhang, J. Tang, Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res (2018). https://doi.org/10.1002/jnr.24327 P. Eser Ocak, U. Ocak, P. Sherchan, J.H. Zhang, J. Tang, Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res (2018). https://​doi.​org/​10.​1002/​jnr.​24327
176.
go back to reference N. McDannold, N. Vykhodtseva, K. Hynynen, Targeted disruption of the blood-brain barrier with focused ultrasound: Association with cavitation activity. Phys Med Biol 51, 793–807 (2006)CrossRefPubMed N. McDannold, N. Vykhodtseva, K. Hynynen, Targeted disruption of the blood-brain barrier with focused ultrasound: Association with cavitation activity. Phys Med Biol 51, 793–807 (2006)CrossRefPubMed
177.
go back to reference K. Hynynen, N. McDannold, N. Vykhodtseva, S. Raymond, R. Weissleder, F.A. Jolesz, N. Sheikov, Focal disruption of the blood-brain barrier due to 260-kHz ultrasound bursts: A method for molecular imaging and targeted drug delivery. J Neurosurg 105, 445–454 (2006)CrossRefPubMed K. Hynynen, N. McDannold, N. Vykhodtseva, S. Raymond, R. Weissleder, F.A. Jolesz, N. Sheikov, Focal disruption of the blood-brain barrier due to 260-kHz ultrasound bursts: A method for molecular imaging and targeted drug delivery. J Neurosurg 105, 445–454 (2006)CrossRefPubMed
178.
go back to reference C.Y. Xia, Y.H. Liu, P. Wang, Y.X. Xue, Low-frequency ultrasound irradiation increases blood-tumor barrier permeability by transcellular pathway in a rat glioma model. J Mol Neurosci 48, 281–290 (2012)CrossRefPubMed C.Y. Xia, Y.H. Liu, P. Wang, Y.X. Xue, Low-frequency ultrasound irradiation increases blood-tumor barrier permeability by transcellular pathway in a rat glioma model. J Mol Neurosci 48, 281–290 (2012)CrossRefPubMed
179.
go back to reference T. Aoki, R. Nomura, T. Fujimoto, Tyrosine phosphorylation of caveolin-1 in the endothelium. Exp Cell Res 253, 629–636 (1999)CrossRefPubMed T. Aoki, R. Nomura, T. Fujimoto, Tyrosine phosphorylation of caveolin-1 in the endothelium. Exp Cell Res 253, 629–636 (1999)CrossRefPubMed
180.
go back to reference T. Inamura, K.L. Black, Bradykinin selectively opens blood-tumor barrier in experimental brain tumors. J Cereb Blood Flow Metab 14, 862–870 (1994)CrossRefPubMed T. Inamura, K.L. Black, Bradykinin selectively opens blood-tumor barrier in experimental brain tumors. J Cereb Blood Flow Metab 14, 862–870 (1994)CrossRefPubMed
181.
go back to reference L.B. Liu, Y.X. Xue, Y.H. Liu, Bradykinin increases the permeability of the blood-tumor barrier by the caveolae-mediated transcellular pathway. J Neuro-Oncol 99, 187–194 (2010)CrossRef L.B. Liu, Y.X. Xue, Y.H. Liu, Bradykinin increases the permeability of the blood-tumor barrier by the caveolae-mediated transcellular pathway. J Neuro-Oncol 99, 187–194 (2010)CrossRef
182.
go back to reference L.B. Liu, Y.X. Xue, Y.H. Liu, Y.B. Wang, Bradykinin increases blood-tumor barrier permeability by down-regulating the expression levels of ZO-1, occludin, and claudin-5 and rearranging actin cytoskeleton. J Neurosci Res 86, 1153–1168 (2008)CrossRefPubMed L.B. Liu, Y.X. Xue, Y.H. Liu, Y.B. Wang, Bradykinin increases blood-tumor barrier permeability by down-regulating the expression levels of ZO-1, occludin, and claudin-5 and rearranging actin cytoskeleton. J Neurosci Res 86, 1153–1168 (2008)CrossRefPubMed
183.
go back to reference C.Y. Xia, Z. Zhang, Y.X. Xue, P. Wang, Y.H. Liu, Mechanisms of the increase in the permeability of the blood-tumor barrier obtained by combining low-frequency ultrasound irradiation with small-dose bradykinin. J Neuro-Oncol 94, 41–50 (2009)CrossRef C.Y. Xia, Z. Zhang, Y.X. Xue, P. Wang, Y.H. Liu, Mechanisms of the increase in the permeability of the blood-tumor barrier obtained by combining low-frequency ultrasound irradiation with small-dose bradykinin. J Neuro-Oncol 94, 41–50 (2009)CrossRef
184.
go back to reference Y.T. Gu, Y.X. Xue, H. Zhang, Y. Li, X.Y. Liang, Adenosine 5′-triphosphate-sensitive potassium channel activator induces the up-regulation of caveolin-1 expression in a rat brain tumor model. Cell Mol Neurobiol 31, 629–634 (2011)CrossRefPubMed Y.T. Gu, Y.X. Xue, H. Zhang, Y. Li, X.Y. Liang, Adenosine 5′-triphosphate-sensitive potassium channel activator induces the up-regulation of caveolin-1 expression in a rat brain tumor model. Cell Mol Neurobiol 31, 629–634 (2011)CrossRefPubMed
185.
go back to reference N.S. Ningaraj, U.T. Sankpal, D. Khaitan, E.A. Meister, T. Vats, Activation of KATP channels increases anticancer drug delivery to brain tumors and survival. Eur J Pharmacol 602, 188–193 (2009)CrossRefPubMed N.S. Ningaraj, U.T. Sankpal, D. Khaitan, E.A. Meister, T. Vats, Activation of KATP channels increases anticancer drug delivery to brain tumors and survival. Eur J Pharmacol 602, 188–193 (2009)CrossRefPubMed
186.
go back to reference A.P. van den Heuvel, A. Schulze, B.M. Burgering, Direct control of caveolin-1 expression by FOXO transcription factors. Biochem J 385, 795–802 (2005)CrossRefPubMedPubMedCentral A.P. van den Heuvel, A. Schulze, B.M. Burgering, Direct control of caveolin-1 expression by FOXO transcription factors. Biochem J 385, 795–802 (2005)CrossRefPubMedPubMedCentral
187.
go back to reference R.P. Cai, Y.X. Xue, J. Huang, J.H. Wang, J.H. Wang, S.Y. Zhao, T.T. Guan, Z. Zhang, Y.T. Gu, NS1619 regulates the expression of caveolin-1 protein in a time-dependent manner via ROS/PI3K/PKB/FoxO1 signaling pathway in brain tumor microvascular endothelial cells. J Neurol Sci 369, 109–118 (2016)CrossRefPubMed R.P. Cai, Y.X. Xue, J. Huang, J.H. Wang, J.H. Wang, S.Y. Zhao, T.T. Guan, Z. Zhang, Y.T. Gu, NS1619 regulates the expression of caveolin-1 protein in a time-dependent manner via ROS/PI3K/PKB/FoxO1 signaling pathway in brain tumor microvascular endothelial cells. J Neurol Sci 369, 109–118 (2016)CrossRefPubMed
188.
go back to reference T.A. Rege, C.Y. Fears, C.L. Gladson, Endogenous inhibitors of angiogenesis in malignant gliomas: nature's antiangiogenic therapy. Neuro-Oncology 7, 106–121 (2005)CrossRefPubMedPubMedCentral T.A. Rege, C.Y. Fears, C.L. Gladson, Endogenous inhibitors of angiogenesis in malignant gliomas: nature's antiangiogenic therapy. Neuro-Oncology 7, 106–121 (2005)CrossRefPubMedPubMedCentral
189.
go back to reference M.R. Gilbert, J.J. Dignam, T.S. Armstrong, J.S. Wefel, D.T. Blumenthal, M.A. Vogelbaum, H. Colman, A. Chakravarti, S. Pugh, M. Won, R. Jeraj, P.D. Brown, K.A. Jaeckle, D. Schiff, V.W. Stieber, D.G. Brachman, M. Werner-Wasik, I.W. Tremont-Lukats, E.P. Sulman, K.D. Aldape, W.J. Curran Jr., M.P. Mehta, A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370, 699–708 (2014)CrossRefPubMedPubMedCentral M.R. Gilbert, J.J. Dignam, T.S. Armstrong, J.S. Wefel, D.T. Blumenthal, M.A. Vogelbaum, H. Colman, A. Chakravarti, S. Pugh, M. Won, R. Jeraj, P.D. Brown, K.A. Jaeckle, D. Schiff, V.W. Stieber, D.G. Brachman, M. Werner-Wasik, I.W. Tremont-Lukats, E.P. Sulman, K.D. Aldape, W.J. Curran Jr., M.P. Mehta, A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370, 699–708 (2014)CrossRefPubMedPubMedCentral
190.
go back to reference O.L. Chinot, W. Wick, W. Mason, R. Henriksson, F. Saran, R. Nishikawa, A.F. Carpentier, K. Hoang-Xuan, P. Kavan, D. Cernea, A.A. Brandes, M. Hilton, L. Abrey, T. Cloughesy, Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370, 709–722 (2014)CrossRefPubMed O.L. Chinot, W. Wick, W. Mason, R. Henriksson, F. Saran, R. Nishikawa, A.F. Carpentier, K. Hoang-Xuan, P. Kavan, D. Cernea, A.A. Brandes, M. Hilton, L. Abrey, T. Cloughesy, Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370, 709–722 (2014)CrossRefPubMed
191.
go back to reference L.N. Zhao, Z.H. Yang, Y.H. Liu, H.Q. Ying, H. Zhang, Y.X. Xue, Vascular endothelial growth factor increases permeability of the blood-tumor barrier via caveolae-mediated transcellular pathway. J Mol Neurosci 44, 122–129 (2011)CrossRefPubMed L.N. Zhao, Z.H. Yang, Y.H. Liu, H.Q. Ying, H. Zhang, Y.X. Xue, Vascular endothelial growth factor increases permeability of the blood-tumor barrier via caveolae-mediated transcellular pathway. J Mol Neurosci 44, 122–129 (2011)CrossRefPubMed
192.
go back to reference Z.H. Yang, L.B. Liu, L.N. Zhao, Y.H. Liu, Y.X. Xue, Permeability of the blood-tumor barrier is enhanced by combining vascular endothelial growth factor with papaverine. J Neurosci Res 92, 703–713 (2014)CrossRefPubMed Z.H. Yang, L.B. Liu, L.N. Zhao, Y.H. Liu, Y.X. Xue, Permeability of the blood-tumor barrier is enhanced by combining vascular endothelial growth factor with papaverine. J Neurosci Res 92, 703–713 (2014)CrossRefPubMed
193.
go back to reference A.C. Berger, H.R. Alexander, G. Tang, P.S. Wu, S.M. Hewitt, E. Turner, E. Kruger, W.D. Figg, A. Grove, E. Kohn, D. Stern, S.K. Libutti, Endothelial monocyte activating polypeptide II induces endothelial cell apoptosis and may inhibit tumor angiogenesis. Microvasc Res 60, 70–80 (2000)CrossRefPubMed A.C. Berger, H.R. Alexander, G. Tang, P.S. Wu, S.M. Hewitt, E. Turner, E. Kruger, W.D. Figg, A. Grove, E. Kohn, D. Stern, S.K. Libutti, Endothelial monocyte activating polypeptide II induces endothelial cell apoptosis and may inhibit tumor angiogenesis. Microvasc Res 60, 70–80 (2000)CrossRefPubMed
194.
go back to reference Z. Li, Y.H. Liu, Y.X. Xue, L.B. Liu, P. Wang, Low-dose endothelial monocyte-activating polypeptide-ii increases permeability of blood-tumor barrier by caveolae-mediated transcellular pathway. J Mol Neurosci 52, 313–322 (2014)CrossRefPubMed Z. Li, Y.H. Liu, Y.X. Xue, L.B. Liu, P. Wang, Low-dose endothelial monocyte-activating polypeptide-ii increases permeability of blood-tumor barrier by caveolae-mediated transcellular pathway. J Mol Neurosci 52, 313–322 (2014)CrossRefPubMed
195.
go back to reference H. Xie, Y.X. Xue, L.B. Liu, Y.H. Liu, Endothelial-monocyte-activating polypeptide II increases blood-tumor barrier permeability by down-regulating the expression levels of tight junction associated proteins. Brain Res 1319, 13–20 (2010)CrossRefPubMed H. Xie, Y.X. Xue, L.B. Liu, Y.H. Liu, Endothelial-monocyte-activating polypeptide II increases blood-tumor barrier permeability by down-regulating the expression levels of tight junction associated proteins. Brain Res 1319, 13–20 (2010)CrossRefPubMed
196.
go back to reference L. Chen, Y. Xue, J. Zheng, X. Liu, J. Liu, J. Chen, Z. Li, Z. Xi, H. Teng, P. Wang, L. Liu, Y. Liu, MiR-429 regulated by endothelial monocyte activating polypeptide-II (EMAP-II) influences blood-tumor barrier permeability by inhibiting the expressions of ZO-1, Occludin and Claudin-5. Front Mol Neurosci 11(35) (2018) L. Chen, Y. Xue, J. Zheng, X. Liu, J. Liu, J. Chen, Z. Li, Z. Xi, H. Teng, P. Wang, L. Liu, Y. Liu, MiR-429 regulated by endothelial monocyte activating polypeptide-II (EMAP-II) influences blood-tumor barrier permeability by inhibiting the expressions of ZO-1, Occludin and Claudin-5. Front Mol Neurosci 11(35) (2018)
197.
go back to reference Y. Lin, P. Wang, Y.H. Liu, X.L. Shang, L.Y. Chen, Y.X. Xue, DT(270-326) , a truncated diphtheria toxin, increases blood-tumor barrier permeability by upregulating the expression of Caveolin-1. CNS Neurosci Ther 22, 477–487 (2016)CrossRefPubMedPubMedCentral Y. Lin, P. Wang, Y.H. Liu, X.L. Shang, L.Y. Chen, Y.X. Xue, DT(270-326) , a truncated diphtheria toxin, increases blood-tumor barrier permeability by upregulating the expression of Caveolin-1. CNS Neurosci Ther 22, 477–487 (2016)CrossRefPubMedPubMedCentral
198.
go back to reference N.R. Parker, N. Correia, B. Crossley, M.E. Buckland, V.M. Howell, H.R. Wheeler, Correlation of MicroRNA 132 up-regulation with an unfavorable clinical outcome in patients with primary glioblastoma Multiforme treated with radiotherapy plus concomitant and adjuvant Temozolomide chemotherapy. Transl Oncol 6, 742–748 (2013)CrossRefPubMedPubMedCentral N.R. Parker, N. Correia, B. Crossley, M.E. Buckland, V.M. Howell, H.R. Wheeler, Correlation of MicroRNA 132 up-regulation with an unfavorable clinical outcome in patients with primary glioblastoma Multiforme treated with radiotherapy plus concomitant and adjuvant Temozolomide chemotherapy. Transl Oncol 6, 742–748 (2013)CrossRefPubMedPubMedCentral
199.
go back to reference N. Stojcheva, G. Schechtmann, S. Sass, P. Roth, A.M. Florea, A. Stefanski, K. Stuhler, M. Wolter, N.S. Muller, F.J. Theis, M. Weller, G. Reifenberger, C. Happold, MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM. Oncotarget 7, 12937–12950 (2016)CrossRefPubMedPubMedCentral N. Stojcheva, G. Schechtmann, S. Sass, P. Roth, A.M. Florea, A. Stefanski, K. Stuhler, M. Wolter, N.S. Muller, F.J. Theis, M. Weller, G. Reifenberger, C. Happold, MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM. Oncotarget 7, 12937–12950 (2016)CrossRefPubMedPubMedCentral
200.
go back to reference G.S. Markopoulos, E. Roupakia, M. Tokamani, E. Chavdoula, M. Hatziapostolou, C. Polytarchou, K.B. Marcu, A.G. Papavassiliou, R. Sandaltzopoulos, E. Kolettas, A step-by-step microRNA guide to cancer development and metastasis. Cell Oncol 40, 303–339 (2017)CrossRef G.S. Markopoulos, E. Roupakia, M. Tokamani, E. Chavdoula, M. Hatziapostolou, C. Polytarchou, K.B. Marcu, A.G. Papavassiliou, R. Sandaltzopoulos, E. Kolettas, A step-by-step microRNA guide to cancer development and metastasis. Cell Oncol 40, 303–339 (2017)CrossRef
201.
go back to reference Y. Gu, R. Cai, C. Zhang, Y. Xue, Y. Pan, J. Wang, Z. Zhang, miR-132-3p boosts caveolae-mediated transcellular transport in glioma endothelial cells by targeting PTEN/PI3K/PKB/Src/Cav-1 signaling pathway. FASEB J 33, 441-454 (2019) Y. Gu, R. Cai, C. Zhang, Y. Xue, Y. Pan, J. Wang, Z. Zhang, miR-132-3p boosts caveolae-mediated transcellular transport in glioma endothelial cells by targeting PTEN/PI3K/PKB/Src/Cav-1 signaling pathway. FASEB J 33, 441-454 (2019)
202.
go back to reference Y. Li, L.B. Liu, T. Ma, P. Wang, Y.X. Xue, Effect of caveolin-1 on the expression of tight junction-associated proteins in rat glioma-derived microvascular endothelial cells. Int J Clin Exp Pathol 8, 13067–13074 (2015)PubMedPubMedCentral Y. Li, L.B. Liu, T. Ma, P. Wang, Y.X. Xue, Effect of caveolin-1 on the expression of tight junction-associated proteins in rat glioma-derived microvascular endothelial cells. Int J Clin Exp Pathol 8, 13067–13074 (2015)PubMedPubMedCentral
203.
go back to reference N. Strazielle, J.F. Ghersi-Egea, Potential pathways for CNS drug delivery across the blood-cerebrospinal fluid barrier. Curr Pharm Des 22, 5463–5476 (2016)CrossRefPubMedPubMedCentral N. Strazielle, J.F. Ghersi-Egea, Potential pathways for CNS drug delivery across the blood-cerebrospinal fluid barrier. Curr Pharm Des 22, 5463–5476 (2016)CrossRefPubMedPubMedCentral
204.
go back to reference S. Yip, J. Miao, D.P. Cahill, A.J. Iafrate, K. Aldape, C.L. Nutt, D.N. Louis, MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin Cancer Res 15, 4622–4629 (2009)CrossRefPubMedPubMedCentral S. Yip, J. Miao, D.P. Cahill, A.J. Iafrate, K. Aldape, C.L. Nutt, D.N. Louis, MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin Cancer Res 15, 4622–4629 (2009)CrossRefPubMedPubMedCentral
205.
go back to reference K. Ujifuku, N. Mitsutake, S. Takakura, M. Matsuse, V. Saenko, K. Suzuki, K. Hayashi, T. Matsuo, K. Kamada, I. Nagata, S. Yamashita, miR-195, miR-455-3p and miR-10a( *) are implicated in acquired temozolomide resistance in glioblastoma multiforme cells. Cancer Lett 296, 241–248 (2010)CrossRefPubMed K. Ujifuku, N. Mitsutake, S. Takakura, M. Matsuse, V. Saenko, K. Suzuki, K. Hayashi, T. Matsuo, K. Kamada, I. Nagata, S. Yamashita, miR-195, miR-455-3p and miR-10a( *) are implicated in acquired temozolomide resistance in glioblastoma multiforme cells. Cancer Lett 296, 241–248 (2010)CrossRefPubMed
206.
go back to reference C. Bruyere, L. Abeloos, D. Lamoral-Theys, R. Senetta, V. Mathieu, M. Le Mercier, R.E. Kast, P. Cassoni, G. Vandenbussche, R. Kiss, F. Lefranc, Temozolomide modifies caveolin-1 expression in experimental malignant gliomas in vitro and in vivo. Transl Oncol 4, 92–100 (2011)CrossRefPubMedPubMedCentral C. Bruyere, L. Abeloos, D. Lamoral-Theys, R. Senetta, V. Mathieu, M. Le Mercier, R.E. Kast, P. Cassoni, G. Vandenbussche, R. Kiss, F. Lefranc, Temozolomide modifies caveolin-1 expression in experimental malignant gliomas in vitro and in vivo. Transl Oncol 4, 92–100 (2011)CrossRefPubMedPubMedCentral
207.
go back to reference K. Quann, D.M. Gonzales, I. Mercier, C. Wang, F. Sotgia, R.G. Pestell, M.P. Lisanti, J.F. Jasmin, Caveolin-1 is a negative regulator of tumor growth in glioblastoma and modulates chemosensitivity to temozolomide. Cell Cycle 12, 1510–1520 (2013)CrossRefPubMedPubMedCentral K. Quann, D.M. Gonzales, I. Mercier, C. Wang, F. Sotgia, R.G. Pestell, M.P. Lisanti, J.F. Jasmin, Caveolin-1 is a negative regulator of tumor growth in glioblastoma and modulates chemosensitivity to temozolomide. Cell Cycle 12, 1510–1520 (2013)CrossRefPubMedPubMedCentral
208.
go back to reference C.A. DI, G. Carrabba, G. Lanfranchi, C. Menghetti, P. Rampini, M. Caroli, Continuous tamoxifen and dose-dense temozolomide in recurrent glioblastoma. Anticancer Res 33, 3383–3389 (2013) C.A. DI, G. Carrabba, G. Lanfranchi, C. Menghetti, P. Rampini, M. Caroli, Continuous tamoxifen and dose-dense temozolomide in recurrent glioblastoma. Anticancer Res 33, 3383–3389 (2013)
209.
go back to reference A.M. Hui, W. Zhang, W. Chen, D. Xi, B. Purow, G.C. Friedman, H.A. Fine, Agents with selective estrogen receptor (ER) modulator activity induce apoptosis in vitro and in vivo in ER-negative glioma cells. Cancer Res 64, 9115–9123 (2004)CrossRefPubMed A.M. Hui, W. Zhang, W. Chen, D. Xi, B. Purow, G.C. Friedman, H.A. Fine, Agents with selective estrogen receptor (ER) modulator activity induce apoptosis in vitro and in vivo in ER-negative glioma cells. Cancer Res 64, 9115–9123 (2004)CrossRefPubMed
210.
211.
go back to reference Z. Wang, X. Zhang, P. Shen, B.W. Loggie, Y. Chang, T.F. Deuel, Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66. Biochem Biophys Res Commun 336, 1023–1027 (2005)CrossRefPubMed Z. Wang, X. Zhang, P. Shen, B.W. Loggie, Y. Chang, T.F. Deuel, Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66. Biochem Biophys Res Commun 336, 1023–1027 (2005)CrossRefPubMed
212.
go back to reference L. Shi, B. Dong, Z. Li, Y. Lu, T. Ouyang, J. Li, T. Wang, Z. Fan, T. Fan, B. Lin, Z. Wang, Y. Xie, Expression of ER-{alpha}36, a novel variant of estrogen receptor {alpha}, and resistance to tamoxifen treatment in breast cancer. J Clin Oncol 27, 3423–3429 (2009)CrossRefPubMedPubMedCentral L. Shi, B. Dong, Z. Li, Y. Lu, T. Ouyang, J. Li, T. Wang, Z. Fan, T. Fan, B. Lin, Z. Wang, Y. Xie, Expression of ER-{alpha}36, a novel variant of estrogen receptor {alpha}, and resistance to tamoxifen treatment in breast cancer. J Clin Oncol 27, 3423–3429 (2009)CrossRefPubMedPubMedCentral
213.
go back to reference Y. Liu, L. Huang, X. Guan, H. Li, Q.Q. Zhang, C. Han, Y.J. Wang, C. Wang, Y. Zhang, C. Qu, J. Liu, W. Zou, ER-alpha36, a novel variant of ERalpha, is involved in the regulation of tamoxifen-sensitivity of glioblastoma cells. Steroids 111, 127–133 (2016)CrossRefPubMed Y. Liu, L. Huang, X. Guan, H. Li, Q.Q. Zhang, C. Han, Y.J. Wang, C. Wang, Y. Zhang, C. Qu, J. Liu, W. Zou, ER-alpha36, a novel variant of ERalpha, is involved in the regulation of tamoxifen-sensitivity of glioblastoma cells. Steroids 111, 127–133 (2016)CrossRefPubMed
214.
go back to reference J.H. Lin, M. Yamazaki, Role of P-glycoprotein in pharmacokinetics: Clinical implications. Clin Pharmacokinet 42, 59–98 (2003)CrossRefPubMed J.H. Lin, M. Yamazaki, Role of P-glycoprotein in pharmacokinetics: Clinical implications. Clin Pharmacokinet 42, 59–98 (2003)CrossRefPubMed
215.
go back to reference J. Jodoin, M. Demeule, L. Fenart, R. Cecchelli, S. Farmer, K.J. Linton, C.F. Higgins, R. Beliveau, P-glycoprotein in blood-brain barrier endothelial cells: Interaction and oligomerization with caveolins. J Neurochem 87, 1010–1023 (2003)CrossRefPubMed J. Jodoin, M. Demeule, L. Fenart, R. Cecchelli, S. Farmer, K.J. Linton, C.F. Higgins, R. Beliveau, P-glycoprotein in blood-brain barrier endothelial cells: Interaction and oligomerization with caveolins. J Neurochem 87, 1010–1023 (2003)CrossRefPubMed
216.
go back to reference P.L. Golden, W.M. Pardridge, P-glycoprotein on astrocyte foot processes of unfixed isolated human brain capillaries. Brain Res 819, 143–146 (1999)CrossRefPubMed P.L. Golden, W.M. Pardridge, P-glycoprotein on astrocyte foot processes of unfixed isolated human brain capillaries. Brain Res 819, 143–146 (1999)CrossRefPubMed
217.
go back to reference Y. Zhang, S.X. Wang, J.W. Ma, H.Y. Li, J.C. Ye, S.M. Xie, B. Du, X.Y. Zhong, EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition. J Neuro-Oncol 121, 41–52 (2015)CrossRef Y. Zhang, S.X. Wang, J.W. Ma, H.Y. Li, J.C. Ye, S.M. Xie, B. Du, X.Y. Zhong, EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition. J Neuro-Oncol 121, 41–52 (2015)CrossRef
218.
go back to reference J.L. Munoz, N.D. Walker, K.W. Scotto, P. Rameshwar, Temozolomide competes for P-glycoprotein and contributes to chemoresistance in glioblastoma cells. Cancer Lett 367, 69–75 (2015)CrossRefPubMed J.L. Munoz, N.D. Walker, K.W. Scotto, P. Rameshwar, Temozolomide competes for P-glycoprotein and contributes to chemoresistance in glioblastoma cells. Cancer Lett 367, 69–75 (2015)CrossRefPubMed
219.
go back to reference P.T. Ronaldson, M. Bendayan, D. Gingras, M. Piquette-Miller, R. Bendayan, Cellular localization and functional expression of P-glycoprotein in rat astrocyte cultures. J Neurochem 89, 788–800 (2004)CrossRefPubMed P.T. Ronaldson, M. Bendayan, D. Gingras, M. Piquette-Miller, R. Bendayan, Cellular localization and functional expression of P-glycoprotein in rat astrocyte cultures. J Neurochem 89, 788–800 (2004)CrossRefPubMed
220.
go back to reference F. Schlachetzki, W.M. Pardridge, P-glycoprotein and caveolin-1alpha in endothelium and astrocytes of primate brain. Neuroreport 14, 2041–2046 (2003)CrossRefPubMed F. Schlachetzki, W.M. Pardridge, P-glycoprotein and caveolin-1alpha in endothelium and astrocytes of primate brain. Neuroreport 14, 2041–2046 (2003)CrossRefPubMed
221.
go back to reference M.D. Walker, S.B. Green, D.P. Byar, E. Alexander Jr., U. Batzdorf, W.H. Brooks, W.E. Hunt, C.S. MacCarty, M.S. Mahaley Jr., J. Mealey Jr., G. Owens, J. Ransohoff 2nd, J.T. Robertson, W.R. Shapiro, K.R. Smith Jr., C.B. Wilson, T.A. Strike, Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med 303, 1323–1329 (1980)CrossRefPubMed M.D. Walker, S.B. Green, D.P. Byar, E. Alexander Jr., U. Batzdorf, W.H. Brooks, W.E. Hunt, C.S. MacCarty, M.S. Mahaley Jr., J. Mealey Jr., G. Owens, J. Ransohoff 2nd, J.T. Robertson, W.R. Shapiro, K.R. Smith Jr., C.B. Wilson, T.A. Strike, Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med 303, 1323–1329 (1980)CrossRefPubMed
222.
go back to reference J. Mahmood, S.R. Zaveri, S.C. Murti, A.A. Alexander, C.Q. Connors, H.D. Shukla, Z. Vujaskovic, Caveolin-1: A novel prognostic biomarker of radioresistance in cancer. Int J Radiat Biol 92, 747–753 (2016)CrossRefPubMed J. Mahmood, S.R. Zaveri, S.C. Murti, A.A. Alexander, C.Q. Connors, H.D. Shukla, Z. Vujaskovic, Caveolin-1: A novel prognostic biomarker of radioresistance in cancer. Int J Radiat Biol 92, 747–753 (2016)CrossRefPubMed
223.
go back to reference H. Zhu, J. Yue, Z. Pan, H. Wu, Y. Cheng, H. Lu, X. Ren, M. Yao, Z. Shen, J.M. Yang, Involvement of Caveolin-1 in repair of DNA damage through both homologous recombination and non-homologous end joining. PLoS One 5, e12055 (2010)CrossRefPubMedPubMedCentral H. Zhu, J. Yue, Z. Pan, H. Wu, Y. Cheng, H. Lu, X. Ren, M. Yao, Z. Shen, J.M. Yang, Involvement of Caveolin-1 in repair of DNA damage through both homologous recombination and non-homologous end joining. PLoS One 5, e12055 (2010)CrossRefPubMedPubMedCentral
224.
go back to reference N. McLaughlin, B. Annabi, M. Bouzeghrane, A. Temme, J.P. Bahary, R. Moumdjian, R. Beliveau, The Survivin-mediated radioresistant phenotype of glioblastomas is regulated by RhoA and inhibited by the green tea polyphenol (−)-epigallocatechin-3-gallate. Brain Res 1071, 1–9 (2006)CrossRefPubMed N. McLaughlin, B. Annabi, M. Bouzeghrane, A. Temme, J.P. Bahary, R. Moumdjian, R. Beliveau, The Survivin-mediated radioresistant phenotype of glioblastomas is regulated by RhoA and inhibited by the green tea polyphenol (−)-epigallocatechin-3-gallate. Brain Res 1071, 1–9 (2006)CrossRefPubMed
225.
go back to reference A. Chakravarti, G.G. Zhai, M. Zhang, R. Malhotra, D.E. Latham, M.A. Delaney, P. Robe, U. Nestler, Q. Song, J. Loeffler, Survivin enhances radiation resistance in primary human glioblastoma cells via caspase-independent mechanisms. Oncogene 23, 7494–7506 (2004)CrossRefPubMed A. Chakravarti, G.G. Zhai, M. Zhang, R. Malhotra, D.E. Latham, M.A. Delaney, P. Robe, U. Nestler, Q. Song, J. Loeffler, Survivin enhances radiation resistance in primary human glioblastoma cells via caspase-independent mechanisms. Oncogene 23, 7494–7506 (2004)CrossRefPubMed
226.
go back to reference P. Dahan, J. Martinez Gala, C. Delmas, S. Monferran, L. Malric, D. Zentkowski, V. Lubrano, C. Toulas, E. Cohen-Jonathan Moyal, A. Lemarie, Ionizing radiations sustain glioblastoma cell dedifferentiation to a stem-like phenotype through survivin: Possible involvement in radioresistance. Cell Death Dis 5, e1543 (2014)CrossRefPubMedPubMedCentral P. Dahan, J. Martinez Gala, C. Delmas, S. Monferran, L. Malric, D. Zentkowski, V. Lubrano, C. Toulas, E. Cohen-Jonathan Moyal, A. Lemarie, Ionizing radiations sustain glioblastoma cell dedifferentiation to a stem-like phenotype through survivin: Possible involvement in radioresistance. Cell Death Dis 5, e1543 (2014)CrossRefPubMedPubMedCentral
227.
go back to reference M.A. Forget, J.L. Voorhees, S.L. Cole, D. Dakhlallah, I.L. Patterson, A.C. Gross, L. Moldovan, X. Mo, R. Evans, C.B. Marsh, T.D. Eubank, Macrophage colony-stimulating factor augments Tie2-expressing monocyte differentiation, angiogenic function, and recruitment in a mouse model of breast cancer. PLoS One 9, e98623 (2014)CrossRefPubMedPubMedCentral M.A. Forget, J.L. Voorhees, S.L. Cole, D. Dakhlallah, I.L. Patterson, A.C. Gross, L. Moldovan, X. Mo, R. Evans, C.B. Marsh, T.D. Eubank, Macrophage colony-stimulating factor augments Tie2-expressing monocyte differentiation, angiogenic function, and recruitment in a mouse model of breast cancer. PLoS One 9, e98623 (2014)CrossRefPubMedPubMedCentral
228.
go back to reference A. Backen, A.G. Renehan, A.R. Clamp, C. Berzuini, C. Zhou, A. Oza, S. Bannoo, S.J. Scherer, R.E. Banks, C. Dive, G.C. Jayson, The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 20, 4549–4558 (2014)CrossRefPubMedPubMedCentral A. Backen, A.G. Renehan, A.R. Clamp, C. Berzuini, C. Zhou, A. Oza, S. Bannoo, S.J. Scherer, R.E. Banks, C. Dive, G.C. Jayson, The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 20, 4549–4558 (2014)CrossRefPubMedPubMedCentral
229.
go back to reference Y. Piao, S.Y. Park, V. Henry, B.D. Smith, N. Tiao, D.L. Flynn, J.F. de Groot, Novel MET/TIE2/VEGFR2 inhibitor altiratinib inhibits tumor growth and invasiveness in bevacizumab-resistant glioblastoma mouse models. Neuro-Oncology 18, 1230–1241 (2016)CrossRefPubMedPubMedCentral Y. Piao, S.Y. Park, V. Henry, B.D. Smith, N. Tiao, D.L. Flynn, J.F. de Groot, Novel MET/TIE2/VEGFR2 inhibitor altiratinib inhibits tumor growth and invasiveness in bevacizumab-resistant glioblastoma mouse models. Neuro-Oncology 18, 1230–1241 (2016)CrossRefPubMedPubMedCentral
230.
go back to reference O.H. Lee, J. Xu, J. Fueyo, G.N. Fuller, K.D. Aldape, M.M. Alonso, Y. Piao, T.J. Liu, F.F. Lang, B.N. Bekele, C. Gomez-Manzano, Expression of the receptor tyrosine kinase Tie2 in neoplastic glial cells is associated with integrin beta1-dependent adhesion to the extracellular matrix. Mol Cancer Res 4, 915–926 (2006)CrossRefPubMed O.H. Lee, J. Xu, J. Fueyo, G.N. Fuller, K.D. Aldape, M.M. Alonso, Y. Piao, T.J. Liu, F.F. Lang, B.N. Bekele, C. Gomez-Manzano, Expression of the receptor tyrosine kinase Tie2 in neoplastic glial cells is associated with integrin beta1-dependent adhesion to the extracellular matrix. Mol Cancer Res 4, 915–926 (2006)CrossRefPubMed
231.
go back to reference E. Bogdanovic, N. Coombs, D.J. Dumont, Oligomerized Tie2 localizes to clathrin-coated pits in response to angiopoietin-1. Histochem Cell Biol 132, 225–237 (2009)CrossRefPubMed E. Bogdanovic, N. Coombs, D.J. Dumont, Oligomerized Tie2 localizes to clathrin-coated pits in response to angiopoietin-1. Histochem Cell Biol 132, 225–237 (2009)CrossRefPubMed
232.
go back to reference M.B. Hossain, R. Shifat, D.G. Johnson, M.T. Bedford, K.R. Gabrusiewicz, N. Cortes-Santiago, X. Luo, Z. Lu, R. Ezhilarasan, E.P. Sulman, H. Jiang, S.S. Li, F.F. Lang, J. Tyler, M.C. Hung, J. Fueyo, C. Gomez-Manzano, TIE2-mediated tyrosine phosphorylation of H4 regulates DNA damage response by recruiting ABL1. Sci Adv 2, e1501290 (2016)CrossRefPubMedPubMedCentral M.B. Hossain, R. Shifat, D.G. Johnson, M.T. Bedford, K.R. Gabrusiewicz, N. Cortes-Santiago, X. Luo, Z. Lu, R. Ezhilarasan, E.P. Sulman, H. Jiang, S.S. Li, F.F. Lang, J. Tyler, M.C. Hung, J. Fueyo, C. Gomez-Manzano, TIE2-mediated tyrosine phosphorylation of H4 regulates DNA damage response by recruiting ABL1. Sci Adv 2, e1501290 (2016)CrossRefPubMedPubMedCentral
233.
go back to reference M.B. Hossain, R. Shifat, J. Li, X. Luo, K.R. Hess, Y. Rivera-Molina, F. Puerta Martinez, H. Jiang, F.F. Lang, M.C. Hung, J. Fueyo, C. Gomez-Manzano, TIE2 associates with Caveolae and regulates Caveolin-1 to promote their nuclear translocation. Mol Cell Biol 37 (2017) M.B. Hossain, R. Shifat, J. Li, X. Luo, K.R. Hess, Y. Rivera-Molina, F. Puerta Martinez, H. Jiang, F.F. Lang, M.C. Hung, J. Fueyo, C. Gomez-Manzano, TIE2 associates with Caveolae and regulates Caveolin-1 to promote their nuclear translocation. Mol Cell Biol 37 (2017)
234.
go back to reference V. Barresi, S. Cerasoli, G. Paioli, E. Vitarelli, G. Giuffre, G. Guiducci, G. Tuccari, G. Barresi, Caveolin-1 in meningiomas: Expression and clinico-pathological correlations. Acta Neuropathol 112, 617–626 (2006)CrossRefPubMed V. Barresi, S. Cerasoli, G. Paioli, E. Vitarelli, G. Giuffre, G. Guiducci, G. Tuccari, G. Barresi, Caveolin-1 in meningiomas: Expression and clinico-pathological correlations. Acta Neuropathol 112, 617–626 (2006)CrossRefPubMed
235.
go back to reference V. Barresi, S. Cerasoli, G. Tuccari, Correlative evidence that tumor cell-derived caveolin-1 mediates angiogenesis in meningiomas. Neuropathology 28, 472–478 (2008)CrossRefPubMed V. Barresi, S. Cerasoli, G. Tuccari, Correlative evidence that tumor cell-derived caveolin-1 mediates angiogenesis in meningiomas. Neuropathology 28, 472–478 (2008)CrossRefPubMed
236.
go back to reference S. Sharma, S. Ray, S. Mukherjee, A. Moiyadi, E. Sridhar, S. Srivastava, Multipronged quantitative proteomic analyses indicate modulation of various signal transduction pathways in human meningiomas. Proteomics 15, 394–407 (2015)CrossRefPubMed S. Sharma, S. Ray, S. Mukherjee, A. Moiyadi, E. Sridhar, S. Srivastava, Multipronged quantitative proteomic analyses indicate modulation of various signal transduction pathways in human meningiomas. Proteomics 15, 394–407 (2015)CrossRefPubMed
237.
go back to reference M. Aarhus, O. Bruland, H.A. Saetran, S.J. Mork, M. Lund-Johansen, P.M. Knappskog, Global gene expression profiling and tissue microarray reveal novel candidate genes and down-regulation of the tumor suppressor gene CAV1 in sporadic vestibular schwannomas. Neurosurgery 67, 998–1019; discussion 1019 (2010)CrossRefPubMed M. Aarhus, O. Bruland, H.A. Saetran, S.J. Mork, M. Lund-Johansen, P.M. Knappskog, Global gene expression profiling and tissue microarray reveal novel candidate genes and down-regulation of the tumor suppressor gene CAV1 in sporadic vestibular schwannomas. Neurosurgery 67, 998–1019; discussion 1019 (2010)CrossRefPubMed
238.
go back to reference M. Torres-Martin, L. Lassaletta, J. San-Roman-Montero, J.M. De Campos, A. Isla, J. Gavilan, B. Melendez, G.R. Pinto, R.R. Burbano, J.S. Castresana, J.A. Rey, Microarray analysis of gene expression in vestibular schwannomas reveals SPP1/MET signaling pathway and androgen receptor deregulation. Int J Oncol 42, 848–862 (2013)CrossRefPubMedPubMedCentral M. Torres-Martin, L. Lassaletta, J. San-Roman-Montero, J.M. De Campos, A. Isla, J. Gavilan, B. Melendez, G.R. Pinto, R.R. Burbano, J.S. Castresana, J.A. Rey, Microarray analysis of gene expression in vestibular schwannomas reveals SPP1/MET signaling pathway and androgen receptor deregulation. Int J Oncol 42, 848–862 (2013)CrossRefPubMedPubMedCentral
239.
go back to reference P. Cassoni, L. Daniele, E. Maldi, L. Righi, V. Tavaglione, S. Novello, M. Volante, G.V. Scagliotti, M. Papotti, Caveolin-1 expression in lung carcinoma varies according to tumour histotype and is acquired de novo in brain metastases. Histopathology 55, 20–27 (2009)CrossRefPubMed P. Cassoni, L. Daniele, E. Maldi, L. Righi, V. Tavaglione, S. Novello, M. Volante, G.V. Scagliotti, M. Papotti, Caveolin-1 expression in lung carcinoma varies according to tumour histotype and is acquired de novo in brain metastases. Histopathology 55, 20–27 (2009)CrossRefPubMed
240.
go back to reference W.T. Chiu, H.T. Lee, F.J. Huang, K.D. Aldape, J. Yao, P.S. Steeg, C.Y. Chou, Z. Lu, K. Xie, S. Huang, Caveolin-1 upregulation mediates suppression of primary breast tumor growth and brain metastases by stat3 inhibition. Cancer Res 71, 4932–4943 (2011)CrossRefPubMedPubMedCentral W.T. Chiu, H.T. Lee, F.J. Huang, K.D. Aldape, J. Yao, P.S. Steeg, C.Y. Chou, Z. Lu, K. Xie, S. Huang, Caveolin-1 upregulation mediates suppression of primary breast tumor growth and brain metastases by stat3 inhibition. Cancer Res 71, 4932–4943 (2011)CrossRefPubMedPubMedCentral
241.
go back to reference E. Duregon, R. Senetta, A. Pittaro, L. Verdun di Cantogno, G. Stella, P. De Blasi, M. Zorzetto, C. Mantovani, M. Papotti, P. Cassoni, CAVEOLIN-1 expression in brain metastasis from lung cancer predicts worse outcome and radioresistance, irrespective of tumor histotype. Oncotarget 6, 29626–29636 (2015)CrossRefPubMedPubMedCentral E. Duregon, R. Senetta, A. Pittaro, L. Verdun di Cantogno, G. Stella, P. De Blasi, M. Zorzetto, C. Mantovani, M. Papotti, P. Cassoni, CAVEOLIN-1 expression in brain metastasis from lung cancer predicts worse outcome and radioresistance, irrespective of tumor histotype. Oncotarget 6, 29626–29636 (2015)CrossRefPubMedPubMedCentral
242.
243.
go back to reference G. Yang, J. Addai, T.M. Wheeler, A. Frolov, B.J. Miles, D. Kadmon, T.C. Thompson, Correlative evidence that prostate cancer cell-derived caveolin-1 mediates angiogenesis. Hum Pathol 38, 1688–1695 (2007)CrossRefPubMed G. Yang, J. Addai, T.M. Wheeler, A. Frolov, B.J. Miles, D. Kadmon, T.C. Thompson, Correlative evidence that prostate cancer cell-derived caveolin-1 mediates angiogenesis. Hum Pathol 38, 1688–1695 (2007)CrossRefPubMed
244.
go back to reference H.J. Joo, D.K. Oh, Y.S. Kim, K.B. Lee, S.J. Kim, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93, 291–296 (2004)CrossRefPubMed H.J. Joo, D.K. Oh, Y.S. Kim, K.B. Lee, S.J. Kim, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93, 291–296 (2004)CrossRefPubMed
246.
go back to reference S.H. Chang, D. Feng, J.A. Nagy, T.E. Sciuto, A.M. Dvorak, H.F. Dvorak, Vascular permeability and pathological angiogenesis in caveolin-1-null mice. Am J Pathol 175, 1768–1776 (2009)CrossRefPubMedPubMedCentral S.H. Chang, D. Feng, J.A. Nagy, T.E. Sciuto, A.M. Dvorak, H.F. Dvorak, Vascular permeability and pathological angiogenesis in caveolin-1-null mice. Am J Pathol 175, 1768–1776 (2009)CrossRefPubMedPubMedCentral
247.
go back to reference K. Podar, R. Shringarpure, Y.T. Tai, M. Simoncini, M. Sattler, K. Ishitsuka, P.G. Richardson, T. Hideshima, D. Chauhan, K.C. Anderson, Caveolin-1 is required for vascular endothelial growth factor-triggered multiple myeloma cell migration and is targeted by bortezomib. Cancer Res 64, 7500–7506 (2004)CrossRefPubMed K. Podar, R. Shringarpure, Y.T. Tai, M. Simoncini, M. Sattler, K. Ishitsuka, P.G. Richardson, T. Hideshima, D. Chauhan, K.C. Anderson, Caveolin-1 is required for vascular endothelial growth factor-triggered multiple myeloma cell migration and is targeted by bortezomib. Cancer Res 64, 7500–7506 (2004)CrossRefPubMed
248.
go back to reference T.D. Anderson, L.A. Loevner, D.C. Bigelow, N. Mirza, Prevalence of unsuspected acoustic neuroma found by magnetic resonance imaging. Otolaryngol Head Neck Surg 122, 643–646 (2000)CrossRefPubMed T.D. Anderson, L.A. Loevner, D.C. Bigelow, N. Mirza, Prevalence of unsuspected acoustic neuroma found by magnetic resonance imaging. Otolaryngol Head Neck Surg 122, 643–646 (2000)CrossRefPubMed
249.
go back to reference J. Sainz, D.P. Huynh, K. Figueroa, N.K. Ragge, M.E. Baser, S.M. Pulst, Mutations of the neurofibromatosis type 2 gene and lack of the gene product in vestibular schwannomas. Hum Mol Genet 3, 885–891 (1994)CrossRefPubMed J. Sainz, D.P. Huynh, K. Figueroa, N.K. Ragge, M.E. Baser, S.M. Pulst, Mutations of the neurofibromatosis type 2 gene and lack of the gene product in vestibular schwannomas. Hum Mol Genet 3, 885–891 (1994)CrossRefPubMed
250.
go back to reference C.O. Hanemann, B. Bartelt-Kirbach, R. Diebold, K. Kampchen, S. Langmesser, T. Utermark, Differential gene expression between human schwannoma and control Schwann cells. Neuropathol Appl Neurobiol 32, 605–614 (2006)CrossRefPubMed C.O. Hanemann, B. Bartelt-Kirbach, R. Diebold, K. Kampchen, S. Langmesser, T. Utermark, Differential gene expression between human schwannoma and control Schwann cells. Neuropathol Appl Neurobiol 32, 605–614 (2006)CrossRefPubMed
251.
go back to reference D.B. Welling, J.M. Lasak, E. Akhmametyeva, B. Ghaheri, L.S. Chang, cDNA microarray analysis of vestibular schwannomas. Otol Neurotol 23, 736–748 (2002)CrossRefPubMed D.B. Welling, J.M. Lasak, E. Akhmametyeva, B. Ghaheri, L.S. Chang, cDNA microarray analysis of vestibular schwannomas. Otol Neurotol 23, 736–748 (2002)CrossRefPubMed
252.
go back to reference P. Eser Ocak, I. Dogan, U. Ocak, C. Dinc, M.K. Baskaya, Facial nerve outcome and extent of resection in cystic versus solid vestibular schwannomas in radiosurgery era. Neurosurg Focus 44, E3 (2018)CrossRefPubMed P. Eser Ocak, I. Dogan, U. Ocak, C. Dinc, M.K. Baskaya, Facial nerve outcome and extent of resection in cystic versus solid vestibular schwannomas in radiosurgery era. Neurosurg Focus 44, E3 (2018)CrossRefPubMed
253.
go back to reference P. Eser Ocak, I. Dogan, S. Sayyahmelli and M.K. Baskaya, in Vestibular Schwannoma Surgery, (2019), p. 105–133 P. Eser Ocak, I. Dogan, S. Sayyahmelli and M.K. Baskaya, in Vestibular Schwannoma Surgery, (2019), p. 105–133
254.
go back to reference A. Grande-Garcia, A. Echarri, J. de Rooij, N.B. Alderson, C.M. Waterman-Storer, J.M. Valdivielso, M.A. del Pozo, Caveolin-1 regulates cell polarization and directional migration through Src kinase and rho GTPases. J Cell Biol 177, 683–694 (2007)CrossRefPubMedPubMedCentral A. Grande-Garcia, A. Echarri, J. de Rooij, N.B. Alderson, C.M. Waterman-Storer, J.M. Valdivielso, M.A. del Pozo, Caveolin-1 regulates cell polarization and directional migration through Src kinase and rho GTPases. J Cell Biol 177, 683–694 (2007)CrossRefPubMedPubMedCentral
Metadata
Title
The role of caveolin-1 in tumors of the brain - functional and clinical implications
Authors
Pinar Eser Ocak
Umut Ocak
Jiping Tang
John H. Zhang
Publication date
01-08-2019
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 4/2019
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00447-x

Other articles of this Issue 4/2019

Cellular Oncology 4/2019 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