Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 3/2012

01.09.2012 | Review Article

Antiangiogenic therapy in the management of brain tumors: a clinical overview

verfasst von: Saroj Kunnakkat, Maya Mathew, Ashwatha Narayana

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 3/2012

Einloggen, um Zugang zu erhalten

Abstract

Central to the process of brain tumor development is angiogenesis, which involves a host of molecules and receptors. In recent years, antiangiogenic therapies have been developed and tested for their effectiveness against these tumors. Among them are inhibitors against vascular endothelial growth factor and its receptors, as well as inhibitors targeting the platelet-derived growth factor family, integrins, and histone deacetylase. While many have been shown to be effective with limited toxicity, some tumors are able to adopt escape mechanisms. Further research is needed in the development of effective multi-targeted agents to reduce these effects.
Literatur
1.
Zurück zum Zitat Fidler IJ (1999) Critical determinants of cancer metastasis: rationale for therapy. Cancer Chemother Pharmaco 43:S3–S10CrossRef Fidler IJ (1999) Critical determinants of cancer metastasis: rationale for therapy. Cancer Chemother Pharmaco 43:S3–S10CrossRef
2.
Zurück zum Zitat Jiménez B, Volpert OV (2001) Mechanistic insights on the inhibition of tumor angiogenesis. J Mol Med 78(12):663–672PubMedCrossRef Jiménez B, Volpert OV (2001) Mechanistic insights on the inhibition of tumor angiogenesis. J Mol Med 78(12):663–672PubMedCrossRef
3.
Zurück zum Zitat Hicklin DJ, Ellis LM (2005) Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23(5):1011–1027PubMedCrossRef Hicklin DJ, Ellis LM (2005) Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23(5):1011–1027PubMedCrossRef
4.
Zurück zum Zitat Holmes K, Roberts OL, Thomas AM, Cross MJ (2007) Vascular endothelial growth factor receptor-2: structure, function, intracellular signalling and therapeutic inhibition. Cell Signal 19(10):2003–2012PubMedCrossRef Holmes K, Roberts OL, Thomas AM, Cross MJ (2007) Vascular endothelial growth factor receptor-2: structure, function, intracellular signalling and therapeutic inhibition. Cell Signal 19(10):2003–2012PubMedCrossRef
5.
Zurück zum Zitat Bautch VL, Redick SD, Scalia A, Harmaty M, Carmeliet P, Rapoport R (2000) Characterization of the vasculogenic block in the absence of vascular endothelial growth factor-A. Blood 95(6):1979–1987PubMed Bautch VL, Redick SD, Scalia A, Harmaty M, Carmeliet P, Rapoport R (2000) Characterization of the vasculogenic block in the absence of vascular endothelial growth factor-A. Blood 95(6):1979–1987PubMed
6.
Zurück zum Zitat Fong G-H, Rossant J, Gertsenstein M, Breitman ML (1995) Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 376(6535):66–70PubMedCrossRef Fong G-H, Rossant J, Gertsenstein M, Breitman ML (1995) Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 376(6535):66–70PubMedCrossRef
7.
Zurück zum Zitat Shalaby F, Rossant J, Yamaguchi TP et al (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535):62–66PubMedCrossRef Shalaby F, Rossant J, Yamaguchi TP et al (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535):62–66PubMedCrossRef
8.
Zurück zum Zitat Zagzag D, Amirnovin R, Greco MA et al (2000) Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis. Lab Invest 80(6):837–849PubMedCrossRef Zagzag D, Amirnovin R, Greco MA et al (2000) Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis. Lab Invest 80(6):837–849PubMedCrossRef
9.
Zurück zum Zitat Zagzag D, Lukyanov Y, Lan L et al (2006) Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Lab Invest 86(12):1221–1232PubMedCrossRef Zagzag D, Lukyanov Y, Lan L et al (2006) Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Lab Invest 86(12):1221–1232PubMedCrossRef
10.
Zurück zum Zitat Vredenburgh J, Desjardins A, Herndon J et al (2007) Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 25:4722–4729PubMedCrossRef Vredenburgh J, Desjardins A, Herndon J et al (2007) Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 25:4722–4729PubMedCrossRef
11.
Zurück zum Zitat Friedman HS, Prados MD, Wen PY et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27(28):4733–4740PubMedCrossRef Friedman HS, Prados MD, Wen PY et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27(28):4733–4740PubMedCrossRef
12.
Zurück zum Zitat Nghiemphu PL, Liu W et al (2009) Bevacizumab and chemotherapy for recurrent glioblastoma. Neurology 72(14):1217–1222PubMedCrossRef Nghiemphu PL, Liu W et al (2009) Bevacizumab and chemotherapy for recurrent glioblastoma. Neurology 72(14):1217–1222PubMedCrossRef
13.
Zurück zum Zitat Kerber M, Reiss Y et al (2008) Flt-1 signaling in macrophages promotes glioma growth in vivo. Cancer Res 68(18):7342–7351PubMedCrossRef Kerber M, Reiss Y et al (2008) Flt-1 signaling in macrophages promotes glioma growth in vivo. Cancer Res 68(18):7342–7351PubMedCrossRef
14.
Zurück zum Zitat Geng L, Donnelly E et al (2001) Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Res 61(6):2413–2419PubMed Geng L, Donnelly E et al (2001) Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Res 61(6):2413–2419PubMed
15.
Zurück zum Zitat Pietsch T, Valter MM, Wolf HK et al (1997) Expression and distribution of vascular endothelial growth factor protein in human brain tumors. Acta Neuropathol 93(2):109–117PubMedCrossRef Pietsch T, Valter MM, Wolf HK et al (1997) Expression and distribution of vascular endothelial growth factor protein in human brain tumors. Acta Neuropathol 93(2):109–117PubMedCrossRef
16.
Zurück zum Zitat Caye-Thomasen P et al (2005) VEGF and VEGF receptor-1 concentration in vestibular schwannoma homogenates correlates to tumor growth rate. [Miscellaneous Article]. Otol Neurotol 26(1):98–101PubMedCrossRef Caye-Thomasen P et al (2005) VEGF and VEGF receptor-1 concentration in vestibular schwannoma homogenates correlates to tumor growth rate. [Miscellaneous Article]. Otol Neurotol 26(1):98–101PubMedCrossRef
17.
Zurück zum Zitat Hoch RV, Soriano P (2003) Roles of PDGF in animal development. Development 130(20):4769–4784PubMedCrossRef Hoch RV, Soriano P (2003) Roles of PDGF in animal development. Development 130(20):4769–4784PubMedCrossRef
18.
Zurück zum Zitat Andrae J, Gallini R et al (2008) Role of platelet-derived growth factors in physiology and medicine. Genes Dev 22(10):1276–1312PubMedCrossRef Andrae J, Gallini R et al (2008) Role of platelet-derived growth factors in physiology and medicine. Genes Dev 22(10):1276–1312PubMedCrossRef
19.
Zurück zum Zitat Charles N, Ozawa T et al (2010) Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell 6(2):141–152PubMedCrossRef Charles N, Ozawa T et al (2010) Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell 6(2):141–152PubMedCrossRef
20.
Zurück zum Zitat Lokker NA, Sullivan CM et al (2002) Platelet-derived growth factor (PDGF) autocrine signaling regulates survival and mitogenic pathways in glioblastoma cells. Cancer Res 62(13):3729–3735PubMed Lokker NA, Sullivan CM et al (2002) Platelet-derived growth factor (PDGF) autocrine signaling regulates survival and mitogenic pathways in glioblastoma cells. Cancer Res 62(13):3729–3735PubMed
21.
Zurück zum Zitat Hynes RO (2002) Integrins: bidirectional, allosteric signaling machines. Cell 110(6):673–687PubMedCrossRef Hynes RO (2002) Integrins: bidirectional, allosteric signaling machines. Cell 110(6):673–687PubMedCrossRef
22.
Zurück zum Zitat Zhong J, Paul A et al (2010) Mesenchymal migration as a therapeutic target in glioblastoma. J Oncol 2010(21):430142PubMed Zhong J, Paul A et al (2010) Mesenchymal migration as a therapeutic target in glioblastoma. J Oncol 2010(21):430142PubMed
23.
Zurück zum Zitat Kanamori M, Kawaguchi T et al (2006) Intracranial microenvironment reveals independent opposing functions of host αVβ3 expression on glioma growth and angiogenesis. J Biol Chem 281(48):37256–37264PubMedCrossRef Kanamori M, Kawaguchi T et al (2006) Intracranial microenvironment reveals independent opposing functions of host αVβ3 expression on glioma growth and angiogenesis. J Biol Chem 281(48):37256–37264PubMedCrossRef
24.
Zurück zum Zitat Schnell O, Krebs B et al (2008) Expression of integrin αvβ3 in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Patho 18(3):378–386CrossRef Schnell O, Krebs B et al (2008) Expression of integrin αvβ3 in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Patho 18(3):378–386CrossRef
25.
Zurück zum Zitat Puduvalli V (2001) Brain metastases: biology and the role of the brain microenvironment. Curr Oncol Rep 3(6):467–475PubMedCrossRef Puduvalli V (2001) Brain metastases: biology and the role of the brain microenvironment. Curr Oncol Rep 3(6):467–475PubMedCrossRef
26.
Zurück zum Zitat Weber GF, Ashkar S (2000) Molecular mechanisms of tumor dissemination in primary and metastatic brain cancers. Brain Res Bull 53(4):421–424PubMedCrossRef Weber GF, Ashkar S (2000) Molecular mechanisms of tumor dissemination in primary and metastatic brain cancers. Brain Res Bull 53(4):421–424PubMedCrossRef
27.
Zurück zum Zitat Ouaissi M, Ouaissi A (2006) Histone deacetylase enzymes as potential drug targets in cancer and parasitic diseases. J Biomed Biotechnol 2006:13474 Ouaissi M, Ouaissi A (2006) Histone deacetylase enzymes as potential drug targets in cancer and parasitic diseases. J Biomed Biotechnol 2006:13474
28.
Zurück zum Zitat Campos B, Bermejo JL et al (2011) Expression of nuclear receptor corepressors and class I histone deacetylases in astrocytic gliomas. Cancer Sci 102(2):387–392PubMedCrossRef Campos B, Bermejo JL et al (2011) Expression of nuclear receptor corepressors and class I histone deacetylases in astrocytic gliomas. Cancer Sci 102(2):387–392PubMedCrossRef
29.
Zurück zum Zitat Sharma V, Koul N et al (2010) HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med 14(8):2151–2161PubMedCrossRef Sharma V, Koul N et al (2010) HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med 14(8):2151–2161PubMedCrossRef
30.
Zurück zum Zitat Thudi NK, Shu ST et al (2011) Development of a brain metastatic canine prostate cancer cell line. Prostate 71(12):1251–1263PubMed Thudi NK, Shu ST et al (2011) Development of a brain metastatic canine prostate cancer cell line. Prostate 71(12):1251–1263PubMed
31.
Zurück zum Zitat Vredenburgh JJ, Desjardins A et al (2007) Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res 13(4):1253–1259PubMedCrossRef Vredenburgh JJ, Desjardins A et al (2007) Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res 13(4):1253–1259PubMedCrossRef
32.
Zurück zum Zitat Kreisl TN, Kim L et al (2009) Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27(5):740–745PubMedCrossRef Kreisl TN, Kim L et al (2009) Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27(5):740–745PubMedCrossRef
33.
Zurück zum Zitat Narayana A, Kelly P et al (2009) Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survival. J Neurosurgery 110(1):173–180CrossRef Narayana A, Kelly P et al (2009) Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survival. J Neurosurgery 110(1):173–180CrossRef
34.
Zurück zum Zitat Norden AD, Young GS et al (2008) Bevacizumab for recurrent malignant gliomas. Neurology 70(10):779–787PubMedCrossRef Norden AD, Young GS et al (2008) Bevacizumab for recurrent malignant gliomas. Neurology 70(10):779–787PubMedCrossRef
35.
Zurück zum Zitat Lai A, Tran A et al (2011) Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol 29(2):142–148PubMedCrossRef Lai A, Tran A et al (2011) Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol 29(2):142–148PubMedCrossRef
36.
Zurück zum Zitat Narayana A, Golfinos JG et al (2008) Feasibility of using bevacizumab with radiation therapy and temozolomide in newly diagnosed high-grade glioma. Int J Rad Oncol Biol Phys 72(2):383–389CrossRef Narayana A, Golfinos JG et al (2008) Feasibility of using bevacizumab with radiation therapy and temozolomide in newly diagnosed high-grade glioma. Int J Rad Oncol Biol Phys 72(2):383–389CrossRef
37.
Zurück zum Zitat Vredenburgh JJ, Desjardins A et al (2012) Addition of bevacizumab to standard radiation therapy and daily temozolomide is associated with minimal toxicity in newly diagnosed glioblastoma multiforme. Int J Rad Oncol Biol Phys 82(1):58–66CrossRef Vredenburgh JJ, Desjardins A et al (2012) Addition of bevacizumab to standard radiation therapy and daily temozolomide is associated with minimal toxicity in newly diagnosed glioblastoma multiforme. Int J Rad Oncol Biol Phys 82(1):58–66CrossRef
38.
Zurück zum Zitat Narayana A, Gruber D et al (2012) A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma. J Neurosurgery 116(2):341–345CrossRef Narayana A, Gruber D et al (2012) A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma. J Neurosurgery 116(2):341–345CrossRef
39.
Zurück zum Zitat Chamberlain MC, Johnston S (2009) Bevacizumab for recurrent alkylator-refractory anaplastic oligodendroglioma. Cancer 115(8):1734–1743PubMedCrossRef Chamberlain MC, Johnston S (2009) Bevacizumab for recurrent alkylator-refractory anaplastic oligodendroglioma. Cancer 115(8):1734–1743PubMedCrossRef
40.
Zurück zum Zitat Mautner V-F, Nguyen R et al (2010) Bevacizumab induces regression of vestibular schwannomas in patients with neurofibromatosis type 2. Neuro-Oncol 12(1):14–18PubMedCrossRef Mautner V-F, Nguyen R et al (2010) Bevacizumab induces regression of vestibular schwannomas in patients with neurofibromatosis type 2. Neuro-Oncol 12(1):14–18PubMedCrossRef
41.
Zurück zum Zitat Plotkin SR, Stemmer-Rachamimov AO et al (2009) Hearing improvement after bevacizumab in patients with neurofibromatosis type 2. N Engl J Med 361(4):358–367PubMedCrossRef Plotkin SR, Stemmer-Rachamimov AO et al (2009) Hearing improvement after bevacizumab in patients with neurofibromatosis type 2. N Engl J Med 361(4):358–367PubMedCrossRef
42.
Zurück zum Zitat Mautner V-F, Nguyen R et al (2010) Radiographic regression of vestibular schwannomas induced by bevacizumab treatment: sustain under continuous drug application and rebound after drug discontinuation. Ann Oncol 21(11):2294–2295PubMedCrossRef Mautner V-F, Nguyen R et al (2010) Radiographic regression of vestibular schwannomas induced by bevacizumab treatment: sustain under continuous drug application and rebound after drug discontinuation. Ann Oncol 21(11):2294–2295PubMedCrossRef
43.
Zurück zum Zitat Puchner MJA, Hans VH et al (2010) Bevacizumab-induced regression of anaplastic meningioma. Ann Oncol 21(12):2445–2446PubMedCrossRef Puchner MJA, Hans VH et al (2010) Bevacizumab-induced regression of anaplastic meningioma. Ann Oncol 21(12):2445–2446PubMedCrossRef
44.
Zurück zum Zitat Goutagny S, Raymond E et al (2011) Radiographic regression of cranial meningioma in a NF2 patient treated by bevacizumab. Ann Oncol 22(4):990–991PubMedCrossRef Goutagny S, Raymond E et al (2011) Radiographic regression of cranial meningioma in a NF2 patient treated by bevacizumab. Ann Oncol 22(4):990–991PubMedCrossRef
45.
Zurück zum Zitat Socinski MA, Langer CJ et al (2009) Safety of bevacizumab in patients with non–small-cell lung cancer and brain metastases. J Clin Oncol 27(31):5255–5261PubMedCrossRef Socinski MA, Langer CJ et al (2009) Safety of bevacizumab in patients with non–small-cell lung cancer and brain metastases. J Clin Oncol 27(31):5255–5261PubMedCrossRef
46.
Zurück zum Zitat Labidi SI, Bachelot T et al (2009) Bevacizumab and paclitaxel for breast cancer patients with central nervous system metastases: a case series. Clin Breast Cancer 9(2):118–121PubMedCrossRef Labidi SI, Bachelot T et al (2009) Bevacizumab and paclitaxel for breast cancer patients with central nervous system metastases: a case series. Clin Breast Cancer 9(2):118–121PubMedCrossRef
47.
Zurück zum Zitat Wong ET, Huberman M et al (2008) Bevacizumab reverses cerebral radiation necrosis. J Clin Oncol 26(34):5649–5650PubMedCrossRef Wong ET, Huberman M et al (2008) Bevacizumab reverses cerebral radiation necrosis. J Clin Oncol 26(34):5649–5650PubMedCrossRef
48.
Zurück zum Zitat Torcuator R, Zuniga R et al (2009) Initial experience with bevacizumab treatment for biopsy confirmed cerebral radiation necrosis. J Neuro-Oncol 94(1):63–68CrossRef Torcuator R, Zuniga R et al (2009) Initial experience with bevacizumab treatment for biopsy confirmed cerebral radiation necrosis. J Neuro-Oncol 94(1):63–68CrossRef
49.
Zurück zum Zitat Spratlin JL, Mulder KE et al (2010) Ramucirumab (IMC-1121B): a novel attack on angiogenesis. Fut Oncol 6(7):1085–1094CrossRef Spratlin JL, Mulder KE et al (2010) Ramucirumab (IMC-1121B): a novel attack on angiogenesis. Fut Oncol 6(7):1085–1094CrossRef
50.
Zurück zum Zitat Spratlin JL, Cohen RB et al (2010) Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol 28(5):780–787PubMedCrossRef Spratlin JL, Cohen RB et al (2010) Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol 28(5):780–787PubMedCrossRef
51.
Zurück zum Zitat Farrar CT, Kamoun WS et al (2011) Sensitivity of MRI tumor biomarkers to VEGFR inhibitor therapy in an orthotopic mouse glioma model. PLoS One 6(3):e17228PubMedCrossRef Farrar CT, Kamoun WS et al (2011) Sensitivity of MRI tumor biomarkers to VEGFR inhibitor therapy in an orthotopic mouse glioma model. PLoS One 6(3):e17228PubMedCrossRef
52.
Zurück zum Zitat Dietrich J, Wang D et al (2009) Cediranib: profile of a novel anti-angiogenic agent in patients with glioblastoma. Expert Opin Investig Drugs 18(10):1549–1557PubMedCrossRef Dietrich J, Wang D et al (2009) Cediranib: profile of a novel anti-angiogenic agent in patients with glioblastoma. Expert Opin Investig Drugs 18(10):1549–1557PubMedCrossRef
53.
Zurück zum Zitat Batchelor TT, Duda DG et al (2010) Phase II study of cediranib, an oral pan–vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol 28(17):2817–2823PubMedCrossRef Batchelor TT, Duda DG et al (2010) Phase II study of cediranib, an oral pan–vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol 28(17):2817–2823PubMedCrossRef
54.
Zurück zum Zitat Wachsberger PR, Lawrence YR et al (2012) Epidermal growth factor receptor expression modulates antitumor efficacy of vandetanib or cediranib combined with radiotherapy in human glioblastoma xenografts. Int J Rad Oncol Biol Phys 82(1):483–491CrossRef Wachsberger PR, Lawrence YR et al (2012) Epidermal growth factor receptor expression modulates antitumor efficacy of vandetanib or cediranib combined with radiotherapy in human glioblastoma xenografts. Int J Rad Oncol Biol Phys 82(1):483–491CrossRef
55.
Zurück zum Zitat Siegelin MD, Raskett CM et al (2010) Sorafenib exerts anti-glioma activity in vitro and in vivo. Neurosci Lett 478(3):165–170PubMedCrossRef Siegelin MD, Raskett CM et al (2010) Sorafenib exerts anti-glioma activity in vitro and in vivo. Neurosci Lett 478(3):165–170PubMedCrossRef
56.
Zurück zum Zitat Navis AC, Hamans BC et al (2011) Effects of targeting the VEGF and PDGF pathways in diffuse orthotopic glioma models. The J Pathol 223(5):626–634CrossRef Navis AC, Hamans BC et al (2011) Effects of targeting the VEGF and PDGF pathways in diffuse orthotopic glioma models. The J Pathol 223(5):626–634CrossRef
57.
Zurück zum Zitat Neyns B, Sadones J et al (2011) Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J Neuro-Oncol 103(3):491–501CrossRef Neyns B, Sadones J et al (2011) Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J Neuro-Oncol 103(3):491–501CrossRef
58.
Zurück zum Zitat Hainsworth JD, Ervin T et al (2010) Concurrent radiotherapy and temozolomide followed by temozolomide and sorafenib in the first-line treatment of patients with glioblastoma multiforme. Cancer 116(15):3663–3669PubMedCrossRef Hainsworth JD, Ervin T et al (2010) Concurrent radiotherapy and temozolomide followed by temozolomide and sorafenib in the first-line treatment of patients with glioblastoma multiforme. Cancer 116(15):3663–3669PubMedCrossRef
59.
Zurück zum Zitat Yoshikawa A, Nakada M et al (2012) Recurrent anaplastic meningioma treated by sunitinib based on results from quantitative proteomics. Neuropathol Appl Neurobiol 38(1):105–110PubMedCrossRef Yoshikawa A, Nakada M et al (2012) Recurrent anaplastic meningioma treated by sunitinib based on results from quantitative proteomics. Neuropathol Appl Neurobiol 38(1):105–110PubMedCrossRef
60.
Zurück zum Zitat Amaravadi RK, Schuchter LM et al (2009) Phase II trial of temozolomide and sorafenib in advanced melanoma patients with or without brain metastases. Clin Cancer Res 15(24):7711–7718PubMedCrossRef Amaravadi RK, Schuchter LM et al (2009) Phase II trial of temozolomide and sorafenib in advanced melanoma patients with or without brain metastases. Clin Cancer Res 15(24):7711–7718PubMedCrossRef
61.
Zurück zum Zitat Gore ME, Hariharan S et al (2011) Sunitinib in metastatic renal cell carcinoma patients with brain metastases. Cancer 117(3):501–509PubMedCrossRef Gore ME, Hariharan S et al (2011) Sunitinib in metastatic renal cell carcinoma patients with brain metastases. Cancer 117(3):501–509PubMedCrossRef
62.
Zurück zum Zitat Wuthrick EJ, Kamrava M et al (2011) A phase 1b trial of the combination of the antiangiogenic agent sunitinib and radiation therapy for patients with primary and metastatic central nervous system malignancies. Cancer 117(24):5548–5559PubMedCrossRef Wuthrick EJ, Kamrava M et al (2011) A phase 1b trial of the combination of the antiangiogenic agent sunitinib and radiation therapy for patients with primary and metastatic central nervous system malignancies. Cancer 117(24):5548–5559PubMedCrossRef
63.
Zurück zum Zitat Maurer GD, Tritschler I et al (2009) Cilengitide modulates attachment and viability of human glioma cells, but not sensitivity to irradiation or temozolomide in vitro. Neuro-Oncol 11(6):747–756PubMedCrossRef Maurer GD, Tritschler I et al (2009) Cilengitide modulates attachment and viability of human glioma cells, but not sensitivity to irradiation or temozolomide in vitro. Neuro-Oncol 11(6):747–756PubMedCrossRef
64.
Zurück zum Zitat Reardon DA, Fink KL et al (2008) Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol 26(34):5610–5617PubMedCrossRef Reardon DA, Fink KL et al (2008) Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol 26(34):5610–5617PubMedCrossRef
65.
Zurück zum Zitat Stupp R, Hegi ME et al (2010) Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J Clin Oncol 28(16):2712–2718PubMedCrossRef Stupp R, Hegi ME et al (2010) Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J Clin Oncol 28(16):2712–2718PubMedCrossRef
66.
Zurück zum Zitat Sawa H, Murakami H et al (2004) Histone deacetylase inhibitor, FK228, induces apoptosis and suppresses cell proliferation of human glioblastoma cells in vitro and in vivo. Acta Neuropathol 107(6):523–531PubMedCrossRef Sawa H, Murakami H et al (2004) Histone deacetylase inhibitor, FK228, induces apoptosis and suppresses cell proliferation of human glioblastoma cells in vitro and in vivo. Acta Neuropathol 107(6):523–531PubMedCrossRef
67.
Zurück zum Zitat Iwamoto FM, Lamborn KR et al (2011) A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: North American brain tumor consortium study 03–03. Neuro-Oncol 13(5):509–516PubMedCrossRef Iwamoto FM, Lamborn KR et al (2011) A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: North American brain tumor consortium study 03–03. Neuro-Oncol 13(5):509–516PubMedCrossRef
68.
Zurück zum Zitat Berg SL, Stone J et al (2004) Plasma and cerebrospinal fluid pharmacokinetics of depsipeptide (FR901228) in nonhuman primates. Cancer Chemother Pharmacol 54(1):85–88PubMedCrossRef Berg SL, Stone J et al (2004) Plasma and cerebrospinal fluid pharmacokinetics of depsipeptide (FR901228) in nonhuman primates. Cancer Chemother Pharmacol 54(1):85–88PubMedCrossRef
69.
Zurück zum Zitat Hariharan S, Gustafson D et al (2007) Assessment of the biological and pharmacological effects of the ανβ3 and ανβ5 integrin receptor antagonist, cilengitide (EMD 121974), in patients with advanced solid tumors. Ann Oncol 18(8):1400–1407PubMedCrossRef Hariharan S, Gustafson D et al (2007) Assessment of the biological and pharmacological effects of the ανβ3 and ανβ5 integrin receptor antagonist, cilengitide (EMD 121974), in patients with advanced solid tumors. Ann Oncol 18(8):1400–1407PubMedCrossRef
70.
Zurück zum Zitat Shah MH, Binkley P et al (2006) Cardio toxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin Cancer Res 12(13):3997–4003PubMedCrossRef Shah MH, Binkley P et al (2006) Cardio toxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin Cancer Res 12(13):3997–4003PubMedCrossRef
71.
Zurück zum Zitat Rini BI, Choueiri TK et al (2008) Sunitinib-induced macrocytosis in patients with metastatic renal cell carcinoma. Cancer 113(6):1309–1314PubMedCrossRef Rini BI, Choueiri TK et al (2008) Sunitinib-induced macrocytosis in patients with metastatic renal cell carcinoma. Cancer 113(6):1309–1314PubMedCrossRef
72.
Zurück zum Zitat Brunello A, Saia G et al (2009) Worsening of osteonecrosis of the jaw during treatment with sunitinib in a patient with metastatic renal cell carcinoma. Bone 44(1):173–175PubMedCrossRef Brunello A, Saia G et al (2009) Worsening of osteonecrosis of the jaw during treatment with sunitinib in a patient with metastatic renal cell carcinoma. Bone 44(1):173–175PubMedCrossRef
73.
Zurück zum Zitat Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603PubMedCrossRef Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603PubMedCrossRef
74.
Zurück zum Zitat Lucio-Eterovic AK, Piao Y et al (2009) Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res 15(14):4589–4599PubMedCrossRef Lucio-Eterovic AK, Piao Y et al (2009) Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res 15(14):4589–4599PubMedCrossRef
75.
Zurück zum Zitat de Groot JF, Fuller G et al (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro-Oncol 12(3):233–242PubMedCrossRef de Groot JF, Fuller G et al (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro-Oncol 12(3):233–242PubMedCrossRef
76.
Zurück zum Zitat Iwamoto FM, Abrey LE et al (2009) Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology 73(15):1200–1206PubMedCrossRef Iwamoto FM, Abrey LE et al (2009) Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology 73(15):1200–1206PubMedCrossRef
77.
Zurück zum Zitat Narayana A, Kunnakkat SD et al (2012) Change in pattern of relapse after antiangiogenic therapy in high-grade glioma. Int J Rad Oncol Biol Phys 82(1):77–82CrossRef Narayana A, Kunnakkat SD et al (2012) Change in pattern of relapse after antiangiogenic therapy in high-grade glioma. Int J Rad Oncol Biol Phys 82(1):77–82CrossRef
78.
Zurück zum Zitat Wen PY, Macdonald DR et al (2010) Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 28(11):1963–1972PubMedCrossRef Wen PY, Macdonald DR et al (2010) Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 28(11):1963–1972PubMedCrossRef
Metadaten
Titel
Antiangiogenic therapy in the management of brain tumors: a clinical overview
verfasst von
Saroj Kunnakkat
Maya Mathew
Ashwatha Narayana
Publikationsdatum
01.09.2012
Verlag
Springer-Verlag
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 3/2012
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-012-1926-7

Weitere Artikel der Ausgabe 3/2012

Cancer Chemotherapy and Pharmacology 3/2012 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

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