Skip to main content
Top
Published in: Journal of Neuro-Oncology 1/2016

01-05-2016 | Clinical Study

Randomized phase II study of axitinib versus physicians best alternative choice of therapy in patients with recurrent glioblastoma

Authors: J. Duerinck, S. Du Four, F. Vandervorst, N. D’Haene, M. Le Mercier, A. Michotte, A. M. Van Binst, H. Everaert, I. Salmon, F. Bouttens, V. Verschaeve, B. Neyns

Published in: Journal of Neuro-Oncology | Issue 1/2016

Login to get access

Abstract

We conducted a randomized, non-comparative, multi center, phase II clinical trial in order to investigate the efficacy of axitinib, an oral small molecule tyrosine kinase inhibitor with high affinity and specificity for the vascular endothelial growth factor receptors, in patients with recurrent glioblastoma following prior treatment with radiation and temozolomide. Forty-four patients were randomly assigned to receive treatment with axitinib (5 mg BID starting dose; N = 22) or “physicians best alternative choice of therapy” that consisted of bevacizumab (N = 20) or lomustine (N = 2). Six-month progression-free survival served as the primary endpoint. The estimated 6-month progression-free survival rate was 34 % (95 % CI 14–54) for patients treated with axitinib and 28 % (95 % CI 8–48) with best alternative treatment; median overall survival was 29 and 17 weeks, respectively. Objective responses according to RANO criteria were documented in 28 % of patients treated with axitinib and 23 % of patients treated with best alternative therapy. A decrease in maximal uptake of 18F-fluoro-ethyl-tyrosine (18F-FET) by the glioblastoma on PET imaging was documented in 85 % of patients at the time of response on axitinib. Corticosteroid treatment could be stopped in four and tapered in seven out of the 15 patients who were treated with steroids at baseline in the axitinib cohort. Most frequent axitinib related grade ≥3 adverse events consisted of fatigue (9 %), diarrhea (9 %), and oral hyperesthesia (4.5 %). We conclude that axitinib has single-agent clinical activity and a manageable toxicity profile in patients with recurrent glioblastoma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Stupp R, Hegi ME, Mason WP et al (2009) 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 10:459–466CrossRefPubMed Stupp R, Hegi ME, Mason WP et al (2009) 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 10:459–466CrossRefPubMed
2.
go back to reference Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA, Yung WK (1999) Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol 17:2572–2578PubMed Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA, Yung WK (1999) Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol 17:2572–2578PubMed
3.
go back to reference Lamborn KR, Yung WKA, Chang SM et al (2008) Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10:162–170CrossRefPubMedPubMedCentral Lamborn KR, Yung WKA, Chang SM et al (2008) Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10:162–170CrossRefPubMedPubMedCentral
4.
go back to reference Puputti M, Tynninen O, Sihto H, Blom T, Mäenpää H, Isola J, Paetau A, Joensuu H, Nupponen NN (2006) Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res 4:927–934CrossRefPubMed Puputti M, Tynninen O, Sihto H, Blom T, Mäenpää H, Isola J, Paetau A, Joensuu H, Nupponen NN (2006) Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res 4:927–934CrossRefPubMed
5.
go back to reference Knizetova P, Darling JL, Bartek J (2008) Vascular endothelial growth factor in astroglioma stem cell biology and response to therapy. J Cell Mol Med 12:111–125CrossRefPubMedPubMedCentral Knizetova P, Darling JL, Bartek J (2008) Vascular endothelial growth factor in astroglioma stem cell biology and response to therapy. J Cell Mol Med 12:111–125CrossRefPubMedPubMedCentral
6.
go back to reference Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN (2006) Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res 66:7843–7848CrossRefPubMed Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN (2006) Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res 66:7843–7848CrossRefPubMed
8.
go back to reference Reardon DA, Turner S, Peters KB et al (2011) A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Cancer Netw 9:414–427 Reardon DA, Turner S, Peters KB et al (2011) A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Cancer Netw 9:414–427
9.
go back to reference Friedman HS, Prados MD, Wen PY et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27:4733–4740CrossRefPubMed Friedman HS, Prados MD, Wen PY et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27:4733–4740CrossRefPubMed
10.
go back to reference Kreisl TN, Kim L, Moore K et al (2009) Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27:740–745CrossRefPubMedPubMedCentral Kreisl TN, Kim L, Moore K et al (2009) Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27:740–745CrossRefPubMedPubMedCentral
11.
go back to reference Batchelor TT, Mulholland P, Neyns B et al (2013) Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol 31:3212–3218CrossRefPubMedPubMedCentral Batchelor TT, Mulholland P, Neyns B et al (2013) Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol 31:3212–3218CrossRefPubMedPubMedCentral
12.
go back to reference Kratzsch T, Gruenwald V, Vajkoczy P, Kuhn S (2013) Use of axitinib, a new-generation tyrosine kinase inhibitor, to decrease glioblastoma growth despite primary resistance to the VEGF-antibody bevacizumab. ASCO Meet Abstr 2013(31):2077 Kratzsch T, Gruenwald V, Vajkoczy P, Kuhn S (2013) Use of axitinib, a new-generation tyrosine kinase inhibitor, to decrease glioblastoma growth despite primary resistance to the VEGF-antibody bevacizumab. ASCO Meet Abstr 2013(31):2077
13.
go back to reference Galldiks N, Rapp M, Stoffels G, Fink GR, Shah NJ, Coenen HH, Sabel M, Langen K-J (2013) Response assessment of bevacizumab in patients with recurrent malignant glioma using [18F]Fluoroethyl-L-tyrosine PET in comparison to MRI. Eur J Nucl Med Mol Imaging 40:22–33CrossRefPubMed Galldiks N, Rapp M, Stoffels G, Fink GR, Shah NJ, Coenen HH, Sabel M, Langen K-J (2013) Response assessment of bevacizumab in patients with recurrent malignant glioma using [18F]Fluoroethyl-L-tyrosine PET in comparison to MRI. Eur J Nucl Med Mol Imaging 40:22–33CrossRefPubMed
14.
go back to reference Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata D, Danenberg PV, Laird PW (2000) MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res 28:E32CrossRefPubMedPubMedCentral Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata D, Danenberg PV, Laird PW (2000) MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res 28:E32CrossRefPubMedPubMedCentral
15.
go back to reference Widschwendter M, Siegmund KD, Müller HM, Fiegl H, Marth C, Müller-Holzner E, Jones PA, Laird PW (2004) Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res 64:3807–3813CrossRefPubMed Widschwendter M, Siegmund KD, Müller HM, Fiegl H, Marth C, Müller-Holzner E, Jones PA, Laird PW (2004) Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res 64:3807–3813CrossRefPubMed
16.
go back to reference Ogino S, Kawasaki T, Brahmandam M et al (2006) Precision and performance characteristics of bisulfite conversion and real-time PCR (MethyLight) for quantitative DNA methylation analysis. J Mol Diagn 8:209–217CrossRefPubMedPubMedCentral Ogino S, Kawasaki T, Brahmandam M et al (2006) Precision and performance characteristics of bisulfite conversion and real-time PCR (MethyLight) for quantitative DNA methylation analysis. J Mol Diagn 8:209–217CrossRefPubMedPubMedCentral
17.
go back to reference D’Haene N, Le Mercier M, De Nève N et al (2015) Clinical validation of targeted next generation sequencing for colon and lung cancers. PLoS One 10:e0138245CrossRefPubMedPubMedCentral D’Haene N, Le Mercier M, De Nève N et al (2015) Clinical validation of targeted next generation sequencing for colon and lung cancers. PLoS One 10:e0138245CrossRefPubMedPubMedCentral
18.
go back to reference Le Mercier M, D’Haene N, De Nève N, Blanchard O, Degand C, Rorive S, Salmon I (2015) Next-generation sequencing improves the diagnosis of thyroid FNA specimens with indeterminate cytology. Histopathology 66:215–224CrossRefPubMed Le Mercier M, D’Haene N, De Nève N, Blanchard O, Degand C, Rorive S, Salmon I (2015) Next-generation sequencing improves the diagnosis of thyroid FNA specimens with indeterminate cytology. Histopathology 66:215–224CrossRefPubMed
19.
go back to reference Thorvaldsdóttir H, Robinson JT, Mesirov JP (2013) Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14:178–192CrossRefPubMedPubMedCentral Thorvaldsdóttir H, Robinson JT, Mesirov JP (2013) Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14:178–192CrossRefPubMedPubMedCentral
20.
go back to reference Hoogstraat M, Hinrichs JWJ, Besselink NJM et al (2015) Simultaneous detection of clinically relevant mutations and amplifications for routine cancer pathology. J Mol Diagn 17:10–18CrossRefPubMed Hoogstraat M, Hinrichs JWJ, Besselink NJM et al (2015) Simultaneous detection of clinically relevant mutations and amplifications for routine cancer pathology. J Mol Diagn 17:10–18CrossRefPubMed
22.
go back to reference Fack F, Espedal H, Keunen O et al (2015) Bevacizumab treatment induces metabolic adaptation toward anaerobic metabolism in glioblastomas. Acta Neuropathol 129:115–131CrossRefPubMedPubMedCentral Fack F, Espedal H, Keunen O et al (2015) Bevacizumab treatment induces metabolic adaptation toward anaerobic metabolism in glioblastomas. Acta Neuropathol 129:115–131CrossRefPubMedPubMedCentral
23.
go back to reference Götz I, Grosu AL (2013) [(18)F]FET-PET imaging for treatment and response monitoring of radiation therapy in malignant glioma patients—a review. Front Oncol 3:104CrossRefPubMedPubMedCentral Götz I, Grosu AL (2013) [(18)F]FET-PET imaging for treatment and response monitoring of radiation therapy in malignant glioma patients—a review. Front Oncol 3:104CrossRefPubMedPubMedCentral
24.
go back to reference Lu-Emerson C, Duda DG, Emblem KE, Taylor JW, Gerstner ER, Loeffler JS, Batchelor TT, Jain RK (2015) Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J Clin Oncol 33:1197–1213CrossRefPubMedPubMedCentral Lu-Emerson C, Duda DG, Emblem KE, Taylor JW, Gerstner ER, Loeffler JS, Batchelor TT, Jain RK (2015) Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J Clin Oncol 33:1197–1213CrossRefPubMedPubMedCentral
25.
go back to reference Zhang X, Fang X, Gao Z et al (2014) Axitinib, a selective inhibitor of vascular endothelial growth factor receptor, exerts an anticancer effect in melanoma through promoting antitumor immunity. Anticancer Drugs 25:204–211CrossRefPubMed Zhang X, Fang X, Gao Z et al (2014) Axitinib, a selective inhibitor of vascular endothelial growth factor receptor, exerts an anticancer effect in melanoma through promoting antitumor immunity. Anticancer Drugs 25:204–211CrossRefPubMed
26.
go back to reference Stehle F, Schulz K, Fahldieck C, Kalich J, Lichtenfels R, Riemann D, Seliger B (2013) Reduced immunosuppressive properties of axitinib in comparison with other tyrosine kinase inhibitors. J Biol Chem 288:16334–16347CrossRefPubMedPubMedCentral Stehle F, Schulz K, Fahldieck C, Kalich J, Lichtenfels R, Riemann D, Seliger B (2013) Reduced immunosuppressive properties of axitinib in comparison with other tyrosine kinase inhibitors. J Biol Chem 288:16334–16347CrossRefPubMedPubMedCentral
27.
go back to reference Bose A, Lowe DB, Rao A, Storkus WJ (2012) Combined vaccine+axitinib therapy yields superior antitumor efficacy in a murine melanoma model. Melanoma Res 22:236–243CrossRefPubMedPubMedCentral Bose A, Lowe DB, Rao A, Storkus WJ (2012) Combined vaccine+axitinib therapy yields superior antitumor efficacy in a murine melanoma model. Melanoma Res 22:236–243CrossRefPubMedPubMedCentral
28.
go back to reference Du Four S, Maenhout SK, De Pierre K, Renmans D, Niclou SP, Thielemans K, Neyns B, Aerts JL (2015) Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology 4:e998107CrossRefPubMedPubMedCentral Du Four S, Maenhout SK, De Pierre K, Renmans D, Niclou SP, Thielemans K, Neyns B, Aerts JL (2015) Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology 4:e998107CrossRefPubMedPubMedCentral
29.
go back to reference Neyns B, Sadones J, Chaskis C et al (2011) Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J Neurooncol 103:491–501CrossRefPubMed Neyns B, Sadones J, Chaskis C et al (2011) Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J Neurooncol 103:491–501CrossRefPubMed
Metadata
Title
Randomized phase II study of axitinib versus physicians best alternative choice of therapy in patients with recurrent glioblastoma
Authors
J. Duerinck
S. Du Four
F. Vandervorst
N. D’Haene
M. Le Mercier
A. Michotte
A. M. Van Binst
H. Everaert
I. Salmon
F. Bouttens
V. Verschaeve
B. Neyns
Publication date
01-05-2016
Publisher
Springer US
Published in
Journal of Neuro-Oncology / Issue 1/2016
Print ISSN: 0167-594X
Electronic ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-016-2092-2

Other articles of this Issue 1/2016

Journal of Neuro-Oncology 1/2016 Go to the issue