Skip to main content
Top
Published in: Current Treatment Options in Oncology 8/2016

01-08-2016 | Neuro-oncology (GJ Lesser, Section Editor)

Targeted Therapeutics in Patients With High-Grade Gliomas: Past, Present, and Future

Authors: Ricky Chen, MD, Adam L. Cohen, MD, Howard Colman, MD, PhD

Published in: Current Treatment Options in Oncology | Issue 8/2016

Login to get access

Opinion statement

High-grade gliomas remain incurable despite current therapies, which are plagued by high morbidity and mortality. Molecular categorization of glioma subtypes using mutations in isocitrate dehydrogenase 1/2 (IDH1/2), TP53, and ATRX; codeletion of chromosomes 1p and 19q; DNA methylation; and amplification of genes such as epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor, alpha polypeptide provides a more accurate prognostication and biologic classification than classical histopathological diagnoses, and a number of molecular markers are being incorporated in the new World Health Organization classification of gliomas. However, despite the improved understanding of the molecular subtypes of gliomas and the underlying alterations in specific signaling pathways, these observations have so far failed to result in the successful application of targeted therapies, as has occurred in other solid tumors. To date, the only targeted therapy for gliomas approved by the US Food and Drug Administration is bevacizumab, which targets vascular endothelial growth factor. EGFR remains a dominant molecular alteration in specific glioma subtypes and represents a potentially promising target, with drugs of multiple types targeting EGFR in development including vaccines, antibody drug conjugates, and chimeric antigen receptor (CAR) T cells, despite the prior failures of EGFR tyrosine kinase inhibitors. Immune therapies under investigation include checkpoint inhibitors, vaccines against tumor-associated antigens and tumor-specific antigens, pulsed dendritic cells, heat shock protein-tumor conjugates, and CAR T cells. Mutations in the IDH1/2 genes are central to gliomagenesis in a high proportion of grade II and III gliomas, and ongoing trials are examining vaccines against IDH1, small molecular inhibitors of IDH1 and IDH2, and metabolic components including NAD+ depletion to target IDH-mutated gliomas. The central role of DNA methylation in a subset of gliomas may be targetable, but better understanding of the relation between epigenetic alterations and resulting tumor biology appears necessary. Ultimately, given the prior failure of single-agent targeted therapy in high-grade gliomas, it appears that novel combinatorial therapy or targeted drugs with immunomodulatory or epigenetic approaches will likely be necessary to successfully combat these challenging tumors.
Literature
1.
go back to reference Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed
2.••
go back to reference Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535–43. Recent phase III trial that demonstrated improvement in progression-free and overall survival in patients treated with tumor treated fields in addition to radiation and adjuvant temozolomide versus radiation and temozolomide alone.CrossRefPubMed Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535–43. Recent phase III trial that demonstrated improvement in progression-free and overall survival in patients treated with tumor treated fields in addition to radiation and adjuvant temozolomide versus radiation and temozolomide alone.CrossRefPubMed
3.
go back to reference Ostrom QT, Gittleman H, de Blank PM, et al. American Brain Tumor Association Adolescent and Young Adult Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008–2012. Neuro-Oncology. 2016;18 Suppl 1:i1–50.CrossRefPubMed Ostrom QT, Gittleman H, de Blank PM, et al. American Brain Tumor Association Adolescent and Young Adult Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008–2012. Neuro-Oncology. 2016;18 Suppl 1:i1–50.CrossRefPubMed
4.
go back to reference Stupp R, Hegi ME, Mason WP, et al. 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. 2009;10(5):459–66.CrossRefPubMed Stupp R, Hegi ME, Mason WP, et al. 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. 2009;10(5):459–66.CrossRefPubMed
5.
6.••
go back to reference Brat DJ, Verhaak RG, Aldape KD, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–98. Comprehensive, multiplatform analysis dividing lower grade gliomas into three distinct molecular classes that were more concordant with IDH mutation, 1p19q loss, and TP53 alteration than with histology. These delineated groups more accurately predict clinical outcomes and help set the stage for molecular - based definition of these tumors in clinical trials and clinical practice.CrossRefPubMed Brat DJ, Verhaak RG, Aldape KD, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–98. Comprehensive, multiplatform analysis dividing lower grade gliomas into three distinct molecular classes that were more concordant with IDH mutation, 1p19q loss, and TP53 alteration than with histology. These delineated groups more accurately predict clinical outcomes and help set the stage for molecular - based definition of these tumors in clinical trials and clinical practice.CrossRefPubMed
7.
go back to reference Eckel-Passow JE, Lachance DH, Molinaro AM, et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med. 2015;372(26):2499–508.CrossRefPubMedPubMedCentral Eckel-Passow JE, Lachance DH, Molinaro AM, et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med. 2015;372(26):2499–508.CrossRefPubMedPubMedCentral
8.
go back to reference Ceccarelli M, Barthel FP, Malta TM, et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell. 2016;164(3):550–63.CrossRefPubMed Ceccarelli M, Barthel FP, Malta TM, et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell. 2016;164(3):550–63.CrossRefPubMed
9.
go back to reference Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef
11.
go back to reference Cairncross G, Berkey B, Shaw E, et al. Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402. J Clin Oncol. 2006;24(18):2707–14.CrossRefPubMed Cairncross G, Berkey B, Shaw E, et al. Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402. J Clin Oncol. 2006;24(18):2707–14.CrossRefPubMed
12.
go back to reference Cairncross JG, Wang M, Jenkins RB, et al. Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH. J Clin Oncol. 2014;32(8):783–90.CrossRefPubMedPubMedCentral Cairncross JG, Wang M, Jenkins RB, et al. Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH. J Clin Oncol. 2014;32(8):783–90.CrossRefPubMedPubMedCentral
13.
go back to reference Sturm D, Witt H, Hovestadt V, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22(4):425–37.CrossRefPubMed Sturm D, Witt H, Hovestadt V, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22(4):425–37.CrossRefPubMed
14.
go back to reference Batchelor TT, Mulholland P, Neyns B, et al. 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. 2013;31(26):3212–8.CrossRefPubMedPubMedCentral Batchelor TT, Mulholland P, Neyns B, et al. 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. 2013;31(26):3212–8.CrossRefPubMedPubMedCentral
15.
go back to reference Lee EQ, Kuhn J, Lamborn KR, et al. Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05–02. Neuro-Oncology. 2012;14(12):1511–8.CrossRefPubMedPubMedCentral Lee EQ, Kuhn J, Lamborn KR, et al. Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05–02. Neuro-Oncology. 2012;14(12):1511–8.CrossRefPubMedPubMedCentral
16.
go back to reference Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun. 2005;333(2):328–35.CrossRefPubMed Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun. 2005;333(2):328–35.CrossRefPubMed
17.
18.
go back to reference Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709–22.CrossRefPubMed Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709–22.CrossRefPubMed
19.
go back to reference Sandmann T, Bourgon R, Garcia J, et al. Patients with proneural glioblastoma may derive overall survival benefit from the addition of bevacizumab to first-line radiotherapy and temozolomide: retrospective analysis of the AVAglio trial. J Clin Oncol. 2015;33(25):2735–44.CrossRefPubMed Sandmann T, Bourgon R, Garcia J, et al. Patients with proneural glioblastoma may derive overall survival benefit from the addition of bevacizumab to first-line radiotherapy and temozolomide: retrospective analysis of the AVAglio trial. J Clin Oncol. 2015;33(25):2735–44.CrossRefPubMed
20.
go back to reference Taal W, Oosterkamp HM, Walenkamp AM, et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014;15(9):943–53.CrossRefPubMed Taal W, Oosterkamp HM, Walenkamp AM, et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014;15(9):943–53.CrossRefPubMed
21.
go back to reference Wick W, Brandes A, Gorlia T, et al. LB-05PHASE III trial exploring the combination of bevacizumab and lomustine in patients with first recurrence of a glioblastoma: the EORTC 26101 trial. Neuro-Oncology. 2015;17 suppl 5:v1.CrossRef Wick W, Brandes A, Gorlia T, et al. LB-05PHASE III trial exploring the combination of bevacizumab and lomustine in patients with first recurrence of a glioblastoma: the EORTC 26101 trial. Neuro-Oncology. 2015;17 suppl 5:v1.CrossRef
22.
go back to reference Swartz AM, Li QJ, Sampson JH. Rindopepimut: a promising immunotherapeutic for the treatment of glioblastoma multiforme. Immunotherapy. 2014;6(6):679–90.CrossRefPubMedPubMedCentral Swartz AM, Li QJ, Sampson JH. Rindopepimut: a promising immunotherapeutic for the treatment of glioblastoma multiforme. Immunotherapy. 2014;6(6):679–90.CrossRefPubMedPubMedCentral
23.
go back to reference Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353(19):2012–24.CrossRefPubMed Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353(19):2012–24.CrossRefPubMed
24.
go back to reference Haas-Kogan DA, Prados MD, Tihan T, et al. Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. J Natl Cancer Inst. 2005;97(12):880–7.CrossRefPubMed Haas-Kogan DA, Prados MD, Tihan T, et al. Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. J Natl Cancer Inst. 2005;97(12):880–7.CrossRefPubMed
25.
go back to reference Reardon DA, Wen PY, Mellinghoff IK. Targeted molecular therapies against epidermal growth factor receptor: past experiences and challenges. Neuro-Oncology. 2014;16 Suppl 8:viii7–13.CrossRefPubMedPubMedCentral Reardon DA, Wen PY, Mellinghoff IK. Targeted molecular therapies against epidermal growth factor receptor: past experiences and challenges. Neuro-Oncology. 2014;16 Suppl 8:viii7–13.CrossRefPubMedPubMedCentral
26.
go back to reference Prados MD, Byron SA, Tran NL, et al. Toward precision medicine in glioblastoma: the promise and the challenges. Neuro-Oncology. 2015;17(8):1051–63.CrossRefPubMed Prados MD, Byron SA, Tran NL, et al. Toward precision medicine in glioblastoma: the promise and the challenges. Neuro-Oncology. 2015;17(8):1051–63.CrossRefPubMed
27.
go back to reference Reardon D, Desjardins A, Schuster J, et al. ReACT: long-term survival from a randomized phase II study of rindopepimut (CDX-110) plus bevacizumab in relapsed glioblastoma. Neuro-Oncoloogy. 2015;17 suppl 5:v109. Reardon D, Desjardins A, Schuster J, et al. ReACT: long-term survival from a randomized phase II study of rindopepimut (CDX-110) plus bevacizumab in relapsed glioblastoma. Neuro-Oncoloogy. 2015;17 suppl 5:v109.
29.
go back to reference Reilly EB, Phillips AC, Boghaert ER, et al. ABT-414, an antibody drug conjugate targeting a tumor-selective EGFR epitope. Mol Cancer Ther. 2016;15(4):661–9. Reilly EB, Phillips AC, Boghaert ER, et al. ABT-414, an antibody drug conjugate targeting a tumor-selective EGFR epitope. Mol Cancer Ther. 2016;15(4):661–9.
30.
go back to reference Gan H, Kumthekar P, Lassman A, et al. ABT-414 mono- or combination therapy with temozolomide (TMZ) rechallenge in patients with recurrent glioblastoma (GBM) and amplified epidermal growth factor receptor (EGFR): a phase I study. Neuro-Oncology. 2015;17 Suppl 5:v10. Gan H, Kumthekar P, Lassman A, et al. ABT-414 mono- or combination therapy with temozolomide (TMZ) rechallenge in patients with recurrent glioblastoma (GBM) and amplified epidermal growth factor receptor (EGFR): a phase I study. Neuro-Oncology. 2015;17 Suppl 5:v10.
31.
go back to reference Sampson JH, Mitchell DA. Vaccination strategies for neuro-oncology. Neuro-Oncology. 2015;17 Suppl 7:vii15–25.CrossRefPubMed Sampson JH, Mitchell DA. Vaccination strategies for neuro-oncology. Neuro-Oncology. 2015;17 Suppl 7:vii15–25.CrossRefPubMed
32.
go back to reference Schumacher T, Bunse L, Pusch S, et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature. 2014;512(7514):324–7.CrossRefPubMed Schumacher T, Bunse L, Pusch S, et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature. 2014;512(7514):324–7.CrossRefPubMed
33.
go back to reference Bregy A, Wong TM, Shah AH, Goldberg JM, Komotar RJ. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev. 2013;39(8):891–907.CrossRefPubMed Bregy A, Wong TM, Shah AH, Goldberg JM, Komotar RJ. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev. 2013;39(8):891–907.CrossRefPubMed
34.
go back to reference Ampie L, Choy W, Lamano JB, Fakurnejad S, Bloch O, Parsa AT. Heat shock protein vaccines against glioblastoma: from bench to bedside. J Neuro-Oncol. 2015;123(3):441–8.CrossRef Ampie L, Choy W, Lamano JB, Fakurnejad S, Bloch O, Parsa AT. Heat shock protein vaccines against glioblastoma: from bench to bedside. J Neuro-Oncol. 2015;123(3):441–8.CrossRef
35.
go back to reference Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology. 2014;16(10):1304–12.CrossRefPubMedPubMedCentral Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology. 2014;16(10):1304–12.CrossRefPubMedPubMedCentral
36.
go back to reference Johnson LA, Scholler J, Ohkuri T, et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med. 2015;7(275):275ra222.CrossRef Johnson LA, Scholler J, Ohkuri T, et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med. 2015;7(275):275ra222.CrossRef
38.
go back to reference Wakimoto H, Tanaka S, Curry WT, et al. Targetable signaling pathway mutations are associated with malignant phenotype in IDH-mutant gliomas. Clin Cancer Res. 2014;20(11):2898–909.CrossRefPubMedPubMedCentral Wakimoto H, Tanaka S, Curry WT, et al. Targetable signaling pathway mutations are associated with malignant phenotype in IDH-mutant gliomas. Clin Cancer Res. 2014;20(11):2898–909.CrossRefPubMedPubMedCentral
39.
go back to reference Chen R, Ravindra VM, Cohen AL, et al. Molecular features assisting in diagnosis, surgery, and treatment decision making in low-grade gliomas. Neurosurg Focus. 2015;38(3):E2.CrossRefPubMed Chen R, Ravindra VM, Cohen AL, et al. Molecular features assisting in diagnosis, surgery, and treatment decision making in low-grade gliomas. Neurosurg Focus. 2015;38(3):E2.CrossRefPubMed
40.
go back to reference Tateishi K, Wakimoto H, Iafrate AJ, et al. Extreme vulnerability of IDH1 mutant cancers to NAD+ depletion. Cancer Cell. 2015;28(6):773–84.CrossRefPubMed Tateishi K, Wakimoto H, Iafrate AJ, et al. Extreme vulnerability of IDH1 mutant cancers to NAD+ depletion. Cancer Cell. 2015;28(6):773–84.CrossRefPubMed
41.
go back to reference Duke Comprehensive Cancer Center, Duke University. Patients with IDH1 positive recurrent grade II glioma enrolled in a safety and immunogenicity study of tumor-specific peptide vaccine. In: ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). 2000. https://clinicaltrials.gov/ct2/show/NCT02193347. Accessed March 23, 2016. Duke Comprehensive Cancer Center, Duke University. Patients with IDH1 positive recurrent grade II glioma enrolled in a safety and immunogenicity study of tumor-specific peptide vaccine. In: ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). 2000. https://​clinicaltrials.​gov/​ct2/​show/​NCT02193347. Accessed March 23, 2016.
42.
go back to reference National Center for Tumor Diseases, Heidelberg. Targeting IDH1R132H in WHO Grade III-IV IDH1R132H-mutated gliomas by a peptide vaccine—a phase I safety, tolerability and immunogenicity multicenter trial (NOA-16). In: ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). 2000. https://clinicaltrials.gov/ct2/show/NCT02454634. Accessed March 23, 2016. National Center for Tumor Diseases, Heidelberg. Targeting IDH1R132H in WHO Grade III-IV IDH1R132H-mutated gliomas by a peptide vaccine—a phase I safety, tolerability and immunogenicity multicenter trial (NOA-16). In: ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). 2000. https://​clinicaltrials.​gov/​ct2/​show/​NCT02454634. Accessed March 23, 2016.
43.
44.
go back to reference Flavahan WA, Drier Y, Liau BB, et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature. 2016;529(7584):110–4.CrossRefPubMed Flavahan WA, Drier Y, Liau BB, et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature. 2016;529(7584):110–4.CrossRefPubMed
45.
go back to reference Selby GB, Upchurch C, Townsend J, Eyre HJ. A phase II evaluation of fazarabine in high-grade gliomas: a Southwest Oncology Group study. Cancer Chemother Pharmacol. 1994;34(2):179–80.CrossRefPubMed Selby GB, Upchurch C, Townsend J, Eyre HJ. A phase II evaluation of fazarabine in high-grade gliomas: a Southwest Oncology Group study. Cancer Chemother Pharmacol. 1994;34(2):179–80.CrossRefPubMed
46.
go back to reference Rheinbay E, Louis DN, Bernstein BE, Suva ML. A tell-tail sign of chromatin: histone mutations drive pediatric glioblastoma. Cancer Cell. 2012;21(3):329–31.CrossRefPubMed Rheinbay E, Louis DN, Bernstein BE, Suva ML. A tell-tail sign of chromatin: histone mutations drive pediatric glioblastoma. Cancer Cell. 2012;21(3):329–31.CrossRefPubMed
47.
go back to reference Majuelos-Melguizo J, Rodriguez MI, Lopez-Jimenez L, et al. PARP targeting counteracts gliomagenesis through induction of mitotic catastrophe and aggravation of deficiency in homologous recombination in PTEN-mutant glioma. Oncotarget. 2015;6(7):4790–803.CrossRefPubMed Majuelos-Melguizo J, Rodriguez MI, Lopez-Jimenez L, et al. PARP targeting counteracts gliomagenesis through induction of mitotic catastrophe and aggravation of deficiency in homologous recombination in PTEN-mutant glioma. Oncotarget. 2015;6(7):4790–803.CrossRefPubMed
48.
go back to reference Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-kappaB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother. 2014;14(11):1293–306.CrossRefPubMedPubMedCentral Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-kappaB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother. 2014;14(11):1293–306.CrossRefPubMedPubMedCentral
49.
go back to reference Wen PY, Chang SM, Lamborn KR, et al. Phase I/II study of erlotinib and temsirolimus for patients with recurrent malignant gliomas: North American Brain Tumor Consortium trial 04–02. Neuro-Oncology. 2014;16(4):567–78.CrossRefPubMedPubMedCentral Wen PY, Chang SM, Lamborn KR, et al. Phase I/II study of erlotinib and temsirolimus for patients with recurrent malignant gliomas: North American Brain Tumor Consortium trial 04–02. Neuro-Oncology. 2014;16(4):567–78.CrossRefPubMedPubMedCentral
50.
go back to reference Chinnaiyan P, Won M, Wen PY, et al. RTOG 0913: a phase 1 study of daily everolimus (RAD001) in combination with radiation therapy and temozolomide in patients with newly diagnosed glioblastoma. Int J Radiat Oncol Biol Phys. 2013;86(5):880–4.CrossRefPubMedPubMedCentral Chinnaiyan P, Won M, Wen PY, et al. RTOG 0913: a phase 1 study of daily everolimus (RAD001) in combination with radiation therapy and temozolomide in patients with newly diagnosed glioblastoma. Int J Radiat Oncol Biol Phys. 2013;86(5):880–4.CrossRefPubMedPubMedCentral
51.
go back to reference Mrugala MM, Engelhard HH, Dinh Tran D, et al. Clinical practice experience with NovoTTF-100A system for glioblastoma: the Patient Registry Dataset (PRiDe). Semin Oncol. 2014;41 Suppl 6:S4–13.CrossRefPubMed Mrugala MM, Engelhard HH, Dinh Tran D, et al. Clinical practice experience with NovoTTF-100A system for glioblastoma: the Patient Registry Dataset (PRiDe). Semin Oncol. 2014;41 Suppl 6:S4–13.CrossRefPubMed
52.••
go back to reference Johnson BE, Mazor T, Hong C, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science. 2014;343(6167):189–93. Study of the clonal evolution of tumor recurrences in low-grade gliomas demonstrating that the sequential recurrences of a tumor may derive phylogenetically from different earlier evolutionary stages of the initial tumor and therefore contain divergent driver mutations that need to be treated differently. This study elucidates one of the great challenges of targeted therapies and may explain one of the reasons single-agent targeted therapies have failed to demonstrate efficacy to date in these tumors.CrossRefPubMed Johnson BE, Mazor T, Hong C, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science. 2014;343(6167):189–93. Study of the clonal evolution of tumor recurrences in low-grade gliomas demonstrating that the sequential recurrences of a tumor may derive phylogenetically from different earlier evolutionary stages of the initial tumor and therefore contain divergent driver mutations that need to be treated differently. This study elucidates one of the great challenges of targeted therapies and may explain one of the reasons single-agent targeted therapies have failed to demonstrate efficacy to date in these tumors.CrossRefPubMed
53.
go back to reference van Thuijl HF, Mazor T, Johnson BE, et al. Evolution of DNA repair defects during malignant progression of low-grade gliomas after temozolomide treatment. Acta Neuropathol. 2015;129(4):597–607.CrossRefPubMedPubMedCentral van Thuijl HF, Mazor T, Johnson BE, et al. Evolution of DNA repair defects during malignant progression of low-grade gliomas after temozolomide treatment. Acta Neuropathol. 2015;129(4):597–607.CrossRefPubMedPubMedCentral
Metadata
Title
Targeted Therapeutics in Patients With High-Grade Gliomas: Past, Present, and Future
Authors
Ricky Chen, MD
Adam L. Cohen, MD
Howard Colman, MD, PhD
Publication date
01-08-2016
Publisher
Springer US
Published in
Current Treatment Options in Oncology / Issue 8/2016
Print ISSN: 1527-2729
Electronic ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-016-0418-0

Other articles of this Issue 8/2016

Current Treatment Options in Oncology 8/2016 Go to the issue

Upper Gastrointestinal Cancers (L Rajdev, Section Editor)

Advanced Hepatocellular Cancer: the Current State of Future Research

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