Defining EGFR amplification status for clinical trial inclusion

Abstract Background Precision medicine trials targeting the epidermal growth factor receptor (EGFR) in glioblastoma patients require selection for EGFR-amplified tumors. However, there is currently no gold standard in determining the amplification status of EGFR or variant III (EGFRvIII) expression. Here, we aimed to determine which technique and which cutoffs are suitable to determine EGFR amplification status. Methods We compared fluorescence in-situ hybridization (FISH) and real-time quantitative (RT-q)PCR data from patients screened for trial inclusion into the Intellance 2 clinical trial, with data from a panel-based next generation sequencing (NGS) platform (both DNA and RNA). Results By using data from >1000 samples, we show that at least 50% of EGFR amplified nuclei should be present to define EGFR gene amplification by FISH. Gene amplification (as determined by FISH) correlates with EGFR expression levels (as determined by RT-qPCR) with receiver operating characteristics analysis showing an area under the curve of up to 0.902. EGFR expression as assessed by RT-qPCR therefore may function as a surrogate marker for EGFR amplification. Our NGS data show that EGFR copy numbers can strongly vary between tumors, with levels ranging from 2 to more than 100 copies per cell. Levels exceeding 5 gene copies can be used to define EGFR-amplification by NGS; below this level, FISH detects very few (if any) EGFR amplified nuclei and none of the samples express EGFRvIII. Conclusion Our data from central laboratories and diagnostic sequencing facilities, using material from patients eligible for clinical trial inclusion, help define the optimal cutoff for various techniques to determine EGFR amplification for diagnostic purposes.

[1]  Jong-Hee Chang,et al.  Targeted next-generation sequencing panel (TruSight Tumor 170) in diffuse glioma: a single institutional experience of 135 cases , 2019, Journal of Neuro-Oncology.

[2]  T. Mikkelsen,et al.  Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial , 2018, Neuro-oncology.

[3]  P. French,et al.  Finding the Right Way to Target EGFR in Glioblastomas; Lessons from Lung Adenocarcinomas , 2018, Cancers.

[4]  M. J. van den Bent,et al.  Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma , 2018, Neuro-oncology.

[5]  A. Tolcher,et al.  Efficacy and safety results of depatuxizumab mafodotin (ABT‐414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor , 2018, Cancer.

[6]  Hao Xiong,et al.  Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: results from a multi-center, international study , 2017, Cancer Chemotherapy and Pharmacology.

[7]  Act Investigators Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial , 2017 .

[8]  G. Reifenberger,et al.  Epidermal Growth Factor Receptor Variant III (EGFRvIII) Positivity in EGFR-Amplified Glioblastomas: Prognostic Role and Comparison between Primary and Recurrent Tumors , 2017, Clinical Cancer Research.

[9]  Jessica M. Rusert,et al.  Extrachromosomal oncogene amplification drives tumor evolution and genetic heterogeneity , 2017, Nature.

[10]  In-Hee Lee,et al.  Clonal evolution of glioblastoma under therapy , 2016, Nature Genetics.

[11]  Steven J. M. Jones,et al.  Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma , 2016, Cell.

[12]  M. J. van den Bent,et al.  Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. , 2015, Neuro-oncology.

[13]  Jun Yao,et al.  Development of Resistance to EGFR-Targeted Therapy in Malignant Glioma Can Occur through EGFR-Dependent and -Independent Mechanisms. , 2015, Cancer research.

[14]  Marc Choisy,et al.  Determination of cut-off cycle threshold values in routine RT–PCR assays to assist differential diagnosis of norovirus in children hospitalized for acute gastroenteritis , 2015, Epidemiology and Infection.

[15]  Na Li,et al.  Rational development and characterization of humanized anti–EGFR variant III chimeric antigen receptor T cells for glioblastoma , 2015, Science Translational Medicine.

[16]  M. J. van den Bent,et al.  A phase 1 study evaluating ABT-414 in combination with temozolomide (TMZ) for subjects with recurrent or unresectable glioblastoma (GBM). , 2014 .

[17]  S. Nelson,et al.  Targeted Therapy Resistance Mediated by Dynamic Regulation of Extrachromosomal Mutant EGFR DNA , 2014, Science.

[18]  D. Haussler,et al.  The Somatic Genomic Landscape of Glioblastoma , 2013, Cell.

[19]  J. Pollack,et al.  EGFRvIII gene rearrangement is an early event in glioblastoma tumorigenesis and expression defines a hierarchy modulated by epigenetic mechanisms , 2013, Oncogene.

[20]  W. Curran,et al.  RTOG 0211: a phase 1/2 study of radiation therapy with concurrent gefitinib for newly diagnosed glioblastoma patients. , 2013, International journal of radiation oncology, biology, physics.

[21]  Paul S Mischel,et al.  Differential sensitivity of glioma- versus lung cancer-specific EGFR mutations to EGFR kinase inhibitors. , 2012, Cancer discovery.

[22]  Rebecca A Betensky,et al.  Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. , 2011, Cancer cell.

[23]  C. James,et al.  Phase II evaluation of gefitinib in patients with newly diagnosed Grade 4 astrocytoma: Mayo/North Central Cancer Treatment Group Study N0074. , 2011, International journal of radiation oncology, biology, physics.

[24]  Susan M. Chang,et al.  A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. , 2010, Neuro-oncology.

[25]  M. J. van den Bent,et al.  Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[26]  S. Gabriel,et al.  Epidermal Growth Factor Receptor Activation in Glioblastoma through Novel Missense Mutations in the Extracellular Domain , 2006, PLoS medicine.

[27]  C. James,et al.  Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas. , 2000, Cancer research.