Skip to main content

Advertisement

Log in

Epidermal growth factor receptor (EGFR) amplification rates observed in screening patients for randomized trials in glioblastoma

  • Clinical Study
  • Published:
Journal of Neuro-Oncology Aims and scope Submit manuscript

Abstract

Purpose

Epidermal growth factor receptor (EGFR) amplification has been reported to occur in ~ 50% of glioblastomas (GBMs). We are conducting several global studies that require central testing for EGFR amplification during screening, representing an opportunity to confirm the frequency of amplification in GBM in a large cohort and to evaluate whether EGFR amplification differs by region of the world.

Methods

EGFR amplification was measured by fluorescence in situ hybridization during screening for therapeutic trials of an EGFR antibody–drug conjugate: two Phase 2/3 global trials (INTELLANCE-1, INTELLANCE-2), and a Japanese Phase 1/2 trial (INTELLANCE-J). We evaluated the proportion of tumor tissue samples harboring EGFR amplification among those tested and differences in amplification frequency by geography.

Results

EGFR was amplified in 54% of 3150 informative cases screened for INTELLANCE-1 and -2, consistent with historic controls, but was significantly lower in patients from Asia versus the rest of the world (35% vs. 56%, P < 0.0030). The independent INTELLANCE-J trial validated this finding (33% amplified of 153 informative cases).

Conclusions

EGFR amplification occurs less frequently in patients from Asia than elsewhere. Further study is required to understand biological differences to optimize treatment in glioblastoma.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, Zou L, Vegesna R, Shukla SA, Ciriello G, Yung WK, Zhang W, Sougnez C, Mikkelsen T, Aldape K, Bigner DD, Van Meir EG, Prados M, Sloan A, Black KL, Eschbacher J, Finocchiaro G, Friedman W, Andrews DW, Guha A, Iacocca M, O'Neill BP, Foltz G, Myers J, Weisenberger DJ, Penny R, Kucherlapati R, Perou CM, Hayes DN, Gibbs R, Marra M, Mills GB, Lander E, Spellman P, Wilson R, Sander C, Weinstein J, Meyerson M, Gabriel S, Laird PW, Haussler D, Getz G, Chin L, Network TR (2013) The somatic genomic landscape of glioblastoma. Cell 155:462–477. https://doi.org/10.1016/j.cell.2013.09.034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gan HK, Cvrljevic AN, Johns TG (2013) The epidermal growth factor receptor variant III (EGFRvIII): where wild things are altered. FEBS J 280:5350–5370. https://doi.org/10.1111/febs.12393

    Article  CAS  PubMed  Google Scholar 

  3. Yoshimoto K, Dang J, Zhu S, Nathanson D, Huang T, Dumont R, Seligson DB, Yong WH, Xiong Z, Rao N, Winther H, Chakravarti A, Bigner DD, Mellinghoff IK, Horvath S, Cavenee WK, Cloughesy TF, Mischel PS (2008) Development of a real-time RT-PCR assay for detecting EGFRvIII in glioblastoma samples. Clin Cancer Res 14:488–493. https://doi.org/10.1158/1078-0432.CCR-07-1966

    Article  CAS  PubMed  Google Scholar 

  4. van den Bent MJ, Gao Y, Kerkhof M, Kros JM, Gorlia T, van Zwieten K, Prince J, van Duinen S, Sillevis Smitt PA, Taphoorn M, French PJ (2015) Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro Oncol 17:935–941. https://doi.org/10.1093/neuonc/nov013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ahluwalia MS, Dimino C, Mansukhani MM, Murty VV, Canoll P, Narita Y, Muragaki Y, Gan HK, Merrell R, van den Bent MJ, Roberts-Rapp L, Jiang F, Guseva M, Bain EE, Ocampo CJ, Ansell PJ, Lassman AB (2018) Effect of therapeutic pressure on stability of EGFR amplification in glioblastoma. J Clin Oncol 36:2033. https://doi.org/10.1200/JCO.2018.36.15_suppl.2033

    Article  Google Scholar 

  6. Phillips AC, Boghaert ER, Vaidya KS, Mitten MJ, Norvell S, Falls HD, DeVries PJ, Cheng D, Meulbroek JA, Buchanan FG, McKay LM, Goodwin NC, Reilly EB (2016) ABT-414, an antibody-drug conjugate targeting a tumor-selective EGFR epitope. Mol Cancer Ther 15:661–669. https://doi.org/10.1158/1535-7163.MCT-15-0901

    Article  CAS  PubMed  Google Scholar 

  7. Li J, Wang M, Won M, Shaw EG, Coughlin C, Curran WJ Jr, Mehta MP (2011) Validation and simplification of the Radiation Therapy Oncology Group recursive partitioning analysis classification for glioblastoma. Int J Radiat Oncol Biol Phys 81:623–630. https://doi.org/10.1016/j.ijrobp.2010.06.012

    Article  PubMed  Google Scholar 

  8. van den Bent M, Eoli M, Sepulveda JM, Smits M, Walenkamp AM, Frenel JS, Brandes AA, Clement P, French P, Dubbink H, Kros J, Weller M, Chinot O, de Vos F, Whenham N, Sanghera P, Looman J, Lee HJ, de Geus JP, Nuyens S, Spruyt M, Brilhante J, Gorlia T, Golfinopoulos V (2017) First results of the randomized Phase II study on depatux-m alone, depatux-m in combination with temozolomide and either temozolomide or lomustine in recurrent EGFR amplified glioblastoma: first report from INTELLANCE 2/EORTC trial 1410. Neuro Oncol 19:vi316. https://doi.org/10.1093/neuonc/nox213

  9. van den Bent MJ, French P, Eoli M, Sepulveda JM, Walenkamp AM, Frenel JS, Franceschi E, Clement PM, Weller M, de Heer I, Looman J, Dey J, Krause S, Xiong H, Ansell PJ, Nuyens S, Spruyt M, Brilhante J, Gorlia T, Golfinopoulos V (2018) Updated results of the INTELLANCE 2/EORTC trial 1410 randomized phase II study on Depatux-M alone, Depatux-M in combination with temozolomide (TMZ) and eitehr TMZ or lomustine (LOM) in recurrent EGFR amplified glioblastoma (NCT02343406). J Clin Oncol 36:2023. https://doi.org/10.1200/JCO.2018.36.15_suppl.2023

    Article  Google Scholar 

  10. Narita Y, Nagane M, Kagawa N, Mishima K, Yamamoto T, Wakabayashi T, Hamada T, Odagawa R, Nishimura Y, Kiriyama T, Xiong H, Ocampo C, Nishikawa R (2016) 146P Tolerability and pharmacokinetics (PK) of ABT-414 in Japanese patients (pts) with recurrent malignant glioma. Ann Oncol 27:mdw578.009–mdw578.009. https://doi.org/10.1093/annonc/mdw578.009

  11. Lassman AB, Roberts-Rapp LA, Sokolova I, Song M, Pestova E, Kular R, Mullen C, Zha Z, Lu X, Gomez E, Bhathena A, Maag D, Kumthekar P, Gan HK, Scott AM, Guseva M, Holen KD, Ansell P, van den Bent MJ (2019) Comparison of biomarker assays for EGFR: implications for precision medicine in patients with glioblastoma. Clin Cancer Res. https://doi.org/10.1158/1078-0432.CCR-18-3034

  12. Qi ST, Yu L, Lu YT, Ou YH, Li ZY, Wu LX, Yao F (2011) IDH mutations occur frequently in Chinese glioma patients and predict longer survival but not response to concomitant chemoradiotherapy in anaplastic gliomas. Oncol Rep 26:1479–1485. https://doi.org/10.3892/or.2011.1428

    Article  CAS  PubMed  Google Scholar 

  13. Yang P, Zhang W, Wang Y, Peng X, Chen B, Qiu X, Li G, Li S, Wu C, Yao K, Li W, Yan W, Li J, You Y, Chen CC, Jiang T (2015) IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry. Oncotarget 6:40896–40906. https://doi.org/10.18632/oncotarget.5683

    Article  PubMed  PubMed Central  Google Scholar 

  14. Yan W, Zhang W, You G, Bao Z, Wang Y, Liu Y, Kang C, You Y, Wang L, Jiang T (2012) Correlation of IDH1 mutation with clinicopathologic factors and prognosis in primary glioblastoma: a report of 118 patients from China. PLoS ONE 7:e30339. https://doi.org/10.1371/journal.pone.0030339

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yan W, Zhang W, You G, Zhang J, Han L, Bao Z, Wang Y, Liu Y, Jiang C, Kang C, You Y, Jiang T (2012) Molecular classification of gliomas based on whole genome gene expression: a systematic report of 225 samples from the Chinese Glioma Cooperative Group. Neuro Oncol 14:1432–1440. https://doi.org/10.1093/neuonc/nos263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lv S, Teugels E, Sadones J, De Brakeleer S, Duerinck J, Du Four S, Michotte A, De Greve J, Neyns B (2012) Correlation of EGFR, IDH1 and PTEN status with the outcome of patients with recurrent glioblastoma treated in a phase II clinical trial with the EGFR-blocking monoclonal antibody cetuximab. Int J Oncol 41:1029–1035. https://doi.org/10.3892/ijo.2012.1539

    Article  CAS  PubMed  Google Scholar 

  17. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu VE, Vogelstein B, Bigner DD (2009) IDH1 and IDH2 mutations in gliomas. N Engl J Med 360:765–773. https://doi.org/10.1056/NEJMoa0808710

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O'Kelly M, Tamayo P, Weir BA, Gabriel S, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN, Cancer Genome Atlas Research N (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17:98–110. https://doi.org/10.1016/j.ccr.2009.12.020

    Article  CAS  Google Scholar 

  19. Kim HR, Shim HS, Chung JH, Lee YJ, Hong YK, Rha SY, Kim SH, Ha SJ, Kim SK, Chung KY, Soo R, Kim JH, Cho BC (2012) Distinct clinical features and outcomes in never-smokers with nonsmall cell lung cancer who harbor EGFR or KRAS mutations or ALK rearrangement. Cancer 118:729–739. https://doi.org/10.1002/cncr.26311

    Article  CAS  PubMed  Google Scholar 

  20. Midha A, Dearden S, McCormack R (2015) EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII). Am J Cancer Res 5:2892–2911

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors and AbbVie would like to thank patients and their families/caregivers; study investigators and staff; Mrinal Y. Shah, PhD for medical writing support, and Yan Sun, PhD for statistical support, both employees of AbbVie, Inc.

Funding

AbbVie provided financial support for these studies (NCT02573324, NCT02343406, NCT02590263) and participated in the design, study conduct, analysis and interpretation of the data, as well as the writing, review, and approval of the manuscript. All authors were involved in the data gathering, analysis, review, interpretation and manuscript preparation and approval. A.B. Lassman was supported in part Voices Against Brain Cancer, the William Rhodes and Louise Tilzer-Rhodes Center for Glioblastoma at NewYork-Presbyterian Hospital, and grants P30CA013696 and UG1CA189960 from the National Cancer Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute/National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Contribution to study design—ABL, PJA, EB, WC, PJF, JL, MM, CO, MAV, TJCW, PZ, MJvdB. Study conduct and patient treatment—ABL, KDA, EB, WC, ME, MK, JL, YM, YN, CO, MAV, AMEW, TJCW, MJvdB. Data collection, analysis, and interpretation—ABL, PJA, LRR, MS, MJvdB. Manuscript review and approval of the final version for submission—All authors.

Corresponding author

Correspondence to Andrew B. Lassman.

Ethics declarations

Conflict of interest

Andrew B. Lassman: In the last 36 months, Dr. Lassman reports grants, personal fees and non-financial support from AbbVie, during the conduct of the study; personal fees and non-financial support from Orbus, grants, personal fees and non-financial support from Karyopharm, personal fees and non-financial support from NW Biotherapeutics, grants and non-financial support from Oncoceutics, personal fees and non-financial support from Agios, personal fees and non-financial support from Celgene, personal fees and non-financial support from Novocure, non-financial support from Tocagen, non-financial support from BMS, grants, personal fees and non-financial support from Kadmon, grants, personal fees and non-financial support from Roche/Genentech, grants and non-financial support from Amgen, grants and non-financial support from Millenium, grants and non-financial support from Celldex, grants and non-financial support from Novartis, grants and non-financial support from Pfizer, non-financial support from Keryx/Aeterna Zentaris, grants and non-financial support from VBI Vaccines, grants and non-financial support from Beigene, personal fees from Bioclinica as an expert blinded independent reviewer of clinical and imaging data for a BMS-sponsored trial, personal fees from prIME Oncology, personal fees from Sapience, personal fees from WebMD, personal fees from Physicians' Education Resource/Chemotherapy Foundation Symposium, personal fees from Astra Zeneca, personal fees from Cortice, outside the submitted work. Kenneth D. Aldape: Nothing to disclose. Peter J. Ansell, Earle Bain, Jim Looman, Christopher Ocampo, Lisa Roberts-Rapp: Employees of AbbVie and may own stock/options. Walter J. Curran: Consultant for AstraZeneca and BMS; ownership shares of Nanthealth. Marica Eoli: Nothing to disclose. Pim J. French: Research funding from AbbVie. Manabu Kinoshita: Honoraria from Eisai, BrainLab, Nihon Medi-Physics, Canon Medical Systems, Integra Japan, Novocure, Daiichi-Sankyo, Chugai; research grants from Chugai, Daiichi-Sankyo, Takeda, Ohtsuka. Minesh Mehta: Consultant for Agenus, Insys, Remedy, IBA, Varian, Celgene, AbbVie, AstraZeneca, Tocagen, Blue Earth Diagnostics; board of directors with stock options, Oncoceutics. Yoshihiro Muragaki: Consulting/advisory role for AbbVie, Daiichi Sankyo; speakers’ bureau for MSD, Daiichi Sankyo, Chugai Pharma, Otsuka, Eisai, Novartis, Hitachi, Bristol-Myers Squibb Japan; research funding to institution from MSD, Daiichi Sankyo, Chugai Pharma, Otsuka, Eisai, Hitachi. Yoshitaka Narita: Grants and lectures fees from Chugai Pharmaceutical Co., Ono Pharmaceutical Co., AbbVie, Dainippon-Sumitomo, Daiichi Sankyo, Eisai, Stella-Pharma, Ohtuka, Meiji-Seika, SBI Pharma. Minghao Song: Employee of Abbott Molecular and may own stock/options. Michael A. Vogelbaum: Indirect equity and royalty interests in Infuseon Therapeutics, Inc. Annemiek Walenkamp: Research grants from Novartis, Ipsen; advisory board member for Ipsen, Novartis, Polyphor, Karyopharm; study budget from Karyopharm, BMS, Genzyme, Roche. Tony J. C. Wang: Personal fees and non-financial support from AbbVie; outside of the submitted work, consultant for Elekta, Merck, Doximity; advisory board and travel expenses for Novocure, AstraZeneca; honoraria from Elekta, Cancer Panels; stock options from Doximity; royalties from Wolters Kluwer. Peixin Zhang: Nothing to disclose. Martin J. van den Bent: Personal fees from AbbVie, Celgene, Genenta, BMS, Agios, Boehringer Ingelheim; research funding from AbbVie.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Electronic supplementary material 1 (DOCX 160 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lassman, A.B., Aldape, K.D., Ansell, P.J. et al. Epidermal growth factor receptor (EGFR) amplification rates observed in screening patients for randomized trials in glioblastoma. J Neurooncol 144, 205–210 (2019). https://doi.org/10.1007/s11060-019-03222-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11060-019-03222-y

Keywords

Navigation