Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 6/2023

28-01-2023 | Glioblastoma | Research

Using EGFR amplification to stratify recurrent glioblastoma treated with immune checkpoint inhibitors

Authors: Joshua S. Friedman, Tomi Jun, Omid Rashidipour, Kuan-lin Huang, Ethan Ellis, Priyanka Kadaba, Puneet Belani, Kambiz Nael, Nadejda M. Tsankova, Robert Sebra, Adília Hormigo

Published in: Cancer Immunology, Immunotherapy | Issue 6/2023

Login to get access

Abstract

Purpose

While immune checkpoint inhibitors (ICI) have had success with various malignancies, their efficacy in brain cancer is still unclear. Retrospective and prospective studies using PD-1 inhibitors for recurrent glioblastoma (GBM) have not established survival benefit. This study evaluated if ICI may be effective for select patients with recurrent GBM.

Methods

This was a single-center retrospective study of adult patients diagnosed with first recurrence GBM and received pembrolizumab or nivolumab with or without concurrent bevacizumab. Archival tissue was used for immunohistochemistry (IHC) and targeted DNA next-generation sequencing (NGS) analysis.

Results

Median overall survival (mOS) from initial diagnosis was 24.5 months (range 10–42). mOS from onset of ICI was 10 months (range 1–31) with 75% surviving > 6 months and 46% > 12 months. Additional IHC analysis on tumors from eight patients demonstrated a trend of longer survival after ICI for those with elevated PD-L1 expression. NGS of samples from 15 patients identified EGFR amplification at initial diagnosis and at any time point to be associated with worse survival after ICI (HR 12.2, 95% CI 1.37–108, p = 0.025 and HR 3.92, 95% CI 1.03–14.9, p = 0.045, respectively). This significance was corroborated with previously tested EGFR amplification via in situ hybridization.

Conclusion

ICI did not extend overall survival for recurrent GBM. However, molecular sequencing identified EGFR amplification as associated with worse survival. Prospective studies can validate if EGFR amplification is a biomarker of ICI resistance and determine if its use can stratify responders from non-responders.
Appendix
Available only for authorised users
Literature
1.
go back to reference Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27(28):4733–4740CrossRefPubMed Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE et al (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27(28):4733–4740CrossRefPubMed
2.
go back to reference Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B et al (2015) Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 16(4):375–384CrossRefPubMed Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B et al (2015) Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 16(4):375–384CrossRefPubMed
3.
go back to reference Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T et al (2018) Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol 20(5):674–686CrossRefPubMed Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T et al (2018) Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol 20(5):674–686CrossRefPubMed
4.
go back to reference Kurz SC, Cabrera LP, Hastie D, Huang R, Unadkat P, Rinne M et al (2018) PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology 91(14):e1355–e1359CrossRefPubMed Kurz SC, Cabrera LP, Hastie D, Huang R, Unadkat P, Rinne M et al (2018) PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology 91(14):e1355–e1359CrossRefPubMed
5.
go back to reference Chamberlain MC, Kim BT (2017) Nivolumab for patients with recurrent glioblastoma progressing on bevacizumab: a retrospective case series. J Neurooncol 133(3):561–569CrossRefPubMed Chamberlain MC, Kim BT (2017) Nivolumab for patients with recurrent glioblastoma progressing on bevacizumab: a retrospective case series. J Neurooncol 133(3):561–569CrossRefPubMed
6.
go back to reference Reiss SN, Yerram P, Modelevsky L, Grommes C (2017) Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J Immunother Cancer 5(1):99CrossRefPubMedPubMedCentral Reiss SN, Yerram P, Modelevsky L, Grommes C (2017) Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J Immunother Cancer 5(1):99CrossRefPubMedPubMedCentral
7.
go back to reference Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A et al (2020) Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the Checkmate 143 phase 3 randomized clinical trial. JAMA Oncol 6(7):1003–1010CrossRefPubMed Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A et al (2020) Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the Checkmate 143 phase 3 randomized clinical trial. JAMA Oncol 6(7):1003–1010CrossRefPubMed
8.
go back to reference Nayak L, Molinaro AM, Peters K, Clarke JL, Jordan JT, de Groot J et al (2021) Randomized phase II and biomarker study of pembrolizumab plus bevacizumab versus pembrolizumab alone for patients with recurrent glioblastoma. Clin Cancer Res 27(4):1048–1057CrossRefPubMed Nayak L, Molinaro AM, Peters K, Clarke JL, Jordan JT, de Groot J et al (2021) Randomized phase II and biomarker study of pembrolizumab plus bevacizumab versus pembrolizumab alone for patients with recurrent glioblastoma. Clin Cancer Res 27(4):1048–1057CrossRefPubMed
9.
go back to reference Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB et al (2019) Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med 25(3):477–486CrossRefPubMedPubMedCentral Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB et al (2019) Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med 25(3):477–486CrossRefPubMedPubMedCentral
10.
go back to reference Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY et al (2019) Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 51(2):202–206CrossRefPubMedPubMedCentral Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY et al (2019) Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 51(2):202–206CrossRefPubMedPubMedCentral
11.
go back to reference Mantica M, Pritchard A, Lieberman F, Drappatz J (2018) Retrospective study of nivolumab for patients with recurrent high grade gliomas. J Neurooncol 139(3):625–631CrossRefPubMed Mantica M, Pritchard A, Lieberman F, Drappatz J (2018) Retrospective study of nivolumab for patients with recurrent high grade gliomas. J Neurooncol 139(3):625–631CrossRefPubMed
12.
go back to reference Ahluwalia MS, Rauf Y, Li H, Wen PY, Peereboom DM, Reardon DA (2021) Randomized phase 2 study of nivolumab (nivo) plus either standard or reduced dose bevacizumab (bev) in recurrent glioblastoma (rGBM). J Clin Oncol. 39(15 suppl):2015–2015CrossRef Ahluwalia MS, Rauf Y, Li H, Wen PY, Peereboom DM, Reardon DA (2021) Randomized phase 2 study of nivolumab (nivo) plus either standard or reduced dose bevacizumab (bev) in recurrent glioblastoma (rGBM). J Clin Oncol. 39(15 suppl):2015–2015CrossRef
13.
go back to reference Reardon DA, Kim TM, Frenel JS, Simonelli M, Lopez J, Subramaniam DS et al (2021) Treatment with pembrolizumab in programmed death ligand 1-positive recurrent glioblastoma: results from the multicohort phase 1 KEYNOTE-028 trial. Cancer 127(10):1620–1629CrossRefPubMed Reardon DA, Kim TM, Frenel JS, Simonelli M, Lopez J, Subramaniam DS et al (2021) Treatment with pembrolizumab in programmed death ligand 1-positive recurrent glioblastoma: results from the multicohort phase 1 KEYNOTE-028 trial. Cancer 127(10):1620–1629CrossRefPubMed
14.
go back to reference Morrison C, Pabla S, Conroy JM, Nesline MK, Glenn ST, Dressman D et al (2018) Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden. J Immunother Cancer 6(1):32CrossRefPubMedPubMedCentral Morrison C, Pabla S, Conroy JM, Nesline MK, Glenn ST, Dressman D et al (2018) Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden. J Immunother Cancer 6(1):32CrossRefPubMedPubMedCentral
15.
go back to reference Restrepo P, Yong R, Laface I, Tsankova N, Nael K, Akturk G et al (2020) Tumoral and immune heterogeneity in an anti-PD-1-responsive glioblastoma: a case study. Cold Spring Harb Mol Case Stud. 6(2):a004762CrossRefPubMedPubMedCentral Restrepo P, Yong R, Laface I, Tsankova N, Nael K, Akturk G et al (2020) Tumoral and immune heterogeneity in an anti-PD-1-responsive glioblastoma: a case study. Cold Spring Harb Mol Case Stud. 6(2):a004762CrossRefPubMedPubMedCentral
16.
go back to reference Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M et al (2016) Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol 34(19):2206–2211CrossRefPubMed Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M et al (2016) Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol 34(19):2206–2211CrossRefPubMed
17.
18.
go back to reference Wang H, Wang X, Xu L, Zhang J, Cao H (2021) Analysis of the EGFR amplification and CDKN2A deletion regulated transcriptomic signatures reveals the prognostic significance of SPATS2L in patients with glioma. Front Oncol. 11:551160CrossRefPubMedPubMedCentral Wang H, Wang X, Xu L, Zhang J, Cao H (2021) Analysis of the EGFR amplification and CDKN2A deletion regulated transcriptomic signatures reveals the prognostic significance of SPATS2L in patients with glioma. Front Oncol. 11:551160CrossRefPubMedPubMedCentral
19.
go back to reference Li J, Liang R, Song C, Xiang Y, Liu Y (2018) Prognostic significance of epidermal growth factor receptor expression in glioma patients. Onco Targets Ther 11:731–742CrossRefPubMedPubMedCentral Li J, Liang R, Song C, Xiang Y, Liu Y (2018) Prognostic significance of epidermal growth factor receptor expression in glioma patients. Onco Targets Ther 11:731–742CrossRefPubMedPubMedCentral
20.
go back to reference Santaniello A, Napolitano F, Servetto A, De Placido P, Silvestris N, Bianco C et al (2019) Tumour microenvironment and immune evasion in EGFR addicted NSCLC: hurdles and possibilities. Cancers Basel 11(10):1419CrossRefPubMedPubMedCentral Santaniello A, Napolitano F, Servetto A, De Placido P, Silvestris N, Bianco C et al (2019) Tumour microenvironment and immune evasion in EGFR addicted NSCLC: hurdles and possibilities. Cancers Basel 11(10):1419CrossRefPubMedPubMedCentral
21.
go back to reference Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z et al (2016) EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res 22(18):4585–4593CrossRefPubMedPubMedCentral Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z et al (2016) EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res 22(18):4585–4593CrossRefPubMedPubMedCentral
22.
go back to reference Lee CK, Man J, Lord S, Links M, Gebski V, Mok T et al (2017) Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer—a meta-analysis. J Thorac Oncol 12(2):403–407CrossRefPubMed Lee CK, Man J, Lord S, Links M, Gebski V, Mok T et al (2017) Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer—a meta-analysis. J Thorac Oncol 12(2):403–407CrossRefPubMed
23.
go back to reference Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE et al (2015) Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373(17):1627–1639CrossRefPubMedPubMedCentral Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE et al (2015) Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373(17):1627–1639CrossRefPubMedPubMedCentral
24.
25.
go back to reference Dong ZY, Zhang JT, Liu SY, Su J, Zhang C, Xie Z et al (2017) EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 6(11):e1356145CrossRefPubMedPubMedCentral Dong ZY, Zhang JT, Liu SY, Su J, Zhang C, Xie Z et al (2017) EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 6(11):e1356145CrossRefPubMedPubMedCentral
26.
go back to reference Shaim H, Shanley M, Basar R, Daher M, Gumin J, Zamler DB et al (2021) Targeting the alphav integrin/TGF-beta axis improves natural killer cell function against glioblastoma stem cells. J Clin Invest. 131(14):9–21CrossRef Shaim H, Shanley M, Basar R, Daher M, Gumin J, Zamler DB et al (2021) Targeting the alphav integrin/TGF-beta axis improves natural killer cell function against glioblastoma stem cells. J Clin Invest. 131(14):9–21CrossRef
27.
go back to reference Liu C, Zheng S, Wang Z, Wang S, Wang X, Yang L et al (2022) KRAS-G12D mutation drives immune suppression and the primary resistance of anti-PD-1/PD-L1 immunotherapy in non-small cell lung cancer. Cancer Commun Lond 42(9):828–847CrossRefPubMedPubMedCentral Liu C, Zheng S, Wang Z, Wang S, Wang X, Yang L et al (2022) KRAS-G12D mutation drives immune suppression and the primary resistance of anti-PD-1/PD-L1 immunotherapy in non-small cell lung cancer. Cancer Commun Lond 42(9):828–847CrossRefPubMedPubMedCentral
28.
go back to reference Chen N, Fang W, Lin Z, Peng P, Wang J, Zhan J et al (2017) KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother 66(9):1175–1187CrossRefPubMedPubMedCentral Chen N, Fang W, Lin Z, Peng P, Wang J, Zhan J et al (2017) KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother 66(9):1175–1187CrossRefPubMedPubMedCentral
30.
go back to reference Lam KHB, Leon AJ, Hui W, Lee SC, Batruch I, Faust K et al (2022) Topographic mapping of the glioblastoma proteome reveals a triple-axis model of intra-tumoral heterogeneity. Nat Commun 13(1):116CrossRefPubMedPubMedCentral Lam KHB, Leon AJ, Hui W, Lee SC, Batruch I, Faust K et al (2022) Topographic mapping of the glioblastoma proteome reveals a triple-axis model of intra-tumoral heterogeneity. Nat Commun 13(1):116CrossRefPubMedPubMedCentral
31.
go back to reference Cancer Genome Atlas Research N (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455(7216):1061–1068CrossRef Cancer Genome Atlas Research N (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455(7216):1061–1068CrossRef
32.
33.
go back to reference Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T et al (2019) Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med 25(3):462–469CrossRefPubMedPubMedCentral Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T et al (2019) Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med 25(3):462–469CrossRefPubMedPubMedCentral
34.
go back to reference Cheng L, Wang Y, Qiu L, Chang Y, Lu H, Liu C et al (2022) mTOR pathway gene mutations predict response to immune checkpoint inhibitors in multiple cancers. J Transl Med 20(1):247CrossRefPubMedPubMedCentral Cheng L, Wang Y, Qiu L, Chang Y, Lu H, Liu C et al (2022) mTOR pathway gene mutations predict response to immune checkpoint inhibitors in multiple cancers. J Transl Med 20(1):247CrossRefPubMedPubMedCentral
35.
go back to reference Lin A, Gu T, Hu X, Zhang J, Luo P (2022) Comprehensive analysis identifies PI3K/Akt pathway alternations as an immune-related prognostic biomarker in colon adenocarcinoma patients receiving immune checkpoint inhibitor treatment. J Immunol Res 2022:8179799CrossRefPubMedPubMedCentral Lin A, Gu T, Hu X, Zhang J, Luo P (2022) Comprehensive analysis identifies PI3K/Akt pathway alternations as an immune-related prognostic biomarker in colon adenocarcinoma patients receiving immune checkpoint inhibitor treatment. J Immunol Res 2022:8179799CrossRefPubMedPubMedCentral
36.
go back to reference Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO et al (2020) Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J 39(15):e103790CrossRefPubMedPubMedCentral Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO et al (2020) Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J 39(15):e103790CrossRefPubMedPubMedCentral
37.
go back to reference Furnari FB, Cloughesy TF, Cavenee WK, Mischel PS (2015) Heterogeneity of epidermal growth factor receptor signalling networks in glioblastoma. Nat Rev Cancer 15(5):302–310CrossRefPubMedPubMedCentral Furnari FB, Cloughesy TF, Cavenee WK, Mischel PS (2015) Heterogeneity of epidermal growth factor receptor signalling networks in glioblastoma. Nat Rev Cancer 15(5):302–310CrossRefPubMedPubMedCentral
38.
go back to reference Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D et al (2021) The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol 23(8):1231–1251CrossRefPubMedPubMedCentral Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D et al (2021) The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol 23(8):1231–1251CrossRefPubMedPubMedCentral
39.
go back to reference Song J, Kadaba P, Kravitz A, Hormigo A, Friedman J, Belani P et al (2020) Multiparametric MRI for early identification of therapeutic response in recurrent glioblastoma treated with immune checkpoint inhibitors. Neuro Oncol 22(11):1658–1666CrossRefPubMedPubMedCentral Song J, Kadaba P, Kravitz A, Hormigo A, Friedman J, Belani P et al (2020) Multiparametric MRI for early identification of therapeutic response in recurrent glioblastoma treated with immune checkpoint inhibitors. Neuro Oncol 22(11):1658–1666CrossRefPubMedPubMedCentral
40.
go back to reference Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I et al (2017) Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med 377(20):1954–1963CrossRefPubMed Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I et al (2017) Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med 377(20):1954–1963CrossRefPubMed
Metadata
Title
Using EGFR amplification to stratify recurrent glioblastoma treated with immune checkpoint inhibitors
Authors
Joshua S. Friedman
Tomi Jun
Omid Rashidipour
Kuan-lin Huang
Ethan Ellis
Priyanka Kadaba
Puneet Belani
Kambiz Nael
Nadejda M. Tsankova
Robert Sebra
Adília Hormigo
Publication date
28-01-2023
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 6/2023
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-023-03381-y

Other articles of this Issue 6/2023

Cancer Immunology, Immunotherapy 6/2023 Go to the issue
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