Skip to main content
Top
Published in: BMC Medicine 1/2021

Open Access 01-12-2021 | Checkpoint Inhibitors | Research article

EPHA7 mutation as a predictive biomarker for immune checkpoint inhibitors in multiple cancers

Authors: Zhen Zhang, Hao-Xiang Wu, Wu-Hao Lin, Zi-Xian Wang, Lu-Ping Yang, Zhao-Lei Zeng, Hui-Yan Luo

Published in: BMC Medicine | Issue 1/2021

Login to get access

Abstract

Background

A critical and challenging process in immunotherapy is to identify cancer patients who could benefit from immune checkpoint inhibitors (ICIs). Exploration of predictive biomarkers could help to maximize the clinical benefits. Eph receptors have been shown to play essential roles in tumor immunity. However, the association between EPH gene mutation and ICI response is lacking.

Methods

Clinical data and whole-exome sequencing (WES) data from published studies were collected and consolidated as a discovery cohort to analyze the association between EPH gene mutation and efficacy of ICI therapy. Another independent cohort from Memorial Sloan Kettering Cancer Center (MSKCC) was adopted to validate our findings. The Cancer Genome Atlas (TCGA) cohort was used to perform anti-tumor immunity and pathway enrichment analysis.

Results

Among fourteen EPH genes, EPHA7-mutant (EPHA7-MUT) was enriched in patients responding to ICI therapy (FDR adjusted P < 0.05). In the discovery cohort (n = 386), significant differences were detected between EPHA7-MUT and EPHA7-wildtype (EPHA7-WT) patients regarding objective response rate (ORR, 52.6% vs 29.1%, FDR adjusted P = 0.0357) and durable clinical benefit (DCB, 70.3% vs 42.7%, FDR adjusted P = 0.0200). In the validation cohort (n = 1144), significant overall survival advantage was observed in EPHA7-MUT patients (HR = 0.62 [95% confidence interval, 0.39 to 0.97], multivariable adjusted P = 0.0367), which was independent of tumor mutational burden (TMB) and copy number alteration (CNA). Notably, EPHA7-MUT patients without ICI therapy had significantly worse overall survival in TCGA cohort (HR = 1.33 [95% confidence interval, 1.06 to 1.67], multivariable adjusted P = 0.0139). Further gene set enrichment analysis revealed enhanced anti-tumor immunity in EPHA7-MUT tumor.

Conclusions

EPHA7-MUT successfully predicted better clinical outcomes in ICI-treated patients across multiple cancer types, indicating that EPHA7-MUT could serve as a potential predictive biomarker for immune checkpoint inhibitors.
Appendix
Available only for authorised users
Literature
1.
go back to reference Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395:1835–44.CrossRef Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395:1835–44.CrossRef
2.
go back to reference Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.CrossRef Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–92.CrossRef
3.
go back to reference Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, de Jesus-Acosta A, Delord JP, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/ mismatch repair–deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1–10.CrossRef Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, de Jesus-Acosta A, Delord JP, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/ mismatch repair–deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1–10.CrossRef
4.
go back to reference Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–23.CrossRef Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–23.CrossRef
5.
go back to reference Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19:133–50.CrossRef Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19:133–50.CrossRef
6.
go back to reference Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322). Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322).
7.
go back to reference Wu HX, Chen YX, Wang ZX, Zhao Q, He MM, Wang YN, et al. Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers. J Immunother Cancer. 2019;7:1–13.CrossRef Wu HX, Chen YX, Wang ZX, Zhao Q, He MM, Wang YN, et al. Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers. J Immunother Cancer. 2019;7:1–13.CrossRef
8.
go back to reference Ott PA, Bang YJ, Piha-Paul SA, Abdul Razak AR, Bennouna J, Soria JC, et al. T-cell–inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37:318–27.CrossRef Ott PA, Bang YJ, Piha-Paul SA, Abdul Razak AR, Bennouna J, Soria JC, et al. T-cell–inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37:318–27.CrossRef
9.
go back to reference Hakimi AA, Voss MH, Kuo F, Sanchez A, Liu M, Nixon BG, et al. Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: data from a randomized phase III trial. Cancer Discov. 2019;9:510–25.CrossRef Hakimi AA, Voss MH, Kuo F, Sanchez A, Liu M, Nixon BG, et al. Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: data from a randomized phase III trial. Cancer Discov. 2019;9:510–25.CrossRef
10.
go back to reference Ready N, Hellmann MD, Awad MM, Otterson GA, Gutierrez M, Gainor JF, et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J Clin Oncol. 2019;37:992–1000.CrossRef Ready N, Hellmann MD, Awad MM, Otterson GA, Gutierrez M, Gainor JF, et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J Clin Oncol. 2019;37:992–1000.CrossRef
11.
go back to reference Zhou H, Liu J, Zhang Y, Huang Y, Shen J, Yang Y, et al. PBRM1 mutation and preliminary response to immune checkpoint blockade treatment in non-small cell lung cancer. NPJ Precis Oncol. 2020;4:4–7.CrossRef Zhou H, Liu J, Zhang Y, Huang Y, Shen J, Yang Y, et al. PBRM1 mutation and preliminary response to immune checkpoint blockade treatment in non-small cell lung cancer. NPJ Precis Oncol. 2020;4:4–7.CrossRef
12.
go back to reference Gonzalez-Cao M, Viteri S, Karachaliou N, Aguilar A, García-Mosquera JJ, Rosell R. Tumor mutational burden as predictive factor of response to immunotherapy. Transl Lung Cancer Res. 2018;7:S358–61.CrossRef Gonzalez-Cao M, Viteri S, Karachaliou N, Aguilar A, García-Mosquera JJ, Rosell R. Tumor mutational burden as predictive factor of response to immunotherapy. Transl Lung Cancer Res. 2018;7:S358–61.CrossRef
13.
go back to reference Funk SD, Orr AW. Ephs and ephrins resurface in inflammation, immunity, and atherosclerosis. Pharmacol Res. 2013;67:42–52.CrossRef Funk SD, Orr AW. Ephs and ephrins resurface in inflammation, immunity, and atherosclerosis. Pharmacol Res. 2013;67:42–52.CrossRef
14.
go back to reference Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer. 2010;10:165–80.CrossRef Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer. 2010;10:165–80.CrossRef
15.
go back to reference Barquilla A, Pasquale EB. Eph receptors and ephrins: therapeutic opportunities. Annu Rev Pharmacol Toxicol. 2015;55:465–87.CrossRef Barquilla A, Pasquale EB. Eph receptors and ephrins: therapeutic opportunities. Annu Rev Pharmacol Toxicol. 2015;55:465–87.CrossRef
16.
go back to reference Song Y, Fu Y, Xie Q, Zhu B, Wang J, Zhang B. Anti-angiogenic agents in combination with immune checkpoint inhibitors: a promising strategy for cancer treatment. Front Immunol. 2020;11:1–17.CrossRef Song Y, Fu Y, Xie Q, Zhu B, Wang J, Zhang B. Anti-angiogenic agents in combination with immune checkpoint inhibitors: a promising strategy for cancer treatment. Front Immunol. 2020;11:1–17.CrossRef
17.
go back to reference Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, et al. Phase Ib/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol. 2020;38:1154–63.CrossRef Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, et al. Phase Ib/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol. 2020;38:1154–63.CrossRef
18.
go back to reference Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38:2053–61.CrossRef Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38:2053–61.CrossRef
19.
go back to reference Zhu AX, Finn RS, Ikeda M, Sung MW, Baron AD, Kudo M, Okusaka T, Kobayashi M, Kumada H, Kaneko S, Pracht M, Mamontov K, Meyer T, Mody K, Kubota T, Dutcus CE, JML K. A phase Ib study of lenvatinib (LEN) plus pembrolizumab (PEMBRO) in unresectable hepatocellular carcinoma (uHCC). J Clin Oncol. 2020;38(15_suppl):4519 (May 20, 2020).CrossRef Zhu AX, Finn RS, Ikeda M, Sung MW, Baron AD, Kudo M, Okusaka T, Kobayashi M, Kumada H, Kaneko S, Pracht M, Mamontov K, Meyer T, Mody K, Kubota T, Dutcus CE, JML K. A phase Ib study of lenvatinib (LEN) plus pembrolizumab (PEMBRO) in unresectable hepatocellular carcinoma (uHCC). J Clin Oncol. 2020;38(15_suppl):4519 (May 20, 2020).CrossRef
20.
go back to reference Shiuan E, Chen J. Eph receptor tyrosine kinases in tumor immunity. Cancer Res. 2016;76:6452–7.CrossRef Shiuan E, Chen J. Eph receptor tyrosine kinases in tumor immunity. Cancer Res. 2016;76:6452–7.CrossRef
21.
go back to reference Yang WH, Cha JH, Xia W, Lee HH, Chan LC, Wang YN, et al. Juxtacrine signaling inhibits antitumor immunity by upregulating PD-L1 expression. Cancer Res. 2018;78:3761–8.CrossRef Yang WH, Cha JH, Xia W, Lee HH, Chan LC, Wang YN, et al. Juxtacrine signaling inhibits antitumor immunity by upregulating PD-L1 expression. Cancer Res. 2018;78:3761–8.CrossRef
22.
go back to reference Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science (80- ). 2015;348:124–8.CrossRef Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science (80- ). 2015;348:124–8.CrossRef
23.
go back to reference Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.CrossRef Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.CrossRef
24.
go back to reference Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science (80- ). 2016;352:207–12.CrossRef Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science (80- ). 2016;352:207–12.CrossRef
25.
go back to reference Miao D, Margolis CA, Vokes NI, Liu D, Taylor-Weiner A, Wankowicz SM, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat Genet. 2018;50:1271–81.CrossRef Miao D, Margolis CA, Vokes NI, Liu D, Taylor-Weiner A, Wankowicz SM, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat Genet. 2018;50:1271–81.CrossRef
26.
go back to reference Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44.
27.
go back to reference Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F, Ahuja A, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33:843–852.e4.CrossRef Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F, Ahuja A, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33:843–852.e4.CrossRef
28.
go back to reference Miao D, Miao D, Margolis CA, Gao W, Voss MH, Li W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science (80- ). 2018;5951:1–11. Miao D, Miao D, Margolis CA, Gao W, Voss MH, Li W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science (80- ). 2018;5951:1–11.
29.
go back to reference Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.CrossRef Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.CrossRef
30.
go back to reference Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA Pan-Cancer Clinical Data Resource to drive high-quality survival outcome analytics. Cell. 2018;173:400–416.e11.CrossRef Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA Pan-Cancer Clinical Data Resource to drive high-quality survival outcome analytics. Cell. 2018;173:400–416.e11.CrossRef
31.
go back to reference Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–830.e14.CrossRef Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–830.e14.CrossRef
32.
go back to reference Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN. Visualizing and interpreting cancer genomics data via the Xena platform. Nature Biotechnology. 2020. p. 1–4. Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN. Visualizing and interpreting cancer genomics data via the Xena platform. Nature Biotechnology. 2020. p. 1–4.
33.
go back to reference Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.CrossRef Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.CrossRef
34.
go back to reference Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:1–14.CrossRef Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:1–14.CrossRef
35.
go back to reference Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12:453–7.CrossRef Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12:453–7.CrossRef
36.
go back to reference Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:1–21.CrossRef Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:1–21.CrossRef
37.
go back to reference Yu G, Wang LG, Han Y, He QY. ClusterProfiler: an R package for comparing biological themes among gene clusters. Omi A J Integr Biol. 2012;16:284–7.CrossRef Yu G, Wang LG, Han Y, He QY. ClusterProfiler: an R package for comparing biological themes among gene clusters. Omi A J Integr Biol. 2012;16:284–7.CrossRef
38.
go back to reference Gao J, Chang MT, Johnsen HC, Gao SP, Sylvester BE, Sumer SO, et al. 3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets. Genome Med. 2017;9:1–13.CrossRef Gao J, Chang MT, Johnsen HC, Gao SP, Sylvester BE, Sumer SO, et al. 3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets. Genome Med. 2017;9:1–13.CrossRef
39.
go back to reference Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol. 2018;15:310–24.CrossRef Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol. 2018;15:310–24.CrossRef
40.
go back to reference Li X, Wenes M, Romero P, Huang SCC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019;16:425–41.CrossRef Li X, Wenes M, Romero P, Huang SCC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019;16:425–41.CrossRef
41.
go back to reference Oricchio E, Nanjangud G, Wolfe AL, Schatz JH, Mavrakis KJ, Jiang M, et al. The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell. 2011;147:554–64.CrossRef Oricchio E, Nanjangud G, Wolfe AL, Schatz JH, Mavrakis KJ, Jiang M, et al. The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell. 2011;147:554–64.CrossRef
42.
go back to reference Li S, Wu Z, Ma P, Xu Y, Chen Y, Wang H, et al. Ligand-dependent EphA7 signaling inhibits prostate tumor growth & progression. Cell Death Dis. 2017;8:e3122.CrossRef Li S, Wu Z, Ma P, Xu Y, Chen Y, Wang H, et al. Ligand-dependent EphA7 signaling inhibits prostate tumor growth & progression. Cell Death Dis. 2017;8:e3122.CrossRef
Metadata
Title
EPHA7 mutation as a predictive biomarker for immune checkpoint inhibitors in multiple cancers
Authors
Zhen Zhang
Hao-Xiang Wu
Wu-Hao Lin
Zi-Xian Wang
Lu-Ping Yang
Zhao-Lei Zeng
Hui-Yan Luo
Publication date
01-12-2021
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2021
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-020-01899-x

Other articles of this Issue 1/2021

BMC Medicine 1/2021 Go to the issue