EGFR amplification and outcome in a randomised phase III trial of chemotherapy alone or chemotherapy plus panitumumab for advanced gastro-oesophageal cancers

Elizabeth C Smyth, Georgios Vlachogiannis, Somaieh Hedayat, Alice Harbery, Sanna Hulkki-Wilson, Massimiliano Salati, Kyriakos Kouvelakis, Javier Fernandez-Mateos, George D Cresswell, Elisa Fontana, Therese Seidlitz, Clare Peckitt, Jens C Hahne, Andrea Lampis, Ruwaida Begum, David Watkins, Sheela Rao, Naureen Starling, Tom Waddell, Alicia Okines, Tom Crosby, Was Mansoor, Jonathan Wadsley, Gary Middleton, Matteo Fassan, Andrew Wotherspoon, Chiara Braconi, Ian Chau, Igor Vivanco, Andrea Sottoriva, Daniel E Stange, David Cunningham, Nicola Valeri, Elizabeth C Smyth, Georgios Vlachogiannis, Somaieh Hedayat, Alice Harbery, Sanna Hulkki-Wilson, Massimiliano Salati, Kyriakos Kouvelakis, Javier Fernandez-Mateos, George D Cresswell, Elisa Fontana, Therese Seidlitz, Clare Peckitt, Jens C Hahne, Andrea Lampis, Ruwaida Begum, David Watkins, Sheela Rao, Naureen Starling, Tom Waddell, Alicia Okines, Tom Crosby, Was Mansoor, Jonathan Wadsley, Gary Middleton, Matteo Fassan, Andrew Wotherspoon, Chiara Braconi, Ian Chau, Igor Vivanco, Andrea Sottoriva, Daniel E Stange, David Cunningham, Nicola Valeri

Abstract

Objective: Epidermal growth factor receptor (EGFR) inhibition may be effective in biomarker-selected populations of advanced gastro-oesophageal adenocarcinoma (aGEA) patients. Here, we tested the association between outcome and EGFR copy number (CN) in pretreatment tissue and plasma cell-free DNA (cfDNA) of patients enrolled in a randomised first-line phase III clinical trial of chemotherapy or chemotherapy plus the anti-EGFR monoclonal antibody panitumumab in aGEA (NCT00824785).

Design: EGFR CN by either fluorescence in situ hybridisation (n=114) or digital-droplet PCR in tissues (n=250) and plasma cfDNAs (n=354) was available for 474 (86%) patients in the intention-to-treat (ITT) population. Tissue and plasma low-pass whole-genome sequencing was used to screen for coamplifications in receptor tyrosine kinases. Interaction between chemotherapy and EGFR inhibitors was modelled in patient-derived organoids (PDOs) from aGEA patients.

Results: EGFR amplification in cfDNA correlated with poor survival in the ITT population and similar trends were observed when the analysis was conducted in tissue and plasma by treatment arm. EGFR inhibition in combination with chemotherapy did not correlate with improved survival, even in patients with significant EGFR CN gains. Addition of anti-EGFR inhibitors to the chemotherapy agent epirubicin in PDOs, resulted in a paradoxical increase in viability and accelerated progression through the cell cycle, associated with p21 and cyclin B1 downregulation and cyclin E1 upregulation, selectively in organoids from EGFR-amplified aGEA.

Conclusion: EGFR CN can be accurately measured in tissue and liquid biopsies and may be used for the selection of aGEA patients. EGFR inhibitors may antagonise the antitumour effect of anthracyclines with important implications for the design of future combinatorial trials.

Keywords: gastric adenocarcinoma; gastrointestinal cancer; molecular oncology; oesophageal cancer.

Conflict of interest statement

Competing interests: ES declares honoraria for advisory role from Astellas, BMS, Celgene, Five Prime, Gritstone Oncology, and Servier. DW has received honoraria from Amgen. NS received research funding from AstraZeneca, Pfizer, BMS and honoraria from Servier, MSD, Merck Serono and AstraZeneca. JW received honoraria from Eisai, AstraZeneca, Sanofi-Genzyme, Ipsen, Novartis, Bayer, Celgene and Advanced Accelerator Applications. IC has had advisory roles with Merck Serono, Roche, Sanofi Oncology, Bristol Myers Squibb, Eli-Lilly, Novartis, Gilead Science. He has received research funding from Merck-Serono, Novartis, Roche and Sanofi Oncology, and honoraria from Roche, Sanofi-Oncology, Eli-Lilly, Taiho, Bayer and Prime Therapeutics. IV has received honoraria from AbbVie. He has received researched funding from Genmab and Basilea. DC received research funding from 4SC, AstraZeneca, Bayer, Celgene, Clovis, Eli Lilly, Janssen, Medimmune, Merck, Merrimack, Amgen, Sanofi. NV received honoraria from Merck Serono, Pfizer, Bayer and Eli-Lilly. All other authors declare no conflict of interest.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
Tissue and liquid biopsy analysis in the REAL3 trial. (A) Diagram shows the number of patients for whom FFPE tissues and plasma cfDNA were available for EGFR testing. (B) Venn diagram shows the number of patients tested for EGFR amplification based on source of material (FFPE tissues vs cfDNA) and method used (ddPCR vs FISH). cfDNA, cell free DNA; ddPCR, digital-droplet PCR; EGFR, epidermal growth factor receptor; FFPE, formalin-fixed paraffin embedded; FISH, fluorescent in situ hybridisation; ITT, intention to treat.
Figure 2
Figure 2
Overall survival (OS) based on EGFR-amplification in the REAL3 trial. EGFR CN was determined using ddPCR on pretreatment cfDNA. Kaplan-Meier curves show the OS of patients based on the presence/absence of EGFR CN. cfDNA=cell free DNA; CN, copy number; ddPCR, digital-droplet PCR; EGFR, epidermal growth factor receptor;
Figure 3
Figure 3
Clinical outcome by treatment arm in EGFR-amplified cases enrolled in the REAL3 trial. (A) Bars show OS and PFS (median±SE) in EGFR-amplified patients treated with chemotherapy alone (EOX) or chemotherapy plus panitumumab (EOX-P). Similar trends are observed when a cut-off of two or five EGFR copies is used. (B) Blue bars indicate PFS, orange bars indicate EGFR copies determined by ddPCR in plasma cfDNA (graph on the top) or tissue (bottom). (C) Plots show copy number changes in EGFR (blue) and other receptor tyrosine kinase genes (red) in patients with PFS greater than 6 months on treatment with EOX-P. Values outside of the y-axis limits are plotted at the limit. cfDNA, cell-free DNA; CN, copy number; ddPCR, digital-droplet PCR; EGFR, epidermal growth factor receptor; EOX, epirubicin+oxaliplatin+capecitabine; OS, overall survival; P, panitumumab; PFS, progression-free survival.
Figure 4
Figure 4
Effect of epirubicin or epirubicin plus EGFR inhibitors in EGFR-amplified and non-amplified patient-derived organoids. (A) EGFR FISH images, demonstrating gain of copies and diploid status in the F-014 BL and DD191 human GEA PDO lines, respectively. (B) Concentration-dependent effect of epirubicin as a monotherapy or in combination with a stable dose of two different anti-EGFR agents (cetuximab, 20 µg/mL; gefitinib, 200 nM) in the EGFR-amplified F-014 BL and EGFR-diploid DD191 human GEA PDO lines. The combination of low epirubicin concentrations with anti-EGFR treatments results in a paradoxical increase in viability selectively in the EGFR-amplified F-014 BL GEA PDO line. Viability data shown are means±SEM of indicated independent experiments. (C) Pathway analysis of RNAseq data from the EGFR-amplified F-014 BL GEA PDO line treated with a low concentration of epirubicin alone or in combination with cetuximab for 24 hours revealed a significant reduction in the expression of cell cycle-related genes associated with the epirubicin and cetuximab combination. RNA from three independent biological replicates were sequenced per condition. (D) Protein analysis of the EGFR-amplified F-014 BL and EGFR-diploid DD191 human GEA PDO lines treated with two low concentrations of epirubicin alone or in combination with cetuximab for 24 hours. In line with RNAseq data, a reduction in p21 and cyclin B1 protein levels was observed when epirubicin was combined with inhibition of EGFR and downstream MAPK and AKT signalling specifically in the EGFR-amplified organoid line. (E) EdU DNA incorporation assay following treatment of the EGFR-amplified F-014 BL and EGFR-diploid DD191 human GEA PDO lines with two low concentrations of epirubicin alone or in combination with cetuximab for 24 hours. The addition of cetuximab antagonises the antiproliferative effect of epirubicin and accelerates DNA synthesis specifically in the EGFR-amplified organoid line. (F) Proposed model of antagonism between epirubicin and anti-EGFR treatments in EGFR-amplified GEA. EGFR, epidermal growth factor receptor; FISH, fluorescent in situ hybridisation; FSC-A; forward scatter-area; GEA, gastro-oesophageal adenocarcinoma; KEGG, Kyoto Encyclopedia of Genes and Genomes; PDO, patient-derived organoid.

References

    1. Cancer Genome Atlas Research Network . Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202–9. 10.1038/nature13480
    1. Maron SB, Alpert L, Kwak HA, et al. . Targeted Therapies for Targeted Populations: Anti-EGFR Treatment for EGFR-Amplified Gastroesophageal Adenocarcinoma. Cancer Discov 2018;8:696–713. 10.1158/-17-1260
    1. Cancer Genome Atlas Research Network . Integrated genomic characterization of oesophageal carcinoma. Nature 2017;541:169–75. 10.1038/nature20805
    1. Bang Y-J, Van Cutsem E, Feyereislova A, et al. . Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–97. 10.1016/S0140-6736(10)61121-X
    1. Romond EH, Perez EA, Bryant J, et al. . Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005;353:1673–84. 10.1056/NEJMoa052122
    1. Slamon DJ, Leyland-Jones B, Shak S, et al. . Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783–92. 10.1056/NEJM200103153441101
    1. Lordick F, Kang Y-K, Chung H-C, et al. . Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (expand): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:490–9. 10.1016/S1470-2045(13)70102-5
    1. Waddell T, Chau I, Cunningham D, et al. . Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:481–9. 10.1016/S1470-2045(13)70096-2
    1. Petty RD, Dahle-Smith A, Stevenson DAJ, et al. . Gefitinib and EGFR gene copy number aberrations in esophageal cancer. J Clin Oncol 2017;35:2279–87. 10.1200/JCO.2016.70.3934
    1. Pectasides E, Stachler MD, Derks S, et al. . Genomic heterogeneity as a barrier to precision medicine in gastroesophageal adenocarcinoma. Cancer Discov 2018;8:37–48. 10.1158/-17-0395
    1. Tomasello G, Valeri N, Ghidini M, et al. . First-Line dose-dense chemotherapy with docetaxel, cisplatin, folinic acid and 5-fluorouracil (dcf) plus panitumumab in patients with locally advanced or metastatic cancer of the stomach or gastroesophageal junction: final results and biomarker analysis from an Italian Oncology group for clinical research (GOIRC) phase II study. Oncotarget 2017;8:111795–806. 10.18632/oncotarget.22909
    1. Seidlitz T, Merker SR, Rothe A, et al. . Human gastric cancer modelling using organoids. Gut 2019;68:207–17. 10.1136/gutjnl-2017-314549
    1. Vlachogiannis G, Hedayat S, Vatsiou A, et al. . Patient-Derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018;359:920–6. 10.1126/science.aao2774
    1. Bartley AN, Washington MK, Colasacco C, et al. . Her2 testing and clinical decision making in gastroesophageal adenocarcinoma: guideline from the College of American pathologists, American Society for clinical pathology, and the American Society of clinical oncology. J Clin Oncol 2017;35:446–64. 10.1200/JCO.2016.69.4836
    1. Cerami E, Gao J, Dogrusoz U, et al. . The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4. 10.1158/-12-0095
    1. Gao J, Aksoy BA, Dogrusoz U, et al. . Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. 10.1126/scisignal.2004088
    1. Sanchez-Vega F, Hechtman JF, Castel P, et al. . EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer. Cancer Discov 2019;9:199–209. 10.1158/-18-0598
    1. Chong CR, Jänne PA. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat Med 2013;19:1389–400. 10.1038/nm.3388
    1. Misale S, Di Nicolantonio F, Sartore-Bianchi A, et al. . Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov 2014;4:1269–80. 10.1158/-14-0462
    1. Ricordel C, Friboulet L, Facchinetti F, et al. . Molecular mechanisms of acquired resistance to third-generation EGFR-TKIs in EGFR T790M-mutant lung cancer. Ann Oncol 2018;29:i28–37. 10.1093/annonc/mdx705
    1. Stahl M, Maderer A, Lordick F, et al. . Perioperative chemotherapy with or without epidermal growth factor receptor blockade in unselected patients with locally advanced oesophagogastric adenocarcinoma: Randomized phase II study with advanced biomarker program of the German Cancer Society (AIO/CAO STO-0801). Eur J Cancer 2018;93:119–26. 10.1016/j.ejca.2018.01.079
    1. Buchholz TA, Tu X, Ang KK, et al. . Epidermal growth factor receptor expression correlates with poor survival in patients who have breast carcinoma treated with doxorubicin-based neoadjuvant chemotherapy. Cancer 2005;104:676–81. 10.1002/cncr.21217
    1. Tjuljandin SA, Doig RG, Sobol MM, et al. . Pharmacokinetics and toxicity of two schedules of high dose epirubicin. Cancer Res 1990;50:5095–101.
    1. Brand TM, Iida M, Luthar N, et al. . Nuclear EGFR as a molecular target in cancer. Radiother Oncol 2013;108:370–7. 10.1016/j.radonc.2013.06.010
    1. Bagheri-Yarmand R, Biernacka A, Hunt KK, et al. . Low molecular weight cyclin E overexpression shortens mitosis, leading to chromosome missegregation and centrosome amplification. Cancer Res 2010;70:5074–84. 10.1158/0008-5472.CAN-09-4094
    1. Birkman E-M, Ålgars A, Lintunen M, et al. . Egfr gene amplification is relatively common and associates with outcome in intestinal adenocarcinoma of the stomach, gastro-oesophageal junction and distal oesophagus. BMC Cancer 2016;16:406. 10.1186/s12885-016-2456-1
    1. Higaki E, Kuwata T, Nagatsuma AK, et al. . Gene copy number gain of EGFR is a poor prognostic biomarker in gastric cancer: evaluation of 855 patients with bright-field dual in situ hybridization (DISH) method. Gastric Cancer 2016;19:63–73. 10.1007/s10120-014-0449-9
    1. Liao J-B, Lee H-P, Fu H-T, et al. . Assessment of EGFR and ErbB2 (HER2) in gastric and gastroesophageal carcinomas: EGFR amplification is associated with a worse prognosis in early stage and well to moderately differentiated carcinoma. Appl Immunohistochem Mol Morphol 2018;26:374–82. 10.1097/PAI.0000000000000437
    1. Lordick F, Kang Y-K, Salman P, et al. . Clinical outcome according to tumor HER2 status and EGFR expression in advanced gastric cancer patients from the expand study. JCO 2013;31:4021. 10.1200/jco.2013.31.15_suppl.4021
    1. Dutton SJ, Ferry DR, Blazeby JM, et al. . Gefitinib for oesophageal cancer progressing after chemotherapy (COG): a phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol 2014;15:894–904. 10.1016/S1470-2045(14)70024-5
    1. Pearson A, Smyth E, Babina IS, et al. . High-Level clonal FGFR amplification and response to FGFR inhibition in a translational clinical trial. Cancer Discov 2016;6:838–51. 10.1158/-15-1246
    1. Van Cutsem E, Bang Y-J, Feng-Yi F, et al. . Her2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 2015;18:476–84. 10.1007/s10120-014-0402-y
    1. Khan KH, Cunningham D, Werner B, et al. . Longitudinal liquid biopsy and mathematical modeling of clonal evolution forecast time to treatment failure in the PROSPECT-C phase II colorectal cancer clinical trial. Cancer Discov 2018;8:1270–85. 10.1158/-17-0891
    1. Gao Y, Zhang K, Xi H, et al. . Diagnostic and prognostic value of circulating tumor DNA in gastric cancer: a meta-analysis. Oncotarget 2017;8:6330–40. 10.18632/oncotarget.14064
    1. Shoda K, Ichikawa D, Fujita Y, et al. . Monitoring the HER2 copy number status in circulating tumor DNA by droplet digital PCR in patients with gastric cancer. Gastric Cancer 2017;20:126–35. 10.1007/s10120-016-0599-z
    1. Cafferkey C, Smyth E, Loehr A, et al. . Genomic loss of heterozygosity (LOH) and survival in patients (PTS) treated with epirubicin, oxaliplatin, capecitabine (EOC) ± panitumumab (P) in the REAL3 trial. Annals of Oncology 2016;27:vi220. 10.1093/annonc/mdw371.41
    1. Okines AFC, Gonzalez de Castro D, Cunningham D, et al. . Biomarker analysis in oesophagogastric cancer: results from the REAL3 and TransMAGIC trials. Eur J Cancer 2013;49:2116–25. 10.1016/j.ejca.2013.02.007
    1. Maughan TS, Adams RA, Smith CG, et al. . Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet 2011;377:2103–14. 10.1016/S0140-6736(11)60613-2
    1. Primrose J, Falk S, Finch-Jones M, et al. . Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis: the new EPOC randomised controlled trial. Lancet Oncol 2014;15:601–11. 10.1016/S1470-2045(14)70105-6
    1. Tveit KM, Guren T, Glimelius B, et al. . Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (Nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: the NORDIC-VII study. J Clin Oncol 2012;30:1755–62. 10.1200/JCO.2011.38.0915
    1. Santoro V, Jia R, Thompson H, et al. . Role of reactive oxygen species in the abrogation of oxaliplatin activity by cetuximab in colorectal cancer. J Natl Cancer Inst 2016;108:djv394. 10.1093/jnci/djv394
    1. Rödel C, Graeven U, Fietkau R, et al. . Oxaliplatin added to fluorouracil-based preoperative chemoradiotherapy and postoperative chemotherapy of locally advanced rectal cancer (the German CAO/ARO/AIO-04 study): final results of the multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2015;16:979–89. 10.1016/S1470-2045(15)00159-X

Source: PubMed

3
Abonnere