Combining gene expression analysis of gastric cancer cell lines and tumor specimens to identify biomarkers for anti-HER therapies-the role of HAS2, SHB and HBEGF

Karolin Ebert, Ivonne Haffner, Gwen Zwingenberger, Simone Keller, Elba Raimúndez, Robert Geffers, Ralph Wirtz, Elena Barbaria, Vanessa Hollerieth, Rouven Arnold, Axel Walch, Jan Hasenauer, Dieter Maier, Florian Lordick, Birgit Luber, Karolin Ebert, Ivonne Haffner, Gwen Zwingenberger, Simone Keller, Elba Raimúndez, Robert Geffers, Ralph Wirtz, Elena Barbaria, Vanessa Hollerieth, Rouven Arnold, Axel Walch, Jan Hasenauer, Dieter Maier, Florian Lordick, Birgit Luber

Abstract

Background: The standard treatment for patients with advanced HER2-positive gastric cancer is a combination of the antibody trastuzumab and platin-fluoropyrimidine chemotherapy. As some patients do not respond to trastuzumab therapy or develop resistance during treatment, the search for alternative treatment options and biomarkers to predict therapy response is the focus of research. We compared the efficacy of trastuzumab and other HER-targeting drugs such as cetuximab and afatinib. We also hypothesized that treatment-dependent regulation of a gene indicates its importance in response and that it can therefore be used as a biomarker for patient stratification.

Methods: A selection of gastric cancer cell lines (Hs746T, MKN1, MKN7 and NCI-N87) was treated with EGF, cetuximab, trastuzumab or afatinib for a period of 4 or 24 h. The effects of treatment on gene expression were measured by RNA sequencing and the resulting biomarker candidates were tested in an available cohort of gastric cancer patients from the VARIANZ trial or functionally analyzed in vitro.

Results: After treatment of the cell lines with afatinib, the highest number of regulated genes was observed, followed by cetuximab and trastuzumab. Although trastuzumab showed only relatively small effects on gene expression, BMF, HAS2 and SHB could be identified as candidate biomarkers for response to trastuzumab. Subsequent studies confirmed HAS2 and SHB as potential predictive markers for response to trastuzumab therapy in clinical samples from the VARIANZ trial. AREG, EREG and HBEGF were identified as candidate biomarkers for treatment with afatinib and cetuximab. Functional analysis confirmed that HBEGF is a resistance factor for cetuximab.

Conclusion: By confirming HAS2, SHB and HBEGF as biomarkers for anti-HER therapies, we provide evidence that the regulation of gene expression after treatment can be used for biomarker discovery.

Trial registration: Clinical specimens of the VARIANZ study (NCT02305043) were used to test biomarker candidates.

Keywords: Biomarker; Gastric cancer; Gene expression; HAS2; HBEGF; SHB.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Workflow for gene expression analysis with identification and validation of candidate biomarkers. Gastric cancer cell lines were treated with EGF, cetuximab, EGF plus cetuximab, trastuzumab, afatinib or trastuzumab plus afatinib. The classification of cell lines into responders and non-responders was carried out previously: MKN1 cells were responsive to cetuximab treatment, Hs746T cells were non-responsive [16, 19]. NCI-N87 cells were trastuzumab-responsive, MKN7 and MKN1 cells were non-responsive. NCI-N87, MKN1 and MKN7 cells were afatinib-responsive, Hs746T cells were non-responsive [17]. Regulated genes and biomarker candidates were identified following gene expression analysis. Biomarker candidates were validated in cell culture or clinical specimens
Fig. 2
Fig. 2
Identification of candidate biomarkers for cetuximab treatment in MKN1 cells. a 49 genes were regulated after 4 h cetuximab and EGF treatment whereas 143 genes were regulated after 24 h cetuximab and EGF treatment. The 22 genes which were regulated after 4 h and 24 h cetuximab and EGF treatment were identified as candidate biomarkers. b The 22 genes which were regulated by cetuximab as well as by EGF treatment after 4 h and 24 h were analyzed using the STRING tool. The colors indicate different functional associations (green: textmining, black: co-expression, pink: experimentally determined)
Fig. 3
Fig. 3
Identification of candidate biomarkers for afatinib treatment in NCI-N87 and MKN1 cells. a 62/335 genes were regulated after 4 h/24 h afatinib treatment in NCI-N87 and MKN1 cells. The 45 genes that were regulated after 4 h and 24 h afatinib treatment in NCI-N87 and MKN1 cells were identified as candidate biomarkers. b The 45 genes which were regulated in NCI-N87 as well as in MKN1 cells after 4 h and 24 h afatinib treatment were analyzed using the STRING Tool. The colors indicate different functional associations (green: textmining, black: co-expression, blue: from curated databases, pink: experimentally determined, purple: protein homology)
Fig. 4
Fig. 4
Confirmation of trastuzumab candidate biomarkers BMF, HAS2, and SHB. MKN1 a cells were treated with EGF, EGF plus cetuximab (EGF + Cet), cetuximab (Cet), trastuzumab (Tra), afatinib (Afa) or trastuzumab plus afatinib (Tra + Afa). NCI-N87 b, c, d were treated with trastuzumab (Tra), afatinib (Afa) or trastuzumab plus afatinib (Tra + Afa). The selected treatment times were 24 h for BMF a, b and HAS2 c and 4 h for SHB d. BMFa, b, HAS2c, SHBd gene expression was measured by RNA Sequencing and qPCR. The mean of three biological experiments with standard deviation is shown. Statistically significant effects compared to untreated are indicated by *p < 0.05, **p < 0.01 or ***p < 0.001 (one-sample t-test)
Fig. 5
Fig. 5
Effects of cetuximab and afatinib on proliferation after HBEGF knockdown or stimulation in MKN1 cells. MKN1 cells were transfected with negative-control (Ctr) or HBEGF (HBEGF KD) siRNA. Non-transfected (NT) cells were used as control. The knockdown was checked on RNA level on day 1 (d1) and day 5 (d5) after transfection a. The metabolic activity was measured by WST-1 proliferation assay for 72 h in the untreated b and treated c state. MKN1 cells were stimulated with 5 ng/ml HBEGF (+ HBEGF 5) or not stimulated (-HBEGF 5). The metabolic activity was measured by WST-1 proliferation assay for 72 h in the untreated d and treated e state. Cells were treated with 1 μg/ml cetuximab (Cet 1), 10 μg/ml cetuximab (Cet 10), 0.5 μM afatinib (Afa) or the corresponding solvents (Cet Solv, Afa Solv) for 72 h. Shown are the mean values ​​from three experiments with standard deviation. Significant effects compared to untreated within a group (HBEGF KD, Ctr, NT (c) or + HBEGF 5, -HBEGF 5 (e)) are indicated by *p < 0.05, a** p < 0.01 or a***p < 0.001 (one-sample t-test). Significant effects compared to Ctr, NT (c) or –HBEGF (e) with the same treatment are indicated by b* p < 0.05 or b** p < 0.01 (two-sample t-test)
Fig. 6
Fig. 6
Identification of predictive biomarkers for overall survival in pre-therapeutic tumor biopsies from trastuzumab-treated patients. a Distribution of gene expression values and Hazard ratios (HR) obtained for SHB (HR 1.5; 95% CI, 1 to 2.31), HAS2 (HR 1.7; 95% CI, 1.01 to 2.73) and BMF (HR 0.8; 95% CI, 0.44 to 1.3). SHB and HAS2 are risk factors for patient overall survival. The error bars show the 95% and boxes the 90% confidence interval. Green coloring shows significance. b Kaplan–Meier curves show the overall survival of patients with respect to HAS2 gene expression levels. Lower HAS2 gene expression measured in tumor biopsies is beneficial, whereas higher HAS2 gene expression is detrimental for patient overall survival under trastuzumab treatment. c Kaplan–Meier curves show the overall survival of patients in relation to SHB gene expression. Lower SHB gene expression measured in tumor biopsies is beneficial, whereas higher SHB gene expression is detrimental for patient overall survival under trastuzumab treatment. The method employed to obtain the p values was the log-rank test

References

    1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660.
    1. Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol. 2016;13(6):348–360. doi: 10.1038/nrclinonc.2016.15.
    1. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki 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(9742):687–697. doi: 10.1016/S0140-6736(10)61121-X.
    1. Lordick F, Kang YK, Chung HC, Salman P, Oh SC, Bodoky G, 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(6):490–499. doi: 10.1016/S1470-2045(13)70102-5.
    1. Yonsei University. An Open-label, Multicenter Phase II Study of Afatinib Plus Weekly Taxol as Second Line Treatment for Advanced/Recurrent Gastric and Gastroesophageal Junction Cancer. ClinicalTrialsgov [Internet]. Bethesda: National Library of Medicine (US). Available from . Accessed 04 December 2021.
    1. Memorial Sloan Kettering Cancer Center. A Phase II Study of Afatinib and Paclitaxel in Patients With Advanced HER2-Positive Trastuzumab Refractory Advanced Esophagogastric Cancer. ClinicalTrialsgov [Internet]. Bethesda: National Library of Medicine (US). Available from . Accessed 04 December 2021.
    1. Yonsei University. The Master Protocol for Biomarker-Integrated Umbrella Trial in Advanced Gastric Cancer. ClinicalTrialsgov [Internet]. Bethesda: National Library of Medicine (US). Available from . Accessed 04 December 2021.
    1. Sanchez-Vega F, Hechtman JF, Castel P, Ku GY, Tuvy Y, Won H, et al. EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer. Cancer Discov. 2019;9(2):199–209. doi: 10.1158/-18-0598.
    1. Kim J, Fox C, Peng S, Pusung M, Pectasides E, Matthee E, et al. Preexisting oncogenic events impact trastuzumab sensitivity in ERBB2-amplified gastroesophageal adenocarcinoma. J Clin Invest. 2014;124(12):5145–5158. doi: 10.1172/JCI75200.
    1. Kim C, Lee CK, Chon HJ, Kim JH, Park HS, Heo SJ, et al. PTEN loss and level of HER2 amplification is associated with trastuzumab resistance and prognosis in HER2-positive gastric cancer. Oncotarget. 2017;8(69):113494–113501. doi: 10.18632/oncotarget.23054.
    1. Battaglin F, Naseem M, Puccini A, Lenz HJ. Molecular biomarkers in gastro-esophageal cancer: recent developments, current trends and future directions. Cancer Cell Int. 2018;18:99. doi: 10.1186/s12935-018-0594-z.
    1. Haffner I, Schierle, K., Luber, B., Maier, D., Geier, B., Raimundez, E., Hasenauer, J., Kretzschmar, A., Fischer von Weikersthal, L., Ahlborn, M., Riera Knorrenschild, J., Rau, B., Siegler, G., Fuxius, S., Decker, T., Wittekind, C., Lordick, F. Central validation of HER2 in gastric cancer: high heterogeneity in HER2 expression and its impact on survival (Abstract # 0783). International Gastric Cancer Congress; 2019; Prague, Czech Republic.
    1. Haffner I, Schierle, K., Maier, D., Geier, B., Raimundez, E., Hasenauer, J., Luber, B., Kretzschmar, A., Fischer von Weikersthal, L., Ahlborn, M., Riera Knorrenschild, J., Rau, B., Siegler, G., Fuxius, S., Decker, T., Wittekind, C., Lordick, F. HER2positive gastric cancer: intermediate HER2 expression levels are related to high test deviation rates between local and central pathology and worse survival (Abstract # V1069). Jahrestagung der Deutschen, Österreichischen und Schweizerischen Gesellschaften für Hämatologie und Medizinische Onkologie; 2019; Berlin, Germany.
    1. Lordick F, Haffner, I., Luber, B., Maier, D., Raimundez, E., Hasenauer, J., Kretzschmar, A., Fischer von Weikersthal, L., Ahlborn, M., Riera Knorrenschild, J., Siegler, G., Rau, B., Fuxius, S., Decker, T., Schierle, K., Wittekind, C. Heterogeneity of HER2 expression in gastric cancer (GC) leads to high deviation rates between local and central testing and hampers effıcacy of anti-HER2 therapy: Survival results from the VARIANZ study (Abstract # 2615). Annual Meeting of the American Association for Cancer Research; 2018; Illinois, USA.
    1. Haffner I, Schierle K, Raimundez E, Geier B, Maier D, Hasenauer J, et al. HER2 Expression, Test Deviations, and Their Impact on Survival in Metastatic Gastric Cancer: Results From the Prospective Multicenter VARIANZ Study. J Clin Oncol. 2021;39(13):1468–1478. doi: 10.1200/JCO.20.02761.
    1. Keller S, Kneissl J, Grabher-Meier V, Heindl S, Hasenauer J, Maier D, et al. Evaluation of epidermal growth factor receptor signaling effects in gastric cancer cell lines by detailed motility-focused phenotypic characterization linked with molecular analysis. BMC Cancer. 2017;17(1):845. doi: 10.1186/s12885-017-3822-3.
    1. Keller S, Zwingenberger G, Ebert K, Hasenauer J, Wasmuth J, Maier D, et al. Effects of trastuzumab and afatinib on kinase activity in gastric cancer cell lines. Mol Oncol. 2018;12(4):441–462. doi: 10.1002/1878-0261.12170.
    1. Ebert K, Zwingenberger G, Barbaria E, Keller S, Heck C, Arnold R, et al. Determining the effects of trastuzumab, cetuximab and afatinib by phosphoprotein, gene expression and phenotypic analysis in gastric cancer cell lines. BMC Cancer. 2020;20(1):1039. doi: 10.1186/s12885-020-07540-7.
    1. Kneissl J, Keller S, Lorber T, Heindl S, Keller G, Drexler I, et al. Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines. Int J Oncol. 2012;41(2):733–744. doi: 10.3892/ijo.2012.1479.
    1. Kauraniemi P, Hautaniemi S, Autio R, Astola J, Monni O, Elkahloun A, et al. Effects of Herceptin treatment on global gene expression patterns in HER2-amplified and nonamplified breast cancer cell lines. Oncogene. 2004;23(4):1010–1013. doi: 10.1038/sj.onc.1207200.
    1. Eckstein M, Wirtz RM, Gross-Weege M, Breyer J, Otto W, Stoehr R, et al. mRNA-Expression of KRT5 and KRT20 Defines Distinct Prognostic Subgroups of Muscle-Invasive Urothelial Bladder Cancer Correlating with Histological Variants. International journal of molecular sciences. 2018;19(11):3396. doi: 10.3390/ijms19113396.
    1. Breyer J, Wirtz RM, Otto W, Erben P, Kriegmair MC, Stoehr R, et al. In stage pT1 non-muscle-invasive bladder cancer (NMIBC), high KRT20 and low KRT5 mRNA expression identify the luminal subtype and predict recurrence and survival. Virchows Arch. 2017;470(3):267–274. doi: 10.1007/s00428-017-2064-8.
    1. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–D613. doi: 10.1093/nar/gky1131.
    1. Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med. 2000;6(4):443–446. doi: 10.1038/74704.
    1. Kono K, Takahashi A, Ichihara F, Sugai H, Fujii H, Matsumoto Y. Impaired antibody-dependent cellular cytotoxicity mediated by herceptin in patients with gastric cancer. Cancer Res. 2002;62(20):5813–5817.
    1. Passi A, Vigetti D, Buraschi S, Iozzo RV. Dissecting the role of hyaluronan synthases in the tumor microenvironment. FEBS J. 2019;286(15):2937–2949. doi: 10.1111/febs.14847.
    1. Kunzke T, Balluff B, Feuchtinger A, Buck A, Langer R, Luber B, et al. Native glycan fragments detected by MALDI-FT-ICR mass spectrometry imaging impact gastric cancer biology and patient outcome. Oncotarget. 2017;8(40):68012–68025. doi: 10.18632/oncotarget.19137.
    1. Welsh M, Jamalpour M, Zang G, Akerblom B. The role of the Src Homology-2 domain containing protein B (SHB) in beta cells. J Mol Endocrinol. 2016;56(1):R21–31. doi: 10.1530/JME-15-0228.
    1. Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget. 2017;8(34):57654–57669. doi: 10.18632/oncotarget.14825.
    1. Sato S, Kamada H, Watanabe T, Tsuji I, Fan J. Identification of the cancer cell proliferation and survival functions of proHB-EGF by using an anti-HB-EGF antibody. PLoS One. 2013;8(1):e54509. doi: 10.1371/journal.pone.0054509.
    1. Kneissl J, Hartmann A, Pfarr N, Erlmeier F, Lorber T, Keller S, et al. Influence of the HER receptor ligand system on sensitivity to cetuximab and trastuzumab in gastric cancer cell lines. J Cancer Res Clin Oncol. 2017;143(4):573–600. doi: 10.1007/s00432-016-2308-z.
    1. Kumar SS, Tomita Y, Wrin J, Bruhn M, Swalling A, Mohammed M, et al. High early growth response 1 (EGR1) expression correlates with resistance to anti-EGFR treatment in vitro and with poorer outcome in metastatic colorectal cancer patients treated with cetuximab. Clin Transl Oncol. 2017;19(6):718–726. doi: 10.1007/s12094-016-1596-8.
    1. Boeckx C, Blockx L, de Beeck KO, Limame R, Camp GV, Peeters M, et al. Establishment and characterization of cetuximab resistant head and neck squamous cell carcinoma cell lines: focus on the contribution of the AP-1 transcription factor. Am J Cancer Res. 2015;5(6):1921–1938.

Source: PubMed

Подписаться