Evaluation of EGFR as a prognostic and diagnostic marker for head and neck squamous cell carcinoma patients

  • Authors:
    • Hana Polanska
    • Martina Raudenska
    • Kristyna Hudcová
    • Jaromir Gumulec
    • Marketa Svobodova
    • Zbynek Heger
    • Michaela Fojtu
    • Hana Binkova
    • Zuzana Horakova
    • Rom Kostrica
    • Vojtech Adam
    • Rene Kizek
    • Michal Masarik
  • View Affiliations

  • Published online on: July 21, 2016     https://doi.org/10.3892/ol.2016.4896
  • Pages: 2127-2132
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Approximately 90% of all head and neck tumors are squamous cell carcinomas. The overall survival of patients with head and neck squamous cell carcinoma (HNSCC) is low (≤50%). A non‑invasive marker of disease progression is sorely required. The present study focused on the plasmatic levels of epidermal growth factor receptor (EGFR) in HNSCC patients (N=92) compared with healthy (N=29) and diabetic [type 2 diabetes mellitus (T2DM); N=26] controls. Enzyme-linked immunosorbent assay using antibodies against the extracellular region of EGFR (L25‑S645) was performed. No significant changes were observed between diabetic and healthy controls. However, there were significantly higher EGFR plasma levels in HNSCC patients compared with both control groups (P=0.001 and 0.005, respectively). Receiver operating characteristic curve analysis identified a sensitivity of 76.09%, a specificity of 67.27% and an area under curve of 0.727 for this comparison. No significant association was observed between EGFR plasma levels and tumor stage, tumor grade, lymph node or distant metastasis occurrence, smoking habit or hypertension. However, the presence of human papillomavirus infection and T2DM in HNSCC patients had borderline effect on the plasma EGFR levels. Survival analysis revealed no significant influence of plasmatic EGFR levels on the overall and disease‑specific survival of HNSCC patients. In conclusion, EGFR plasma levels appear to be a relatively promising diagnostic, but poor prognostic, HNSCC marker.

Introduction

Epidermal growth factor receptor (EGFR), also known as ErbB1, is a 170-kDa transmembrane glycoprotein belonging to the ErbB/human epidermal growth factor receptor family of receptor tyrosine kinases (13). EGFR is composed of an extracellular highly glycosylated ligand-binding domain (ECD) comprising amino acids 1–621, a hydrophobic transmembrane domain (amino acids 622–644) and an intracellular domain with tyrosine kinase activity for signal transduction (amino acids 645–1,186) (Fig. 1) (13). Upon binding of a ligand-like amphiregulin, EGF or transforming growth factor α (TGFα) undergoes a conformational change by homo-dimerization or hetero-dimerization with another member of the erbB family, followed by auto-phosphorylation (4,5). This results in tyrosine kinase activation and triggering of signaling cascades. Activation of EGFR leads to the activation of intracellular signaling pathways that regulate cell proliferation, invasion, angiogenesis and metastasis (4,5). EGFR has been selected as a target of anticancer treatments due to its critical roles in cell survival and proliferation (6). EGFR is a strong prognostic marker in head and neck, ovarian and cervical cancer (79). EGFR expression has been associated with a higher proliferative index, advanced tumor stage and increased tumor angiogenesis in HNSCC (9). Overexpression of EGFR and TGFα significantly predicted a shorter disease-free and overall survival (9). EGFR activation also resulted into increased cell invasiveness and motility (10) via the induction of epithelial-to-mesenchymal transition (11,12). Furthermore, EGFR can interact with the receptor cluster of differentiation 44, resulting in a migratory cell phenotype (13). In addition to membrane-bound EGFR, tumor cells express soluble EGFR proteins that can be produced by alternative messenger (m)RNA splicing events, aberrant translocation or disintegration of circulating tumour cells (14,15). Another 110-kDa soluble EGFR isoform, termed proteolytic isoform-soluble (PI-s)EGFR, is disengaged by proteolytic cleavage partially caused by metalloproteases (16,17). Sanderson et al (18) have also reported two soluble isoforms of EGFR (150 and 100-kDa) within exosomes.

The present study focused on plasmatic EGFR levels of HNSCC patients, which were analyzed by enzyme-linked immunosorbent assay (ELISA) using anti-EGFR antibodies raised against the L25-S645 region of full-length EGFR. Notably, information about binding sites of ELISA antibodies are often not provided in the literature, despite that it could be very important for interpretation of the results obtained. Blood markers are less invasive than tissue biopsies, and sample collection can be repeated, which enables real-time monitoring of disease progression and treatment response in patients. As a control group, a gender- and age-matched healthy cohort, and a gender- and age-matched cohort of patients with type 2 diabetes mellitus (T2DM), were used. The T2DM group was included because a proportion of the present HNSCC patients also exhibited T2DM, and certain studies have shown that diabetes suppresses the expression of EGFR (19). Since EGFR is affected by both female estrogen receptors (20,21) and male androgen receptors (22), EGFR may be a potential mediator of gender-related differences in HNSCC. Based on these facts, female HNSCC patients were excluded from the current study.

Materials and methods

Samples preparation

The present study was approved by the ethical committee of St. Anne's Faculty Hospital (Brno, Czech Republic). All surgical tissue samples were obtained from male HNSCC patients treated at St. Anne's Faculty Hospital between April 2013 and June 2015 upon providing informed consent. Histologically verified primary HNSCC carcinoma tissues were collected (92 samples). The tissue material harvested at surgery was placed into RNAlater® solution for RNA stabilization and storage (Ambion; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The material was maintained cold, and RNA was isolated within 24 h. Additional information about the patients and controls is presented in Table I.

Table I.

Characterization of patients and controls.

Table I.

Characterization of patients and controls.

GroupFactorNumber of casesAge, years (range)
HNSCC patients 9262.90 (44–89)
TNM T1-23962.42 (44–89)
TNM T3-45263.17 (47–87)
TNM N04265.04 (44–89)
TNM N14961.26 (44–77)
TNM M08662.86 (44–89)
TNM M1  562.12 (55–71)
Grade 1  663.46 (53–79)
Grade 25063.26 (44–89)
Grade 33261.49 (47–75)
Non-smoker2866.99 (46–89)
Smoker5962.01 (44–78)
Healthy controls 2964.38 (54–69)
Diabetic controls 2656.73 (50–83)

[i] TNM, tumor-node-metastasis; HNSCC, head and neck squamous cell carcinoma.

Blood samples from HNSCC patients and healthy (N=29) and T2DM (N=26) controls were obtained by venipuncture, and 5 ml was placed into an S-Monovette® 4.9 ml, K3EDTA test-tube (Sarstedt AG & Co., Nümbrecht, Germany) for plasma preparation. The blood samples were centrifuged at 1,200 × g at 4°C for 10 min within 60 min of collection. Plasma was aliquoted and stored at −80°C until analysis.

ELISA analysis

Plasma levels of EGFR were determined with a commercial ELISA kit (RayBiotech, Inc., Norcross, GA, USA) according to the manufacturer's protocol. The ELISA was designed to detect human EGFR in plasma or serum with a detection limit of 4 pg/ml, a 10% intra-assay varaibility and a 12% inter-assay variability, as described in the manufacturer's instructions. For the assay, plasma samples were diluted 100-fold, and evaluated with anti-EGFR antibodies raised against the L25-S645 region of EGFR.

RNA isolation and reverse transcription (RT)

TriPure Isolation reagent (Roche Diagnostics, Basel, Switzerland) was used for RNA isolation. The isolated RNA was used for complementary (c)DNA synthesis. RNA (1,000 ng) was reverse transcribed using Transcriptor First Strand cDNA Synthesis kit (Roche Diagnostics) according to manufacturer's protocol. The cDNA (20 µl) prepared from total RNA was diluted with RNase-free water to 100 µl, and 5 µl cDNA was directly analyzed using the LightCycler® 480 II System (Roche Diagnostics).

RT-quantitative polymerase chain reaction (qPCR)

RT-qPCR was performed using TaqMan® Gene Expression Assays (Life Technologies; Thermo Fisher Scientific, Inc.) with the LightCycler® 480 II System, and the amplified DNA was analyzed by the comparative ΔΔCq calculation (23) using β-actin as an endogenous control. The primer and probe sets for β-actin (Hs99999903_m1), metallothionein (MT)2 (Hs02379661_g1), MT1 (Hs00831826_s1), tumor protein p53 (TP53) (Hs01034249_m1), B-cell lymphoma (BCL)-2 associated X protein (BAX) (Hs00180269_m1), BCL-2 (Hs00608023_m1), vascular endothelial growth factor A (VEGFA) (Hs00900055_m1), fms-related tyrosine kinase 1 (FLT1) (Hs01052961_m1), matrix metalloproteinase 2 (MMP2) (Hs01548727_m1), MMP9 (Hs00234579_m1), proto-oncogene c-Fos (FOS) (Hs00170630_m1), c-Jun (JUN) (Hs00277190_s1), marker of proliferation Ki-67 (MKI67) (Hs00606991_m1), EGF (Hs01099999_m1) and EGFR (Hs01076078_m1) were selected from TaqMan® Gene Expression Assays. RT-qPCR was performed under the following amplification conditions in a total volume of 20 µl (5 µl cDNA, 10 µl TaqMan® Gene Expression Master Mix, 4 µl molecular-grade water and 1 µl TaqMan Gene Expression Assay): Initial incubation, 50°C for 2 min, followed by denaturation at 95°C for 10 min and then 45 cycles of 95°C for 15 sec and 60°C for 1 min.

Human papillomavirus (HPV) detection

The 142 bp-long sequence of the conservative major capsid protein L1 gene were amplified using general primers, GP5 and GP6, for non-specific identification of HPV-positive subjects. The PCR mixture from New England BioLabs, Inc. (Ipswich, MA, USA) contained PCR buffer (10 mM Tris HCl, pH 8.3, 50 mM KCl and 2.5 mM MgCl2), 0.05 mM of each deoxynucleotide, and 0.05 mM of GP5 (5′-TTTGTTACTGTGGTAGATAC-3′) and GP6 (5′-GAAAAATAAACTGTAAATCA-3′) primers. The DNA amplification was performed during 40 cycles that included a denaturation step at 94°C for 30 sec, annealing at 45°C for 30 sec and extension at 72°C for 30 sec.

As internal quality control of the isolated DNA, the β-actin gene (600 bp) was amplified (forward primer 5′-CCTGAACCCTAAGGCCAACC-3′ and reverse primer 5′-GCAATGCCTGGGTACATGGT-3′). Each PCR product was analyzed using electrophoresis on 1% agarose gels stained with ethidium bromide.

Data analysis

Differences between the two groups were calculated using the t-test. Survival analysis was conducted using Cox proportional hazard regression analysis with plasma EGFR levels as covariates. Receiver operating characteristic (ROC) curves were calculated using the DeLong methodology. Subsequently, Kaplan-Meier analysis was used with continuous data being divided into two groups as follows: Low expression (<mean values) and high expression (≥mean values) groups. The associations between the continuous variables were analyzed using Pearson's correlations. Unless noted otherwise, P<0.05 was considered to indicate a statistically significant difference. Software STATISTICA 12 (StatSoft, Inc., Tulsa, OK, USA) and MedCalc 15.8 (MedCalc Software bvba, Ostend, Belgium) were used for analysis.

Results

Association between plasma levels of EGFR and HNSCC occurrence

No significant changes in EGFR plasma levels were observed between diabetic and healthy controls (P=0.690). However, there was a significant difference between EGFR plasma levels in HNSCC patients and in both control groups (P=0.001 and 0.005, respectively) (Fig. 2A and Table II). If both control groups were assessed together, the statistical significance was P=0.0001. ROC curve analysis identified a sensitivity of 76.09%, a specificity of 67.27% and an area under the curve (AUC) of 0.727 for this comparison (Fig. 2B). Additional information about the patients and controls is contained in Table I.

Table II.

Plasma levels of EGFR and clinical characteristics in HNSCC patients.

Table II.

Plasma levels of EGFR and clinical characteristics in HNSCC patients.

FactorStatus (number of cases)EGFR levels, ng/ml mean ± standard deviationP-value
Cases vs. controlsHNSCC patients (92)33.1±8.3
Healthy controls (29)28.2±6.40.001
Diabetic controls (26)27.7±2.90.005
SmokingYes (59)31.8±7.70.150
No (28)34.5±8.5
HypertensionYes (28)34.4±8.80.380
No (58)32.7±8.0
Diabetes mellitusYes (10)37.4±9.20.085
No (76)32.6±8.0
TNM T-stagingT1-2 (39)32.5±8.10.580
T3-4 (52)33.4±8.5
TNM N-stagingN+ (49)33.7±8.20.430
N- (42)32.3±8.5
TNM M-stagingM+ (5)31.3±8.10.640
M- (86)33.1±8.3
Tumor gradeHigh (82)32.8±8.10.270
Low (6)   36.8±12.3
HPV statusHPV+ (49)32.0±7.00.084
HPV- (18)35.5±8.3

[i] EGFR, epidermal growth factor receptor; HNSCC, head and neck squamous cell carcinoma; TNM, tumor-node-metastasis; HPV, human papillomavirus.

Correlation between tumor gene expression and EGFR plasma levels

Correlations between plasma EGFR levels and expression of genes in tumor tissues of HNSCC patients were examined. There was no significant correlation between plasma EGFR levels and tumor tissue EGFR mRNA expression, and only a weak negative correlation with MMP9 mRNA was observed (r=−0.21, P=0.040). Gene expression analyses of EGF, EGFR, MKI67, BCL-2, BAX, FOS, JUN, TP53, VEGFA, FLT1, MMP2, MMP9, MT1A and MT2A genes in HNSCC tumor tissue compared with tumor adjacent tissue and tonsillectomies have been published elsewhere (24).

Association between plasma levels of EGFR and clinicopathological characteristics

By examining the associations between plasma EGFR levels and clinicopathological characteristics of HNSCC patients, no significant association was identified for smoking habit, T2DM, hypertension, HPV infection, tumor stage, tumor grade, or lymph node or distant metastasis occurrence. However, the presence of HPV infection and T2DM in HNSCC patients had a borderline effect on the plasma EGFR levels (Table II and Fig. 2C and D).

Association between plasma levels of EGFR and disease-free and overall survival

The prognostic value of EGFR plasma levels on overall and disease-free survival was studied by Cox proportional hazard regression analysis and Kaplan-Meier curves. Survival analysis revealed no significant influence of plasmatic EGFR levels on overall or disease-specific survival in the present cohort of HNSCC patients [hazard ratio (HR)=0.97; 95% confidence interval (CI)=0.92–1.01; P=0.200, and HR=0.96; 95 CI=0.91–1.01; P=0.130 for overall and disease-free survival, respectively] (Fig. 3).

Discussion

Numerous studies have shown that EGFR is overexpressed in HNSCC tumor tissue, but only few studies focused on soluble EGFR levels (2426). There are contradictory studies on soluble EGFR levels, which could be either decreased or elevated in cancer patients compared with a healthy cohort. For example, Partanen et al reported that patients with asbestosis-induced lung cancer have elevated serum soluble EGFR ECD levels (27). Increased soluble EGFR ECD levels were also reported in the urine of patients with squamous cell carcinomas of the lung, head and neck (28), whereas patients with ovarian cancer had decreased levels of serum p110 EGFR compared with the normal population (29). In the present study, ELISA using antibodies against the L25-S645 region of EGFR was used to measure the levels of EGFR in plasma samples of HNSCC patients. Female patients were excluded from the present study due to possible gender-specific EGFR interactions with estrogen or androgen receptors (2022). Significantly higher EGFR plasma levels were detected in HNSCC patients compared with the healthy cohort and the diabetic control group (P=0.001 and 0.005, respectively). This finding is in accordance with that of Perez-Torres et al (17), who suggested that the mechanism of proteolytic cleavage of EGFR and shedding of PI-sEGFR into the plasma may be activated in malignant cells that overexpress the full-length receptor. The cleavage of EGFR probably occurs in the transmembrane domain between G625 and M626 (17). In HNSCC patients, EGFR expression is supposed to be higher in tumor tissues compared with tonsillectomy samples and tumor-adjacent tissues (24). Furthermore, the release of two soluble EGFR isoforms within the exosomes is activated by EGF (17), which is highly produced by HNSCC tumor-adjacent tissues (24).

No significant changes in EGFR plasma levels were observed between diabetic and healthy controls, which is not in accordance with the Vairaktaris et al hypothesis that diabetes suppresses the expression of EGFR (19). However, a slight change on the borderline of statistical significance was observed between HNSCC patients with or without diabetes (P=0.085), while HNSCC patients with diabetes tended to have higher EGFR plasma levels. Borderline changes in EGFR plasma levels were also noticed between the HPV-positive and HPV-negative groups of HNSCC patients (slightly higher levels of plasmatic EGFR were detected in the HPV-negative cohort), although these changes were not significant.

Survival analysis revealed no significant influence of the plasmatic EGFR levels on overall and disease-specific survival in the present cohort of HNSCC patients. By contrast, Ye et al demonstrated that non-small-cell lung cancer patients with lower plasma EGFR concentrations (<27.24 ng/ml) had a significantly shorter overall survival compared with patients who had higher plasma EGFR concentrations (≥27.24 ng/ml) (18.2 vs. 33.4 months, P=0.021) (30).

In conclusion, EGFR plasma levels appear to be a relatively promising diagnostic, but poor prognostic, HNSCC marker. However, further studies are required to determine the clinical value of plasmatic EGFR levels in HNSCC patients. The next important step in soluble EGFR research should be a precise distinction between 110-kDa PI-sEGFR originating from full-length EGFR protein cleavage and EGFR isoforms originating from alternative splicing of EGFR gene transcripts. These EGFR isoforms could readily have slightly different functions. For example, 110-kDa PI-sEGFR originating from full-length EGFR protein cleavage could reflect the presence of malignant cells that overexpress the full-length receptor (17) or a necrotic disintegration of tumor cells. Such form of soluble EGFR was probably originally involved in a proliferative signaling pathway, and could be marker of poor prognosis, while the soluble EGFR isoform originating from alternative splicing was probably not an activator of these proliferative signaling pathways due to the missing intracellular domain, and could exhibit a high affinity binding for EGF, which should result in decreased proliferative signaling and better prognosis.

Acknowledgements

The present study was supported by the Ministry of Health of the Czech Republic (Prague, Czech Republic; grant no. IGA MZ NT 14337-3/2013) and by Specific University Research Grants (grant nos. MUNI/A/1365/2015 and MUNI/A/1426/2015) provided by the Ministry of Education, Youth and Sports of the Czech Republic (Prague, Czech Republic) in 2016 and by Czech Science Foundation (GACR GA16-12454S).

References

1 

Albitar L, Pickett G, Morgan M, Wilken JA, Maihle NJ and Leslie KK: EGFR isoforms and gene regulation in human endometrial cancer cells. Mol Cancer. 9:1662010. View Article : Google Scholar : PubMed/NCBI

2 

Ullrich A, Coussens L, Hayflick JS, Dull TJ, Gray A, Tam AW, Lee J, Yarden Y, Libermann TA, Schlessinger J, et al: Human epidermal growth-factor receptor cDNA sequence and aberrant expression of the amplified gene in A431 epidermoid carcinoma-cells. Nature. 309:418–425. 1984. View Article : Google Scholar : PubMed/NCBI

3 

Ciardiello F and Tortora G: Epidermal growth factor receptor (EGFR) as a target in cancer therapy: Understanding the role of receptor expression and other molecular determinants that could influence the response to anti-EGFR drugs. Eur J Cancer. 39:1348–1354. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Psyrri A, Seiwert TY and Jimeno A: Molecular pathways in head and neck cancer: EGFR, PI3K, and more. Am Soc Clin Oncol Educ Book. 246–255. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Sartor CI: Biological modifiers as potential radiosensitizers: Targeting the epidermal growth factor receptor family. Semin Oncol. 27(6 Suppl 11): S15–S20; discussion 92–100. 2000.

6 

Woodburn JR: The epidermal growth factor receptor and its inhibition in cancer therapy. Pharmacol Ther. 82:241–250. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Nicholson RI, Gee JM and Harper ME: EGFR and cancer prognosis. Eur J Cancer. 37(Suppl 4): S9–S15. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Salomon DS, Brandt R, Ciardiello F and Normanno N: Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 19:183–232. 1995. View Article : Google Scholar : PubMed/NCBI

9 

Grandis Rubin J, Melhem MF, Gooding WE, Day R, Holst VA, Wagener MM, Drenning SD and Tweardy DJ: Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 90:824–832. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Box C, Rogers SJ, Mendiola M and Eccles SA: Tumour-microenvironmental interactions: Paths to progression and targets for treatment. Semin Cancer Biol. 20:128–138. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Zuo JH, Zhu W, Li MY, Li XH, Yi H, Zeng GQ, Wan XX, He QY, Li JH, Qu JQ, et al: Activation of EGFR promotes squamous carcinoma SCC10A cell migration and invasion via inducing EMT-Like phenotype change and MMP-9-mediated degradation of E-cadherin. J Cell Biochem. 112:2508–2517. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Holz C, Niehr F, Boyko M, Hristozova T, Distel L, Budach V and Tinhofer I: Epithelial-mesenchymal-transition induced by EGFR activation interferes with cell migration and response to irradiation and cetuximab in head and neck cancer cells. Radiother Oncol. 101:158–164. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Wang SJ and Bourguignon LY: Hyaluronan and the interaction between CD44 and epidermal growth factor receptor in oncogenic signaling and chemotherapy resistance in head and neck cancer. Arch Otolaryngol Head Neck Surg. 132:771–778. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Hunts JH, Shimizu N, Yamamoto T, Toyoshima K, Merlino GT, Xu YH and Pastan I: Translocation chromosome 7 of A431 cells contains amplification and rearrangement of EGF receptor gene responsible for production of variant mRNA. Somat Cell Mol Genet. 11:477–484. 1985. View Article : Google Scholar : PubMed/NCBI

15 

Kulasinghe A, Perry C, Jovanovic L, Nelson C and Punyadeera C: Circulating tumour cells in metastatic head and neck cancers. Int J Cancer. 136:2515–2523. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Ancot F, Foveau B, Lefebvre J, Leroy C and Tulasne D: Proteolytic cleavages give receptor tyrosine kinases the gift of ubiquity. Oncogene. 28:2185–2195. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Perez-Torres M, Valle BL, Maihle NJ, Negron-Vega L, Nieves-Alicea R and Cora EM: Shedding of epidermal growth factor receptor is a regulated process that occurs with overexpression in malignant cells. Exp Cell Res. 314:2907–2918. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Sanderson MP, Keller S, Alonso A, Riedle S, Dempsey PJ and Altevogt P: Generation of novel, secreted epidermal growth factor receptor (EGFR/ErbB1) isoforms via metal loprotease-dependent ectodomain shedding and exosome secretion. J Cell Biochem. 103:1783–1797. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Vairaktaris E, Goutzanis L, Yapijakis C, Vassiliou S, Spyridonidou S, Vylliotis A, Nkenke E, Lazaris AC, Strantzias P and Patsouris E: Diabetes enhances the expression of H-ras and suppresses the expression of EGFR leading to increased cell proliferation. Histol Histopathol. 24:531–539. 2009.PubMed/NCBI

20 

Britton DJ, Hutcheson IR, Knowlden JM, Barrow D, Giles M, McClelland RA, Gee JM and Nicholson RI: Bidirectional cross talk between ERalpha and EGFR signalling pathways regulates tamoxifen-resistant growth. Breast Cancer Res Treat. 96:131–146. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Levin ER: Bidirectional signaling between the estrogen receptor and the epidermal growth factor receptor. Mol Endocrinol. 17:309–317. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Bonaccorsi L, Muratori A, Carloni V, Marchiani S, Formigli L, Forti G and Baldi E: The androgen receptor associates with the epidermal growth factor receptor in androgen-sensitive prostate cancer cells. Steroids. 69:549–552. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Raudenska M, Sztalmachova M, Gumulec J, Fojtu M, Polanska H, Balvan J, Feith M, Binkova H, Horakova Z, Kostrica R, et al: Prognostic significance of the tumour-adjacent tissue in head and neck cancers. Tumour Biol. 36:9929–9939. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Polanska H, Raudenska M, Gumulec J, Sztalmachova M, Adam V, Kizek R and Masarik M: Clinical significance of head and neck squamous cell cancer biomarkers. Oral Oncol. 50:168–177. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Grandis JR and Tweardy DJ: Elevated levels of transforming growth-factor-alpha and epidermal growth-factor receptor messenger RNA are early markers of carcinogenesis in head and neck-cancer. Cancer Res. 53:3579–3584. 1993.PubMed/NCBI

27 

Partanen R, Hemminki K, Koskinen H, Luo JC, Carney WP and Brandtrauf PW: The detection of increased amounts of the extracellular domain of the epidermal growth-factor receptor in serum during carcinogenesis in asbestosis patients. J Occup Med. 36:1324–1328. 1994. View Article : Google Scholar : PubMed/NCBI

28 

Witters LM, Curley EM, Kumar R, Chinchilli VM, Harvey JP, Crebbin V, Harvey HA and Lipton A: Epidermal growth factor receptor ectodomain in the urine of patients with squamous cell carcinoma. Clin Cancer Res. 1:551–557. 1995.PubMed/NCBI

29 

Carney WP: Circulating oncoproteins HER2/neu, EGFR and CAIX (MN) as novel cancer biomarkers. Expert Rev Mol Diagn. 7:309–319. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Ye P, Zhao J, Wang S and Kong FM: The plasma level of soluble epidermal growth factor Receptor (EGFR) and overall survival (OS) in non-small-cell lung cancer (NSCLC) patients. Annual Meeting of the American Society of Clinical Oncology (ASCO). J Clin Oncol. 33:e190912015.

Related Articles

Journal Cover

September-2016
Volume 12 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Polanska H, Raudenska M, Hudcová K, Gumulec J, Svobodova M, Heger Z, Fojtu M, Binkova H, Horakova Z, Kostrica R, Kostrica R, et al: Evaluation of EGFR as a prognostic and diagnostic marker for head and neck squamous cell carcinoma patients. Oncol Lett 12: 2127-2132, 2016
APA
Polanska, H., Raudenska, M., Hudcová, K., Gumulec, J., Svobodova, M., Heger, Z. ... Masarik, M. (2016). Evaluation of EGFR as a prognostic and diagnostic marker for head and neck squamous cell carcinoma patients. Oncology Letters, 12, 2127-2132. https://doi.org/10.3892/ol.2016.4896
MLA
Polanska, H., Raudenska, M., Hudcová, K., Gumulec, J., Svobodova, M., Heger, Z., Fojtu, M., Binkova, H., Horakova, Z., Kostrica, R., Adam, V., Kizek, R., Masarik, M."Evaluation of EGFR as a prognostic and diagnostic marker for head and neck squamous cell carcinoma patients". Oncology Letters 12.3 (2016): 2127-2132.
Chicago
Polanska, H., Raudenska, M., Hudcová, K., Gumulec, J., Svobodova, M., Heger, Z., Fojtu, M., Binkova, H., Horakova, Z., Kostrica, R., Adam, V., Kizek, R., Masarik, M."Evaluation of EGFR as a prognostic and diagnostic marker for head and neck squamous cell carcinoma patients". Oncology Letters 12, no. 3 (2016): 2127-2132. https://doi.org/10.3892/ol.2016.4896