Open Access

EGFR and Cortactin: Markers for potential double target therapy in oral squamous cell carcinoma

  • Authors:
    • Oliver Bissinger
    • Andreas Kolk
    • Enken Drecoll
    • Melanie Straub
    • Christina Lutz
    • Klaus‑Dietrich Wolff
    • Carolin Götz
  • View Affiliations

  • Published online on: September 18, 2017     https://doi.org/10.3892/etm.2017.5120
  • Pages: 4620-4626
  • Copyright: © Bissinger et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Survival periods of patients following surgical therapy of oral squamous cell carcinoma (OSCC) have previously been demonstrated to decrease over recent decades. Epidermal growth factor receptor (EGFR) and Cortactin are molecular markers that are important in tumour progression and development, and interact within the EGF pathway. Although EGFR antibody therapy exists, sufficient efforts for increased survival are still lacking due to the present limited response rates. The aim of the present study was to examine the association between EGFR and Cortactin expression on survival rates of OSCC patients and to determine whether EGFR and Cortactin expression levels are associated with advanced tumor sizes and lymphnode‑metastases. In total, 222 OSCC patients were included in the study. EGFR and Cortactin expression in tumor tissue was evaluated by immunohistochemistry. Cox regression was used for survival analysis. Categories were tested for associations by using cross tabs (Chi‑square test). Groups were compared by the non‑parametric Mann Whitney U‑test. Probabilities of less than 0.05 were considered significant and significant expression of Cortactin was observed in Advanced Union Internationale Contre le Cancer stage (P=0.032), including advanced tumour stage (P=0.021) and lymph node metastasis (P=0.049). High Cortactin expression was significantly associated with poorer survival rates (P=0.037). Further Cortactin expression was not associated with extracapsular spread, however EGFR exhibited a significant association (P=0.034). Neither EGFR nor Cortactin expression was correlated to grading. EGFR and Cortactin co‑expression was demonstrated to be significantly associated with poorer survival rates in OSCC patients, suggesting that identification of predictive biomarkers for adjuvant therapies are of primary concern in OSCC. In particular, efficient dual‑target therapy may act as an appropriate therapy to improve survival time for patients at advanced OSCC tumor stages.

Introduction

Oral cancer is one of the most common cancers. Male patients between the fifth and sixth decade are mainly affected by oral squamous cell carcinoma (OSCC) (1). Advanced stages are often present at primary diagnosis. Unfortunately, new insights into the understanding and further treatment options of OSCC are still lacking (1). Surgery has advanced opportunities for good functional and reconstructive results (2). Nevertheless, recurrence and second tumours are dominating and influencing survival rates (3). TNM classification and grading were discussed as having a high impact on patients outcomes. However, even recent studies indicate that this is not a sufficient explanation system for predicting recurrence (4). Poor survival rates reflect the status quo of OSCC and further therapy is frustrating in many cases. The need to identify molecular markers with a therapeutical impact on metastasis and recurrence is urgent. The correlation between tumour biology and survival rates is becoming more relevant. Further, tumour biology might lead to suitable therapeutic strategies. The creation of a variable patient-specific key target therapy with acceptable side effects is a main goal of today's research (5). Various molecular markers have emerged and have provided a new understanding of pathogenesis in OSCC. Epidermal growth factor receptor (EGFR) is one well studied target. The EGFR tyrosin kinase and its signal transduction pathway is a key route for distinct molecular interactions. Intracellular signalling chains, e.g., Ras/mitogen-activated protein kinase (MAPK) and the activation of transcription and extracellular chains, e.g., extracellular signal-regulated kinase (ERK) are activated through the EGFR. Endpoints of the signalling chain are supporting tumour growth, invasion, angiogenesis, metastases and interactions with lymph nodes. Essential research was carried out on the EGFR pathways with the emergence of EGFR antibody therapies (6).

Recently, three EGFR antibodies were developed. Cetuximab is a monoclonal immunglobulin G1 antibody inhibiting the receptor, whereas Erlotinib and Afatinib are blocking proteins of the ErbB family (7) and inhibit the tyrosin kinase activity of the receptor. To date, Cetuximab is the only EGFR antibody used in OSCC. EGFR overexpression was associated with poor prognosis and decreased survival rates in several OSCC studies. The first clinical study use of Cetuximab therapy in combination with radiotherapy in advanced OSCC was assessed in the often-cited study of Bonner et al (8). Effects of EGFR antibody therapy on survival rates were reported within this trial, but with a limited time benefit for the patients.

Despite these milestones at the beginning of the clinical use, the treatment response to EGFR inhibitors is not always sufficient resulting in a low or lack of impact on survival rates and is also dependent on the amount of EGFR expression (6). Further, up to 12 potential ligands next to EGF exist that could interact with the receptor and mutations of the receptor are not rare and could negatively interact with the response (9). Therefore improvements in therapy influencing survival rates are necessary. One option to maximise therapeutical effects is to block more targets than one in a single signal transduction pathway. Dual-blocking or dual-targeting was also considered as a method to enhance the anti-tumour response and is a topic of high clinical impact (10). The combination of antibodies targeting two patterns offers the chance to strengthen effiacacy without increasing side-effects. One substrate of the EGFR cascade is Cortactin (11). Cortactin is located within the cytoplasm and around the nucleus. It also co-localises with actin in the plasma membrane and at peripheral adhesion sites (12,13). Currently, the activity of Cortactin presents many unsolved questions because of its complexity (14). Important actions of Cortactin include cell spreading and adhesion (15). Hence, Cortactin is clearly also essential in tumour progress. An interpretation of the intensity of staining, of the proportion of stained cells, of the score of positivity and of the use of recommended scores is possible and has to be carried out carefully and independently (16). Therefore, our aim was, to use immunohistochemistry (IHC) indexing to create subgroups with meaningful numbers of patient samples in order to avoid overlaps, any interference of subgroups with insufficient numbers and any lack of clarity. Our objective was further to investigate the clinicopathological and prognostic significance of the co-expression of EGFR and Cortactin via immunohistochemical staining and to determine whether a collective of OSCC patients had sufficient numbers for evaluation.

Patients and methods

Patients

In total, 222 patients were included in the current study. They were treated between 2009 and 2011 at our maxillofacial surgery department. Relevant data (Table I) from patients diagnosed with OSCC for statistical evaluation and formalin fixed and paraffin embedded tissue (FFPE) for laboratory use were available in every single case. Regular follow up examinations of every included patient were held at our department according to the German guidelines of oral cancer (17). All included patients received regular follow up. In the first 2 years after the diagnosis the follow up was done every 3 months, after 2 years the follow up was done every 6 months until the fifth year. After the fifth year our follow up was completed.

Table I.

Clinical Parameters of the cohort-not subdivided.

Table I.

Clinical Parameters of the cohort-not subdivided.

Clinical ParametersTotal (n=222)
Median age in years (range)60.1 (49.2–69.7)
Gender
  Male/female175/47
UICC stage
  I  25
  II  40
  III  42
  IVa118
Tumour size
  T1  46
  T2  92
  T3  34
  T4a/b  50
N Stage
  N0  97
  N1  37
  N2  88
  Extracapsular spread  24
Grading
  G1  12
  G2113
  G3  97

The therapy regimes of the included patients were primary surgery, with intra-operative margin control via the help of frozen sections and with neck dissection with the intention of curative treatment. All tumour tissues were collected at the main tumour operation, which also included neck dissection. The tumour was operated by excisional biopsy of the whole tumour. Postoperative adjuvant cisplatin-based chemoradiation was performed in cases of pN1, pN2 or tumour infiltration of the jaw or locally infiltrating tumour growth of the oral cavity (T4a/b) and of positive microscopic resection margins and/or extracapsular spread, also according to the German guidelines for oral cancer as previously described (18).

Exclusion criteria were death resulting from a cause other than OSCC, distant metastasis at primary diagnosis and the use of primary radiochemotherapy before operation. The methods were approved by the ethics committee of the Technische Universität München (no. 212108) and are in accordance with the Declaration of Helsinki.

Tissue microarray (TMA) construction

Two independent pathologists defined the centre of the tumour and the invasion front of every study patient. The tissue was formalin-fixed and paraffin-embedded in blocks. The pathologists then marked the areas to be represented in the TMA. A minimum of two tumour cores from the centre of the tumour, the invasion front and the corresponding lymph nodes with a 6-mm core size were assembled into the TMA by using a Tissue Microarrayer (Beecher Instruments, Inc., Sun Prairie, WI, USA) as previously described (18,19). All lymph nodes, used for the TMA, were positive lymph nodes if the patient had positive lymph nodes. If the patient had no positive lymph nodes, negative lymph nodes were taken. Therefore lymph nodes of every patient were presented in the TMA.

Immunohistochemistry

Immunohistochemical staining was performed as described previously (20) by using 4-µm-thick sections of the TMA. The sections were incubated with primary antibodies against EGFR (1:50; Dako, Hamburg, Germany); and Cortactin (1:100; BD Bioscience, Heidelberg; Germany) overnight according to the manufacturers' recommendations.

Scoring

Immunohistochemical samples were blind-scored by two investigators and checked by one pathologist. EGFR and Cortactin staining was evaluated under a light microscope (magnification, ×200). The immunostaining intensity and positive cell proportion were assessed for both markers. Further, the staining was evaluated via an immunoreactive score (IRS) (21). We also evaluated the EGFR expression of both the cell cytoplasm and the cell membrane independently, since EGFR has two cellular loci of expression.

The staining intensity score was adjusted on a scale of 0-1-2-3: no staining was scored as 0; weak staining as 1; intermediate staining as 2; and strong staining as 3. Positive cell proportion was also assigned (0<25%; 1 if 25–50%; 2 if 50–75%; and 3 if >75%) as previously described (22). For a combination of quality and quantity, scores of intensity and quantity were multiplied (IRS results: 0-1-2-4-6-9). For the evaluated markers of the current cohort a final cut off score was determined as I (low expression: 0–4) and II (high expression: 6–9).

Statistical analysis

Data was analyzed with the SPSS for Windows, release 24.0.0, 2016 (SPSS, Inc., Chicago, IL, USA) and results were presented as figures. Cox regression and Kaplan Meier curves were used for survival analysis. Categories were tested for associations by using cross tabs (Chi-square test). To compare groups, the non-parametric Mann Whitney U-test was used. P<0.05 was considered to indicate a statistically significant difference.

Results

IHC scoring system of EGFR and Cortactin

We analysed staining as described in the methods section for cytoplasmic EGFR, membrane EGFR and Cortactin. The staining results of the cut off value groups I and II for the evaluated markers of the current cohort are shown in Fig. 1 (cytoplasmic EGFR, membrane EGFR and Cortactin).

Association of Cortactin expression to the survival rates

We used the Chi-square test to compare the clinicopathological parameters between the Cortactin low expression (I) and high expression (II) group. We did these tests for every TMA localisation. All the following results are only valid for the expression of the central tumour area. The invasion front and lymph nodes had no impact on the evaluation of expression. The analysis showed that overall survival was significantly poorer (P=0.037) in the case of Cortactin II: 50.3 months [SD 3.59; 95% confidence interval (CI): 43.28–57.36] compared with Cortactin I: 63.7 months (SD 4.82; 95% CI: 54.22–73.13: Kaplan Meier curves of Cortactin). Remarkably, during the analysis, Cortactin with a high expression score had an influence on clinicopathological data (Table II). Cortactin II was significantly associated with advanced UICC stages, especially III and IV (P=0.032). T1 stages were rare in Cortactin II (P=0.021). The incidence of lymphatic invasion (P=0.049) also dominated in Cortactin II and showed significantly more N2 stages in this cohort. Grading (P=0.057) and extracapsular spread (P=0.15) had no influence.

Table II.

Clinical parameters of the cohort-subdivided to EGFR and Cortactin expression.

Table II.

Clinical parameters of the cohort-subdivided to EGFR and Cortactin expression.

Clinical parametersEGFRCortactin
Median age in years (range)57.6 (49.5–64.8)57.4 (44.3–65.8)
Gender
Male/female84/1591/32
UICC stage
  I1510
  II1426
  III1923
  IVa4966
Tumour size
  T12818
  T23359
  T31222
  T4a/b2525
N Stage
  N04453
  N12116
  N23058
  Extracapsular spread  816
Grading
  G1  7  5
  G24745
  G36652

[i] EGFR, epidermal growth factor receptor.

Association of EGFR expression to the survival rates

The Chi-square test was also used to compare the clinicopathological parameters between the EGFR low expression (I) and high expression (II) group. We performed these tests for every TMA localisation as for the Cortactin cohort. We evaluated cytoplasmic EGFR and membrane EGFR. All the following results are only valid for the expression of the central tumour area. The invasion front and lymph nodes had no impact on the evaluation of expression. The analysis showed that overall survival did not differ in dependence on EGFR expression (cytoplasmic EGFR, P=0.636; membrane EGFR, P=0.978). The average survival of the cytoplasmic EGFR cohort for I was: 84.5 months (SD 7.98; 95% CI: 68.92–100.18) and for II was 89.6 months [SD 7.49; 95% CI: 74.92–104.30, Fig. 2A (Kaplan Meier curves of cytoplasmic EGFR)]. The membrane EGFR cohort had an average survival for I of 99.5 months (SD 13.29; 95% CI: 73.43–125.50) and for II of 99.5 months [SD 13.29; 95% CI: 73.43–125.50, Fig. 2B (Kaplan Meier curves of membrane EGFR)]. Furthermore, cytoplasmic EGFR and membrane EGFR in an high expression score did not have an influence on clinicopathological data (Table II). EGFR was not significantly associated with advanced UICC stages (cytoplasmic EGFR, P=0.094; membrane EGFR, P=0.113) nor T stage (cytoplasmic EGFR, P=0.670; membrane EGFR, P=0.439) or N stage (cytoplasmic EGFR, P=0.473; membrane EGFR, P=0.113). Moreover, grading (P=0.33) had no influence. Remarkably, extracapsular spread was significantly associated with high cytoplasmic EGFR expression (P=0.034).

Association of EGFR expression to Cortactin expression and the survival rates

Interestingly, a strong co-expression in the tumour centre of EGFR II and Cortactin II led significantly to reduced survival rates (P=0.04) with a median of a reduction in survival by 8 months.

Clinical data

Relevant clinical data from the 222 included patients with an OSCC diagnosis are listed in Table I. The average survival of the cohort was 88.4 months (SD 4.8; 95% CI: 78.84–90.04). Risk factors such as smoking and alcohol consumption were evaluated, with approximately 50% of the patients having a positive anamnesis. Expression of EGFR II and Cortactin II in combination of smoking and regular alcohol consumption was observed in 10% of all patients.

Lymph node recurrence played a major role in survival (P=0.028), whereas local recurrence did not (P=0.128). Lymph node recurrence occurred in 21 patients, which means that recurrence of lymph nodes metastasis occured after surgical removal and neck dissection. Local recurrence was evaluated in 35 patients.

The UICC stage (P=0.031), age of patients (P=0.012) and lymph node metastasis (P=0.003) at the time of primary diagnosis had a significant influence on overall survival rates.

In contrast, patient gender, T category, extra capsular spread and tumour grading were not significantly associated with overall tumour-related survival (P>0.05) and were independent of the marker expression status.

Discussion

The EGF cascade is an important pathway that is upregulated in a high percentage of human tumours (23). EGF and its receptor have complex influences on cell signalling and are key targets in oncology. EGFR expression has been studied in various malignomas (24) and EGFR interaction was often correlated with survival rates (25). Several studies emphasised that expression level of EGFR is proportional to recurrence, therapy failure and worse overall survival in OSCC (26). However, on the other hand, various authors have argued that this does not reflect reality and, to date, many trials are questioning the statement of the proportionality of EGFR to worse survival rates (27). Therefore, one of our aims was to do further research in the field of EGFR expression in a cohort of OSCC with a large number of patients. Our results from the current study do not confirm the unlimited correlation of high EGFR expression to lower survival rates. Monoclonal antibody therapy is linked to specific targets such as glycoproteins, vascular targets, growth factors, stromal antigens and the cluster of differentiation antigens (28). These therapies are applied only in well-defined special clinical cases. Currently, these individual target therapies are rescue therapies in OSCC and other malignancies and are applied after the first- or second-line therapy failed or in the case of recurrence after the first line therapy protocol has been administered (29). Further, these antibody therapies depend on the expression of the molecular target in order to be started. In the case of OSCC, EGFR antibody therapy is selected if cisplatin-based radiotherapy was unable to lead the tumour into remission (6). Cetuximab is the antibody of choice in the therapy of OSCC. The tyrosin kinase inhibitors Erlotinib and Afatinib are developed e.g., for use in lung cancer and gastric cancer (30,31). Yet these tyrosin kinase inhibiting antibodies are not authorised for the clinical use in OSCC. Due to our results showing the lack of influence of EGFR expression on survival rates, as previously suggested by other studies, we emphasise hereby the importance of double-target blocking with additional key targets as EGFR monotherapy might not be sufficient to eliminate EGFR-positive tumours (32). Another important issue is that resistance of EGFR to the antibody therapy are emerging, and could cause an altered therapy response. In particular, associations to the expression of multi-drug resistance proteins are newly being discussed (33,34). Molecular cross-talk offers options for identifying targets for future therapies (35). Further, a strong clinical correlation of every target is necessary. Several co-targets are considered in the literature. Cortactin plays a major role in cell interactions and Cortactin influences survival in OSCC in a significant way according to our current results. We could set a proof-of-principle in our cohort with regard to the influence of Cortactin in OSCC. To the best of our knowledge, the expression of both EGFR and Cortactin was not evaluated previously in OSCC. Our results show, that these interactions should not be disregarded. Discordance to previous published results regarding Cortactin expression can be explained on the basis of the use od smaller cohorts (36). Evaluation methods such as IHC scoring must be extremely detailed and well thought out to provide safe prognostic values of potential biomarkers (16,37). Therefore, we conducted IRS scoring and further divided the collective into the score cohorts I and II to avoid any interferences of subgroups with insufficient numbers. Moreover, we evaluated distinct localisations of the tumour, as also heed in the TMA: the centre of the tumour, the invasion front and the corresponding lymph nodes because of the potential differential expression of the biomarkers (38). Hence, we evaluated the mentioned tumour regions independently. Our results showed that significant interactions of Cortactin occur in the central tumour area. The area of tumour invasion and the lymph nodes play no significant role in Cortactin expression and have no influence on clinical features. In previous studies, Cortactin expression was reported in advanced stages of OSCC (39). Nevertheless, none of these few studies evaluated distinct tumour areas separately for Cortactin (40) as it was conducted successfully in the present study. EGFR staining is very common in clinical routine and is the basis of several studies. However, to our knowledge, studies having the topic EGFR and OSCC did not differ between the two expression sites of EGFR as we have for cytoplasmic EGFR and membrane EGFR (41). In the present study we were able to evaluate differences in these localisations. We found a significant correlation of high cytoplasmic EGFR expression and extracapsular spread in the central tumour area. In the literature, this interesting fact was not reported before. Only the general presence of EGFR expression, rather than its detailed cellular localisation and increased extracapsular spread were reported (42). According to the present understanding of molecular oncology, the differential results regarding the localisations of EGFR expressions might lead to significantly different outcomes and should be taken into consideration. The results of our current study based on a cohort with a large size and the complete availability of after-care data, suggest that the staining and evaluation of Cortactin expression have translational clinical impact and the results of our study are of high relevance. In particular, the majority of included patients were primarily diagnosed with advanced UICC stages (III and IV). Curative surgical treatment is often not possible for these stages and further therapy strategies are all the more important for these cohorts. Our results indicate for the first time that Cortactin is a protein having a concomitant and not a compensatory pathway next to EGFR. This result is essential, since cross-talk therapy is based on molecules that are independent of each other in expression. In summary, we showed that the dual-antibody-therapy targeting EGFR and Cortactin is superior to EGFR targeting alone in OSCC (43). Cortactin might represent an important molecule for the therapeutic approaches urgently needed to solve the problems of mutations and therapy resistances (44) and of recurrence. Regarding other malignancies, for example lymphocytic leukaemia, Cortactin plays also an important role as a checkpoint molecule (45). In colon cancer, Cortactin promotes cell migration and invasion (46). These findings are showing the importance of further studies with the subject Cortactin.

Immunohistochemical evaluations have their limitations. Because only protein expression can be evaluated by IHC, genetic profiles and further cellular interactions remain unknown. Further studies are needed to answer these open molecular questions.

Our results indicate that Cortactin could be a prognostic marker for OSCC and also that the co-expression of EGFR and Cortactin could have a clinical impact on survival rates. The development of a Cortactin antibody to improve the stagnated survival rates of OSCC patients is worthy of further studies. Mainly in advanced UICC stages (III and IV) this cross link antibody therapy could be the future therapy of choice, since conventional therapies have only a limited range. The genetic regulations of these markers should now be evaluated to substantiate the findings of the current study.

Acknowledgements

The authors thank Daniela Hellmann for excellent technical support. This study was supported by the German Research Foundation (DFG) and the Technical University of Munich (TUM) in the framework of the Open Access Publishing Program.

References

1 

Scully C and Bagan JV: Recent advances in oral oncology. Oral Oncol. 43:107–115. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Breeze J, Morrison A, Dawson D, Tipper J, Rehman K, Grew N and Pigadas N: Health-related quality of life after treatment for neoplasia of the major salivary glands: A pilot study. Br J Oral Maxillofac Surg. 54:806–811. 2016. View Article : Google Scholar : PubMed/NCBI

3 

González-García R, Naval-Gías L, Román-Romero L, Sastre-Pérez J and Rodríguez-Campo FJ: Local recurrences and second primary tumors from squamous cell carcinoma of the oral cavity: A retrospective analytic study of 500 patients. Head Neck. 31:1168–1180. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Lindenblatt Rde C, Martinez GL, Silva LE, Faria PS, Camisasca DR and Lourenço Sde Q: Oral squamous cell carcinoma grading systems-analysis of the best survival predictor. J Oral Pathol Med. 41:34–39. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Gupta S, Khan H, Kushwaha VS, Husain N, Negi M, Ghatak A and Bhatt M: Impact of EGFR and p53 expressions on survival and quality of life in locally advanced oral squamous cell carcinoma patients treated with chemoradiation. Cancer Biol Ther. 16:1269–1280. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Argiris A: EGFR inhibition for recurrent or metastatic HNSCC. Lancet Oncol. 16:488–489. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Cho HS and Leahy DJ: Structure of the extracellular region of HER3 reveals an interdomain tether. Science. 297:1330–1333. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, Jones CU, Sur R, Raben D, Jassem J, et al: Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med. 354:567–578. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Nakamura H, Koizumi H, Kimura H, Marushima H, Saji H and Takagi M: Epidermal growth factor receptor mutations in adenocarcinoma in situ and minimally invasive adenocarcinoma detected using mutation-specific monoclonal antibodies. Lung Cancer. 99:143–147. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Mabry R, Gilbertson DG, Frank A, Vu T, Ardourel D, Ostrander C, Stevens B, Julien S, Franke S, Meengs B, et al: A dual-targeting PDGFRbeta/VEGF-A molecule assembled from stable antibody fragments demonstrates anti-angiogenic activity in vitro and in vivo. MAbs. 2:20–34. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Buday L and Downward J: Roles of cortactin in tumor pathogenesis. Biochim Biophys Acta. 1775:263–273. 2007.PubMed/NCBI

12 

Wu H, Reynolds AB, Kanner SB, Vines RR and Parsons JT: Identification and characterization of a novel cytoskeleton-associated pp60src substrate. Mol Cell Biol. 11:5113–5124. 1991. View Article : Google Scholar : PubMed/NCBI

13 

Belsches AP, Haskell MD and Parsons SJ: Role of c-Src tyrosine kinase in EGF-induced mitogenesis. Front Biosci. 2:d501–d518. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Meiler E, Nieto-Pelegrín E and Martinez-Quiles N: Cortactin tyrosine phosphorylation promotes its deacetylation and inhibits cell spreading. PLoS One. 7:e336622012. View Article : Google Scholar : PubMed/NCBI

15 

Kruchten AE, Krueger EW, Wang Y and McNiven MA: Distinct phospho-forms of cortactin differentially regulate actin polymerization and focal adhesions. Am J Physiol Cell Physiol. 295:C1113–C1122. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Halon A, Donizy P, Biecek P, Rudno-Rudzinska J, Kielan W and Matkowski R: HER-2 expression in immunohistochemistry has no prognostic significance in gastric cancer patients. ScientificWorldJournal. 2012:9412592012. View Article : Google Scholar : PubMed/NCBI

17 

Wolff KD, Follmann M and Nast A: The diagnosis and treatment of oral cavity cancer. Dtsch Arztebl Int. 109:829–835. 2012.PubMed/NCBI

18 

Götz C, Drecoll E, Straub M, Bissinger O, Wolff KD and Kolk A: Impact of HPV in oral squamous cell carcinoma. Oncotarget. 7:76704–76712. 2016.PubMed/NCBI

19 

Young RJ, Urban D, Angel C, Corry J, Lyons B, Vallance N, Kleid S, Iseli TA, Solomon B and Rischin D: Frequency and prognostic significance of p16 (INK4A) protein overexpression and transcriptionally active human papillomavirus infection in laryngeal squamous cell carcinoma. Br J Cancer. 112:1098–1104. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Kolk A, Jubitz N, Mengele K, Mantwill K, Bissinger O, Schmitt M, Kremer M and Holm PS: Expression of Y-box-binding protein YB-1 allows stratification into long- and short-term survivorsof head and neck cancer patients. Br J Cancer. 105:1864–1873. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Fedchenko N and Reifenrath J: Different approaches for interpretation and reporting of immunohistochemistry analysis results in the bone tissue - a review. Diagn Pathol. 9:2212014. View Article : Google Scholar : PubMed/NCBI

22 

Bondarenko A, Angrisani N, Meyer-Lindenberg A, Seitz JM, Waizy H and Reifenrath J: Magnesium-based bone implants: Immunohistochemical analysis of peri-implant osteogenesis by evaluation of osteopontin and osteocalcin expression. J Biomed Mater Res A. 102:1449–1457. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Su CM, Chang TY, Hsu HP, Lai HH, Li JN, Lyu YJ, Kuo KT, Huang MT, Su JL and Chen PS: A novel application of E1A in combination therapy with EGFR-TKI treatment in breast cancer. Oncotarget. 7:63924–63936. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Xue ZX, Wen WX, Zhuang Y, Hua ZJ and Xia YN: Comparison of the efficacy of icotinib in patients with non-small-cell lung cancer according to the type of epidermal growth factor receptor mutation. Mol Clin Oncol. 5:265–268. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Grandis JR, Zeng Q, Drenning SD and Tweardy DJ: Normalization of EGFR mRNA levels following restoration of wild-type p53 in a head and neck squamous cell carcinoma cell line. Int J Oncol. 13:375–378. 1998.PubMed/NCBI

26 

Baschnagel AM, Tonlaar N, Eskandari M, Kumar T, Williams L, Hanna A, Pruetz BL and Wilson GD: Combined CD44, c-MET, and EGFR expression in p16-positive and p16-negative head and neck squamous cell carcinomas. J Oral Pathol Med. 46:208–213. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Hrustanovic G, Lee BJ and Bivona TG: Mechanisms of resistance to EGFR targeted therapies. Cancer Biol Ther. 14:304–314. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Scott AM, Allison JP and Wolchok JD: Monoclonal antibodies in cancer therapy. Cancer Immun. 12:142012.PubMed/NCBI

29 

Andreadis C, Vahtsevanos K, Sidiras T, Thomaidis I, Antoniadis K and Mouratidou D: 5-Fluorouracil and cisplatin in the treatment of advanced oral cancer. Oral Oncol. 39:380–385. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Pasini F, Fraccon AP, Modena Y, Bencivenga M, Giacopuzzi S, La Russa F, Gusella M and de Manzoni G: Targeted therapies for advanced and metastatic adenocarcinoma of the gastroesophageal junction: Is there something new? Gastric Cancer. 20:31–42. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Neumair P, Joos L, Warschkow R, Dutly A, Ess S, Hitz F, Früh M, Brutsche M, Baty F, Krähenbühl S, et al: Erlotinib has comparable clinical efficacy to chemotherapy in pretreated patients with advanced non-small cell lung cancer (NSCLC): A propensity-adjusted, outcomes research-based study. Lung Cancer. 100:38–44. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Oliveira-Silva RJ, de Carvalho Carolina A, de Souza Viana L, Carvalho AL and Reis RM: Anti-EGFR therapy: Strategies in head and neck squamous cell carcinoma. Recent Pat Anticancer Drug Discov. 11:170–183. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ma L, Zou B and Yan H: Identifying EGFR mutation-induced drug resistance based on alpha shape model analysis of the dynamics. Proteome Sci. 14:122016. View Article : Google Scholar : PubMed/NCBI

34 

Jin Y, Zhang W, Wang H, Zhang Z, Chu C, Liu X and Zou Q: EGFR/HER2 inhibitors effectively reduce the malignant potential of MDR breast cancer evoked by P-gp substrates in vitro and in vivo. Oncol Rep. 35:771–778. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Steinway SN, Dang H, You H, Rountree CB and Ding W: The EGFR/ErbB3 pathway acts as a compensatory survival mechanism upon c-met inhibition in human c-Met+ hepatocellular carcinoma. PLoS One. 10:e01281592015. View Article : Google Scholar : PubMed/NCBI

36 

Sato H, Hatanaka KC, Hatanaka Y, Hatakeyama H, Hashimoto A, Matsuno Y, Fukuda S and Sabe H: High level expression of AMAP1 protein correlates with poor prognosis and survival after surgery of head and neck squamous cell carcinoma patients. Cell Commun Signal. 12:172014. View Article : Google Scholar : PubMed/NCBI

37 

Aboshanif M, Kawasaki Y, Omori Y, Suzuki S, Honda K, Motoyama S and Ishikawa K: Prognostic role of regenerating gene-I in patients with stage-IV head and neck squamous cell carcinoma. Diagn Pathol. 11:792016. View Article : Google Scholar : PubMed/NCBI

38 

Gupta R, Chetty C, Bhoopathi P, Lakka S, Mohanam S, Rao JS and Dinh DE: Downregulation of uPA/uPAR inhibits intermittent hypoxia-induced epithelial-mesenchymal transition (EMT) in DAOY and D283 medulloblastoma cells. Int J Oncol. 38:733–744. 2011.PubMed/NCBI

39 

Yamada S, Yanamoto S, Kawasaki G, Mizuno A and Nemoto TK: Overexpression of cortactin increases invasion potential in oral squamous cell carcinoma. Pathol Oncol Res. 16:523–531. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Liu HS, Lu HH, Lui MT, Yu EH, Shen W, Chen YP, Chang KW and Tu HF: Detection of copy number amplification of cyclin D1 (CCND1) and cortactin (CTTN) in oral carcinoma and oral brushed samples from areca chewers. Oral Oncol. 45:1032–1065. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Pu YS, Huang CY, Kuo YZ, Kang WY, Liu GY, Huang AM, Yu HJ, Lai MK, Huang SP, Wu WJ, et al: Characterization of membranous and cytoplasmic EGFR expression in human normal renal cortex and renal cell carcinoma. J Biomed Sci. 16:822009. View Article : Google Scholar : PubMed/NCBI

42 

Michikawa C, Uzawa N, Sato H, Ohyama Y, Okada N and Amagasa T: Epidermal growth factor receptor gene copy number aberration at the primary tumour is significantly associated with extracapsular spread in oral cancer. Br J Cancer. 104:850–855. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Gonzales CB, De La Chapa JJ, Saikumar P, Singha PK, Dybdal-Hargreaves NF, Chavez J, Horning AM, Parra J and Kirma NB: Co-targeting ALK and EGFR parallel signaling in oral squamous cell carcinoma. Oral Oncol. 59:12–19. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Stewart EL, Tan SZ, Liu G and Tsao MS: Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-a review. Transl Lung Cancer Res. 4:67–81. 2015.PubMed/NCBI

45 

Martini V, Gattazzo C, Frezzato F, Trimarco V, Pizzi M, Chiodin G, Severin F, Scomazzon E, Guzzardo V, Saraggi D, et al: Cortactin, a Lyn substrate, is a checkpoint molecule at the intersection of BCR and CXCR4 signalling pathway in chronic lymphocytic leukaemia cells. Br J Haematol. 178:81–93. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Wang ZN, Liu D, Yin B, Ju WY, Qiu HZ, Xiao Y, Chen YJ, Peng XZ and Lu CM: High expression of PTBP1 promote invasion of colorectal cancer by alternative splicing of cortactin. Oncotarget. 8:36185–36202. 2017.PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bissinger O, Kolk A, Drecoll E, Straub M, Lutz C, Wolff KD and Götz C: EGFR and Cortactin: Markers for potential double target therapy in oral squamous cell carcinoma. Exp Ther Med 14: 4620-4626, 2017
APA
Bissinger, O., Kolk, A., Drecoll, E., Straub, M., Lutz, C., Wolff, K., & Götz, C. (2017). EGFR and Cortactin: Markers for potential double target therapy in oral squamous cell carcinoma. Experimental and Therapeutic Medicine, 14, 4620-4626. https://doi.org/10.3892/etm.2017.5120
MLA
Bissinger, O., Kolk, A., Drecoll, E., Straub, M., Lutz, C., Wolff, K., Götz, C."EGFR and Cortactin: Markers for potential double target therapy in oral squamous cell carcinoma". Experimental and Therapeutic Medicine 14.5 (2017): 4620-4626.
Chicago
Bissinger, O., Kolk, A., Drecoll, E., Straub, M., Lutz, C., Wolff, K., Götz, C."EGFR and Cortactin: Markers for potential double target therapy in oral squamous cell carcinoma". Experimental and Therapeutic Medicine 14, no. 5 (2017): 4620-4626. https://doi.org/10.3892/etm.2017.5120