Open Access

EGFR mutations subset in Chinese lung squamous cell carcinoma patients

  • Authors:
    • Ying Sun
    • Xin Yin
    • Miao‑Miao Wen
    • Jiao Zhang
    • Xue‑Jiao Wang
    • Jing‑Hua Xia
    • Yan‑Ning Zhang
    • Zhi‑Pei Zhang
    • Xiao‑Fei Li
  • View Affiliations

  • Published online on: April 5, 2018     https://doi.org/10.3892/mmr.2018.8859
  • Pages: 7575-7584
  • Copyright: © Sun 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

Abstract. Research has identified that epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) possess large benefits for adenocarcinoma (ADC), although little benefit for squamous cell carcinoma (SCC). The aim of the present study was to investigate the percentage of patients with SCC with the EGFR mutations subset and the benefits of EGFR TKIs in SCC. In the present study, the EGFR mutations subset was detected with an amplification refractory mutation system in 1,359 clinical SCC tissues. The association of the EGFR mutations subset with clinicopathological parameters was evaluated using the Mann‑Whitney U test, and Kruskal‑Wallis H. Kaplan‑Meier survival analysis was used to estimate the effect of the EGFR mutations subset on SCC patient survival rates. A total of 94 out of 1,359 SCC patients were identified as having EGFR mutations, an EGFR mutation rate of 6.92%. The EGFR mutations subset in the 94 cases was identified as follows: 37.2% (35/94) in exon 19; 39.4% (37/94) in L858R; 5.3% (5/94) in T790M; 4.3% (4/94) in G719X; 2.1% (2/94) in L861Q; and 11.7% (11/94) in other mutations. Kaplan‑Meier survival analysis identified that the differentiation, pathological tumor, node, metastasis stage, lymph node metastasis and distant metastases were significantly associated with patients' survival (P>0.05; log‑rank test), and no significant difference was observed between TKI therapy and chemotherapy in terms of patient survival rates (P>0.05). In addition, the overall discordant rate of the EGFR mutations subset in SCC patients was relatively low. Due to the non‑significant difference between TKI therapy and chemotherapy in terms of patient survival and the lower discordance rate of the EGFR mutations subset in SCC patients, EGFR TKIs could be a recommended treatment for SCC.

Introduction

Epidermal growth factor receptor (EGFR), a trans-membrane glycoprotein, may be distinguished into three parts: an extracellular domain, a transmembrane domain and an intracellular domain, which possesses tyrosine kinase (TK) activity. Particular mutations of EGFR, for example EGFR exon 19 deletion and exon 21 point mutation, serve a key role in the development of non-small cell lung cancer (NSCLC), such as promoting cancer cell proliferation, differentiation, revascularization and metastasis (1). EGFR TK inhibitors (TKIs), mainly targeting EGFR TK activities, are able to effectively prolong the survival time of patients with cancer. In particular, a number of studies have demonstrated the efficacy of the two most widely available TKIs, erlotinib and gefitinib, in the first-line, maintenance and relapsed settings (24).

The response rate of EGFR mutation activation to EGFR TKI treatment reaches 70%; TKI treatment may additionally prolong the NSCLC patients' progression free survival (PFS) (58). EGFR exon 19 deletion (Del 19) and exon 21 point mutation (L858R), the main EGFR mutation activation, primarily exist in females, never-smokers, East Asians (~50%) and patients with lung adenocarcinoma (ADC, ~30%) (912). However, activation of EGFR mutations are rare in patients with squamous cell carcinoma (SCC) (<3%); the lack of reported mutations may limit the use of EGFR-TKIs in lung cancer patients with SCC (1317). To date, the benefits of EGFR-TKIs in EGFR-mutated patients with SCC have not been well-studied.

In the present study, the mutation rate of SCC in all EGFR mutations was analyzed in our laboratory and the correlation of EGFR-mutations associated with SCC under clinicopathological parameters was conducted. A Kaplan-Meier survival analysis performed to estimate the effect of the EGFR mutations in the survival rates of patient with SCC. Additionally, the potential implications of EGFR TKI treatment and the tumor biology of EGFR mutations in patients with SCC was investigated. The aim of the present study was to investigate the mutations of EGFR in lung SCC; the results of the present study may contribute to developments in treatments for lung SCC.

Materials and methods

Patients and samples

All SCC samples (tumor tissues, blood and pleural effusion) used in the present study were collected between 2010 and 2016 from clinical data or an archived thoracic oncology tissue repository at the Department of Thoracic Surgery of Tangdu Hospital affiliated with The Fourth Military Medical University (Xi'an, China; Fig. 1). Patients who had received preoperative chemotherapy, radiotherapy or EGFR-targeted therapy were excluded from the present study. Detailed clinicopathological information, including patient ages, sex, smoking history, tumor status, histological differentiation, nodal status, clinical manifestation, surgical method, postoperative treatment and follow-up information were collected and completed. The surgery day was considered to be the starting day for estimating postoperative survival time. The follow-up lasted until August 13, 2016, with a median follow-up period of 39.62 months (range, 2–63.28 months). The histological classification of the tumors was reviewed by pathologists. And all tumors were staged according to the pathological tumor, node, metastasis (pTNM) classification (7th edition) of the International Union against Cancer (18). The study protocol was approved by the Regional Ethics Committee for Clinical Research of The Fourth Military Medical University. All patients provided written informed consent for use of their medical records and tumor specimens for research purposes.

EGFR mutation testing

Genomic DNA was isolated and purified from fresh tumor specimens using TIANamp Genomic DNA kit (Taingen Biotech, Beijing, People's Republic of China) according to the manufacturer's instructions.

EGFR mutation analysis of DNA was performed using ADx-ARMS® technology, a technology based on amplified refractory mutation system (ARMS) (19). Quantitative polymerase chain reaction (qPCR) was conducted on the MX3005P qPCR system (Stratagene California; Agilent Technologies, Inc., Santa Clara, CA, USA) using the AmoyDx human EGFR Gene Mutation Detection kit (Amoy Diagnostics Co., Ltd., Xiamen, China) according to the manufacturer's protocols.

A total of 26 mutations in exon 18 (G719A, G719S, G719C), exon 20 (T790M, S768I), and exon 21 (L858R, L861Q), and exon 19 (deletions, n=19) were detected. The primer sequences of EGFR mutation testing were obtained from The Primer Express® Software v3.0.1 (Thermo Fisher Scientific, MA, Waltham, USA) was applied to design specific primers for these common mutations of the EGFR gene; the primer catalogue numbers of the 26 mutations of EGFR gene provided by the AmoyDx human EGFR Gene Mutation Detection kit are presented in Table I.

Table I.

Primer sequences for quantitative polymerase chain reaction.

Table I.

Primer sequences for quantitative polymerase chain reaction.

PrimerCatalogue number
18-F1SEQ ID NO:1
18-F2SEQ ID NO:2
18-F3SEQ ID NO:3
18-RSEQ ID NO:4
18-PSEQ ID NO:5
19-F1SEQ ID NO:6
19-F2SEQ ID NO:7
19-F3SEQ ID NO:8
19-F4SEQ ID NO:9
19-F5SEQ ID NO:10
19-PSEQ ID NO:11
19-RSEQ ID NO:12
20-F1SEQ ID NO:13
20-F2SEQ ID NO:14
20-F3SEQ ID NO:15
20-F4SEQ ID NO:16
20-F5SEQ ID NO:17
20-PSEQ ID NO:18
20-RSEQ ID NO:19
21-F1SEQ ID NO:20
21-F2SEQ ID NO:21
21-PSEQ ID NO:22
21-RSEQ ID NO:23
CTRL-FSEQ ID NO:24
CTRL-RSEQ ID NO:25
CTRL-PSEQ ID NO:26

The qPCR amplification program was performed as follows: Initial denaturation at 95°C for 5 min, 15 cycles of amplification (at 95°C for 25 sec, 64°C for 20 sec, and 72°C for 20 sec) and a final denaturation followed by 31 cycles of amplification (at 93°C for 25 sec, 60°C for 35 sec, and 72°C for 20 sec), and the FAM and HEX signals were collected at 60°C.

According to the manufacturer's protocols of the AmoyDx human EGFR Gene Mutation Detection kit described that samples were defined as EGFR mutation-negative when the Cq value≥34; when the sample mutation Cq value<31, the samples were defined as EGFR mutation-positive. When the calculated ΔCq value [ΔCq=Cq (sample)-Ct (control)], when ΔCq value <ΔCt (Cut-off) was 31≤ the sample mutation Cq value ≤33, the samples were defined as EGFR mutation-positive, and when the ΔCq value ≥ΔCq (Cut-off), the samples were defined as EGFR mutation-negative.

Discordance rate of EGFR mutations analysis

Patients with lung SCC (n=14) with EGFR activating mutations were used for discordance rate of EGFR mutations analysis. The tumor samples were fixed with 10% formaldehyde for 24 h at room temperature and embedded with paraffin. Sections were sliced to 4-µm thickness, deparaffinized with a series of xylene and rehydrated with a graded alcohol series. The sections were stained with hematoxylin for 40 sec, then counterstained with eosin for 1 min at room temperature. Finally, the sections were dehydrated with a graded alcohol series and embedded in paraffin. Inclusion in the study was based on the presence of morphologically different tumor areas within the same tumor and a sufficient cancer cell content (>30%) in each defined area. Subsequently, three parts of each individual tumor were selected, and EGFR mutation detection was performed. For every tumor, three areas were identified by three pathologists to represent the most distinct and variable histological patterns (Fig. 2).

Similarly, each of five such SCCs was divided into >100 segments. The present study used two sections from each tumor. Thus, each section yielded 50 or more pieces, which were 3×3 mm on average (Fig. 2). The EGFR mutations of each piece were detected independently. To ensure that the results were collected in an unbiased manner, mutations were analyzed by a different technician from the one who had scratched the tissues (20).

Statistical analyses

The correlations of EGFR mutations with clinicopathological parameters were statistically analyzed using the Mann-Whitney U test, and Kruskal-Wallis H (mainly used to detect pathological differentiation). Kaplan-Meier survival analysis was used to estimate the effect of the type of EGFR mutation on the survival of patients with SCC. Statistical analyses were performed using SPSS software version 18.0 (SPSS, Inc., Chicago, IL, USA). P-values were adjusted for multiple testing, and P>0.05 was considered to indicate a statistically significant difference.

Results

EGFR mutation

A total of 94 out of 1,359 patients with lung SCC had EGFR mutations (6.92%), and 1,265 patients did not. All EGFR mutations identified are present in Table II: Exon 19 (n=35, 37.2%); L858R (n=37, 39.4%); T790M (n=5, 5.3%); G719X (n=4, 4.3%); L861Q (n=2, 2.1%); and other mutations (n=11, 11.7%).

Table II.

Tumors examined in the present study.

Table II.

Tumors examined in the present study.

VariableNo. of tumors (n, %)Exon 19 (%)L858R (%)T790M (%)G719X (%)L861Q (%)Others (%)
EGFR-mutated SCC94 (1,359, 6.92)35 (37.2)37 (39.4)5 (5.3)4 (4.3)2 (2.1)11 (11.7)
Trans-sectional analysis of number of areas in a tumor
  314 (94, 14.9)4 (28.6)7 (50)2 (14.3)001 (7.1)
  1005 (14, 35.7)2 (40)3 (60)0000

[i] EGFR, epidermal growth factor receptor; SCC, squamous cell carcinoma.

Patient characteristics

The clinicopathological characteristics of the patients are summarized in Table III. In 94 SCCs with EGFR mutations, there were 18 female and 76 male patients, with a median age of 59 years (range, 36–84 years). Histopathological diagnoses included well-differentiated (10, 10.64%), moderate differentiation (60, 63.83%), and poor differentiation (24, 25.53%). Postoperative staging evaluation demonstrated stage I disease in 24 patients, stage II disease in 22 patients, stage III disease in 36 patients and stage IV disease in 12 patients. Metastatic sites included five brain metastases (5.3%), one liver metastasis (1.1%), eight bone metastases (8.5%), and one kidney metastasis (1.1%).

Table III.

Patient characteristics.

Table III.

Patient characteristics.

Patients (n=94)
Age, years
Median59
Range36–84

No.%

Sex
  Male7680.9
  Female1819.1
Smoking history
  Smoker6973.4
  Non-smoker2526.6
Differentiation
  Well1010.64
  Moderate6063.83
  Poor2425.53
pTNM stage
  I–II4648.9
  III–IV4851.1
Primary tumor location
  Central6973.4
  Peripheral2526.6
Primary tumor size, cm
  <4.24547.9
  ≥4.24952.1
Lymph node metastasis
  Yes4446.8
  No5053.2
No. of metastatic sites analyzed
  Brain  55.3
  Liver  11.1
  Bone  88.5
  Kidney  11.1

[i] pTNM, pathological tumor, node, metastasis classification.

Clinical characteristics in smoker and non-smoker patients with lung SCC with EGFR mutation

When comparing smoker and non-smoker patients in terms of baseline clinical characteristics, significant differences were identified in sex (smoker vs. non-smoker: 100% male vs. 28%, respectively P>0.001), differentiation (smoker vs. non-smoker: 71.01% moderate vs. 44%; 18.84% poor vs. 44%, P=0.036) and pTNM stage (smoker vs. non-smoker: 55.1% I–II vs. 32%, P=0.049). There was no significant difference in age and lymph node metastasis (Table IV).

Table IV.

Clinical characteristics in smoker and non-smoker patients with lung squamous cell carcinoma with EGFR mutation.

Table IV.

Clinical characteristics in smoker and non-smoker patients with lung squamous cell carcinoma with EGFR mutation.

VariablesNo. of cases, n=94 (%)Smoker, n=69 (%)Non-smoker, n=25 (%)P-value
Age, years   0.913
  <5946 (48.9)34 (49.3)12 (48)
  ≥5948 (51.1)35 (50.7)13 (52)
Sex <0.001
  Male76 (80.9)69 (100)  7 (28)
  Female18 (19.1)0 (0)18 (72)
Differentiation   0.036
  Well10 (10.64)7 (10.15)  3 (12)
  Moderate60 (63.83)49 (71.01)11 (44)
  Poor24 (25.53)13 (18.84)11 (44)
pTNM stage   0.049
  I–II46 (48.9)38 (55.1)  8 (32)
  III–IV48 (51.1)31 (44.9)17 (68)
Lymph node metastasis   0.546
  Yes44 (46.8)31 (44.9)13 (52)
  No50 (53.2)38 (55.1)12 (48)

[i] EGFR, epidermal growth factor receptor; pTNM, pathological tumor, node, metastasis classification.

Clinical characteristics in patients with early- and advanced-stage lung SCC with EGFR mutation

When comparing patients with early (I–II) and advanced (III–IV) stage in baseline clinical characteristics, significant differences were identified in smoking history (patients with early vs. advanced stage: 82.6% smoker vs. 64.6%, P=0.049), differentiation (patients with early vs. advanced stage: 71.74% moderate vs. 56.25%; 8.7% poor vs. 41.67%, P>0.001) and lymph node metastasis (patients with early vs. advanced stage: 21.74% metastasis vs. 70.83%, P>0.001). There were no significant differences in age and sex (Table V).

Table V.

Clinical characteristics in early stage and advanced stage lung squamous cell carcinoma patients with EGFR mutation.

Table V.

Clinical characteristics in early stage and advanced stage lung squamous cell carcinoma patients with EGFR mutation.

VariablesNo. of cases, n=94 (%)TNM I–II, n=46 (%)TNM III–IV, n=48 (%)P-value
Age, years 0.149
  <5946 (48.9)19 (41.3)27 (56.3)
  ≥5948 (51.1)27 (58.7)21 (43.7)
Sex 0.143
  Male76 (80.9)40 (87)36 (75)
  Female18 (19.1)6 (13)12 (25)
Smoking history 0.049
  Smoker69 (73.4)38 (82.6)31 (64.6)
  Non-smoker25 (26.6)8 (17.4)17 (35.4)
Differentiation <0.001
  Well10 (10.64)9 (19.56)1 (2.08)
  Moderate60 (63.83)33 (71.74)27 (56.25)
  Poor24 (25.53)4 (8.70)20 (41.67)
Lymph node metastasis <0.001
  Yes44 (46.8)10 (21.74)34 (70.83)
  No50 (53.2)36 (78.26)14 (29.17)

[i] EGFR, epidermal growth factor receptor; TNM, tumor, node, metastasis classification.

Clinical characteristics in young, middle-aged, and elderly patients with lung SCC with EGFR mutation

When comparing young, middle-aged and elderly patients in terms of baseline clinical characteristics, no significant differences were identified in the clinical characteristics of the patients (Table VI).

Table VI.

Clinical characteristics in young and elderly patients with lung squamous cell carcinoma with EGFR mutation.

Table VI.

Clinical characteristics in young and elderly patients with lung squamous cell carcinoma with EGFR mutation.

VariablesNo. of cases, n-94 (%)≤40 year of age, n=2 (%)41–60 years of age, n=56 (%)61–80 years of age, n=33 (%)>80 years of age, n=3 (%)P-value
Sex 0.929
  Male76 (80.9)0 (0)48 (85.7)25 (75.8)3 (100)
  Female18 (19.1)2 (100)8 (14.3)8 (24.2)0 (0)
Smoking history 0.613
  Smoker69 (73.4)0 (0)45 (80.4)21 (63.6)3 (100)
  Non-smoker25 (26.6)2 (100)11 (19.6)12 (36.4)0 (0)
Differentiation 0.797
  Well10 (10.64)0 (0)7 (12.5)3 (9.1)0 (0)
  Moderate60 (63.83)1 (50)37 (66.1)19 (57.6)3 (100)
  Poor24 (25.53)1 (50)12 (21.4)11 (33.3)0 (0)
pTNM stage 0.303
  I–II46 (48.9)0 (0)27 (48.2)16 (48.5)3 (100)
  III–IV48 (51.1)2 (100)29 (51.8)17 (51.5)0 (0)
Lymph node metastasis 0.108
  Yes44 (46.8)2 (100)28 (50)14 (42.4)0 (0)
  No50 (53.2)0 (0)28 (50)19 (57.6)3 (100)

[i] EGFR, epidermal growth factor receptor; pTNM, pathological tumor, node, metastasis classification.

Overall discordance rate of EGFR mutations in lung SCC

To determine the overall discordance rate of EGFR mutations, EGFR mutations in 14 SCCs were detected and analyzed. Three parts of each individual tumor were selected and examined for the EGFR mutations subset, and identical mutations were demonstrated in the three morphologically different tumor areas (Table VII).

Table VII.

Tumor cell content and epidermal growth factor receptor mutation status detected in the three histologically distinct tumor areas from each patient.

Table VII.

Tumor cell content and epidermal growth factor receptor mutation status detected in the three histologically distinct tumor areas from each patient.

Case no.AreaTumor cell content, %Predominant growth patternaMutation
1Tumor area 160KeratinizingExon 19 del E746-A750
Tumor area 260KeratinizingExon 19 del E746-A750
Tumor area 370KeratinizingExon 19 del E746-A750
2Tumor area 180 Non-keratinizingExon 21 L858R
Tumor area 285 Non-keratinizingExon 21 L858R
Tumor area 390KeratinizingExon 21 L858R
3Tumor area 155BasaloidExon 21 L858R
Tumor area 255WartyExon 21 L858R
Tumor area 350WartyExon 21 L858R
4Tumor area 160KeratinizingExon 21 L858R
Tumor area 265KeratinizingExon 21 L858R
Tumor area 365KeratinizingExon 21 L858R
5Tumor area 170BasaloidExon 21 L858R
Tumor area 275BasaloidExon 21 L858R
Tumor area 380BasaloidExon 21 L858R
6Tumor area 140BasaloidExon 21 L858R
Tumor area 250WartyExon 21 L858R
Tumor area 360BasaloidExon 21 L858R
7Tumor area 180KeratinizingExon 19 del E746-A750
Tumor area 270KeratinizingExon 19 del E746-A750
Tumor area 370 Non-keratinizingExon 19 del E746-A750
8Tumor area 155BasaloidExon 21 L858R
Tumor area 245WartyExon 21 L858R
Tumor area 350BasaloidExon 21 L858R
9Tumor area 145KeratinizingExon 20 Ins
Tumor area 235KeratinizingExon 20 Ins
Tumor area 340KeratinizingExon 20 Ins
10Tumor area 170BasaloidExon 21 L858R
Tumor area 270BasaloidExon 21 L858R
Tumor area 370BasaloidExon 21 L858R
11Tumor area 155KeratinizingExon 19 del E746-A750
Tumor area 260KeratinizingExon 19 del E746-A750
Tumor area 360BasaloidExon 19 del E746-A750
12Tumor area 140 Non-keratinizingExon 19 del E746-A750
Tumor area 230KeratinizingExon 19 del E746-A750
Tumor area 350KeratinizingExon 19 del E746-A750
13Tumor area 185WartyExon 20 T790M
Tumor area 285BasaloidExon 20 T790M
Tumor area 370BasaloidExon 20 T790M
14Tumor area 160 Non-keratinizingExon 20 T790M
Tumor area 260KeratinizingExon 20 T790M
Tumor area 360KeratinizingExon 20 T790M

a Predominant growth pattern in analyzed area in accordance with the International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society lung squamous cell carcinoma classification 2011 (21).

As three parts may be insufficient to detect the discordance rate of the EGFR mutations, five tumors were dissected into >100 pieces, and each piece was examined for EGFR mutations (Fig. 2). The results additionally revealed identical mutations throughout each individual tumor.

Clinical characteristic-associated prognosis of patients with lung SCC with EGFR mutations

Among clinicopathological factors, including age, sex, smoking history, et al differentiation, pTNM stage and lymph node metastasis were significantly associated with patient survival rates. Patients with well or moderately differentiated tumors [n=70; 95% confidence interval (CI), 45.036–56.253 months] exhibited longer durations of survival compared with those with poorly differentiated tumors (n=24; 95% CI, 20.905–43.613 months; P=0.005) (Fig. 3A). Patients with pTNM I–II tumors (n=46; 95% CI, 49.091–60.002 months) exhibited a longer duration of survival compared with those with pTNM III–IV tumors (n=48; 95% CI, 29.621–45.614 months; P<0.001; Fig. 3B). Patients with no lymph node metastasis (n=50; 95% CI, 46.783–58.485 months) exhibited a longer duration of survival compared with those with lymph node metastasis (n=44; 95% CI, 30.236–46.535 months; P=0.005; Fig. 3C).

The prognosis of patients with lung SCC with EGFR mutations associated with distant metastases, EGFR mutations, and postoperative treatment (chemotherapy and EGFR TKI) were subsequently investigated. Patients with non-distant metastasis (n=79; 95% CI, 42.350–53.076 months) exhibited a longer duration of survival compared with those with distant metastasis (n=15; 95% CI, 19.069–47.515 months; P=0.014; Fig. 3D). A significant difference was not observed between patients with L858R (n=37; 95% CI, 41.678–57.284 months) and patients with Del 19 (n=35; 95% CI, 28.587–45.703 months; P>0.05; Fig. 3E). Additionally, a significant difference between patients with aged 41–60 years (n=56; 95% CI, 37.213–51.322 months) and patients with aged 61–80 years was not observed (n=33; 95% CI, 40.064–56.205 months; P>0.05; Fig. 3F). Furthermore, a significant difference was observed between patients treated with TKI (n=24; 95% CI, 33.099–51.624 months) and patients treated with chemotherapy (n=66; 95% CI, 38.160–51.387 months; P>0.05; Fig. 3G).

Discussion

ADC, SCC, and large-cell undifferentiated carcinoma are the principal subsets of non-small cell lung cancer (NSCLC), and approximately 20–30% of cases of NSCLC are SCC (22). Historically, the subtype of NSCLC has not been a major factor in determining patient therapy management, and there is not been well established regarding the fundamental difference in the molecular pathogenesis of ADC and SCC (23). It is only in recent years that driver oncogenes, including EGFR-activating mutations, and subsequent corresponding therapies have been identified (7,2426). The majority of patients with NSCLC with EGFR mutations respond well to EGFR TKIs (including gefitinib and erlotinib). EGFR mutations are frequently observed in female, non-smoking, ADC and Asian patients, but rare in SCC (912). Research has identified that in pure SCC, there is the presence of fibroblast growth factor receptor 1, phosphatase and tensin homolog and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit á/AKT serine/threonine kinase 1 mutations, and an absence of EGFR and KRAS proto-oncogene GTPase mutations (27). Compared with lung ADC, evidence about the efficacy of EGFR TKIs and treatment progress in patients with lung SCC is limited and controversial (4,2830).

The present study performed ARMS analysis to investigate the EGFR mutations subset in clinical lung SCC samples. Statistical analysis revealed that 6.9% (94/1,359) of the tumor samples were EGFR-activating mutations. The EGFR mutated SCC samples were identified as follows: 37.2% (35/94) in exon 19; 39.4% (37/94) in L858R; 5.3% (5/94) in T790M; 4.3% (4/94) in G719X; 2.1% (2/94) in L861Q; and 11.7% (11/94) in other mutations (Table II). Due to the limited number in the EGFR mutations subset, only the proportions of EGFR mutations in exon 19 (Del 19) and exon 21 (L858R) were larger (~76.6% of the total), although no significant difference in prognosis was observed between the EGFR Del 19 and L858R groups in SCC.

In the present study, there were significant differences between the smoking group, pTNM stage group and baseline clinical characteristics. Recently, along with extended life span, patients >80 years of age are increasing in number, and differences in prognosis are significant in the age range 28–30 years (3133). However, due to the limited sample size, no significant difference was observed between very young and very elderly patients.

Previous studies on the role of EGFR TKIs in SCC have identified that EGFR TKIs may be an option for the treatment of SCC, and the EGFR mutations subset may help to select a subgroup of patients with best response to TKIs (3436). In the present study, among clinical characteristics, only the differentiation, pTNM stage, lymph node metastasis and distant metastases were significantly associated with patients' survival (P>0.05; log-rank test). The SCC patients identified as having EGFR activating mutations following surgery were treated as follows: 70.2% (66/94) with chemotherapy; 25.5% (24/94) with EGFR TKIs; 3.2% (3/94) with radiotherapy; and 1.1% (1/94) with chemotherapy and EGFR TKIs. However, the difference in prognosis was not marked between the chemotherapy and TKI therapy groups.

There may be specific reasons to explain these results, and it was hypothesized that EGFR TKIs may prolong patient survival in a way comparable to the function of chemotherapy. However, the present study used a limited sample size, thus a study with an expanded sample size is required. In addition, EGFR TKIs are used for patients with EGFR mutation. Whether used in ADC or SCC, EGFR TKIs are recommended as long as the EGFR site is mutated. In the present study, the prognostic difference was not marked between the chemotherapy and the TKIs therapy groups, which indicated that EGFR TKIs were able to prolong patient survival in way comparable to the function of chemotherapy; therefore, it was hypothesized that EGFR TKIs may be an option for the treatment of SCC with EGFR mutations.

In certain individual tumors, EGFR mutations were not evenly distributed, and this may be one of the causes of drug-resistance to EGFR TKIs. However, in previous studies, the opposite results have been demonstrated in lung ADC (18,26). In the present study, identical EGFR mutations were identified throughout individual tumors by examining 14 tumors divided into three parts and five tumors divided into 100 parts. However, the limited sample size is a shortcoming of the present study, thus it is intended to expand the sample size in the future.

The results of the present study suggested that EGFR Del 19/L858R may be the main EGFR mutations subset in SCC. The effect of EGFR TKIs on SCC patients' prognoses is the same as the effect of chemotherapy, showing fewer complications and a higher quality of life, so EGFR TKIs could be a worthwhile option for the treatment of SCC. In addition, the heterogeneous distribution of EGFR mutations in SCC is extremely rare.

Acknowledgements

The authors wish to thank Dr Xiao-li Li for assistance with software assays, who from the laboratory of Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University (Xi'an, China).

Funding

No funding was received.

Availability of data and materials

All data generated and analyzed during this study are included in this published article.

Authors' contributions

YS, XY and MW conducted the EGFR mutation test; JZ contributed to the follow-up; XW and JX interpreted the clinicopathological information statistics; YZ conducted the discordance rate of EGFR mutations analysis; and ZZ and XL performed statistical analysis. The manuscript was drafted by YS and edited by XY. All authors read and approved the manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Regional Ethics Committee for Clinical Research of the Fourth Military Medical University. All patients provided written informed consent for use of their medical records and tumor specimens for research purposes.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EGFR

epidermal growth factor receptor

TKIs

tyrosine kinase inhibitors

SCC

squamous cell carcinoma

ADC

lung adenocarcinoma

PFS

progression free survival

OS

overall survival

NSCLC

non-small cell lung cancer

References

1 

Cohen S: Purification of the receptor for epidermal growth factor from A-431 cells: Its function as a tyrosyl kinase. Methods Enzymol. 99:379–387. 1983. View Article : Google Scholar : PubMed/NCBI

2 

Mendelsohn J and Baselga J: Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol. 21:2787–2799. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Herbst RS, Maddox AM, Rothenberg ML, Small EJ, Rubin EH, Baselga J, Rojo F, Hong WK, Swaisland H, Averbuch SD, et al: Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: Results of a phase I trial. J Clin Oncol. 20:3815–3825. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Thatcher N, Chang A, Parikh P, Pereira Rodrigues J, Ciuleanu T, von Pawel J, Thongprasert S, Tan EH, Pemberton K, Archer V and Carroll K: Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: Results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet. 366:1527–1537. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Han SW, Kim TY, Hwang PG, Jeong S, Kim J, Choi IS, Oh DY, Kim JH, Kim DW, Chung DH, et al: Predictive and prognostic impact of epidermal growth factor receptor mutation in non-small-cell lung cancer patients treated with gefitinib. J Clin Oncol. 23:2493–2501. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Huang SF, Liu HP, Li LH, Ku YC, Fu YN, Tsai HY, Chen YT, Lin YF, Chang WC, Kuo HP, et al: High frequency of epidermal growth factor receptor mutations with complex patterns in non-small cell lung cancers related to gefitinib responsiveness in Taiwan. Clin Cancer Res. 10:8195–8203. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, et al: Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 350:2129–2139. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, et al: EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science. 304:1497–1500. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Pao W and Miller VA: Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: Current knowledge and future directions. J Clin Oncol. 23:2556–2568. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, Varella-Garcia M, Franklin WA, Aronson SL, Su PF, et al: Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 311:1998–2006. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Jackman DM, Miller VA, Cioffredi LA, Yeap BY, Jänne PA, Riely GJ, Ruiz MG, Giaccone G, Sequist LV and Johnson BE: Impact of epidermal growth factor receptor and KRAS mutations on clinical outcomes in previously untreated non-small cell lung cancer patients: Results of an online tumor registry of clinical trials. Clin Cancer Res. 15:5267–5273. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, et al: Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 361:958–967. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Jukna A, Montanari G, Mengoli MC, Cavazza A, Covi M, Barbieri F, Bertolini F and Rossi G: Squamous cell carcinoma ‘Transformation’ concurrent with secondary T790M mutaton in resistant EGFR-mutated adenocarcinomas. J Thorac Oncol. 11:e49–e51. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Kuiper JL, Ronden MI, Becker A, Heideman DA, van Hengel P, Ylstra B, Thunnissen E and Smit EF: Transformation to a squamous cell carcinoma phenotype of an EGFR-mutated NSCLC patient after treatment with an EGFR-tyrosine kinse inhibitor. J Clin Pathol. 68:320–321. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Levin PA, Mayer M, Hoskin S, Sailors J, Oliver DH and Gerber DE: Histologic transformation from adenocarcinoma to squamous cell carcinoma as a mechanism of resistance to EGFR inhibition. J Thorac Oncol. 10:e86–e88. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Billah S, Stewart J, Staerkel G, Chen S, Gong Y and Guo M: EGFR and KRAS mutations in lung carcinoma: Molecular testing by using cytology specimens. Cancer Cytopathology. 119:111–117. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Pao W, Kris MG, Iafrate AJ, Ladanyi M, Jänne PA, Wistuba II, Miake-Lye R, Herbst RS, Carbone DP, Johnson BE and Lynch TJ: Integration of molecular profiling into the lung cancer clinic. Clin Cancer Res. 15:5317–5322. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA, Rami-Porta R, Postmus PE, Rusch V and Sobin L: International Association for the Study of Lung Cancer International Staging Committee; Participating Institutions: The IASLC lung cancer staging project: Proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM classification of malignant tumours. J Thorac Oncol. 2:706–714. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Liu J, Zhao R and Zhang J and Zhang J: ARMS for EGFR mutation analysis of cytologic and corresponding lung adenocarcinoma histologic specimens. J Cancer Res Clin Oncol. 141:221–227. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Yatabe Y, Matsuo K and Mitsudomi T: Heterogeneous distribution of EGFR mutations is extremely rare in lung adenocarcinoma. J Clin Oncol. 29:2972–2977. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Travis WD: Pathology of lung cancer. Clin Chest Med. 32:669–692. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Travis WD, Brambilla E, Noguchi M, Nicholson AG, Geisinger KR, Yatabe Y, Beer DG, Powell CA, Riely GJ, Van Schil PE, et al: International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 6:244–285. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Lu J, Wang W, Xu M, Li Y, Chen C and Wang X: A global view of regulatory networks in lung cancer: An approach to understand homogeneity and heterogeneity. Semin Cancer Biol. 42:31–38. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, et al: Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 361:947–957. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, Gemma A, Harada M, Yoshizawa H, Kinoshita I, et al: Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 362:2380–2388. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Petrelli F, Borgonovo K, Cabiddu M and Barni S: Efficacy of EGFR tyrosine kinase inhibitors in patients with EGFR-mutated non-small-cell lung cancer: A meta-analysis of 13 randomized trials. Clin Lung Cancer. 13:107–114. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Rekhtman N, Paik PK, Arcila ME, Tafe LJ, Oxnard GR, Moreira AL, Travis WD, Zakowski MF, Kris MG and Ladanyi M: Clarifying the spectrum of driver oncogene mutations in biomarker-verified squamous carcinoma of lung: Lack of EGFR/KRAS and presence of PIK3CA/AKT1 mutations. Clin Cancer Res. 18:1167–1176. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Zhang L, Ma S, Song X, Han B, Cheng Y, Huang C, Yang S, Liu X, Liu Y, Lu S, et al: Gefitinib versus placebo as maintenance therapy in patients with locally advanced or metastatic non-small-cell lung cancer (INFORM; C-TONG 0804): A multicentre, double-blind randomised phase 3 trial. Lancet Oncol. 13:466–475. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Clark GM, Zborowski DM, Santabarbara P, Ding K, Whitehead M, Seymour L and Shepherd FA: National Cancer Institute of Canada Clinical Trials Group: Smoking history and epidermal growth factor receptor expression as predictors of survival benefit from erlotinib for patients with non-small-cell lung cancer in the National Cancer Institute of Canada Clinical Trials Group study BR.21. Clin Lung Cancer. 7:389–394. 2016. View Article : Google Scholar

30 

Cappuzzo F, Ciuleanu T, Stelmakh L, Cicenas S, Szczésna A, Juhász E, Esteban E, Molinier O, Brugger W, Melezínek I, et al: Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: A multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol. 11:521–529. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Owonikoko TK, Ragin CC, Belani CP, Oton AB, Gooding WE, Taioli E and Ramalingam SS: Lung cancer in elderly patients: An analysis of the surveillance, epidemiology, and end results database. J Clin Oncol. 25:5570–5577. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Thomas A, Chen Y, Yu T, Jakopovic M and Giaccone G: Trends and characteristics of young non-small cell lung cancer patients in the United States. Front Oncol. 5:1132015. View Article : Google Scholar : PubMed/NCBI

33 

Miyoshi S, Hamada H, Ito R, Hamaguchi N, Kadowaki T and Higaki J: Successful treatment of non-small cell lung cancer by gefitinib in an elderly patient with poor performance status. Nihon Ronen Igakkai Zasshi. 45:338–342. 2008.(In Japanese). View Article : Google Scholar : PubMed/NCBI

34 

Xu J, Zhang Y, Jin B, Chu T, Dong X, Yang H, Wu D, Lou Y, Zhang X, Wang H and Han B: Efficacy of EGFR tyrosine kinase inhibitors for non-adenocarcinoma lung cancer patients harboring EGFR-sensitizing mutations in China. J Cancer Res Clin Oncol. 142:1325–1330. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Ameratunga M, Pavlakis N, Gebski V, Broad A and Khasraw M: Epidermal growth factor receptor-tyrosine kinase inhibitors in advanced squamous cell carcinoma of the lung: A meta-analysis. Asia Pac J Clin Oncol. 10:273–278. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Cobo M, Gutiérrez V, Rodelo L, López O, Ruiz M and Godoy A: Afatinib in patients with squamous cell carcinoma of the lung: Current context and the option of oral treatment. Med Clin (Barc). 146 Suppl 1:S25–S29. 2016.(In Spanish). View Article : Google Scholar

Related Articles

Journal Cover

June-2018
Volume 17 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun Y, Yin X, Wen MM, Zhang J, Wang XJ, Xia JH, Zhang YN, Zhang ZP and Li XF: EGFR mutations subset in Chinese lung squamous cell carcinoma patients. Mol Med Rep 17: 7575-7584, 2018
APA
Sun, Y., Yin, X., Wen, M., Zhang, J., Wang, X., Xia, J. ... Li, X. (2018). EGFR mutations subset in Chinese lung squamous cell carcinoma patients. Molecular Medicine Reports, 17, 7575-7584. https://doi.org/10.3892/mmr.2018.8859
MLA
Sun, Y., Yin, X., Wen, M., Zhang, J., Wang, X., Xia, J., Zhang, Y., Zhang, Z., Li, X."EGFR mutations subset in Chinese lung squamous cell carcinoma patients". Molecular Medicine Reports 17.6 (2018): 7575-7584.
Chicago
Sun, Y., Yin, X., Wen, M., Zhang, J., Wang, X., Xia, J., Zhang, Y., Zhang, Z., Li, X."EGFR mutations subset in Chinese lung squamous cell carcinoma patients". Molecular Medicine Reports 17, no. 6 (2018): 7575-7584. https://doi.org/10.3892/mmr.2018.8859