Skip to main content

Quantification of EGFR mutations in primary and metastatic tumors in non-small cell lung cancer

Abstract

Background

EGFR mutation detection has been widely applied in the prediction of TKIs therapy in Non-Small Cell Lung Cancer (NSCLC). Metastatic tumors rather than primary tumors were usually assayed for those patients in advanced stages. Although the difference of EGFR mutation status in primary and metastatic tumors has been reported, the quantitative difference (ratio of mutated EGFR among total EGFR) in primary and metastatic tumors as well as in different sites of primary tumors was not clear.

Methods

Genomic DNA in Formalin Fixed-Paraffin Embedded samples of primary and metastatic tumors of 50 NSCLC patients was extracted. Real-time fluorescent PCR was performed to quantify the EGFR mutation ratios.

Results

The EGFR mutation ratios detected in different sites of primary tumors were highly concordant, whereas the EGFR mutation ratios in metastatic tumors were lower than those in primary tumors.

Conclusions

Randomly chosen sample may reliably represent the type and ratio of mutations of EGFR in primary tumors. EGFR mutation ratios in primary tumors and metastatic tumors are different. If metastatic tumors are used for the detection of EGFR mutation, the sensitivity of the detection assay must be considered.

Background

Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase encoded by the c-erb-B1 proto-oncogene. Multiple studies showed that the efficacy of tyrosine kinase inhibitors (TKIs) in the treatment of Non-Small Cell Lung Cancer (NSCLC) is highly correlated with EGFR mutation status in exon 18–21 [1–4].

EGFR mutations have been detected in 30-50% of NSCLC patients in China [5, 6]. The detection methods include PCR-sequencing, Taqman real-time PCR, DHPLC, and SARMS [6–12]. For some of the NSCLC patients, especially those with metastatic cancer, the primary tumor specimen may not be available; therefore EGFR mutations in metastases are often analyzed. However, the molecular nature of the tumors may change during metastasis, and currently it is unclear whether the mutations detected in primary tumors correlate with those in metastases. It has been reported that EGFR mutations detected in metastases are 10-60% inconsistent with those in primary tumors [13, 14]. It is worth noting that gefitinib has been reported to be beneficial for patients in which EGFR mutations were detected in metastases but not primary tumors [15]. However, since these studies used qualitative detection of EGFR mutations, it is impossible to quantitatively evaluate the abundance of EGFR mutations in the primary tumor and metastases.

Real-time fluorescent PCR detection of mutations is a straightforward method with high sensitivity and reliability. In this study, we used real-time PCR to quantitatively detect EGFR mutations in primary and metastatic tumors. Fifty Chinese NSCLC patients that harbor EGFR mutations in their primary tumors were identified. EGFR mutation status and abundance were compared among different areas of a primary tumor and its corresponding metastatic tumor of the same individual. Our study provides new insights on clinical interpretation of EGFR mutation status in different specimens.

Methods

Patients and Clinical Characteristics

From the patients who visited Henan Cancer Hospital between January 2010 and December 2012, those diagnosed with NSCLC by histological examination were tested for EGFR mutations, and 50 patients that were positive for EGFR mutations in the primary tumor samples were randomly selected for further evaluation. Their clinical and pathological characteristics are listed in Table 1. All study subjects never received TKI treatment before the study, and the formalin-fixed paraffin-embedded (FFPE) specimens were available for both the primary and metastatic tumors. Patients consented to tissue specimen collection prospectively, and the study was approved by the ethics committee of Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University.

Table 1 Clinical characteristics of 50 advanced NSCLC cases and the detection of EGFR mutations in primary tumors and metastases

Clinical specimens

Pathological diagnosis was established as NSCLC by assessing the HE stained sections of formalin-fixed paraffin-embedded primary tumors. The tumor contents was >50% for slides prepared from primary tumors, and >20% for those from lymph node metastases. For each subject, four DNA samples corresponding to the two lateral regions and one center region of the primary tumor specimen, as well as one from lymph node metastases were prepared. For each sample, DNA was isolated from no less than 5 pieces of consecutive 5 μm slides of Formalin-fixed paraffin-embedded (FFPE) specimens that had been stored at room temperature for less than 5 years.

Isolation of genomic DNA

Genomic DNA from the FFPE samples was isolated by using QIAamp DNA FFPE Tissue Kit (Qiagen) according to the manufacturer’s instructions. The DNA concentration was measured by UV spectrometer and adjusted to 20 ~ 50 ng/μl. DNA samples were stored at -20°C before use.

Quantitative measurement of EGFR mutation ratio

45 types of EGFR mutations corresponding to hotspots in exons 18, 19, 20 and 21 were detected by using Human EGFR mutation quantitative PCR detection kit (ACCB, Beijing, China) according to the manufacturer’s instructions. Real-time PCR were performed on Stratagene Mx3000P PCR machine with the following settings: 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. The mutant and wild-type alleles were amplified separately, and the levels of each mutation in the sample were calculated by normalizing to standard curves. The mutation ratio was defined as [mutation ratio % = level of mutants/(level of mutants + level of wild type allele) × 100%].

Statistical analysis

Statistical analysis was carried out using SPSS version 16.0 software (SPSS Inc., Chicago, IL, US). Fisher’s exact test was used to analyze whether the different categories had different positive rates. Kappa test was used to analyze whether the two sampling regions had consistent outcomes. Wilcoxon matched pairs test was used to compare the mutation ratios from the two regions. Two-sided p < 0.05 was considered statistically significant.

Results

EGFR mutations in primary tumors and metastases

Of the 50 cases of NSCLC that had EGFR mutations in primary tumors, exon 19 mutations (in-frame deletions only) were present in 28 cases (56%), and exon 21 (L858R point mutations only) mutations were detected in 22 cases (44%). Mutations in exon 19 and 21 were mutually exclusive and no multiple mutations were found. Of the metastases samples, 47 were positive for EGFR mutation (94% concordance with the detection in primary tumors), and exon 19 and exon 21 mutations were detected in 26 cases (55%, 93% concordance) and 21 cases (45%, 95% concordance), respectively. Notably, all cases presented the same mutation type in the matching primary and metastatic tumors. EGFR mutation detection and the clinical characteristics were listed in Table 1. Among the 50 subjects, only 3 (6%) had different test results for EGFR mutations in primary tumor and metastases, however, the difference was insignificant (P = 0.242) as analyzed by Fisher’s exact test.

EGFR mutations at different sites of primary tumors of the same patient

We performed quantitative measurement of EGFR mutations at different sites of primary tumors (Table 2). The median mutation deviation for different primary sites (see footnote of Table 2 for the formula of calculation) was 18.3% (with a range of 0.0% ~ 54.3%), indicating that the results of the quantitative measurement of EGFR mutations in different sites of primary tumor in the same patient have a high level of concordance.

Table 2 Quantitative measurement of EGFR mutation ratios in 3 primary tumor sites and one metastases of the same patient

Quantitative measurement of EGFR mutations in primary tumors and metastases of the same patient

Although the qualitative measurement of EGFR mutations in primary sites and metastases showed a high level of concordance (94%), the quantitative measurements had significant difference (Tables 2 and 3). The Kappa value of the two groups was 0.615 (P < 0.01), indicating that different sampling sites only had moderate concordance. Overall, the mutation ratios of metastases is significantly lower than those of primary tumors (P < 0.01) as analyzed by Wilcoxon matched pairs test.

Table 3 EGFR mutation ratios in primary tumor and metastases of the same patients

Discussion

NSCLC patients carrying EGFR mutations often benefit from TKI treatments with reduced sizes of primary tumors and metastases visualized by medical imaging. For a subset of patients, however, TKIs treatment only diminished the primary tumors but had no effect on metastases, and occasionally the metastases even became enlarged or more numerous over time. It raises the questions whether the abundance of EGFR mutations are different in different primary tumor sites, and whether the abundance and type of mutations are the same for primary tumors and metastases. Our study revealed the following characteristics of EGFR mutations.

First of all, although the mutation ratio in different primary tumor sites varied (the median value ranged from <10% to 85.9%) (Table 2), the deviation of the mutation ratio in different primary sites was limited (median was 18.3% with a range of 0.0% ~ 54.3%) (Table 2), indicating that different sites of primary tumor in the same patient have a high level of concordance. During the routine pathological evaluation of FFPE specimens of primary tumors, EGFR mutations were often tested only in one randomly chosen sample. Our study showed that when the area of cancerous cells were greater than 50%, a randomly chosen sample may reliably represent the type and ratio of mutations of EGFR in primary tumors.

Secondly, when the EGFR mutations were present in primary tumors, they could be detected in metastases with a high concordance regardless of the mutation ratios. The concordance of EGFR mutations in primary tumor and metastases is 94%, and that for mutation ratios is 84%. Moreover, different types of mutations, such as those in exon 19 and 21, were also identified with high concordances (93% and 95%, respectively), suggesting that the type of mutation did not affect the detection rates.

In addition, mutation detection is also affected by the proportion of cancerous cells in a sample. Therefore, for metastases with a lower number of cancerous cells, highly sensitive methods such as real-time PCR are highly recommended.

Moreover, in comparison to those in primary tumor sites, the mutation ratios in metastases were reduced and occasionally undetectable (16% samples had reduced or negative detection). These results suggest that the use of metastases specimens might generate false negative diagnosis for EGFR mutations that could have been present in primary tumors. The decreased EGFR mutation ratios in metastases suggest that EGFR mutations may not be essential for metastasis, which may underlie the lack of response to TKIs in metastases despite an positive outcome for the primary tumors. Notably, in this study we had one case of squamous cell carcinoma that harbors EGFR exon 19 mutation in the primary tumor, but the mutation was undetectable in metastases. It is unclear if it is due to the nature of squamous cell carcinoma.

In addition to the different pathological nature of primary tumor and metastases, the inconsistency in the identification of EGFR mutation may also be due to the sensitivity of the detection methods. For instance, Sanger sequencing may give a negative calling for samples with a mutation ratio of <10%, and therefore leads to low concordance for EGFR mutations in different samples of the same patients. Our study showed that by using quantitative real-time PCR, the positive identification of mutations in primary tumor and metastases had a 94% concordance, and for the quantitative measurement of mutation ratio the concordance was 84%. Hence, metastases specimens could be used for mutation assessment if the specimen for primary tumors is lacking, but the detection methods must be of high sensitivity. Recently, the abundance of mutations of predictive biomarkers, such as EGFR and KRAS, has drawn more attention [16–18]. It has been shown that the abundance of EGFR mutations predicts benefit from EGFR-TKI treatment for NSCLC [16]. Similarly, colorectal cancer patients with low abundance of KRAS mutation have been reported to benefit from EGFR antibody therapy [17]. Precise quantification of EGFR mutation abundance may become a trend in clinic to help with a better patient selection and better treatment strategies. To enable precise quantification of mutation ratio, real time PCR with a standard curve such as the method applied in this report serves as one of the optimal options.

In this study, only subjects with EGFR mutations in primary tumors were included, but it did not address the issues of positive mutation detection in metastases but negative in primary sites. Future studies should combine the prognosis data of the patients that received TKIs therapy and analyze the correlation between the quantitative measurement of EGFR mutations in primary and metastatic tumors and their response to TKIs, especially those with inconsistent measurement of EGFR mutation status in those sites. These studies could provide guidance for doctors to make informed decision in NSCLC treatment.

Conclusions

Randomly chosen sample may reliably represent the type and ratio of EGFR mutations in primary tumors. EGFR mutation ratios in primary and metastatic tumors are different. If metastatic tumors are used for EGFR mutation detection, the sensitivity of the detection assay must be taken into consideration.

References

  1. Paez JG, Janne 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 (New York, NY). 2004, 304: 1497-1500. 10.1126/science.1099314.

    Article  CAS  Google Scholar 

  2. 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. 2004, 350: 2129-2139. 10.1056/NEJMoa040938.

    Article  CAS  PubMed  Google Scholar 

  3. 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. 2009, 361: 947-957. 10.1056/NEJMoa0810699.

    Article  CAS  PubMed  Google Scholar 

  4. Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M, Johnson BE, Eck MJ, Tenen DG, Halmos B: EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005, 352: 786-792. 10.1056/NEJMoa044238.

    Article  CAS  PubMed  Google Scholar 

  5. Song G, Di L, Ren J, Zhang L, Yu J: Analysis of EGFR mutation in Chinese non-small cell lung cancer patients. J Mod Oncol. 2008, 16: 553-556.

    CAS  Google Scholar 

  6. Feng Q, Li X, Chen Z, He J, Wang C, Zhou L, Xue W: Epidermal growth factor receptor gene mutations and clinicopathologic correlation in 309 patients with non-small cell lung cancer. Chin J Pathol. 2011, 40: 660-663.

    CAS  Google Scholar 

  7. Abo-Elwafa HA, Attia FM, Sharaf AE: The prognostic value of p53 mutation in pediatric marrow hypoplasia. Diagn Pathol. 2011, 6: 58-10.1186/1746-1596-6-58.

    Article  PubMed Central  PubMed  Google Scholar 

  8. Carbonell P, Turpin MC, Torres-Moreno D, Molina-Martinez I, Garcia-Solano J, Perez-Guillermo M, Conesa-Zamora P: Comparison of allelic discrimination by dHPLC, HRM, and TaqMan in the detection of BRAF mutation V600E. J Mol Diagn. 2011, 13: 467-473. 10.1016/j.jmoldx.2011.03.009.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Didelot A, Le Corre D, Luscan A, Cazes A, Pallier K, Emile JF, Laurent-Puig P, Blons H: Competitive allele specific TaqMan PCR for KRAS, BRAF and EGFR mutation detection in clinical formalin fixed paraffin embedded samples. Exp Mol Pathol. 2012, 92: 275-280. 10.1016/j.yexmp.2012.03.001.

    Article  CAS  PubMed  Google Scholar 

  10. Endo K, Konishi A, Sasaki H, Takada M, Tanaka H, Okumura M, Kawahara M, Sugiura H, Kuwabara Y, Fukai I, et al: Epidermal growth factor receptor gene mutation in non-small cell lung cancer using highly sensitive and fast TaqMan PCR assay. Lung Cancer. 2005, 50: 375-384. 10.1016/j.lungcan.2005.08.009.

    Article  PubMed  Google Scholar 

  11. Hamfjord J, Stangeland AM, Skrede ML, Tveit KM, Ikdahl T, Kure EH: Wobble-enhanced ARMS method for detection of KRAS and BRAF mutations. Diagn Mol Pathol. 2011, 20: 158-165. 10.1097/PDM.0b013e31820b49e2.

    Article  CAS  PubMed  Google Scholar 

  12. Liu Y, Liu B, Li XY, Li JJ, Qin HF, Tang CH, Guo WF, Hu HX, Li S, Chen CJ, et al: A comparison of ARMS and direct sequencing for EGFR mutation analysis and tyrosine kinase Inhibitors treatment prediction in body fluid samples of non-small-cell lung cancer patients. J Exp Clin Cancer Res. 2011, 30: 111-10.1186/1756-9966-30-111.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Sun L, Zhang Q, Luan H, Zhan Z, Wang C, Sun B: Comparison of KRAS and EGFR gene status between primary non-small cell lung cancer and local lymph node metastases: implications for clinical practice. J Exp Clin Cancer Res. 2011, 30: 30-10.1186/1756-9966-30-30.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Monaco SE, Nikiforova MN, Cieply K, Teot LA, Khalbuss WE, Dacic S: A comparison of EGFR and KRAS status in primary lung carcinoma and matched metastases. Hum Pathol. 2010, 41: 94-102. 10.1016/j.humpath.2009.06.019.

    Article  CAS  PubMed  Google Scholar 

  15. Sun L, Zhang Q, Li H, Zhan Z, Sun B: Comparison of KRAS and EGFR gene statuses between primary non-small cell lung cancer and local lymph node metastases and their clinical significance. Chin J Clin Oncol. 2012, 39: 970-973.

    CAS  Google Scholar 

  16. Zhou Q, Zhang XC, Chen ZH, Yin XL, Yang JJ, Xu CR, Yan HH, Chen HJ, Su J, Zhong WZ, et al: Relative abundance of EGFR mutations predicts benefit from gefitinib treatment for advanced non-small-cell lung cancer. J Clin Oncol. 2011, 29: 3316-3321. 10.1200/JCO.2010.33.3757.

    Article  CAS  PubMed  Google Scholar 

  17. Yu S, Xiao X, Lu J, Qian X, Liu B, Feng J: Colorectal cancer patients with low abundance of KRAS mutation may benefit from EGFR antibody therapy. PloS One. 2013, 8: e68022-10.1371/journal.pone.0068022.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Wu YC, Chang IC, Wang CL, Chen TD, Chen YT, Liu HP, Chu Y, Chiu YT, Wu TH, Chou LH, et al: Comparison of IHC, FISH and RT-PCR methods for detection of ALK rearrangements in 312 non-small cell lung cancer patients in Taiwan. PloS One. 2013, 8: e70839-10.1371/journal.pone.0070839.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgement

This work was supported by a Research Plan of Medical Science and Technology Project of Henan Province (No. 201204105) (to JM).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jie Ma.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

BW, KY and JZ carried out the DNA isolation. BW, YC, ZM, BD and YG performed real time PCR for quantification of EGFR mutation. BW and JM performed the statistical analysis. BW and JM designed the study and drafted the manuscript. All authors read and approved the final manuscript.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Cite this article

Wei, B., Yang, K., Zhao, J. et al. Quantification of EGFR mutations in primary and metastatic tumors in non-small cell lung cancer. J Exp Clin Cancer Res 33, 5 (2014). https://doi.org/10.1186/1756-9966-33-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1756-9966-33-5

Keywords