Genetic alterations in epidermal growth factor receptor‑tyrosine kinase inhibitor‑naïve non‑small cell lung carcinoma

  • Authors:
    • Takumi Yamaura
    • Satoshi Muto
    • Hayato Mine
    • Hironori Takagi
    • Masayuki Watanabe
    • Yuki Ozaki
    • Takuya Inoue
    • Mitsuro Fukuhara
    • Naoyuki Okabe
    • Yuki Matsumura
    • Takeo Hasegawa
    • Jun Osugi
    • Mika Hoshino
    • Mitsunori Higuchi
    • Yutaka Shio
    • Hiroyuki Suzuki
  • View Affiliations

  • Published online on: April 8, 2020     https://doi.org/10.3892/ol.2020.11524
  • Pages: 4169-4176
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) are an approved first-line therapy against unresectable or advanced non‑small cell lung cancer (NSCLC) harboring EGFR gene activating mutations. However, the majority of tumors develop acquired resistance against EGFR‑TKIs and some tumors exhibit natural resistance. A number of resistance mechanisms against the latest third‑generation EGFR‑TKIs have been reported, including tertiary EGFR C797S mutation and several gene alterations activating EGFR or other signaling pathways. The current study aimed to identify the frequency of natural EGFR‑TKI resistance in pretreatment NSCLC and to predict the therapeutic effect of EGFR‑TKIs. A total of 246 EGFR‑TKI‑naïve NSCLC patients harboring known EGFR gene mutations were identified. The presence of EGFR C797S and T790M mutations were determined using the peptide nucleic acid‑locked nucleic acid PCR clamp method. ERBB2, MET, EGFR, ALK, BRAF, FGFR1, MYC, RET, CCND1, CCND2, CDK4, CDK6, MDM2 and MDM4 gene amplification, which can lead to resistance against any generation EGFR‑TKIs, was determined using the multiplex ligation‑dependent probe amplification assay. No concurrent C797S mutation with known EGFR mutations were identified. T790M mutation was identified in 12 patients (4.9%). ERBB2 or MET gene amplification was found in some patients (0.0‑0.4%). MDM2 gene amplification was associated with tumor recurrence and shorter progression‑free survival (PFS) for first‑ or second‑generation EGFR‑TKIs. De novo EGFR C797S mutation was not identified. Other resistance mechanisms against EGFR‑TKIs were indicated in some patients with EGFR‑TKI‑naïve NSCLC. MDM2 gene amplification, which can lead to altered cell cycle, was associated with tumor recurrence and shorter PFS in EGFR‑TKI therapy.

Introduction

Precision molecular targeted agents in non-small cell lung cancer (NSCLC) have improved survival of patients harboring driver gene mutations. Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) improves progression-free survival (PFS) of NSCLC patients with EGFR mutations compared with traditional platinum-based doublet chemotherapy (1,2). Furthermore, osimertinib, a third-generation EGFR-TKI, is promising as first-line treatment for EGFR mutant NSCLC (3,4). Although good responses to EGFR-TKI therapy have been shown, tumor cells can acquire resistance through several methods, in particular, secondary gene mutations that cause structural changes in the ATP binding site of the EGFR tyrosine kinase domain. EGFR T790M mutation occurs in almost half of patients following first- or second-generation EGFR-TKI therapy (5), and EGFR C797S mutation is the most common mechanism of acquired resistance against third-generation EGFR-TKIs (6).

Approximately 0.4–8% of NSCLC patients harboring de novo or germline T790M mutations are resistant to first- or second-generation EGFR-TKIs (7). However, the frequency of EGFR C797S gene mutation remains unclear. To the best of our knowledge, only one case of an NSCLC patient harboring concurrent C797S and L858R mutations prior to receiving EGFR-TKI treatment has been reported (8).

Several other mechanisms of resistance against all generation EGFR-TKIs have been identified including tertiary gene mutations other than EGFR C797S mutation (911), activation of bypass signaling by gene amplification (e.g., ERBB2 (12) and MET (13,14), driver gene mutations (e.g., RAS, RAF and PIK3CA) (3,15), gene alteration in cell cycle genes (14), and transformation to mesenchyme, small cell carcinoma (SCC), or squamous cell carcinoma (SqCC) (2,16,17). These described EGFR-TKI resistance mechanisms may also be expressed during the pre-TKI NSCLC state (6,15) and can be a challenge for cancer treatment of NSCLC patients with EGFR mutations.

In this retrospective study, we assessed potential resistance against third-generation EGFR-TKI therapy, such as EGFR C797S mutation, and gene amplification in EGFR-TKI-naïve surgical specimens from patients harboring known EGFR mutations.

Materials and methods

Patient selection

Consecutive patients who underwent initial lung resection or surgical tumor biopsy in Fukushima Medical University Hospital and were diagnosed with NSCLC harboring a known EGFR gene activating mutation (e.g., exon 19 deletion, L858R, T790M, S768I, G719X and L861Q) at the time samples were collected, and whose specimens were available for gene examination described below, were included in this study. Patients who had received systemic treatment or irradiation therapy before surgery were excluded.

Ethics statement

This study was conducted with approval of the ethics board at Fukushima Medical University (approval no. 2955). Human rights and welfare of participants were protected in accordance with the Declaration of Helsinki, and written informed consent was obtained from participants.

Preparation of genomic DNA

Tumor DNA was extracted from macro-dissected tumor tissue of formalin-fixed paraffin-embedded surgical specimens using the QIAamp DNA FFPE Tissue kit (Qiagen) according to the manufacturer's instructions. Tumor DNA quantity was assessed using the Qubit dsDNA HS Assay kit (Thermo Fisher Scientific, Waltham, MA, USA) and Qubit 2.0 Fluorometer (Thermo Fisher Scientific).

Peptide nucleic acid-locked nucleic acid PCR clamp method

Premix Ex Taq (Takara Bio Inc.), 200 nM of primer, 100 nM of mutation LNA probe, 100 nM of total probe, 250–2,500 nM of clamp probe, and sample DNA were mixed in a total reaction volume of 25 µl and analyzed as described previously (18). Real-time PCR was performed in 50 cycles using Light Cycler480 II (Roche; denaturation: 5 sec at 95°C; annealing and extension: 30 sec at 62°C). EGFR C797S and T790M mutations were judged using LightCycler 480 software (Roche).

Multiplex ligation-dependent probe amplification (MLPA) Assay

Gene amplification was analyzed according to the standard protocol for MLPA (19) using SALSA MLPA Probemix P175 Tumor Gain (MRC-Holland). ERBB2, MET, EGFR, ALK, BRAF, FGFR1, MYC, RET, CCND1, CCND2, CDK4, CDK6, MDM2 and MDM4 gene amplification was assessed. Fragments of PCR products were analyzed using the 3130×l Genetic Analyzer (Thermo Fisher Scientific) and amplification was judged by Coffalyser Data analysis software (MRC-Holland).

Statistical analysis

Statistical analysis was performed using SPSS 21.0 (IBM; SPSS). Continuous variables were compared by two-tailed t-tests or one-way ANOVA, and categorical variables were compared by the chi-squared test or Fisher's exact test. Multivariate analyses using a binary logistic regression model were performed to evaluate independent predictors, and hazard ratio (HR) and confidence interval (CI) were calculated. PFS was estimated using the Kaplan-Meier method, and survival curves were compared using log-rank tests. P-values of less than 0.05 were considered statistically significant. Some of the statistical analysis was performed by AC Medical Inc. (Tokyo, Japan).

Results

Patient characteristics

Between January 2007 and December 2015, we identified 248 patients, of which 246 patients were eligible for this study and included in the analyses. Of the 232 patients who had undergone complete or microscopically incomplete resection, recurrence was found in 49 patients. Surgery with incomplete resection or tumor biopsy was performed in 14 patients. Of the 63 advanced or recurrent NSCLC patients, 54 patients received EGFR-TKI therapy (Fig. 1). Demographic data and clinicopathological characteristics of patients are presented in Table I: Age at surgery or biopsy ranged from 37 to 90 years (median age: 67 years). There were 159 (64.6%) women, 166 (67.4%) never smokers, 240 (97.6%) patients with adenocarcinoma, and 193 (78.4%) patients with pathological stage I (Table I).

Table I.

Clinicopathologic, genetic and histologic characteristics of patients.

Table I.

Clinicopathologic, genetic and histologic characteristics of patients.

A, Clinicopathologic characteristic

CharacteristicTotal (n=246)Received EGFR-TKI therapy(n=44)
Sex
  Male87 (35.4)14 (31.8)
Age (y)
  Median (range)67.0 (37–90)65.5 (37–82)
Smoking status
  Never166 (67.4)30 (68.2)
  Former67 (27.2)11 (25.0)
  Current13 (5.3)3 (6.8)
Pathological subtypes
  Adeno240 (97.6)42 (95.5)
  AdSq2 (0.8)2 (4.5)
  Sq4 (1.6)0 (0.0)
p-Stage at initial surgery
  I193 (78.4)
  II16 (6.5)
  III24 (9.7)
  IV12 (4.9)

B, EGFR mutation

CharacteristicTotal (n=246)Received EGFR-TKI therapy(n=44)

C797S0 (0.0)0 (0.0)
Ex19del86 (35.0)21 (47.7)
Ex19del+T790M1 (0.4)0 (0.0)
L858R134 (54.5)20 (45.5)
L858R+G719S1 (0.4)1 (2.3)
L858R+T790M4 (1.6)0 (0.0)
T790M7 (2.8)0 (0.0)
G719A3 (1.2)0 (0.0)
G719C1 (0.4)0 (0.0)
L861Q1 (0.4)1 (2.3)
G719A+S768I2 (0.8)0 (0.0)
G719A+L861Q2 (0.8)1 (2.3)
G719C+L861Q1 (0.4)0 (0.0)

C, Gene amplification

CharacteristicTotal (n=246)Received EGFR-TKI therapy (n=44)

ERBB21 (0.4)1 (2.3)
MET0 (0.0)0 (0.0)
EGFR5 (2.0)3 (6.8)
ALK0 (0.0)0 (0.0)
RET0 (0.0)0 (0.0)
BRAF0 (0.0)0 (0.0)
FGFR11 (0.4)0 (0.0)
MYC2 (0.8)1 (2.3)
CCND12 (0.8)2 (4.5)
CCND21 (0.4)1 (2.3)
CDK415 (6.0)4 (9.1)
MDM217 (6.8)6 (13.6)
MDM42 (0.8)1 (2.3)

[i] Data are presented as n (%) unless otherwise indicated. AdSq, adenosquamous; Sq, squamous; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor.

EGFR C797S and T790M mutations

No patients harbored concurrent C797S mutation with known EGFR mutations. T790M mutation was identified in 12 patients (4.9%); 5 patients had the deletion in exon19 or L858R and 7 patients had T790M mutation alone (Table I).

Gene amplification

EGFR gene amplification was found in five patients. All patients with EGFR gene amplification had advanced disease or recurrence (9.8 vs. 0.0%, P<0.01). ERBB2 gene amplification was found in only one patient (0.4%) who developed recurrence. No patients harbored MET gene amplification. MDM2 gene amplification was found in 17 patients (7.1%) and CDK4 gene amplification in 14 patients (5.8%); duplication of both MDM2 and CDK4 was observed in 11 patients (4.6%). MDM2 gene amplification was significantly associated with tumor recurrence (16.7 vs. 5.1%, P=0.032). Patients with tumors with any gene amplification had significantly more advanced disease or developed recurrence compared with patients without recurrence after surgery (33.3 vs. 8.9%, P=0.002) (Fig. 2).

We next evaluated associations between clinicopathological parameters and/or gene amplification and progress of first- and second-generation EGFR-TKI treatment. Fifty-four patients had received EGFR-TKI therapy, and 44 patients underwent gene amplification analysis and were followed up. Patients who developed central nervous system metastasis (HR, 2.259, 95% CI, 1.150–4.436) and had MDM2 gene amplification (HR, 3.405, 95% CI, 1.209–9.591) exhibited significantly shorter PFS (Figs. 3,4). Patients who had EGFR gene amplification (HR, 0.656, 95% CI, 0.195–2.210), ERBB2 gene amplification (HR, 0.801, 95% CI, incalculable), CDK4 gene amplification (HR, 0.194, 95% CI, 0.660–7.802) (Figs. 3,4), CCND1 gene amplification (HR, 2.823 95% CI, 0.653–12.203), CCND2 gene amplification (HR, 0.801, 95% CI, incalculable), CDK4 gene amplification (HR, 0.194, 95% CI, 0.660–7.802), and MDM4 gene amplification (HR, 0.465, 95% CI, 0.063–3.441) (data not shown) showed no significant difference in PFS (Figs. 3,4).

Discussion

We herein showed that in our study cohort no EGFR-TKI-naïve NSCLC patients harbored concurrent EGFR C797S mutation with known EGFR gene mutation. Amplification of several genes was found before EGFR-TKI therapy, and MDM2 gene amplification was associated with resistance to first-generation EGFR-TKIs.

C797S mutation, which leads to acquired resistance to third-generation EGFR TKIs, occurs in 5.3–40.0% of NSCLC patients following osimertinib treatment (2,3,6,11,20) and is the most common resistance mechanism against osimertinib. No concurrent C797S mutation was found in this study, and, to our knowledge, only one case of de novo somatic L858R and C797S mutations has been reported (8). C797S mutation is an important challenge for EGFR mutant NSCLC therapy. A recent report suggests that therapeutic efficacy is dependent on allelic context of common EGFR mutations, C797S and T790M. If T790M and C797S mutations occur on separate alleles, then combination therapy comprising first- and third-generation EGFR-TKIs can restore EGFR inhibition in vitro (21). Unfortunately, these mutations almost always occur on the same allele (22). Conversely, combination therapy composed of osimertinib, bevacizumab, and brigatinib is effective for NSCLC with T790M and C797S mutations occurring on the same allele (23). Combination therapy comprising an allosteric inhibitor and cetuximab was also shown to be effective in a murine model of NSCLC driven by EGFR L858R, T790M, and/or C797S mutation (24). Furthermore, other tertiary mutations of EGFR [L792X (11), G796D (9), L718Q, and L844V (5)] in the EGFR tyrosine kinase domain were reported, and novel therapies for tumors with these mutations are required.

Other resistance mechanisms against third-generation EGFR-TKIs involving activation of signaling pathways have been reported. Lin et al (2) and Ramalingam et al (3) carried out plasma-based analyses after development of osimertinib resistance in NSCLC. The following gene alterations were noted: EGFR C797S mutation in 5.2–16.7%, KRAS mutation in 2.6–7.7%, BRAF mutation in 7.7%, PIK3CA mutation in 2.6%, MEK mutation in 2.6%, JAK2 mutation in 2.6%, MET amplification in 2.6–50%, KRAS amplification in 2.6%, ERBB2 insertion in 2.6%, transformation to SCC in 9.1%, and transformation to SqCC in 4.5% of patients (2,3). Furthermore, analysis of matched samples of pre- and post-administration of first- or second-generation EGFR-TKIs showed acquired resistance in addition to T790M mutation in lung adenocarcinoma, MET amplification in 8%, ERBB2 amplification in 5%, and EGFR amplification in 16% of patients (15). Transformation to SCC was noted in 2.6–5.0% of patients (15), and another report demonstrated SqCC rarely occurred (17).

The genetic alterations described above occurred in some pretreatment EGFR mutant NSCLC tumors and patients with these mutations can exhibit resistance to osimertinib therapy. Concurrent gene alterations in EGFR-TKI-naïve, EGFR mutant NSCLC have been reported. Yu et al, conducted next-generation sequencing analysis of tissue samples and revealed EGFR mutant NSCLC had concurrent alteration of TP53 mutation in 60%, RB1 mutation in 10%, PIK3CA mutation in 12%, CTNNB1 mutation in 18.9%, EGFR amplification in 22%, MDM2 mutation in 12%, CDK4 mutation in 10%, ERBB2 amplification in 8.4%, and MET amplification in 2% of patients before EGFR-TKI treatment (15).

Gene amplification of ERBB2, MET, MDM2 or CDK4 alone leads to poor prognosis for NSCLC regardless of EGFR gene mutation (2527). Furthermore, Le et al (11) and Blakely et al (14) reported that cell cycle gene alteration, such as CDK4 or CDK6, shortens PFS following osimertinib therapy. Patients with EGFR mutant NSCLC accompanied by TP53 mutation, ERBB2 amplification, or MET amplification before first- or second-generation EGFR-TKI treatment exhibited a shorter time to progression and also showed resistance to third-generation EGFR-TKIs (12,13,28).

In this study, MDM2 gene amplification was shown to be associated with shorter PFS for first- or second-generation EGFR-TKIs. MDM2 is a proto-oncogene that is often coexpressed with CDK4 in liposarcoma (29). MDM2 binds to TP53 to downregulate transcription, leading to proteasome degradation by ubiquitination (30).

The limitations of this study are as follows: First, we did not evaluate EGFR mutations associated with acquired resistance to EGFR-TKIs other than C797S (e.g., L792X, G796X and L718Q), exon 20 insertion, mutation in driver genes KRAS, BRAF and PIK3CA, or transformation to mesenchyme or other pathological subtypes. Furthermore, the study cohort was limited to NSCLC patients harboring known EGFR gene mutations. Therefore, whether C797S mutation alone could be an oncogenic mechanism remains unclear. Second, this study did not evaluate single nucleotide polymorphisms or germline gene mutations. As only seven patients had EGFR T790M mutation, they could have germline mutations (7) rather than somatic mutations.

Finally, because our survival analysis was conducted in a small cohort, it is possible that amplification of genes other than MDM2, such as MDM4 and ERBB2 evaluated in this study, could be biomarkers for EGFR-mutant NSCLC.

In conclusion, no concurrent EGFR-C797S with known EGFR mutant NSCLC was identified in our study cohort. This result confirms the efficacy of third-generation EGFR-TKIs; however, it is important to remain aware of how genetic alterations can affect EGFR-TKI responses. Therefore, further studies in a large cohort are required to completely elucidate resistance mechanisms against third-generation EGFR-TKIs.

Acknowledgements

The authors would like to thank Ms. Mie Otsuki, Ms. Eiko Otomo and Ms. Yukiko Kikuta (Department of Chest Surgery, Fukushima Medical University) for their technical supports

Funding

This work was supported by grant from AstraZeneca. K.K. (Osaka, Japan; grant no. ESR-16-12328).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TY, SM and HS designed the study. TY, HM, HT, MW, YO, TI, MF, NO, YM, TH, JO, MHo, MHi and YS collected and analyzed the data. TY and HS wrote and revised the manuscript. All the authors approved the final manuscript.

Ethics approval and consent to participate

The current study was conducted with approval of the ethics board at Fukushima Medical University (approval no. 2955). Human rights and welfare of participants were protected in accordance with the Declaration of Helsinki, and written informed consent was obtained from participants.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang JC, Ahn MJ, Kim DW, Ramalingam SS, Sequist LV, Su WC, Kim SW, Kim JH, Planchard D, Felip E, et al: Osimertinib in pretreated T790M-Positive advanced non-small-cell lung cancer: AURA study phase II extension component. J Clin Oncol. 35:1288–1296. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Lin CC, Shih JY, Yu CJ, Ho CC, Liao WY, Lee JH, Tsai TH, Su KY, Hsieh MS, Chang YL, et al: Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: A genomic study. Lancet Respir Med. 6:107–116. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Ramalingam SS, Yang JC, Lee CK, Kurata T, Kim DW, John T, Nogami N, Ohe Y, Mann H, Rukazenkov Y, et al: Osimertinib as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer. J Clin Oncol. 36:841–849. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, Dechaphunkul A, Imamura F, Nogami N, Kurata T, et al: Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 378:113–125. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, Johnson BE, Eck MJ, Tenen DG and Halmos B: EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 352:786–792. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, et al: Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med. 21:560–562. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Su KY, Chen HY, Li KC, Kuo ML, Yang JC, Chan WK, Ho BC, Chang GC, Shih JY, Yu SL and Yang PC: Pretreatment epidermal growth factor receptor (EGFR) T790M mutation predicts shorter EGFR tyrosine kinase inhibitor response duration in patients with non-small-cell lung cancer. J Clin Oncol. 30:433–440. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Lee JS, Hur JY, Kim HJ, Lee KY and Kim WS: A case of concurrent de novo C797S and L858R EGFR mutation detected in stage IA non-small cell lung cancer patient. J Thorac Oncol. 12:e179–e181. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Zheng D, Hu M, Bai Y, Zhu X, Lu X, Wu C, Wang J, Liu L, Wang Z, Ni J, et al: EGFR G796D mutation mediates resistance to osimertinib. Oncotarget. 8:49671–49679. 2017.PubMed/NCBI

10 

Ercan D, Choi HG, Yun CH, Capelletti M, Xie T, Eck MJ, Gray NS and Jänne PA: EGFR mutations and resistance to irreversible pyrimidine-based EGFR inhibitors. Clin Cancer Res. 21:3913–3923. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Le X, Puri S, Negrao MV, Nilsson MB, Robichaux J, Boyle T, Hicks JK, Lovinger KL, Roarty E, Rinsurongkawong W, et al: Landscape of EGFR-Dependent and -independent resistance mechanisms to osimertinib and continuation therapy beyond progression in EGFR-Mutant NSCLC. Clin Cancer Res. 24:6195–6203. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, Ohashi K, Janjigian YY, Spitzler PJ, Melnick MA, et al: HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2:922–933. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Ou SI, Agarwal N and Ali SM: High MET amplification level as a resistance mechanism to osimertinib (AZD9291) in a patient that symptomatically responded to crizotinib treatment post-osimertinib progression. Lung Cancer. 98:59–61. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Blakely CM, Watkins TBK, Wu W, Gini B, Chabon JJ, McCoach CE, McGranahan N, Wilson GA, Birkbak NJ, Olivas VR, et al: Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet. 49:1693–1704. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Yu HA, Suzawa K, Jordan E, Zehir A, Ni A, Kim R, Kris MG, Hellmann MD, Li BT, Somwar R, et al: Concurrent alterations in EGFR-Mutant lung cancers associated with resistance to EGFR kinase inhibitors and characterization of MTOR as a mediator of resistance. Clin Cancer Res. 24:3108–3118. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, Bergethon K, Shaw AT, Gettinger S, Cosper AK, et al: Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 3:75ra262011. View Article : Google Scholar : PubMed/NCBI

17 

Okabe N, Takagi H, Mine H, Fukai S, Minemura H and Suzuki H: Osimertinib for epidermal growth factor receptor mutation-positive lung adenocarcinoma that transformed to T790M-Positive squamous cell carcinoma. J Thorac Oncol. 12:e167–e169. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Watanabe K, Fukuhara T, Tsukita Y, Morita M, Suzuki A, Tanaka N, Terasaki H, Nukiwa T and Maemondo M: EGFR mutation analysis of circulating tumor DNA using an improved PNA-LNA PCR clamp method. Can Respir J. 2016:52973292016. View Article : Google Scholar : PubMed/NCBI

19 

Schouten JP, McElgunn CJ, Waaijer R, Zwijnenburg D, Diepvens F and Pals G: Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res. 30:e572002. View Article : Google Scholar : PubMed/NCBI

20 

Oxnard GR, Hu Y, Mileham KF, Husain H, Costa DB, Tracy P, Feeney N, Sholl LM, Dahlberg SE, Redig AJ, et al: Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-Positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 4:1527–1534. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Niederst MJ, Hu H, Mulvey HE, Lockerman EL, Garcia AR, Piotrowska Z, Sequist LV and Engelman JA: The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies. Clin Cancer Res. 21:3924–3933. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Yatabe Y, Takahashi T and Mitsudomi T: Epidermal growth factor receptor gene amplification is acquired in association with tumor progression of EGFR-mutated lung cancer. Cancer Res. 68:2106–2111. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Zhao J, Zou M, Lv J, Han Y and Wang G and Wang G: Effective treatment of pulmonary adenocarcinoma harboring triple EGFR mutations of L858R, T790M, and cis-C797S by osimertinib, bevacizumab, and brigatinib combination therapy: A case report. Onco Targets Ther. 11:5545–5550. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Jia Y, Yun CH, Park E, Ercan D, Manuia M, Juarez J, Xu C, Rhee K, Chen T, Zhang H, et al: Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature. 534:129–132. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Sholl LM, Yeap BY, Iafrate AJ, Holmes-Tisch AJ, Chou YP, Wu MT, Goan YG, Su L, Benedettini E, Yu J, et al: Lung adenocarcinoma with EGFR amplification has distinct clinicopathologic and molecular features in never-smokers. Cancer Res. 69:8341–8348. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Dworakowska D, Jassem E, Jassem J, Peters B, Dziadziuszko R, Zylicz M, Jakóbkiewicz-Banecka J, Kobierska-Gulida G, Szymanowska A, Skokowski J, et al: MDM2 gene amplification: A new independent factor of adverse prognosis in non-small cell lung cancer (NSCLC). Lung Cancer. 43:285–295. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al: Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 17:77–88. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Planchard D, Loriot Y, André F, Gobert A, Auger N, Lacroix L and Soria JC: EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann Oncol. 26:2073–2078. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Dei Tos AP, Doglioni C, Piccinin S, Sciot R, Furlanetto A, Boiocchi M, Dal Cin P, Maestro R, Fletcher CD and Tallini G: Coordinated expression and amplification of the MDM2, CDK4, and HMGI-C genes in atypical lipomatous tumours. J Pathol. 190:531–536. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Deben C, Deschoolmeester V, Lardon F, Rolfo C and Pauwels P: TP53 and MDM2 genetic alterations in non-small cell lung cancer: Evaluating their prognostic and predictive value. Crit Rev Oncol Hematol. 99:63–73. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2020
Volume 19 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamaura T, Muto S, Mine H, Takagi H, Watanabe M, Ozaki Y, Inoue T, Fukuhara M, Okabe N, Matsumura Y, Matsumura Y, et al: Genetic alterations in epidermal growth factor receptor‑tyrosine kinase inhibitor‑naïve non‑small cell lung carcinoma. Oncol Lett 19: 4169-4176, 2020
APA
Yamaura, T., Muto, S., Mine, H., Takagi, H., Watanabe, M., Ozaki, Y. ... Suzuki, H. (2020). Genetic alterations in epidermal growth factor receptor‑tyrosine kinase inhibitor‑naïve non‑small cell lung carcinoma. Oncology Letters, 19, 4169-4176. https://doi.org/10.3892/ol.2020.11524
MLA
Yamaura, T., Muto, S., Mine, H., Takagi, H., Watanabe, M., Ozaki, Y., Inoue, T., Fukuhara, M., Okabe, N., Matsumura, Y., Hasegawa, T., Osugi, J., Hoshino, M., Higuchi, M., Shio, Y., Suzuki, H."Genetic alterations in epidermal growth factor receptor‑tyrosine kinase inhibitor‑naïve non‑small cell lung carcinoma". Oncology Letters 19.6 (2020): 4169-4176.
Chicago
Yamaura, T., Muto, S., Mine, H., Takagi, H., Watanabe, M., Ozaki, Y., Inoue, T., Fukuhara, M., Okabe, N., Matsumura, Y., Hasegawa, T., Osugi, J., Hoshino, M., Higuchi, M., Shio, Y., Suzuki, H."Genetic alterations in epidermal growth factor receptor‑tyrosine kinase inhibitor‑naïve non‑small cell lung carcinoma". Oncology Letters 19, no. 6 (2020): 4169-4176. https://doi.org/10.3892/ol.2020.11524