1. Introduction
Overexpression of epidermal growth factor receptor (EGFR) has been well known to be implicated in the pathogenesis of lung cancer [
1]. EGFR tyrosine kinase inhibitors (TKIs) were the first molecular targeted drugs leading to a paradigm shift in the management of patients with advanced non-small cell lung cancer (NSCLC) [
2]. Patients with lung cancer harboring drug-sensitive mutations on the
EGFR gene, such as exon 19 deletion or exon 21 L858R mutations, show dramatic and durable responses to EGFR-TKIs [
3]. However, almost all lung cancer cases that initially respond well to these drugs ultimately develop drug resistance and disease progression. In the last decade, several major mechanisms to drive resistance to EGFR-TKIs have been identified through translational research. The most common resistance mechanism of first- or second-generation EGFR-TKIs is a secondary T790M mutation in exon 20 of
EGFR, as detected in 50–60% of resistant tumors [
4,
5,
6].
C-MET amplification is the second most common mechanism, ranging from 5% to 22% of tumors resistant to EGFR-TKIs [
7,
8]. Other minor resistance mechanisms include mutations in
PIK3CA MAPK1 HER2, and
BRAF, epithelial–mesenchymal transition (EMT), and small cell lung cancer transformation [
9].
Although the mechanisms of acquired resistance to EGFR-TKI are well known, it remains a challenge to predict the development of specific resistance mechanisms upon EGFR-TKI treatment. Identifying EGFR-TKI resistance before or shortly after the treatment would be helpful to prevent or delay the resistance that leads to disease progression. Many preclinical and clinical studies suggest that some drug-resistant cancer cell clones already exist before exposure to the anti-cancer drug and survive during the course of treatment [
8,
10,
11,
12,
13,
14,
15]. In accordance with previous studies, we also reported that 25% of patients with
EGFR-mutated lung cancer had the T790M resistance mutation before EGFR-TKI treatment, and this pre-existing T790M mutation may negatively affect the drug’s efficacy [
12]. However, it is challenging for a conventional sequencing method to detect these latent resistance mutations that exist at a minor frequency. Additionally, if multiple resistance-associated gene alterations exist simultaneously, knowing which resistance mechanism will be finally selected is challenging. Thus, we hypothesized that among several pre-existing cancer cell clones carrying different resistance mechanisms, some resistant cell clones that expand shortly after EGFR-TKIs would eventually cause the resistant phenotype. In this study, we investigated whether resistance clones significantly increase shortly after exposure to EGFR-TKIs and determine the final phenotype of the resistance to EGFR-TKIs using in vitro, in vivo, and ex vivo
EGFR-mutant tumor models. We further tested this hypothesis in patients who were diagnosed with advanced
EGFR-mutated NSCLC and treated with EGFR-TKIs by monitoring early dynamic changes of resistance-associated gene alterations during treatment using plasma circulating tumor DNA (ctDNA). Our study focused on developing two main resistance mechanisms:
EGFR T790M mutation and
C-MET amplification.
2. Materials and Methods
2.1. Cell Line and Reagents
HCC827 and PC9 cell lines were purchased from the Korean Cell Line Bank (Seoul, Korea) and RIKEN BioResource Center cell bank (Ibaraki, Japan), respectively. H4006, A549, and H1975 cell lines were obtained from American Type Culture Collection (Manassas, VA, USA). Cells were maintained in RPMI-1640 supplemented with 10% fetal bovine serum. Gefitinib was purchased from LC Laboratories (Woburn, MA, USA). AZD9291, PHA665752, and Afatinib were purchased from Selleckchem (Houston, TX, USA).
2.2. Establishment of Gefitinib-Resistant Cell Lines
Gefitinib-resistant cells were established by continuously exposing parent cells to increasing drug concentrations. Beginning at 0.01 μM, the exposure drug dose was doubled until a final concentration of 1.0 μM. Drug-resistant phenotypes were confirmed using cell viability assays. PC9GR and HCC827GR cells were the resistant cells from PC9 and HCC827 cells, respectively.
2.3. Establishment of Patient-Derived Cells (PDCs)
Pleural effusions were obtained from patients (female, 65 years old) and collected in sterile sample cups. Samples were transferred to conical tubes and centrifuged at 120× g for 10 min at room temperature (RT). The cell pellet was resuspended in RPMI and carefully layered onto LSM (lymphocyte separation medium; #091692249; MP Biomedicals, Illkirch, France). After centrifugation at 400× g for 30 min at RT, the interphase layer between RPMI and LSM including tumor cells was harvested and washed with RPMI. The washed cells were cultured in AR-5 medium (5% fetal bovine serum, 1× GlutaMAX (Thermo Fisher Scientific, Waltham, MA, USA), 1× ITS (insulin–transferrin–selenium, Thermo Fisher Scientific), 1% penicillin/streptomycin, 50 nM hydrocortisone, 1 mM sodium pyruvate and 1 ng/mL EGF in RPMI) at 37 °C in a 5% CO2 atmosphere.
2.4. Cell Viability Assay
The cells were cultured in gefitinib-free medium for 2 weeks before testing. The cells were then seeded at a density of 4 × 103 cells/well in 96-well plates. After 24 h, the cells were exposed to different concentrations of gefitinib and further incubated for 48 h. The cells were then washed with phosphate-buffered saline, and cell viability was measured using the CellTiter 96® AQueous One Solution Cell Proliferation Assay (Promega, Madison, WI, USA) according to the manufacturer’s instructions.
2.5. Direct Sequencing
Genomic DNA was extracted from cells using the phenol-chloroform method. Polymerase chain reaction (PCR) was performed using 1 μL of the extracted genomic DNA, SG-PCR Premix-EX mix (SG-P004EX; SG-Bio), and 10 pmol of the primers in a final volume of 20 μL. The primers are shown in
Table S1. The PCR cycling parameters were as follows: 95 °C for 5 min, 35 cycles at 95 °C for 30 s, 60 °C for 30 s, and 72 °C for 30 s, followed by a final step at 72 °C for 10 min. After the PCR products were purified, they were directly sequenced by Cosmogenetech, Inc. (Seoul, Korea) according to the manufacturer’s instructions.
2.6. Droplet Digital PCR
Droplet digital PCR (ddPCR) was performed using a QX200 Droplet Digital PCR system (Bio-Rad, Hercules, CA, USA). Reactions were performed in 10 μL of ddPCR 2× Master mix, 1 μL of 20× primer, and TaqMan Probe mix (Bio-Rad) (for the T790M mutation: #1863103; MET CNV FAM: #10031240; HEX: #10031243), 8 μL of nuclease-free water, and 1 μL of extracted genomic DNA in a final volume of 20 μL. Each sample was transferred to the middle wells of the cartridge, and 70 μL of droplet generation oil was added to the lower wells. Next, the cartridge was placed into the droplet generator to generate each sample-containing oil droplet; 40 μL droplets of each sample were transferred to the wells of a 96-well PCR plate. The PCR cycling parameters were as follows: 95 °C for 10 min, 40 cycles at 94 °C for 30 s, 58 °C for 1 min, and 98 °C for 10 min. After PCR was complete, the 96-well PCR plate was loaded into the droplet reader to read TaqMan Probe fluorescence in individual droplets. After that, we used QuantaSoft software to analyze the data based on the results from no template control wells with a threshold. Based on a previous study, a limit of detection (LOD) of the ddPCR assays for T790M mutation was determined as 0.05% [
16].
2.7. Quantitative Real-Time PCR (RT-PCR)
Total RNA was prepared using TRIzol (Ambion, Austin, TX, USA) according to the manufacturer’s protocol. cDNA was synthesized from total RNA using Superscript II (Invitrogen, Carlsbad, CA, USA) (
Table S2). Gene expression was investigated using quantitative real-time PCR (qPCR) on a Lightcycler
® 480 instrument (Roche, Basel, Switzerland), and all the reactions were run in triplicate. Reactions were performed in 2× Sensi FAST SYBR No-ROX mix (Bioline, London, UK), 8 μL of nuclease-free water, 1 μg of cDNA, and 10 pmol of the primers in a final volume of 20 μL. All the primers were ordered from Cosmogenetech. Gene expression was normalized to the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase.
2.8. Immunoblotting
Cells were washed with cold phosphate-buffered saline (PBS) and harvested with radioimmunoprecipitation assay lysis buffer containing phosphatase inhibitor cocktail set V (#524629; MERCK, Kenilworth, NJ, USA) and protease inhibitor cocktail set III (#535140; MERCK). Whole-cell lysates were separated using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and were blotted onto polyvinylidene fluoride membranes (#10600030; Amersham, Little Chalfont, UK). After blocking with 5% skim milk in Tris-buffered saline buffer (pH 8.0) with 0.1% Tween-20, the membrane was incubated with the primary antibody overnight (Cell Signaling Technology, Danvers, MA, USA; anti-pEGFR: #3777S; anti-EGFR: #4267S; anti-pMET: #3077S; anti-MET: #8198S; anti-pAKT: #9271S; anti-AKT: #9272S; anti-pERK: #4370S;anti-ERK: #4695S; anti-E-CADHERIN: #3195S; anti-VIMENTIN: #5741S). After rinsing with Tris-buffered saline buffer, the membrane was incubated with a secondary antibody for 1 h and washed, followed by visualization using electrochemiluminescence (SuperSignal™ West Femto Maximum Sensitivity Substrate; Thermo Fisher Scientific; #34095) and a LAS-3000 detection system (Fujifilm, Tokyo, Japan).
2.9. Colorimetric Detection of ctDNA Mutations
The probe sequences were designed using the UCSC Genome Browser. Biotinylated probes were synthesized by Macrogen (Seoul, Korea) (
Table S3). Initially, positively charged polyethyleneimine-conjugated nanowires (PEI-mNWs) (Genopsy Inc., Seoul, Korea; 5 µg/mL) were added to 200 µL fragmented DNA samples and mixed for 30 min at RT to allow DNA-NW complex formation. DNA-NW complexes were used after heating at 95 °C for 1 min. Biotin-labelled probe mix (1 pM) was added to the resulting solution and incubated additionally for 20 min at RT, followed by the addition of 2 μg/mL of horseradish peroxidase (HRP)-labelled streptavidin (st)-conjugated nanoparticles (NPs) (HRP-st NPs) (Genopsy Inc., ) and incubation for 5 min at RT. After precipitation, 25 µL of 10 mM 3,3′,5,5′-tetramethylbenzidine, 25 µL of 0.1 M H
2O
2, and 200 µL of 0.2 M sodium acetate trihydrate buffer (pH 5.0) were added to the resulting sample. The optical densities (ODs) of the samples were measured using absorbance at 490–800 nm and an Epoch UV-Vis spectrophotometer (BioTek, Winooski, VT, USA). The signal associated with mutation was determined by subtracting the absorbance at 500 nm from the absorbance at 650 nm (ΔOD 650−500).
2.10. Synthesis of Anti-C-MET Antibody-Fluorophore Conjugate
For the analysis of C-MET expression on the surface of the cells, anti-C-MET antibody-fluorophore conjugate was prepared as below. Onartuzumab (Anti-C-MET antibody BSA, azide free) was purchased from Evitria (Schlieren, Switzerland). Alexa Fluor 647-NHS ester was obtained from Thermo Fisher Scientific (Waltham, MA, USA).
Ornatuzumab (0.5 mg, 3.33 nmol) was reacted with AF647-NHS ester (antibody: dye mole ratio = 1:3.2) in phosphate buffered saline solution (PBS, pH 7.4) for 1 h at 25 °C. The reaction was performed under the light-protected condition with gentle shaking. The unreacted AF647-NHS ester was removed using a PD-10 column (GE healthcare, Chicago, IL, USA). The eluted antibody-dye conjugates were concentrated using Amicon Ultra-0.5 mL centrifugal filter (MWCO 50K, Millipore, Burlington, VT, USA) and stored at 4 °C till further analysis. The number of conjugated dyes per antibody was analyzed to be 1.5.
2.11. Flow Cytometry Staining and Cell Sorting
To detect apoptosis and C-MET expression at the same time, we used Annexin-V-FITC apoptosis detection kit (BD Biosciences, San Diego, CA, USA) and C-MET-Alexa 647 fluorescent dye described in
Section 2.10 (Materials and Methods). HCC827 and PC9 cells are grown in 100 mm plates and treated with 0.1 μM of Gefitinib and 0.01 μM Paclitaxel for 48 h. After drug treatment, cells were collected with trypsin-EDTA, washed with phosphate-buffered saline and then suspended in Annexin-V binding buffer solution. After being suspended in 200 μL binding buffer, cells were double stained with 3 μL Annexin-V-FITC and 1 μL C-MET-Alexa 647 fluorescent dyes. Cells were incubated for 30 min at RT in the dark and analyzed by FACs. We gated cells according to the expressions of fluorescent dyes and sorted the cells as Annexin-V positive (Annexin-V
+/C-MET
High+low) and Annexin-V negative (Annexin-V
−/C-MET
High). We centrifuged the sorted cells and extracted DNA for ddPCR experiment.
2.12. Mouse Xenograft Studies
We received approval of protocol from the National Cancer Center for mouse xenograft studies with accession number NCC-19-482. We used only 6-week-old BALB/c-nu female mice (n = 48) (Orientbio, Korea). The mice were randomly divided into three groups according to weight (average 20–22 g). They had an adaptation period of one week before the injection of cancer cells. We provided cozy bedding to reduce stress for mouse adaptation. Two cell lines (HCC827 and PC9) were injected and three drugs (dimethyl sulfoxide (DMSO), paclitaxel, and gefitinib) were used for treatment. We injected 1 × 106 cells within 50% Matrigel and measured the tumor volume using a caliper twice a week. Tumor sampling was performed when the volume reached approximately 300 mm3. We resected some tumors to obtain pre-treatment samples (1st operation) before starting drug treatment. After closely observing the mice for two days for operation site healing, the mice were randomized to receive one of three drug treatments. The mice that died after the 1st operation were not included in the subsequent experiment. Gefitinib was dissolved in DMSO and orally administered 5 days per week at a dose of 20 mg/kg. Paclitaxel was dissolved in DMSO and intraperitoneally injected once a week at a dose of 30 mg/kg. The tumor volume measurement time, intraperitoneal injection or oral administration time of the drug occurred between 3:30 pm and 6:30 pm. The test sequence was randomized each time, and each animal was tested at different times each day. After 1 week of treatment, the tumors were resected (2nd operation) and lysed in lysis buffers for RT-PCR and ddPCR. We injected continuous carbon dioxide (CO2) flow into the cage to induce euthanasia. We injected CO2 flow rapidly to induce painless death of the mice. After confirming the mouse’s breathing had stopped, the injection of CO2 was stopped. All the animal studies were conducted under the guidance of the Institutional Animal Care and Use Committee, and all relevant ethical regulations were followed.
2.13. Patient and Tumor Samples
We recruited four patients who were diagnosed with metastatic NSCLC with EGFR-sensitive mutations and had started EGFR-TKIs as a first-line treatment at the National Cancer Center (Goyang, Korea). These four patients with a median age of 58 years (range, 46–68) consisted of 2 males and 2 females. Blood samples were collected at baseline and every 8 to 12 weeks during EGFR-TKI treatment. Tumor response was evaluated every 8 to 12 weeks. Tissue samples were also collected to evaluate the acquired resistance mechanism after disease progression. To evaluate the EGFR T790M mutation levels and C-MET expression in plasma ctDNA, the nanowire-based colorimetric assay was performed. PEI-NWs (5 μg/mL) and diluted plasma (150 μL) were combined and mixed for 20 min at RT to form DNA-NW complexes. The colorimetric assay was performed as described above.
2.14. Ethics Approval and Consent to Participate
All the patients provided written informed consent. This study was performed with approval from the National Cancer Center Institutional Review Board (approval number NCC2011-0547). The study was conducted in compliance with the principles of the Declaration of Helsinki, the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use guidelines and local ethical and legal requirements. The protocol and IC document were approved by the independent institutional review boards of all the participating institutions.
2.15. Statistical Analysis
Each experiment was performed at least three times, and all data are presented as the mean ± SEM. Statistical analysis was performed using two-sample t-test. p-values ≤ 0.05 were considered significant (**, p ≤ 0.05; ns, not significant).
4. Discussion
We first evaluated whether the mechanism of acquired EGFR-TKI resistance varies among individual tumors using two lung cancer cell lines harboring the same
EGFR mutation. Although two gefitinib-resistant cancer cell lines were established by the same drug and treatment method during the same period, they showed different drug resistance mechanisms. This finding was consistent with that in a study by Shien et al. in which three lung cancer cell lines with
EGFR mutations became gefitinib-resistant with different mechanisms after stepwise escalation of chronic exposure to gefitinib [
25]. In that study, PC9, HCC827, and H4006 cells that acquired gefitinib resistance harbored a secondary T790M mutation,
C-MET amplification, and EMT features, respectively [
25]. However, how the different resistance mechanisms developed during treatment with the same EGFR-TKI was not revealed. One possible explanation is that each tumor with
EGFR mutations has different innate drug-resistant cell clone profiles. The selection of pre-existing drug-resistant cancer cell clones during treatment is a mechanism of acquired resistance to molecularly targeted drugs [
8,
10]. A preclinical study by Turke et al. identified
C-MET amplification as the mechanism of EGFR-TKI resistance and reported subclones of cancer cells carrying
C-MET amplification before drug exposure [
8]. Additionally, one whole-genome sequencing study analyzing several
EGFR-mutant lung cancers transforming into small cell lung cancer after an initial response to EGFR-TKI demonstrated that small cell lung cancer precursors were already present before treatment [
11]. Several clinical studies demonstrated the presence of low-abundance
EGFR T790M mutations in pre-treatment tumor tissue of patients with NSCLC harboring
EGFR mutations [
13,
14,
15]. However, the latent drug-resistant cell theory cannot fully explain why different drug resistance mechanisms occur. We found no significant difference in the baseline levels of
EGFR T790M mutations and
C-MET gene copy numbers in two parental cancer cells that subsequently developed different gefitinib resistance mechanisms after treatment. Further investigation is needed to evaluate the fundamental causes of different drug resistance mechanisms.
Most pre-existing drug-resistant subclones are present at very low frequencies within a tumor before treatment, and they cannot be detected using standard sequencing methods [
8,
12]. In addition, multiple resistant clones with different molecular mechanisms may exist within a tumor, and which resistance-associated clones will survive drug treatment cannot be predicted. Thus, we assumed that measuring changes in drug-resistant clone proportions after short-term drug exposure would be more useful to predict the final resistance mechanism than measuring their initial proportion before drug exposure. Consequently, we observed that the patterns in resistance-related genes or gene products after short-term exposure to EGFR-TKI varied depending on
EGFR-mutant lung cancer cell lines with a different EGFR-TKI resistance mechanism. This finding suggests that early emergent resistance clones can reflect the final molecular mechanism for EGFR-TKI failure.
To validate the preclinical findings, we performed serial plasma ctDNA analysis in four patients during EGFR-TKI treatment. One reason for ctDNA monitoring is that multiple tissue sampling over time is not feasible in most lung cancer patients. In addition, accurately measuring the proportion of gene alterations within a tumor is challenging because the biopsy sample can contain varying numbers of non-tumor cells. Thus, liquid biopsy, such as sampling circulating tumor cells or ctDNA, is more feasible and accurate in tracking molecular changes during anti-cancer treatment. We applied a new ctDNA analysis method, the nanowire-based colorimetric assay, which is an accurate, cost-effective, and rapid method for oncogenic mutations or amplification using small amounts of plasma in patients with several tumor types [
18,
21]. Increasing levels of
EGFR T790M or
C-MET expression in plasma ctDNA after short-term EGFR-TKI treatment was closely related to
EGFR T790M or
C-MET amplification in the post-treatment tumor tissue. However, this finding requires further validation studies because of our small sample size. Moreover, further technological improvement is needed to fully use liquid biopsy samples for the early prediction of the EGFR-TKI resistance mechanism. First, multiple specific gene alterations relevant to EGFR-TKI resistance must be simultaneously assessed despite using small biological specimen amounts. Second, very small fractional variations of gene variants should be detected with high accuracy. This approach requires analysis of transcriptomic data as well as genomic data to recognize nongenetic mechanisms related to resistance (e.g., EMT and transformation to small cell lung cancer). Technological progress in both ultra-deep sequencing methods and non-invasive sampling will improve the ability to predict the drug-resistant mechanism during EGFR-TKI treatment. Another promising method for the early prediction of drug-resistant mechanisms in patients who start EGFR-TKI treatment is to use PDC models. PDCs can provide more material to perform comprehensive genetic tests than blood samples. If the time to establish PDC lines can be shortened, their clinical application will be further expanded.
Since EGFR-TKIs were established as a first-line standard of care for
EGFR-mutant NSCLC patients, several randomized clinical trials in the last decade have evaluated the efficacy of combination treatment with EGFR-TKIs and other drugs [
26,
27]. In the NEJ009 study, the survival benefit of concurrent treatment with gefitinib and pemetrexed-carboplatin chemotherapy with gefitinib alone as a first-line treatment was compared in Japanese patients with advanced NSCLC with
EGFR mutations [
26]. An unprecedented median overall survival (OS) of 52.2 months after EGFR-TKI gefitinib and chemotherapy combination was reported, significantly longer than the 38.8-month median OS after treatment with gefitinib alone. In addition, a combination of erlotinib plus the anti-angiogenesis agent bevacizumab showed excellent outcomes compared with erlotinib alone (median progression-free survival (PFS), 16.9 vs. 13.3 months; hazard ratio (HR) = 0.605;
p = 0.016) [
27]. This combination strategy demonstrated survival outcomes comparable to the outstanding results of the third-generation EGFR-TKI osimertinib as a first-line standard treatment in the FLAURA study (median PFS, 18.9 vs. 10.2 months, HR = 0.46,
p < 0.001; median OS, 38.6 vs. 31.8 months, HR = 0.80,
p = 0.046) [
28]. However, these combination treatment strategies do not reflect individual biological characteristics of lung cancer with
EGFR mutations. A combination treatment that simultaneously targets
EGFR mutation and emerging resistance mechanisms is a reasonable personalized strategy to further improve patient outcomes. The early prediction of resistance mechanisms would be essential for the success of this treatment strategy.