EGFR-mutated lung cancers account for a significant subgroup of non–small cell lung cancers overall. Third-generation EGFR tyrosine kinase inhibitors (TKI) are mutation-selective inhibitors with minimal effects on wild-type EGFR. Acquired resistance develops to these agents, however, the mechanisms are as yet uncharacterized. In this study, we report that the Src–AKT pathway contributes to acquired resistance to these TKI. In addition, amplification of EGFR wild-type alleles but not mutant alleles was sufficient to confer acquired resistance. These findings underscore the importance of signals from wild-type EGFR alleles in acquiring resistance to mutant-selective EGFR-TKI. Our data provide evidence of wild-type allele-mediated resistance, a novel concept of acquired resistance in response to mutation-selective inhibitor therapy in cancer treatment. Cancer Res; 77(8); 2078–89. ©2017 AACR.

EGFR-mutated lung cancers comprise a significant subgroup of non–small cell lung cancer (NSCLC; refs. 1–3). In general, EGFR tyrosine kinase domain somatic mutations activate EGFR by promoting the active conformation of EGFR (4–7). Activated EGFR transduces signals to downstream pathways, such as the phosphoinositide 3-kinase (PI3K)–AKT and MEK–ERK mitogen-activated protein kinase (MAPK) pathways. After the identification of EGFR-activating mutations and response to first-generation EGFR tyrosine kinase inhibitors (EGFR-TKI; refs. 8–10), the treatment strategy for NSCLC patients harboring these mutations changed dramatically. The significant improvement of prognosis of lung cancer patients with EGFR mutations by first- (gefitinib and erlotinib) and second- (afatinib) generation EGFR-TKIs has been repeatedly demonstrated by multiple clinical trials (11–13). However, lung cancer cells inevitably acquire resistance to these inhibitors after approximately one year (14–16).

Multiple mechanisms of acquired resistance to first- and second-generation EGFR-TKIs have been identified to date—including the EGFR T790M gatekeeper mutation (17, 18), transformation to small cell lung cancer (SCLC; refs. 19, 20), and adaptive bypass pathway activation through the MET (21), AXL (22), and FGFR1 (23) axes. Of these, the EGFR T790M mutation is responsible for acquired resistance in approximately 50% of cases. The T790 residue is located at the entrance to a hydrophobic pocket on the posterior side of the ATP-binding cleft. As such, the T790M mutation induces a conformational change within the EGFR ATP-binding pocket, resulting in the steric hindrance of first or second-generation EGFR-TKIs (17). Coincidentally, EGFR T790M also enhances the affinity between ATP and its binding pocket (24), thereby synergistically conferring resistance to EGFR-TKIs. To address this issue, third-generation EGFR-TKIs, such as osimertinib (AZD9291; ref. 25), rociletinib (CO-1686; ref. 26), and nazartinib (EGF816; ref. 27), have been developed that irreversibly bind the EGFR ATP-binding pocket by forming a covalent bond with the C797 residue at pocket periphery, simultaneously blocking ATP binding due to the increased affinity. Interestingly, these third-generation EGFR-TKIs are also effective for the majority of EGFR-activating mutations, such as in-frame deletions in exon 19 and the L858R point mutation in exon 21 (28). Furthermore, mutation selectivity of third-generation EGFR-TKIs has been reported in preclinical and clinical models (25–27). The wide therapeutic window of third-generation EGFR-TKIs mirrors the significant efficacy and safety of these agents performed in multiple clinical trials (29, 30). Third-generation EGFR-TKIs are also subject to acquired resistance. It is important to fully clarify the mechanisms of acquired resistance to third-generation EGFR-TKIs to improve the prognosis of lung cancer patients harboring EGFR mutations. The mechanisms clarified to date include C797S or L798I mutation (31, 32), constitutive MAPK pathway activation by mutated KRAS or MEK (33), or bypass pathway activation via MET or ERBB2 (32, 34–36). However, the mechanisms underlying acquired resistance to third-generation EGFR-TKIs are not fully clarified.

In this study, we elucidated novel mechanisms underlying acquired resistance to third-generation EGFR-TKIs by whole-exome sequencing analysis. Notably, we determined that Src–AKT pathway activation and EGFR wild-type allele amplification can both contribute to EGFR-TKI acquired resistance, the latter of which might possibly be due to the decreased inhibitory pressure for EGFR wild-type by mutation selective third-generation EGFR-TKIs. Our findings highlight the importance of EGFR wild-type allele-mediated signaling in acquired resistance to mutant-selective EGFR-TKIs.

Reagents

Erlotinib, afatinib, rociletinib, and wortmannin were purchased from LC Laboratories. Osimertinib was purchased from Selleck Chemicals (Houston). Nazartinib was purchased from ApexBio. Bosutinib was purchased from Cayman Chemical). Cetuximab was purchased from Keio University Hospital (Tokyo, Japan). Total EGFR antibody (#2232), EGFR E746-A750del–specific antibody (#2085), total AKT antibody (#9272), phospho-AKT (S473/D9E) antibody (#4060), total p44/42 MAPK antibody (#9102S), phospho-p44/42 MAPK (T202/204) antibody (#9101S), E-cadherin antibody (#3195S), vimentin antibody (#5741S), total Src antibody (#2108), Phospho-(Tyr416)-Src antibody (#2101S), PTEN antibody (#9559), and PI3 kinase p110 alpha antibody (#4255S) were purchased from Cell Signaling Technology). The HA-tag (ab18181) and integrin beta 1 (ab52971) antibodies were purchased from Abcam). Phospho-EGFR (Y1068) antibody (44788G) was purchased from Invitrogen/Life Technologies. Actin antibody was purchased from Sigma-Aldrich.

Cell lines

The PC9 (EGFR E746-A750del) and H1975 (EGFR L858R + T790M) human NSCLC cell lines were obtained from Dr. Susumu Kobayashi (Beth Israel Deaconess Medical Center, Boston, MA) and the ATCC, respectively. Cells were cultured in RPMI1640 growth medium supplemented with 10% FBS at 37°C in a humidified 5% CO2 incubator. Cell authentication for PC9 and H1975 was performed by the authors in June 2015 using genetic profiling of polymorphic short tandem repeat (STR) loci (Takara).

Cell proliferation assay

MTS cell proliferation assays were performed as previously described (37). Briefly, 2 × 103 cells/well were seeded in 96-well plates and treated with EGFR-TKI or dimethyl sulfoxide (DMSO) vehicle 24 hours later. Absorbance was measured 72 hours after treatment. All experiments were performed at least three times.

Phospho-receptor tyrosine kinase array

The human phospho-receptor tyrosine kinase (phospho-RTK) array kit was purchased from R&D Systems) and screened according to the manufacturer's protocol with 150 μg of protein for each experiment. Signal intensity was calculated using LumiVision Analyzer software.

Establishment of the H1975 EGFR stable cell line

H1975 cells with stable EGFR wild-type or C797S overexpression were generated by retroviral infection using MigR1-EGFR constructs that harbor a GFP as previously described (5). Briefly, Phoenix-Ampho cells were transfected with 15 μg of wild-type or C797S EGFR using Lipofectamine 2000. Viral supernatant was collected 2 days after transfection and applied to H1975 cells on retronectin-coated plates (Takara) for 24 hours. EGFR-overexpressing H1975 cells were sorted by a FACS MoFlo XDP (Beckman Coulter) based on GFP expression. Purified cells were then grown in RPMI with 10% FBS.

Western blot analysis

Cells were treated with increasing concentrations of EGFR-TKIs (0.1–1 μmol/L). Cetuximab was used at concentrations of 10 μg/mL. Cells were lysed in Cell Lysis Buffer (Cell Signaling Technology), and equivalent amounts of protein were separated by SDS-PAGE and transferred to polyvinylidene fluoride membranes. The membranes were incubated overnight with primary antibodies at 4°C and then with secondary antibodies for 1 hour. Immunoreactive proteins were visualized with LumiGLO reagent and peroxide (Cell Signaling Technology), then exposed to X-ray film.

Apoptosis assay

Cells were seeded in 6-well plates (50,000/well) and were treated with rociletinib (1 μmol/L) and cetuximab (10 μg/mL) individually or in combination for 72 hours. Control cells were treated with DMSO. Apoptosis was monitored using the TACS Annexin V–FITC Apoptosis Detection Kit (R&D Systems) according to the manufacturer's protocol. The proportion of apoptotic cells was evaluated by flow cytometric analysis using a Gallios Flow Cytometer system (Beckman Coulter).

Quantitative RT-PCR and quantitative PCR

Total RNA was isolated from cultured cells using an RNeasy Mini Kit (Qiagen) and genomic DNA was isolated using a DNeasy Blood & Tissue Kit (Qiagen). RNA was subjected to reverse transcription using the High-Capacity RNA-to-cDNA Kit (Life Technologies) according to the manufacturer's protocol. Quantitative RT-PCR was performed using fluorescent SYBR Green and an ABI Prism 7000 Sequence Detection System (Life Technologies). Human GAPDH was used to normalize input cDNA. The LINE1 repetitive element was used as a reference gene for EGFR copy-number analysis. The primers used in this study are shown in Supplementary Table S1.

FISH

EGFR and chromosome 7 FISH analysis was performed by Genetic lab Co., Ltd. using Vysis LSI EGFR SpectrumOrange and CEP 7 SpectrumGreen probes, respectively.

Standard Sanger sequencing of PIK3CA

Isolated cDNA from H1975 cells was used as template in PCR reactions for PIK3CA. Amplified PIK3CA was sequenced and compared with the National Center for Biotechnology Information reference sequence NM_006218.3.

PIK3CA siRNA knockdown

Cells were transfected with PIK3CA-specific siRNA (#S10520; Life Technologies) or negative control siRNA (Ambion Silencer Select Negative Control mix, Life Technologies) using siLentFect transfection reagent (Bio-Rad) according to the manufacturers’ protocols. PIK3CA qRT-PCR was used to confirm gene knockdown.

Mouse xenograft model

All animal experiments were approved by the Laboratory Animal Center, Keio University School of Medicine (Tokyo, Japan). Female BALB/c-nu mice were purchased from Charles River. Mice were anesthetized with ketamine and PC9-COR#9 cells were subcutaneously injected in a Matrigel suspension. Calipers were used to measure tumor volume. Once average tumor volume reached 150 mm3, mice were randomized to receive vehicle alone, cetuximab (1 mg/mouse twice per week, intraperitoneally), rociletinib (30 mg/kg daily, orally), or a combination of both. Animals were humanely sacrificed and tumor tissues were harvested.

Whole-exome sequencing

Whole-exome sequencing libraries were prepared with 3 μg of DNA. The exomes were captured using the SureSelect Human All Exon V5+UTRs Kit (Agilent Technologies) according to the manufacturer's instructions and then sequenced using a HiSeq 1500 system (Illumina) to generate 100-bp paired-end data. The whole-exome sequencing data were deposited in the DDBJ database. Accession numbers: DRA004904, PRJDB5021, and SAMD00056514-SAMD00056521

Variant calling

Sequence reads were aligned to the human reference genome UCSC hg19 using the Burrows-Wheeler Aligner program (BWA, http://bio-bwa.sourceforge.net/). Single-nucleotide variants (SNV) and insertions and deletions (INDEL) were called and annotated using the Genome Analysis Toolkit software package (GATK, http://www.broadinstitute.org/gatk/). Sequencing artifacts were filtered out using custom filters (GATK confidence score, ≥50; number of variant reads in each direction, ≥1; variant allele frequency, ≥10%). Known germline variants were filtered out using data from dbSNP build 131, the 1,000 Genomes Project (Phase 1 exome data, released May 21, 2011), 1 Japanese genome, and 299 in-house Japanese exomes.

Copy number analysis

CNVs in resistant cells were analyzed from exome sequence data. The log ratio of depth coverage between parental and resistant cells was calculated using the GATK Depth of Coverage tool. CNV segments were then called from the log ratio of depth of coverage using the ExomeCNV package (38).

Statistical analysis

Statistical analysis was performed using GraphPad Prism software, version 4.0 (GraphPad Software). Two-sided Student t tests were used for comparisons, with P < 0.05 regarded as statistically significant.

Establishment of third-generation EGFR-TKI–resistant cells

To clarify the mechanisms underlying acquired resistance to third-generation EGFR-TKIs, such as osimertinib (AZD9291) and rociletinib (CO-1686), we established third-generation EGFR-TKI–resistant cells using a dose-escalation method. PC9 and H1975 cells were cultured with rociletinib or osimertinib. The initial concentration was 0.03 μmol/L, and this was incrementally increased to 1 μmol/L. After several months of exposure, resistant cell lines, rociletinib-resistant PC9 (PC9-COR), osimertinib-resistant PC9 (PC9-AZDR), rociletinib-resistant H1975 (H1975-COR), and osimertinib-resistant H1975 (H1975 AZDR) were established. Their resistance to a first-(erlotinib), third-generation (rociletinib, osimertinib, or nazartinib) EGFR-TKIs was confirmed by the MTS cell proliferation assay (Fig. 1). The calculated IC50 values are summarized in Supplementary Table S2. Notably, the resistant cells displayed cross-resistance to all third-generation EGFR-TKIs examined in this study, suggesting that the mechanisms of resistance to third-generation drugs may not be inhibitor specific. Further, resistant cells also showed a diminished propensity to undergo treatment-induced apoptosis (Supplementary Fig. S1), indicating that the cells acquired resistance to third-generation EGFR-TKIs.

Figure 1.

Establishment of third-generation EGFR-TKI–resistant cells. MTS cell proliferation assays for PC9 and H1975 parental and resistant cells treated with increasing concentrations of the indicated EGFR-TKIs for 72 hours. Error bars, SD.

Figure 1.

Establishment of third-generation EGFR-TKI–resistant cells. MTS cell proliferation assays for PC9 and H1975 parental and resistant cells treated with increasing concentrations of the indicated EGFR-TKIs for 72 hours. Error bars, SD.

Close modal

Whole-exome sequencing of parental and EGFR-TKI–resistant cells

Bypass pathway activation is consistently reported as one of the mechanisms by which cells acquire resistance to EGFR-TKIs. As such, we first examined the phosphorylation levels of multiple receptor tyrosine kinases in our resistant PC9 and H1975 cells with a human phospho-RTK array kit, but did not observe any activation of reported bypass pathways (HER2, HGFR, IGF-1R, and AXL signaling; Supplementary Fig. S2).

To clarify the heterogeneity of potential resistance mechanisms, the PC9-COR, PC9-AZDR, H1975-COR, and H1975-AZDR lines were subcloned to isolate resistant clones (Supplementary Fig. S3). DNA isolated from the parental and resistant cell clones was then subjected to whole-exome sequencing. Quality of the whole-exome sequencing results and copy number alterations are summarized in Supplementary Table S3 and Supplementary Fig. S4. Several genetic alterations potentially relevant to EGFR-TKIs sensitivity were detected (Table 1), including some already reported. For instance, osimertinib resistance can occur via oncogenic KRAS signaling (33). In our model, KRAS G13D was identified in PC9-AZDR clones (Supplementary Fig. S5), supporting the validity of our findings. EGFR C797S or L798I was not found in the resistant cells.

Table 1.

Results from whole-exome sequencing show several genetic alterations potentially relevant to EGFR-TKI sensitivity

Cell lineGenetic alterations
PC9 EGFR E746_A750del 
PC9-COR#9 EGFR E746_A750del+EGFR wild-type amplification 
PC9-AZDR#4 EGFR E746_A750del+KRAS G13D 
PC9-AZDR#5 EGFR E746_A750del+KRAS G13D 
H1975 EGFR L858R/T790M+PIK3CA G118D (heterozygosity) 
H1975-COR#3 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 
H1975-AZDR#1 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 
H1975-AZDR#3 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 
Cell lineGenetic alterations
PC9 EGFR E746_A750del 
PC9-COR#9 EGFR E746_A750del+EGFR wild-type amplification 
PC9-AZDR#4 EGFR E746_A750del+KRAS G13D 
PC9-AZDR#5 EGFR E746_A750del+KRAS G13D 
H1975 EGFR L858R/T790M+PIK3CA G118D (heterozygosity) 
H1975-COR#3 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 
H1975-AZDR#1 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 
H1975-AZDR#3 EGFR L858R/T790M+PIK3CA G118D (homozygosity) 

Epithelial-to-mesenchymal transition phenotype and AKT pathway activation in third-generation EGFR-TKI–resistant cells

Parental H1975 cells harbor a heterozygous PIK3CA G118D mutation. Interestingly, the PIK3CA wild-type allele was lost in three of the resistant clones—H1975-AZDR#1 and #3, and H1975-COR#3, leading to a loss of heterozygosity (Supplementary Table S4; Supplementary Fig. S6A and S6B). The activating PIK3CA G118D mutation is found in various cancer types, including endometrium (39) and colorectal (40, 41) cancers. In order to investigate whether acquired homozygosity of PIK3CA G118D could functionally induce resistance to EGFR-TKIs, we examined the impact of PIK3CA inhibition on EGFR-TKI resistant cells using the PIK3CA inhibitor wortmannin. Although effective when used in combination with EGFR-TKI in parental H1975 cells, wortmannin was unable to restore EGFR-TKI sensitivity in the H1975-resistant clones (Supplementary Fig. S6C and S6D). Further, the siRNA-mediated knockdown of PIK3CA gene expression in the H1975-resistant clones also failed to restore EGFR-TKI sensitivity (Supplementary Fig. S6E and S6F). These data indicate that acquired homozygosity of the PIK3CA G118D mutation did not contribute to the acquired resistance to EGFR-TKI in H1975-resistant cells.

While performing these experiments, we noticed a change in cell morphology where the H1975-resistant cells exhibited a spindle cell-like morphology (Fig. 2A). Epithelial-to-mesenchymal transition (EMT) is reported to occur in EGFR-resistant NSCLC (42). To confirm whether H1975 cell clones acquired a more mesenchymal phenotype, we analyzed EMT markers by Western blotting. Consistently, expression of the mesenchymal marker vimentin and the epithelial E-cadherin was upregulated and downregulated in the resistant clones at the protein level, respectively (Fig. 2B). Moreover, both total and phosphorylated Src were elevated in H1975-resistant cells. Next, we examined the effect of EGFR-TKI treatment on EGFR downstream pathway activation, including AKT and MAPK, in H1975-resistant cells. EGFR and ERK1/2 phosphorylation were efficiently inhibited by rociletinib and osimertinib treatment; however, limited effects were observed in AKT phosphorylation in H1975-resistant cells when compared with H1975 parental counterparts (Fig. 2C). Integrin β1/Src/AKT-mediated bypass signaling has previously been reported in erlotinib-resistant NSCLC (43). Consistent with this report, we found that integrin β1 and phospho-Src were increased at the RNA and protein levels, respectively, in H1975-resistant clones (Fig. 2B and D). A loss of PTEN and subsequent AKT pathway activation facilitate EGFR-TKI resistance in mutant lung cancer (44); however, PTEN expression was intact in H1975-resistant cells (Fig. 2B and E). To examine whether Src–AKT pathway activation contributed to the acquired resistance to third-generation EGFR-TKI resistance, we performed MTS proliferation assays on H1975 clones treated with third-generation EGFR-TKIs in the presence or absence of the Src inhibitors dasatinib or bosutinib. Interestingly, both dasatinib and bosutinib partially restored sensitivity to third-generation EGFR-TKIs (Fig. 2F and G), suggesting that Src–AKT pathway activation may contribute to the acquired resistance to third-generation EGFR-TKIs.

Figure 2.

EMT and Src–AKT pathway activation in third-generation EGFR-TKI–resistant cells. A, Representative pictures of parental and resistant H1975 cells. B, Western blot analysis of H1975 parental and resistant (COR#3, AZDR#1) cells for E-cadherin, vimentin, integrin β1, total (t-) and phosphorylated (p-) Src, PTEN, and β-actin. C, Western blot analysis for total (t-) and phosphorylated (p-) EGFR, AKT, ERK1/2, and β actin in H1975 parental and resistant (COR#3, AZDR#1) cells. D, Integrin β1 (ITGB1) gene expression relative to that of GAPDH in H1975 parental cells and resistant clones. Error bars, SD. E, PTEN gene expression relative to that of GAPDH in H1975 parental cells and resistant clones. Error bars, SD. F, MTS cell proliferation assays following treatment with the indicated EGFR-TKI concentrations with or without bosutinib or dasatinib in H1975 parental cells and resistant clones (H1975-COR#3, H1975-AZDR#1). Error bars, SD. G, Western blot analysis for total (t-) and phosphorylated (p-) Src and β-actin in H1975 parental cells treated with the indicated concentrations of bosutinib or dasatinib.

Figure 2.

EMT and Src–AKT pathway activation in third-generation EGFR-TKI–resistant cells. A, Representative pictures of parental and resistant H1975 cells. B, Western blot analysis of H1975 parental and resistant (COR#3, AZDR#1) cells for E-cadherin, vimentin, integrin β1, total (t-) and phosphorylated (p-) Src, PTEN, and β-actin. C, Western blot analysis for total (t-) and phosphorylated (p-) EGFR, AKT, ERK1/2, and β actin in H1975 parental and resistant (COR#3, AZDR#1) cells. D, Integrin β1 (ITGB1) gene expression relative to that of GAPDH in H1975 parental cells and resistant clones. Error bars, SD. E, PTEN gene expression relative to that of GAPDH in H1975 parental cells and resistant clones. Error bars, SD. F, MTS cell proliferation assays following treatment with the indicated EGFR-TKI concentrations with or without bosutinib or dasatinib in H1975 parental cells and resistant clones (H1975-COR#3, H1975-AZDR#1). Error bars, SD. G, Western blot analysis for total (t-) and phosphorylated (p-) Src and β-actin in H1975 parental cells treated with the indicated concentrations of bosutinib or dasatinib.

Close modal

EGFR wild-type allele amplification induces resistance to third-generation EGFR-TKIs

Notably, our exome sequencing data revealed an amplification of the EGFR gene locus on chromosome 7 (Fig. 3A). To confirm this finding, chromosome 7 and the EGFR gene were analyzed by FISH and scored using the ratio EGFR signal to that of chromosome 7 (Fig. 3B). PC9 parental cells harbor EGFR gene amplification; however, the ratio of signals became higher in PC9-COR#9 clones when compared with parental counterparts (Supplementary Table S5). In addition, genomic DNA qPCR for the EGFR loci in the PC9-COR clones confirmed the amplification (Fig. 3C). Furthermore, the EGFR wild-type copy number and total EGFR expression were markedly increased in PC9-COR#9 cells (Supplementary Table S6; Fig. 3D). The relative number of tags for the wild-type EGFR allele (31 tags) was about one fourth (0.25 times) that of the mutant allele (127 tags) in PC9 parental cells; however, the wild-type allele was approximately 8.5-fold more prevalent in PC9-COR#9 cells (512 vs. 60 tags for the wild-type and mutant alleles, respectively). These data indicate that the amplified EGFR wild-type allele likely conveyed resistance to third-generation EGFR-TKIs.

Figure 3.

EGFR wild-type allele amplification in rociletinib-resistant clones. A, Copy-number variants (CNV) in PC9-COR#9 cells analyzed from exome sequence data. B, Two-color FISH analysis of PC9 parental and PC9-COR#9 cells. Green and red signals indicate CEP7 and EGFR, respectively. C, Relative gene copy number of EGFR when compared with LINE1. D, Western blot analysis of total (t-) EGFR, exon 19 deletion-specific EGFR (EGFRdel19), and β-actin in PC9 parental and resistant cells. E, Relative gene expression in PC9 parental and resistant cells. Error bars, SD. F, Western blot analysis for total (t-) and phosphorylated (p-) EGFR, exon 19 deletion–specific EGFR (EGFRdel19), total (t-), and phosphorylated (p-) AKT, ERK1/2, and β-actin in PC9 parental and resistant cells.

Figure 3.

EGFR wild-type allele amplification in rociletinib-resistant clones. A, Copy-number variants (CNV) in PC9-COR#9 cells analyzed from exome sequence data. B, Two-color FISH analysis of PC9 parental and PC9-COR#9 cells. Green and red signals indicate CEP7 and EGFR, respectively. C, Relative gene copy number of EGFR when compared with LINE1. D, Western blot analysis of total (t-) EGFR, exon 19 deletion-specific EGFR (EGFRdel19), and β-actin in PC9 parental and resistant cells. E, Relative gene expression in PC9 parental and resistant cells. Error bars, SD. F, Western blot analysis for total (t-) and phosphorylated (p-) EGFR, exon 19 deletion–specific EGFR (EGFRdel19), total (t-), and phosphorylated (p-) AKT, ERK1/2, and β-actin in PC9 parental and resistant cells.

Close modal

We next examined EGFR ligand expression to evaluate the possibility of ligand-induced activation of EGFR (45). Until now, seven EGFR ligands have been identified: epithelial growth factor (EGF), amphiregulin, heparin-binding EGF-like growth factor (HB-EGF), epiregulin, TGFα, epigen, and betacellulin. Quantitative RT-PCR analysis revealed an increased expression of all seven ligands in PC9-COR cells and PC9-COR#9 cells compared with PC9 parental cells (Fig. 3E). As expected, the inhibition of phosphorylation of EGFR, AKT, and ERK by rociletinib was attenuated in the PC9-COR#9 clone when compared with the PC9 parental cells (Fig. 3F). These results indicated that EGFR wild-type allele amplification likely induces the acquired resistance to third-generation EGFR-TKIs through EGFR ligand-induced activation.

To ascertain the plausibility of the aforementioned situation, we expressed HA-tagged wild-type EGFR in H1975 parental cells and evaluated their sensitivity to rociletinib and osimertinib in the presence of EGF. When compared with mock-transfected H1975 cells, wild-type EGFR-overexpressing H1975 cells developed resistance to rociletinib and osimertinib upon EGF administration (Fig. 4A and B), and rescued EGF-induced EGFR, AKT, and ERK1/2 phosphorylation (Fig. 4C). Thus, these data indicate that EGFR wild-type allele amplification and concomitant EGFR ligand expression contribute to acquired resistance to third-generation EGFR-TKIs.

Figure 4.

EGFR wild-type allele amplification contributes to third-generation EGFR-TKI resistance. A, Western blot analysis of H1975 cells transduced with empty vector (mock) or HA-tagged EGFR. B, MTS cell proliferation assays for H1975 mock and H1975 EGFR cells treated with the indicated EGFR-TKI concentrations with or without EGF (10 ng/mL, 100 ng/mL) for 72 hours. Error bars, SD. C, Western blot analysis of H1975 mock and H1975 EGFR cells treated with the indicated concentrations of EGF and rociletinib for phosphorylated (p-) and total (t-) EGFR, AKT, ERK1/2, and β-actin.

Figure 4.

EGFR wild-type allele amplification contributes to third-generation EGFR-TKI resistance. A, Western blot analysis of H1975 cells transduced with empty vector (mock) or HA-tagged EGFR. B, MTS cell proliferation assays for H1975 mock and H1975 EGFR cells treated with the indicated EGFR-TKI concentrations with or without EGF (10 ng/mL, 100 ng/mL) for 72 hours. Error bars, SD. C, Western blot analysis of H1975 mock and H1975 EGFR cells treated with the indicated concentrations of EGF and rociletinib for phosphorylated (p-) and total (t-) EGFR, AKT, ERK1/2, and β-actin.

Close modal

Rociletinib/cetuximab cotreatment overcomes EGFR wild-type–mediated acquired resistance in vitro and in vivo

Concomitant EGFR wild-type amplification and ligand expression promoted us to determine whether the inhibition of EGFR ligand binding would be sufficient to restore sensitivity to rociletinib in PC9-COR#9 cells. For this, we performed MTS assays in the presence of cetuximab, an anti-EGFR antibody that blocks ligand binding. As a single agent treatment, cetuximab had no effect on cell proliferation in PC9 parental cells and PC9-COR#9 cells (Fig. 5A), nor did rociletinib/cetuximab combination therapy show a synergistic effect in PC9 parental cells and PC9-AZDR#5 cells that harbor a KRAS G13D mutation (Fig. 5B). However, combination therapy effectively restored rociletinib sensitivity in PC9-COR#9 cells (Fig. 5C). We used Western blotting to investigate whether rociletinib/cetuximab cotreatment suppressed pathway activation downstream of EGFR in PC9-COR#9 cells and found that it efficiently inhibited both AKT and ERK1/2 phosphorylation (Fig. 5D). Analysis of apoptosis by FACS revealed an increase in Annexin V–positive cells with the subsequent addition of cetuximab (12.5 % with 1 μmol/L rociletinib alone, 32.7% with rociletinib/cetuximab combination; Fig. 5E), indicating that ligand-mediated EGFR activation contributed to the acquired resistance to third-generation EGFR-TKIs. We evaluated the effect of afatinib on PC9-COR#9 cells by MTS assay because we previously reported the decreased mutation specificity of afatinib (28). The proliferation of PC9-COR#9 cells was inhibited by lower concentrations of afatinib than rociletinib or nazartinib (Fig. 5F). These findings also indicate that EGFR wild-type mediated signaling contributes to PC9-COR#9 cell resistance.

Figure 5.

Rociletinib and cetuximab combination treatment overcomes EGFR wild–type–mediated acquired resistance in vitro. A, MTS cell proliferation assays for PC9 and PC9-COR#9 cells treated with the indicated concentrations of cetuximab for 72 hours. Error bars, SD. B, MTS cell proliferation assays of PC9-AZDR#5 cells treated with the indicated concentrations of cetuximab and osimertinib for 72 hours. Error bars, SD. C, MTS cell proliferation assays for PC9-COR#9 cells treated with the indicated concentrations of rociletinib and cetuximab for 72 hours. Error bars, SD. D, Western blot analysis of phosphorylated (p-) and total (t-) EGFR, AKT, ERK1/2, and β-actin in PC9-COR#9 cells treated with the indicated concentrations of rociletinib and cetuximab for 8 hours. E, Flow cytometric data for PC9-COR#9 cells treated with DMSO, rociletinib, cetuximab, and rociletinb/cetuximab in combination for 72 hours. The numbers (%) indicate the proportion of Annexin V–FITC- and/or propidium iodide-stained cells. F, MTS cell proliferation assays for PC9-COR#9 cells treated with the indicated concentrations of rociletinib, nazartinib, or afatinib for 72 hours. Error bars, SD.

Figure 5.

Rociletinib and cetuximab combination treatment overcomes EGFR wild–type–mediated acquired resistance in vitro. A, MTS cell proliferation assays for PC9 and PC9-COR#9 cells treated with the indicated concentrations of cetuximab for 72 hours. Error bars, SD. B, MTS cell proliferation assays of PC9-AZDR#5 cells treated with the indicated concentrations of cetuximab and osimertinib for 72 hours. Error bars, SD. C, MTS cell proliferation assays for PC9-COR#9 cells treated with the indicated concentrations of rociletinib and cetuximab for 72 hours. Error bars, SD. D, Western blot analysis of phosphorylated (p-) and total (t-) EGFR, AKT, ERK1/2, and β-actin in PC9-COR#9 cells treated with the indicated concentrations of rociletinib and cetuximab for 8 hours. E, Flow cytometric data for PC9-COR#9 cells treated with DMSO, rociletinib, cetuximab, and rociletinb/cetuximab in combination for 72 hours. The numbers (%) indicate the proportion of Annexin V–FITC- and/or propidium iodide-stained cells. F, MTS cell proliferation assays for PC9-COR#9 cells treated with the indicated concentrations of rociletinib, nazartinib, or afatinib for 72 hours. Error bars, SD.

Close modal

In addition, we evaluated the efficacy of rociletinib and cetuximab or afatinib cotreatment with H1975 cells expressing wild-type EGFR (H1975 EGFR) or C797S EGFR (H1975 C797S; Supplementary Fig. S7). As observed previously, wild-type EGFR overexpression induced rociletinib resistance following EGF stimulation. The addition of afatinib or cetuximab restored rociletinib sensitivity to both transduced lines. However, the restoration of afatinib was more significant in H1975 EGFR cells compared with H1975 C797S cells.

Finally, mouse xenograft models were used to examine whether the roclietinib/cetuximab combination treatment could suppress EGFR wild-type-mediated resistance in vivo. During treatment, the body weight of the mice and PC9-COR#9 cells derived tumor volumes were monitored (Fig. 6A–C). Notably, tumor volumes increased in the control and rociletinib groups; however, roclietinib/cetuximab cotreatment significantly inhibited tumor growth. Collectively, these data support the possibility that combination therapy may be an effective approach to overcome EGFR wild-type-dependent acquired resistance.

Figure 6.

Overcoming EGFR wild-type–mediated acquired resistance by rociletinib and cetuximab combination treatment in vivo. A and B, PC9-COR#9–derived tumor-bearing mice were randomized into control, rociletinib, or rociletinib/cetuximab combination treatment groups. The body weight of the mice was monitored. Tumor size was measured to calculate tumor volume. Values indicate average tumor volume in each group. **, P < 0.01 for the combination of rociletinib/cetuximab combination versus rociletinib alone. Error bars, SD. C, Representative images of tumor-bearing mice and tumors. D, Proposed model of EGFR wild-type–mediated acquired resistance.

Figure 6.

Overcoming EGFR wild-type–mediated acquired resistance by rociletinib and cetuximab combination treatment in vivo. A and B, PC9-COR#9–derived tumor-bearing mice were randomized into control, rociletinib, or rociletinib/cetuximab combination treatment groups. The body weight of the mice was monitored. Tumor size was measured to calculate tumor volume. Values indicate average tumor volume in each group. **, P < 0.01 for the combination of rociletinib/cetuximab combination versus rociletinib alone. Error bars, SD. C, Representative images of tumor-bearing mice and tumors. D, Proposed model of EGFR wild-type–mediated acquired resistance.

Close modal

In NSCLC, EGFR mutations activate EGFR, which induce the transformation of lung epithelial cells. EGFR is ubiquitously expressed in organ epithelial cells, including lung, gastrointestinal tract, and skin. As such, it is essential to selectively inhibit mutant EGFR-derived signals to minimize adverse effects of EGFR-TKIs, such as diarrhea and skin rash. Third-generation EGFR-TKIs selectively inhibit EGFR mutants and minimally affect the wild-type EGFR (25, 26). This mirrors the significant efficacy and safety of third-generation EGFR-TKIs observed in recent clinical trials. Specifically, osimertinib and rociletinib showed significant efficacy and safety for previously treated NSCLC patients harboring EGFR-activating mutations, although rociletinib is no longer in development. The response rate and progression-free survival of osimertinib for EGFR T790M-positive patients were 61% and 9.6 months, respectively (29). The response rate of rociletinib for EGFR T790M-positive patients was 45% (46). Neither osimertinib nor rociletinib exhibited a dose-limiting toxicity; therefore, a maximum tolerated dose was not determined (29, 30). Thus, mutation-selective inhibitors offer a clinically relevant advantage of EGFR-TKI treatment with minimal adverse effect.

In this study, we identified several novel mechanisms of acquired resistance to third-generation EGFR-TKIs, including Src–AKT pathway activation and EGFR wild-type allele amplification (Fig. 6D and Supplementary Fig. S8). Most cancer-specific somatic driver mutations, such as those in KRAS and EGFR, exist in heterozygous. Until today, a limited number of studies have focused on the function of the wild-type allele in cancer cells. Here, we show that cancer cells exploit this decreased inhibitory pressure for the wild-type allele.

The mechanisms underlying acquired resistance to third-generation EGFR-TKIs have already been partially clarified. These include the C797S (26) and L798I (32) mutations that presumably prevent the covalent binding of EGFR-TKIs to EGFR, bypass pathway activation (33–35, 47), and mutant EGFR gene amplification (48, 49). Specifically, Piotrowska and colleagues reported EGFR T790M allele amplification in rociletinib-resistant clones (49). To our knowledge, this is the first report describing a role for wild-type EGFR in acquired resistance to mutant-selective third-generation EGFR-TKIs.

Recently, circulating tumor DNA in blood is becoming a study sample for clarification of mechanisms underlying acquired resistance to EGFR-TKIs (26, 32). By using recent next-generation sequencing technology, the identification of cancer cells derived genetic alterations including mutations and gene copy-number alterations become possible. Considering the convenient access to blood samples and sensitivity and specificity of circulating tumor DNA, it will become commonly used as reliable samples in the clinics. However, the contamination of nonmalignant cells derived DNA is inevitable and may limit the accuracy of gene copy-number analysis in cancer cells. Thus, to identify the EGFR wild-type allele amplification, we believe biopsies from either primary tumors or metastatic lesions are necessary.

In summary, the present study characterized novel mechanisms of acquired resistance to mutant-selective third-generation EGFR-TKIs. We propose a novel concept of acquired resistance to mutation selective inhibitors, wild-type allele mediated resistance. In addition, we propose a preclinical rationale for the use of more promiscuous EGFR-TKI, afatinib, or cetuximab combination therapy in EGFR inhibitor–resistant cancers. While these findings underscore the importance of EGFR wild-type–mediated signals in acquired resistance, additional preclinical and clinical trials are necessary to evaluate the efficacy and safety of these treatments to avoid adverse side effects related to EGFR pathway inhibition in nonmalignant cells.

K. Soejima has received speakers bureau honoraria from Chugai, Ono, Taiho, Eli Lilly, AstraZeneca, Pfizer, and Shionogi. No potential conflicts of interest were disclosed by the others.

Conception and design: S. Nukaga, H. Yasuda, K. Tsuchihara, J. Hamamoto, I. Kawada, S. Ikemura, K. Goto, K. Soejima

Development of methodology: S. Nukaga, H. Yasuda, K. Tsuchihara, J. Hamamoto, I. Kawada, S. Matsumoto, S. Ikemura, K. Soejima

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Nukaga, H. Yasuda, K. Tsuchihara, J. Hamamoto, K. Masuzawa, K. Naoki, S. Mimaki, S. Ikemura

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Nukaga, H. Yasuda, K. Tsuchihara, J. Hamamoto, K. Masuzawa, K. Naoki, S. Ikemura

Writing, review, and/or revision of the manuscript: S. Nukaga, H. Yasuda, K. Tsuchihara, I. Kawada, K. Naoki, S. Matsumoto, S. Ikemura, K. Goto, T. Betsuyaku, K. Soejima

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Yasuda, J. Hamamoto, S. Ikemura, T. Betsuyaku

Study supervision: H. Yasuda, K. Tsuchihara, I. Kawada, S. Ikemura, K. Goto, T. Betsuyaku, K. Soejima

We thank Ms. Mikiko Shibuya for her excellent technical assistance. We also thank the Collaborative Research Resources, Keio University, School of Medicine, for cell sorting.

This work was supported in part by Grants-in-Aid for Scientific Research on Priority Areas from the Ministry of Education, Culture, Sports, Science, and Technology of Japan to S. Nukaga (Grant #16K19465), T. Betsuyaku (Grant #15H04833), K. Soejima (Grant #22590870), H. Yasuda (Grant #25860656, 15H05666, and 15K14398) and the Practical Research for Innovative Cancer Control from Japan Agency for Medical Research and Development to S. Matsumoto (16ck0106012h0003).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sharma
SV
,
Bell
DW
,
Settleman
J
,
Haber
DA
. 
Epidermal growth factor receptor mutations in lung cancer
.
Nat Rev Cancer
2007
;
7
:
169
81
.
2.
Kosaka
T
,
Yatabe
Y
,
Endoh
H
,
Kuwano
H
,
Takahashi
T
,
Mitsudomi
T
. 
Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications
.
Cancer Res
2004
;
64
:
8919
23
.
3.
The Cancer Genome Atlas Research Network. 
Comprehensive molecular profiling of lung adenocarcinoma
.
Nature
2014
;
511
:
543
50
.
4.
Yun
CH
,
Boggon
TJ
,
Li
Y
,
Woo
MS
,
Greulich
H
,
Meyerson
M
, et al
Structures of lung cancer-derived EGFR mutants and inhibitor complexes: mechanism of activation and insights into differential inhibitor sensitivity
.
Cancer Cell
2007
;
11
:
217
27
.
5.
Yasuda
H
,
Park
E
,
Yun
CH
,
Sng
NJ
,
Lucena-Araujo
AR
,
Yeo
WL
, et al
Structural, biochemical, and clinical characterization of epidermal growth factor receptor (EGFR) exon 20 insertion mutations in lung cancer
.
Sci Transl Med
2013
;
5
:
216ra177
.
6.
Jura
N
,
Zhang
X
,
Endres
NF
,
Seeliger
MA
,
Schindler
T
,
Kuriyan
J
. 
Catalytic control in the EGF receptor and its connection to general kinase regulatory mechanisms
.
Mol Cell
2011
;
42
:
9
22
.
7.
Zhang
X
,
Gureasko
J
,
Shen
K
,
Cole
PA
,
Kuriyan
J
. 
An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor
.
Cell
2006
;
125
:
1137
49
.
8.
Paez
JG
,
Janne
PA
,
Lee
JC
,
Tracy
S
,
Greulich
H
,
Gabriel
S
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.
9.
Lynch
TJ
,
Bell
DW
,
Sordella
R
,
Gurubhagavatula
S
,
Okimoto
RA
,
Brannigan
BW
, 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
39
.
10.
Pao
W
,
Miller
V
,
Zakowski
M
,
Doherty
J
,
Politi
K
,
Sarkaria
I
, et al
EGF receptor gene mutations are common in lung cancers from "never smokers" and are associated with sensitivity of tumors to gefitinib and erlotinib
.
Proc Natl Acad Sci U S A
2004
;
101
:
13306
11
.
11.
Shepherd
FA
,
Rodrigues Pereira
J
,
Ciuleanu
T
,
Tan
EH
,
Hirsh
V
,
Thongprasert
S
, et al
Erlotinib in previously treated non–small-cell lung cancer
.
N Engl J Med
2005
;
353
:
123
32
.
12.
Mok
TS
,
Wu
YL
,
Thongprasert
S
,
Yang
CH
,
Chu
DT
,
Saijo
N
, et al
Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
2009
;
361
:
947
57
.
13.
Sequist
LV
,
Yang
JC
,
Yamamoto
N
,
O'Byrne
K
,
Hirsh
V
,
Mok
T
, et al
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
2013
;
31
:
3327
34
.
14.
Maemondo
M
,
Inoue
A
,
Kobayashi
K
,
Sugawara
S
,
Oizumi
S
,
Isobe
H
, et al
Gefitinib or chemotherapy for non–small-cell lung cancer with mutated EGFR
.
N Engl J Med
2010
;
362
:
2380
8
.
15.
Rosell
R
,
Carcereny
E
,
Gervais
R
,
Vergnenegre
A
,
Massuti
B
,
Felip
E
, et al
Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non–small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial
.
Lancet Oncol
2012
;
13
:
239
46
.
16.
Miller
VA
,
Hirsh
V
,
Cadranel
J
,
Chen
YM
,
Park
K
,
Kim
SW
, et al
Afatinib versus placebo for patients with advanced, metastatic non–small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial
.
Lancet Oncol
2012
;
13
:
528
38
.
17.
Kobayashi
S
,
Boggon
TJ
,
Dayaram
T
,
Janne
PA
,
Kocher
O
,
Meyerson
M
, et al
EGFR mutation and resistance of non-small-cell lung cancer to gefitinib
.
N Engl J Med
2005
;
352
:
786
92
.
18.
Pao
W
,
Miller
VA
,
Politi
KA
,
Riely
GJ
,
Somwar
R
,
Zakowski
MF
, et al
Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain
.
PLoS Med
2005
;
2
:
e73
.
19.
Niederst
MJ
,
Sequist
LV
,
Poirier
JT
,
Mermel
CH
,
Lockerman
EL
,
Garcia
AR
, et al
RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer
.
Nat Commun
2015
;
6
:
6377
.
20.
Okamoto
I
,
Araki
J
,
Suto
R
,
Shimada
M
,
Nakagawa
K
,
Fukuoka
M
. 
EGFR mutation in gefitinib-responsive small-cell lung cancer
.
Ann Oncol
2006
;
17
:
1028
9
.
21.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
22.
Zhang
Z
,
Lee
JC
,
Lin
L
,
Olivas
V
,
Au
V
,
LaFramboise
T
, et al
Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer
.
Nat Genet
2012
;
44
:
852
60
.
23.
Terai
H
,
Soejima
K
,
Yasuda
H
,
Nakayama
S
,
Hamamoto
J
,
Arai
D
, et al
Activation of the FGF2-FGFR1 autocrine pathway: a novel mechanism of acquired resistance to gefitinib in NSCLC
.
Mol Cancer Res
2013
;
11
:
759
67
.
24.
Yun
CH
,
Mengwasser
KE
,
Toms
AV
,
Woo
MS
,
Greulich
H
,
Wong
KK
, et al
The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP
.
Proc Natl Acad Sci U S A
2008
;
105
:
2070
5
.
25.
Cross
DA
,
Ashton
SE
,
Ghiorghiu
S
,
Eberlein
C
,
Nebhan
CA
,
Spitzler
PJ
, et al
AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer
.
Cancer Discov
2014
;
4
:
1046
61
.
26.
Walter
AO
,
Sjin
RT
,
Haringsma
HJ
,
Ohashi
K
,
Sun
J
,
Lee
K
, et al
Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC
.
Cancer Discov
2013
;
3
:
1404
15
.
27.
Jia
Y
,
Juarez
J
,
Li
J
,
Manuia
M
,
Niederst
MJ
,
Tompkins
C
, et al
EGF816 exerts anticancer effects in non-small cell lung cancer by irreversibly and selectively targeting primary and acquired activating mutations in the EGF receptor
.
Cancer Res
2016
;
76
:
1591
602
.
28.
Hirano
T
,
Yasuda
H
,
Tani
T
,
Hamamoto
J
,
Oashi
A
,
Ishioka
K
, et al
In vitro modeling to determine mutation specificity of EGFR tyrosine kinase inhibitors against clinically relevant EGFR mutants in non–small-cell lung cancer
.
Oncotarget
2015
;
6
:
38789
803
.
29.
Janne
PA
,
Yang
JC
,
Kim
DW
,
Planchard
D
,
Ohe
Y
,
Ramalingam
SS
, et al
AZD9291 in EGFR inhibitor-resistant non–small-cell lung cancer
.
N Engl J Med
2015
;
372
:
1689
99
.
30.
Sequist
LV
,
Soria
JC
,
Goldman
JW
,
Wakelee
HA
,
Gadgeel
SM
,
Varga
A
, et al
Rociletinib in EGFR-mutated non–small-cell lung cancer
.
N Engl J Med
2015
;
372
:
1700
9
.
31.
Thress
KS
,
Paweletz
CP
,
Felip
E
,
Cho
BC
,
Stetson
D
,
Dougherty
B
, et al
Acquired EGFR C797S mutation mediates resistance to AZD9291 in non–small cell lung cancer harboring EGFR T790M
.
Nat Med
2015
;
21
:
560
2
.
32.
Chabon
JJ
,
Simmons
AD
,
Lovejoy
AF
,
Esfahani
MS
,
Newman
AM
,
Haringsma
HJ
, et al
Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients
.
Nat Commun
2016
;
7
:
11815
.
33.
Eberlein
CA
,
Stetson
D
,
Markovets
AA
,
Al-Kadhimi
KJ
,
Lai
Z
,
Fisher
PR
, et al
Acquired Resistance to the Mutant-Selective EGFR Inhibitor AZD9291 Is Associated with Increased Dependence on RAS Signaling in Preclinical Models
.
Cancer Res
2015
;
75
:
2489
500
.
34.
Planchard
D
,
Loriot
Y
,
Andre
F
,
Gobert
A
,
Auger
N
,
Lacroix
L
, et al
EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients
.
Ann Oncol
2015
;
26
:
2073
8
.
35.
Ortiz-Cuaran
S
,
Scheffler
M
,
Plenker
D
,
Dahmen
I
,
Scheel
A
,
Fernandez-Cuesta
L
, et al
Heterogeneous mechanisms of primary and acquired resistance to third-generation EGFR inhibitors
.
Clin Cancer Res
2016
;
22
:
4837
47
36.
Mizuuchi
H
,
Suda
K
,
Murakami
I
,
Sakai
K
,
Sato
K
,
Kobayashi
Y
, et al
Oncogene swap as a novel mechanism of acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitor in lung cancer
.
Cancer Sci
2016
;
107
:
461
8
.
37.
Tanizaki
J
,
Okamoto
I
,
Okabe
T
,
Sakai
K
,
Tanaka
K
,
Hayashi
H
, et al
Activation of HER family signaling as a mechanism of acquired resistance to ALK inhibitors in EML4-ALK-positive non–small cell lung cancer
.
Clin Cancer Res
2012
;
18
:
6219
26
.
38.
Sathirapongsasuti
JF
,
Lee
H
,
Horst
BA
,
Brunner
G
,
Cochran
AJ
,
Binder
S
, et al
Exome sequencing-based copy-number variation and loss of heterozygosity detection: ExomeCNV
.
Bioinformatics
2011
;
27
:
2648
54
.
39.
Burke
JE
,
Perisic
O
,
Masson
GR
,
Vadas
O
,
Williams
RL
. 
Oncogenic mutations mimic and enhance dynamic events in the natural activation of phosphoinositide 3-kinase p110alpha (PIK3CA)
.
Proc Natl Acad Sci U S A
2012
;
109
:
15259
64
.
40.
Samuels
Y
,
Wang
Z
,
Bardelli
A
,
Silliman
N
,
Ptak
J
,
Szabo
S
, et al
High frequency of mutations of the PIK3CA gene in human cancers
.
Science
2004
;
304
:
554
.
41.
Araya
CL
,
Cenik
C
,
Reuter
JA
,
Kiss
G
,
Pande
VS
,
Snyder
MP
, et al
Identification of significantly mutated regions across cancer types highlights a rich landscape of functional molecular alterations
.
Nat Genet
2016
;
48
:
117
25
.
42.
Sequist
LV
,
Waltman
BA
,
Dias-Santagata
D
,
Digumarthy
S
,
Turke
AB
,
Fidias
P
, et al
Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors
.
Sci Transl Med
2011
;
3
:
75ra26
.
43.
Kanda
R
,
Kawahara
A
,
Watari
K
,
Murakami
Y
,
Sonoda
K
,
Maeda
M
, et al
Erlotinib resistance in lung cancer cells mediated by integrin beta1/Src/Akt-driven bypass signaling
.
Cancer Res
2013
;
73
:
6243
53
.
44.
Sos
ML
,
Koker
M
,
Weir
BA
,
Heynck
S
,
Rabinovsky
R
,
Zander
T
, et al
PTEN loss contributes to erlotinib resistance in EGFR-mutant lung cancer by activation of Akt and EGFR
.
Cancer Res
2009
;
69
:
3256
61
.
45.
Roskoski
R
 Jr
. 
The ErbB/HER family of protein-tyrosine kinases and cancer
.
Pharmacol Res
2014
;
79
:
34
74
.
46.
Sequist
LV
,
Soria
JC
,
Camidge
DR
. 
Update to rociletinib data with the RECIST confirmed response rate
.
N Engl J Med
2016
;
374
:
2296
7
.
47.
Kim
TM
,
Song
A
,
Kim
DW
,
Kim
S
,
Ahn
YO
,
Keam
B
, et al
Mechanisms of acquired resistance to AZD9291: a mutation-selective, irreversible EGFR inhibitor
.
J Thorac Oncol
2015
;
10
:
1736
44
.
48.
Ercan
D
,
Zejnullahu
K
,
Yonesaka
K
,
Xiao
Y
,
Capelletti
M
,
Rogers
A
, et al
Amplification of EGFR T790M causes resistance to an irreversible EGFR inhibitor
.
Oncogene
2010
;
29
:
2346
56
.
49.
Piotrowska
Z
,
Niederst
MJ
,
Karlovich
CA
,
Wakelee
HA
,
Neal
JW
,
Mino-Kenudson
M
, et al
Heterogeneity underlies the emergence of EGFRT790 wild-type clones following treatment of T790M-positive cancers with a third-generation EGFR inhibitor
.
Cancer Discov
2015
;
5
:
713
22
.

Supplementary data