Lung Cancer 83 (2014) 44–50

Contents lists available at ScienceDirect

Lung Cancer journal homepage: www.elsevier.com/locate/lungcan

Loss of an EGFR-amplified chromosome 7 as a novel mechanism of acquired resistance to EGFR-TKIs in EGFR-mutated NSCLC cells夽 Koh Furugaki, Toshiki Iwai, Yoichiro Moriya ∗ , Naoki Harada, Kaori Fujimoto-Ouchi Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Japan

a r t i c l e

i n f o

Article history: Received 11 June 2013 Received in revised form 10 September 2013 Accepted 5 October 2013 Keywords: Erlotinib EGFR-TKI Mutation Resistance Non-small cell lung cancer NSCLC

a b s t r a c t Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) show notable effects against non-small cell lung cancers (NSCLCs) harboring EGFR-activating mutations. However, almost all patients eventually acquire resistance to EGFR-TKIs. In this study, we established novel erlotinib resistant NSCLC cells and examined their resistant mechanisms. Resistant cells were established in 14, 3, and 0 wells exposed to 0.1, 1, and 10 ␮M erlotinib, respectively. The IC50 values of these cells were 47- to 1209-fold higher than that of the parent cells. No secondary T790M mutation was detected in any resistant cells. However, in 13/17 resistant cells, EGFR copy number was reduced less than approximately one-eighth of parent cells, and in one resistant cell (B10), >99.99% of the population was EGFR-unamplified cells. Most (97.5%) parent cells showed EGFR amplification, but 2.5% of the population comprised EGFR-unamplified cells. An EGFR-unamplified clone (4D8) isolated from parent cells in erlotinib-free normal medium also showed erlotinib resistance comparable to the resistant B10 cells. Loss of an EGFR-amplified chromosome 7 (EGFR-ampch7) was observed in 4D8 and B10 cells. EGFR-unamplified cells were constantly maintained as a minor population of the parent cells under normal cell culture conditions. In conclusion, loss of an EGFR-ampch7 causes acquired resistance in EGFR-mutated HCC827 cells exposed to a relatively low concentration of erlotinib, but a high concentration prevents the emergence of resistance. © 2013 The Authors. Published by Elsevier Ireland Ltd. All rights reserved.

1. Introduction Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for approximately 80% of lung cancers. NSCLC is attributed in part to somatic mutations of the epidermal growth factor receptor gene (EGFR) [1]. The most common mutations are an in-frame E746-A750 deletion in exon 19 and a single-point substitutional L858R mutation in exon 21, both of which are located in the tyrosine kinase domain of EGFR. These two mutations are observed in approximately 90% of EGFR mutations and are termed “activating mutations” [2]. EGFR-TKIs, such as gefitinib and erlotinib, block autophosphorylation of EGFR with subsequent inhibition of the downstream signaling pathways involving RAS/extracellular signal regulated kinase (ERK)1/2 and phosphoinositide 3-kinase (PI3K)/AKT, and show favorable activity in NSCLC patients with activating mutations of EGFR [3]. However,

夽 This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-No Derivative Works License, which permits non-commercial use, distribution, and reproduction in any medium, provided the original author and source are credited. ∗ Corresponding author at: Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan. Tel.: +81 467 47 6356; fax: +81 467 45 7643. E-mail address: [email protected] (Y. Moriya).

almost all patients eventually develop acquired resistance to EGFRTKIs within several years [4]. Two genetic mechanisms of acquired resistance to EGFR-TKIs have been identified in EGFR-mutated NSCLC. A secondary mutation of T790M in exon 20 of EGFR and amplification of the MET oncogene are observed in approximately 50% and 5% of resistant cases, respectively [5–8]. Moreover, Yano et al. showed that overexpression of hepatocyte growth factor (HGF), a ligand for MET, induces acquired resistance by activating MET signals [9]. However, the mechanisms underlying acquired resistance to EGFR-TKIs are unknown in approximately 30% of EGFR-mutated NSCLC patients [8]. Further characterization of these unknown mechanisms is important to develop novel strategies for overcoming acquired resistance to EGFR-TKIs. In the present study, we established novel erlotinib-resistant NSCLC cells with exon 19 deletion of EGFR, and investigated their acquired resistance mechanisms. 2. Materials and methods 2.1. Cell culture and reagents The human NSCLC cell line HCC827 harboring E746-A750 deletion in exon 19 of EGFR was purchased from the American Type Culture Collection (ATCC, Manassas, VA) and maintained in

0169-5002/$ – see front matter © 2013 The Authors. Published by Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.lungcan.2013.10.003

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

RPMI1640 (Sigma-Aldrich Co., Ltd., St. Louis, MO) supplemented with 10% FBS (Japan Bio Serum Co., Ltd., Fukuyama, Japan) at 37 ◦ C in 5% CO2 . Erlotinib was provided by F. Hoffman-La Roche Ltd. (Basel, Switzerland) and was dissolved in DMSO. A single cell was isolated from a cell suspension under a light microscope using Picopipet (Altair, Tokyo, Japan) according to the manufacturer’s instructions and expanded for further analysis. 2.2. Cell proliferation assay Cells were seeded in 96-well plates and the following day erlotinib was added at the indicated concentrations. After 4 days, the viability was determined by crystal violet assay, as described previously [10].

45

examine the HGF mRNA expression level using the LightCycler system and Applied Biosystems Assay-on-Demand primer probe sets, Mm00441908m1 (Life Technologies). 2.9. Fluorescence in situ hybridization Fluorescence in situ hybridization (FISH) for EGFR and the centromere of chromosome 7 was performed using the VysisTM LSI EGFR SpectrumOrange/CEP7 SpectrumGreen Probe (Abott, Princeton, NJ) according to the manufacturer’s instructions. This LSI EGFR probe detects wild-type as well as E746-A750 mutations of EGFR. To determine the ratio of EGFR-unamplified cells to total cells, 1000 cells were analyzed and counted. Data are shown as the mean of triplicate replications.

2.3. Immunoblotting 2.10. G-banded chromosome 7 analysis Immunoblotting was performed as described previously [11]. Briefly, cells were lysed in lysis buffer, and the 20 ␮g protein lysates were separated on a 7.5% SDS-PAGE gel and then transferred onto the membrane. Antibodies against EGFR, phospho-EGFR (Y1068), ERK, phospho-ERK, AKT, phospho-AKT (S473) (Cell Signaling Technology, Inc., Danvers, MA) and ␤-actin (Sigma–Aldrich) were used. 2.4. Generation of erlotinib-resistant cells The 3 × 102 cells/well were seeded in 96-well plates, and were cultured in the presence of 0.1, 1, or 10 ␮M erlotinib for 3 months. The resistant cells in each well were isolated and maintained in culture medium supplemented with the corresponding concentration of erlotinib. 2.5. Copy number analysis Genomic DNA was obtained from the cells using a DNeasy Blood&Tissue kit (QIAGEN, Valencia, CA). Copy numbers of EGFR and MET were determined using quantitative real-time PCR analysis with a LightCycler 480 System (Roche Diagnostics, Ltd., Basel, Switzerland) and LightCycler 480 SYBR Green I Master (Roche Diagnostics) in accordance with the manufacturer’s instructions and normalized with ˇ-globin. Human genomic DNA (Promega, Madison, WI) was used as diploid control DNA. 2.6. Direct sequencing analysis PCR primers and sequencing primers for exon 19 and exon 20 of EGFR are listed in Supplementary Table 1. The PCR products were sequenced directly using a BigDye Terminator v3.1 Cycle Sequencing kit (Life Technologies Co., Ltd., Carlsbad, CA) with an ABI PRISM 3100 genetic analyzer according to the manufacturer’s instructions. 2.7. Melting curve analysis Melting curve analysis was performed as described previously [12]. In brief, to analyze the E746-A750 mutation status, exon 19 of EGFR was amplified by PCR from DNA using the appropriate primers and the LightCycler 480 Genotyping Master (Roche Diagnostics), and then hybridized using sensor and anchor probes. The primers and probes are listed in Supplementary Table 1. Plasmids containing wild-type or E746-A750 mutation sequences were used as standard DNA. 2.8. HGF expression analysis cDNA was synthesized by using a CellAmp Direct RNA Prep Kit (TAKARA, Shiga, Japan). Real-time RT-PCR was performed to

Karyotypes of chromosome 7 were analyzed using Giemsa staining of metaphase spreads using standard methods [13]. Chromosomal identification and karyotypic designation were performed in accordance with ISCN 2009 guidelines [14]. 3. Results 3.1. Exposure to a low concentration of erlotinib generates erlotinib-resistant HCC827 cells Erlotinib inhibited HCC827 cell proliferation in a dosedependent manner, with the IC50 value of 0.0071 ␮M (Fig. 1A). Erlotinib markedly blocked not only EGFR phosphorylation, but also ERK and AKT phosphorylation; the downstream kinases of EGFR signaling cascades (Fig. 1B). To examine the effect of erlotinib concentrations on the acquisition of resistance, HCC827 cells were exposed to 0.1, 1, or 10 ␮M of erlotinib for 3 months in 96-well plates, as described in the Materials and Methods section. Erlotinibresistant cells were generated in 14 out of 96 wells by exposure to 0.1 ␮M erlotinib and in 3 out of 96 wells by exposure to 1 ␮M erlotinib (Supplementary Fig. 1A and Table 1). The IC50 values of the resistant cells were 47- to 1209-fold higher than that of the parent cells. In addition, the induction of apoptosis by erlotinib was markedly decreased compared with that of the parent cells (Supplementary Fig. 1B). No resistant cells appeared in wells exposed to 10 ␮M erlotinib. Next, we investigated the mechanisms by which the parent cells acquired resistance to erlotinib (Table 1). No secondary mutation of T790M in exon 20 of EGFR or no expression of HGF mRNA was detected in any of the erlotinib-resistant cells. An approximately 3-fold amplification of MET was detected in E10 resistant cells compared with the parent cells. In addition, we found that the parent cells had 82 copies of EGFR, whereas 13 out of the 17 resistant cells had less than 10 copies of EGFR. 3.2. EGFR-unamplified clone 4D8 isolated from parent cells in normal cell culture medium without erlotinib is resistant to erlotinib We found that HCC827 parent cells were classified into two populations: 97.5% were EGFR-amplified cells and 2.5% were EGFRunamplified cells (Fig. 2A). We next isolated the EGFR-unamplified clone 4D8 from parent cells cultured in normal medium without erlotinib by single cell cloning. The clone 4D8 had 3.3 copies of EGFR and was resistant to erlotinib (IC50 : 0.76 ␮M) (Fig. 2B and C). In addition, the levels of expression and phosphorylation of EGFR in 4D8 were markedly decreased compared with the parent cells, and

46

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

Fig. 1. Generation of erlotinib-resistant cells from HCC827 cells. (A) Parent cells were cultured in the presence of erlotinib for 4 days, and the proliferation was determined by Crystal Violet assay. Each point represents the mean + SD of triplicates. (B) Immunoblots of cell lysates from cells treated with or without erlotinib for 3 h.

the phosphorylation of AKT and ERK was not completely inhibited even by treatment with 1 ␮M of erlotinib (Figs. 1B and 2D).

3.3. Loss of an EGFR-amplified chromosome 7 (EGFR-ampch7) occurs constantly and leads to the emergence of EGFR-unamplified cells in the population of parent cells Metaphase analysis demonstrated that almost all EGFRamplified parent cells had four chromosome 7 s. Three of them contained a single copy of EGFR and the other contained multiple copies of EGFR (EGFR-ampch7) (Fig. 3A). By G-banded karyotype analysis of chromosome 7, we found that the EGFR-amplified parent cells had four different type of chromosome 7 s (n, a, b and c) and clone 4D8 had three different type of chromosome 7 s (n, b, and c) (Fig. 3B). Since the chromosome 7 s (n, b and c) other than EGFR-ampch7 (a) were shared with both parent cells and clone

4D8, it can be considered that clone 4D8 was emerged by loss of an EGFR-ampch7 in EGFR-amplified parent cells. Next, we determined whether the EGFR-unamplified cells were originally present in the parent cell population and evenly proliferated as EGFR-amplified cells, or whether these emerged constantly as part of the parent cell population under normal cell culture conditions. For this purpose, we isolated and expanded two EGFR-amplified clones, 3B4 and 4F7, from the parent cells, and found that these clones contain 2.5% and 1.0% of EGFR-unamplified cells, respectively (Fig. 3C and Supplementary Table 2). Furthermore, we isolated two EGFR-amplified clones from each of 3B4 and 4F7. These four clones again had 0.6–2.4% of EGFR-unamplified cells (Supplementary Table 2). These findings indicate that a small population of EGFR-unamplified cells emerges constantly in parent cells under normal cell culture conditions (without erlotinib) by means of the loss of an EGFR-ampch7 in EGFR-amplified cells.

Table 1 Characterization of parent cells and erlotinib-resistant cells. Cells Parent cells 0.1 ␮M erlotinib-resistant cells

1 ␮M erlotinib-resistant cells

Erlotinib IC50 (␮M)

Exon20 of EGFR sequence

HGF mRNA expression

MET copy number

Exon19 of EGFR sequence

EGFR copy number

F5 E6 A5 B10 D11 E10 G1 B4 G3 B7 G4 G9 E9 A2

0.0071 1.5 0.47 0.98 0.68 2.0 7.3 0.33 2.0 0.47 0.34 4.0 0.40 1.7 0.51

WT WT WT WT WT WT WT WT WT WT WT WT WT WT WT

N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D. N.D.

3.9 2.1 4.8 3 3.8 2.2 13 3.3 1.9 5.7 2 2.6 2.1 2.7 2.2

DEL DEL/WT DEL/WT DEL/WT DEL/WT DEL/WT DEL DEL/WT DEL/WT DEL/WT DEL/WT DEL DEL/WT DEL/WT DEL/WT

82 3.4 6.2 3.0 3.0 3.1 52 9.7 2.5 6.5 3.1 21 3.8 3.6 3.4

A10 F9 G7

1.8 8.5 1.8

WT WT WT

N.D. N.D. N.D.

4.3 3 3.2

DEL DEL DEL/WT

54 67 3.6

Cells were evaluated for erlotinib sensitivity, T790M mutational status, HGF mRNA expression, and MET and EGFR copy number. N.D.: not detected. DEL: E746-A750 deletion of exon 19, WT: wild-type. Each copy number represents the mean of triplicate assays.

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

47

Fig. 2. Characteristics of the EGFR-unamplified clone 4D8 isolated from HCC827 parent cells cultured in normal culture medium without erlotinib. (A) FISH analysis of the EGFR/CEP7 probe set. EGFR, red; CEP7, green. A subset (2.5%) of cells had unamplified copies of EGFR (EGFR-unamplified cells; arrow) in parent cells. (B) EGFR copy number analysis was performed using quantitative real-time PCR. Each bar represents the mean + SD of triplicates. (C) Proliferation of parent cells and clone 4D8 treated with erlotinib for 4 days was determined by crystal violet assay. Each point represents the mean + SD of triplicates. (D) Immunoblots of cell lysates from clone 4D8 treated with or without erlotinib for 3 h.

3.4. Erlotinib-resistant B10 and D11 cell populations were EGFR-unamplified cells The IC50 values of resistant cells B10 and D11 to erlotinib (0.68 and 2.0 ␮M, respectively) were approximately the same as that of clone 4D8 (0.76 ␮M). The level of expression and phosphorylation of EGFR in B10 cells were markedly decreased, but the phosphorylation of AKT and ERK were not completely inhibited by 1 ␮M of erlotinib (Fig. 4A) as with clone 4D8. Both of these resistant cells had three copies of EGFR, and >99.99% of their populations were classified as EGFR-unamplified because no EGFR-amplified cells were detected in more than 10,000 cells (Fig. 4B, C and Supplementary Fig. 2A and B). By direct sequencing analysis, the parent cells were shown to have only the E746-A750 deletion in exon 19, as described previously [15], whereas clone 4D8 and B10 and D11 resistant cells contained both the wild-type and the E746-A750 deletional sequences (Fig. 4D). However, by melting curve analysis, we found that approximately 2% of the parent cell population had the wildtype allele and 98% had the E746-A750 deletion allele, whereas in clone 4D8 and B10, D11 resistant cells, approximately 60% of the population had the wild-type allele and 40% had the E746-A750 deletion allele (Fig. 4E). 4. Discussion Human NSCLC HCC827 cells have an E746-A750 deletion of exon 19 and are highly sensitive to EGFR-TKIs. In this study, to

examine novel mechanisms of acquired resistance to EGFR-TKIs, erlotinib-resistant cells were established by continuously exposing HCC827 cells to 0.1, 1, or 10 ␮M of erlotinib. Since clinically applicable erlotinib doses, 25, 100, or 150 mg, lead to maximum plasma concentrations of 0.8, 1.9, and 5.6 ␮M, respectively [16,17], the exposure concentrations were selected to cover the achievable plasma concentrations of erlotinib (0.8–5.6 ␮M) in examining the relationship between concentration and resistance acquisition to erlotinib. Erlotinib inhibited the generation of resistant cells in a dose-dependent manner. Resistant cells were generated by exposure to 0.1 and 1 ␮M of erlotinib in 14/96 wells and 3/96 wells, respectively. No resistant cells appeared in wells exposed to 10 ␮M erlotinib. These results suggest that, to prevent acquired resistance to erlotinib, it is important to keep the plasma concentration as high as possible by treating patients with the highest recommended dose (150 mg) of erlotinib as far as it can be tolerated. We found that 17 resistant cells obtained were classified into three groups based on the change in MET or EGFR copy number compared with the parent cells: (1) cells having more than 3-fold increase in MET copy number, (2) cells having nearly-unchanged MET and EGFR copy numbers, (3) cells having less than a half decrease in EGFR copy number. The first group included one resistant cell (E10) having more than 3-fold increase in MET copy number. Engelman et al. reported that HCC827 cells developed resistance to gefitinib in vitro as a result of focal amplification of MET in all six clones isolated [7]. The discrepancy in the incidence of MET amplified cells between our study (1/17) and Engelman’s

48

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

Fig. 3. Loss of an EGFR-ampch7 in EGFR-amplified cells. (A) Metaphase FISH analysis of the EGFR/CEP7 probe set. EGFR-amplified cells had an EGFR-ampch7 (arrow). (B) G-banded karyotype analysis of chromosome 7. n, normal chromosome 7; a, add(7) (p11.2); b and c, add(7) (q11.2) with different additional material of unknown origin. (C) FISH analysis of the EGFR/CEP7 probe set of clone 3B4 isolated from EGFR-amplified parent cells. A subset (2.5%) of cells was EGFR-unamplified cells (arrow).

study (6/6) may be caused by the different methods for generating resistant cells. In Engelman’s study cells were exposed to stepwiseincreased concentration (0.001–0.1 ␮M) of gefitinib. In contrast, our method, exposing cells to fixed concentrations of erlotinib (0.1 or 1.0 ␮M), is considered to better mimic clinical settings because patients are constantly treated with the recommended dose of an EGFR-TKI during the therapy. The second group included 2 resistant cells (A10 and F9). The MET and EGFR copy numbers of these cells were the closest to the parent cells in the three groups. No secondary mutation of T790M, HGF mRNA over-expression, or KRAS mutations were detected in these cells (data not shown). Thus, we did not identify the resistance mechanism in this group so far. Further studies are needed to elucidate the mechanism associated with resistance. Several known mechanisms such as insulin-like growth factor I receptor (IGF1R) expression, HER2/HER3 expression, PIK3CA mutations, epithelial–mesenchymal transition (EMT), and small cell lung cancer (SCLC) transformation [8,18] may be candidates. The third group included 14 resistant cells (other than E10, A10, and F9). We proposed a model for the acquired resistance to erlotinib in this group (Fig. 5). Even in usual culture condition without erlotinib, HCC827 parent cells maintain EGFR-unamplified cells by a constant fraction. These cells were generated by the loss of an EGFR-ampch7 in EGFR-amplified cells. The levels of expression and phosphorylation of EGFR in EGFR-unamplified cells, such as clone 4D8 and resistant cell B10, were drastically decreased compared with the parent cells, whereas the downstream AKT/ERK phosphorylation was not decreased (Supplementary Fig. 3). When exposed to relatively low concentrations of erlotinib (0.1 and 1 ␮M), the resistant cells, namely, the pre-existing EGFR-unamplified cells survived and proliferated in the parent cell population (Fig. 5).

Whether this phenomenon can be found in other cell lines is of interest. We found that EGFR exon 19 deleted NSCLC cell line B901L has two EGFR-ampch7 and has pre-existing EGFR-unamplified cells (about 0.2%) under normal culture conditions (Supplementary Fig. 4). Although the mechanism associated with the loss of an EGFR-ampch7 with exon 19 deletion in EGFR-amplified cells under normal culture conditions is unclear, the mutation of EGFR and multiple centromeres in EGFR-ampch7 may cause genetic instability. Copy number gains and mutant allele-specific imbalances such as amplification, polysomy, or uniparental disomy, occur frequently in tumor cells with EGFR mutations [19]. In fission yeast, abnormal centromere function results in a highly elevated rate of chromosome loss and chromosome missegregation [20]. Furthermore, the proportion of EGFR-unamplified cells in the parent cell population was unchanged for 9 months under normal cell culture conditions (2.5% at the start and 2.1% after 9 months; data not shown). These findings indicate that the abnormality of the EGFR-ampch7 may lead to uneven distribution of the chromosome during mitosis not frequently but constantly. Although we did not identify the novel addicted oncogene in EGFR-unamplified resistant cells (4D8, B10 or D11), wild-type EGFR may be a candidate because the proliferation of these cells was still inhibited by more than approximately 1 ␮M of erlotinib (Supplementary Fig. 5A). In addition, the erlotinib of corresponding concentration completely blocks the phosphorylation of wild-type EGFR [21]. Furthermore, the IC50 value of irreversible EGFR-TKI, afatinib, to 4D8 and D11 cells was approximately 25-fold higher than that of the parent cells (Supplementary Fig. 5B). In addition, EGFR knockdown by siRNA partially but significantly inhibited cell proliferation in all of three resistant cells (Supplementary Fig. 5 C). These results indicate that EGFR-unamplified resistant cells could

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

49

Fig. 4. Characteristics of the erlotinib-resistant B10 and D11 cell populations. (A) Immunoblots of cell lysates from erlotinib-resistant B10 cells treated with or without erlotinib for 3 h. (B and C) FISH analysis of the EGFR/CEP7 probe set of control (B) and metaphase spreads (C). EGFR, red; CEP7, green. (D) Direct sequencing analysis of EGFR exon 19. The underlined sequences of the 4D8 clone, and the B10 and D11 cells contained both the E746-A750 deletion and the wild-type alleles. (E) Melting curve analysis of standard deletion/wild-type plasmids and DNA from cells. Left peaks indicate the wild-type EGFR (arrow). The wild-type peaks of the parent cells and the 4D8 clone and B10 and D11 cells were nearly identical to those of the 2% and 60% wild-type/E746-A750 deletion plasmids, respectively.

Parent cells

Without EGFR-TKIs

EGFR-TKIs resistant cells

Low concentration of EGFR-TKIs

Amplified EGFR exon 19 deletion EGFR exon 19 deletion

Not acquired resistant cells

EGFR wild-type

Fig. 5. Model for acquired resistance to EGFR-TKIs in EGFR-mutated HCC827 cells. A small population of EGFR-unamplified cells is constantly maintained in the parent cell population by the loss of an EGFR-ampch7 with exon 19 deletion in EGFR-amplified cells under normal cell culture conditions in HCC827 cells (without EGFR-TKIs). Resistant cells are generated by pre-existing EGFR-unamplified cells being selected at low concentrations of EGFR-TKIs. High concentrations of EGFR-TKIs completely inhibit the proliferation of not only EGFR-amplified cells, but also EGFR-unamplified cells. EGFR-amplified cells have three types of chromosome 7 s; approximately 80 copies of exon 19 deletion EGFR (EGFR-ampch7), single-copy of exon 19 deletion EGFR and wild-type EGFR. EGFR-unamplified cells have two types of chromosome 7 s; single-copy of exon 19 deletion EGFR and wild-type EGFR.

50

K. Furugaki et al. / Lung Cancer 83 (2014) 44–50

favorably change the addiction from delE746-A750 EGFR to the other growth drivers including wild-type EGFR even in the presence of one or two copies of delE746-A750 EGFR. HER family receptor kinases may be another candidates. Tabara et al. have shown that complete loss of the activating EGFR mutant gene results in the gain of a novel addiction to HER2/HER3 signaling and the acquisition of EGFR-TKI resistance in vitro [22]. In our resistant cells (4D8 and B10), cell proliferation was partially blocked by HER2 or HER3 knockdown (Supplementary Fig. 6). These findings indicate that the EGFR-unamplified resistant cells partially depend on not only EGFR but also HER2/HER3 signaling for survival. Compared with other solid tumors, NSCLC is well known for the heterogeneity of the cell populations in individual lesions [23]. Heterogeneous distribution of EGFR mutations in individual tumors has also been reported [24–26]. In addition, loss of an EGFR mutation is reported in 3 out of 11 EGFR-mutated NSCLC patients with progressive disease after gefitinib treatment [22]. These findings indicate that some NSCLCs are genetically heterogeneous and concurrently have tumor cell populations with either mutant or wild-type EGFR, and that the EGFR genetic heterogeneity might contribute to acquired resistance to EGFR-TKIs. Our results strongly support this mechanism of resistance, because we have clearly shown that the genetic heterogeneity of EGFR is constantly maintained by the loss of an EGFR-ampch7 in NSCLC cells with EGFR mutations. In conclusion, we demonstrated that loss of amplified EGFRmutated genes causes acquired resistance in HCC827 cells when the cells are exposed to a relatively low concentration of erlotinib, whereas high concentration of erlotinib prevents the emergence of resistance. In addition to the major known mechanisms of acquired resistance to EGFR-TKIs, including secondary mutation of T790M, amplification of MET, mutations of PIK3CA, EMT, and transformation to SCLC [8], our findings propose a novel acquired resistant mechanism, namely, the selection of preexisting EGFR-unamplified cells, which are generated by the loss of an amplified EGFR-mutated gene, may contribute to the acquired resistance to EGFR-TKIs. Further studies are needed to identify alternative addictive signal pathway(s) after the loss of amplified EGFR with mutation and to lead to the development of a novel molecular targeted therapy against EGFR-TKI-refractory NSCLC. Conflict of interest statement None. Acknowledgements The authors thank Kumiko Kondoh, Hiromi Sawamura and Masako Takahashi for technical assistance in the experiments, and also thank Kazushige Mori, Naohito Inagaki, Masamichi Sugimoto and Keiji Kosaka for support and special advice in this study. Appendix A. Supplementary data Supplementary material related to this article can be found, in the online version, at http://dx.doi.org/10.1016/j.lungcan. 2013.10.003. References [1] Mitsudomi T. Advances in target therapy for lung cancer. Japanese Journal of Clinical Oncology 2010;40:101–6.

[2] Mitsudomi T, Yatabe Y. Mutations of the epidermal growth factor receptor gene and related genes as determinants of epidermal growth factor receptor tyrosine kinase inhibitors sensitivity in lung cancer. Cancer Science 2007;98:1817–24. [3] Pao W, Miller VA. Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: current knowledge and future directions. Journal of Clinical Oncology 2005;23:2556–68. [4] 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 Medicine 2005;2:e73. [5] Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF, et al. Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clinical Cancer Research 2006;12:6494–501. [6] Kosaka T, Yatabe Y, Endoh H, Yoshida K, Hida T, Tsuboi M, et al. Analysis of epidermal growth factor receptor gene mutation in patients with non small cell lung cancer and acquired resistance to gefitinib. Clinical Cancer Research 2006;12:5764–9. [7] 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. [8] 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. Science Translational Medicine 2011;3:75ra26. [9] Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura T, et al. Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor activating mutations. Cancer Research 2008;68:9479–87. [10] Yamashita-Kashima Y, Iijima S, Yorozu K, Furugaki K, Kurasawa M, Ohta M, et al. Pertuzumab in combination with trastuzumab shows significantly enhanced antitumor activity in HER2-positive human gastric cancer xenograft models. Clinical Cancer Research 2011;17:5060–70. [11] Furugaki K, Iwai T, Kondoh K, Moriya Y, Mori K. Antitumor activity of erlotinib in combination with gemcitabine in in vitro and in vivo models of KRAS-mutated pancreatic cancers. Oncology Letters 2010;1:231–5. [12] Tsiatis AC, Norris-Kirby A, Rich RG, Hafez MJ, Gocke CD, Eshleman JR, et al. Comparison of Sanger sequencing, pyrosequencing, and melting curve analysis for the detection of KRAS mutations: diagnostic and clinical implications. Journal of Molecular Diagnostics 2010;12:425–32. [13] Seabright M. A rapid banding technique for human chromosomes. Lancet 1971;2:971–2. [14] Brothman AR, Persons DL, Shaffer LG. Nomenclature evolution: changes in the ISCN from the 2005 to the 2009 edition. Cytogenetic and Genome Research 2009;127:1–4. [15] Kubo T, Yamamoto H, Lockwood WW, Valencia I, Soh J, Peyton M, et al. MET gene amplification or EGFR mutation activate MET in lung cancers untreated with EGFR tyrosine kinase inhibitors. International Journal of Cancer 2009;124:1778–84. [16] Costa DB, Kobayashi S, Yeo W-L, Hamada A. Serum concentrations of erlotinib at a dose of 25 mg daily. Journal of Thoracic Oncology 2010;5:1311–2. [17] Yamamoto N, Horiike A, Fujisaka Y, Murakami H, Shimoyama T, Yamada Y, et al. Phase I dose-finding and pharmacokinetic study of the oral epidermal growth factor receptor tyrosine kinase inhibitor Ro50-8231 (erlotinib) in Japanese patients with solid tumors. Cancer Chemotherapy and Pharmacology 2008;61:489–96. [18] Wheeler DL, Dunn EF, Harari PM. Understanding resistance to EGFR inhibitorsimpact on future treatment strategies. Nature Reviews Clinical Oncology 2010;7:493–507. [19] Soh J, Okumura N, Lockwood WW, Yamamoto H, Shigematsu H, Zhang W, et al. Oncogene mutations, copy number gains and mutant allele specific imbalance (MASI) frequently occur together in tumor cells. PLoS ONE 2009;4:e7464. [20] Ekwall K, Olsson T, Turner BM, Cranston G, Allshire RC. Transient inhibition of histone deacetylation alters the structural and functional imprint at fission yeast centromeres. Cell 1997;91:1021–32. [21] Carey KD, Garton AJ, Romero MS, Kahler J, Thomson S, Ross S, et al. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer Research 2006;66:8163–71. [22] Tabara K, Kanda R, Sonoda K, Kubo T, Murakami Y, Kawahara A, et al. Loss of activating EGFR mutant gene contributes to acquired resistance to EGFR tyrosine kinase inhibitors in lung cancer cells. PLoS ONE 2012;7:e41017. [23] Dunnill MS, Gatter KC. Cellular heterogeneity in lung cancer. Histopathology 1986;10:461–75. [24] Taniguchi K, Okami J, Kodama K, Higashiyama M, Kato K. Intratumor heterogeneity of epidermal growth factor receptor mutations in lung cancer and its correlation to the response to gefitinib. Cancer Science 2008;99:929–35. [25] Sakurada A, Lara-Guerra H, Liu N, Shepherd FA, Tsao MS. Tissue heterogeneity of EGFR mutation in lung adenocarcinoma. Journal of Thoracic Oncology 2008;3:527–9. [26] Nakano H, Soda H, Takasu M, Tomonaga N, Yamaguchi H, Nakatomi K, et al. Heterogeneity of epidermal growth factor receptor mutations within a mixed adenocarcinoma lung nodule. Lung Cancer 2008;60:136–40.

Loss of an EGFR-amplified chromosome 7 as a novel mechanism of acquired resistance to EGFR-TKIs in EGFR-mutated NSCLC cells.

Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) show notable effects against non-small cell lung cancers (NSCLCs) harboring EG...
2MB Sizes 0 Downloads 0 Views