Overcoming cellular mechanisms of de novo and acquired resistance to drug therapy remains a central challenge in the clinical management of many cancers, including non–small cell lung cancer (NSCLC). Although much work has linked the epithelial–mesenchymal transition (EMT) in cancer cells to the emergence of drug resistance, it is less clear where tractable routes may exist to reverse or inhibit EMT as a strategy for drug sensitization. Here, we demonstrate that extracellular signal-regulated kinase (ERK) 1/2 (mitogen-activated protein kinase 3/1, MAPK3/1) signaling plays a key role in directing the mesenchymal character of NSCLC cells and that blocking ERK signaling is sufficient to heighten therapeutic responses to EGF receptor (EGFR) inhibitors. MEK1/2 (MAPKK1/2) inhibition promoted an epithelial phenotype in NSCLC cells, preventing induction of EMT by exogenous TGF-β. Moreover, in cells exhibiting de novo or acquired resistance to the EGFR inhibitor gefitinib, MEK inhibition enhanced the sensitivity to gefitinib and slowed cell migration. These effects only occurred, however, if MEK was inhibited for a period sufficient to trigger changes in EMT marker expression. Consistent with these findings, changes in EMT phenotypes and markers were also induced by the expression of mutant KRAS in a MEK-dependent manner. Our results suggest that prolonged exposure to MEK or ERK inhibitors may not only restrain EMT but also overcome naïve or acquired resistance of NSCLC to EGFR-targeted therapy in the clinic. Cancer Res; 74(1); 309–19. ©2013 AACR.

EGF receptor (EGFR) overexpression and activation are hallmarks of many cancers, including non–small cell lung cancer (NSCLC). Consequently, a number of inhibitors and monoclonal antibodies targeting EGFR have been developed and approved for various cancers. Unfortunately, these drugs are generally ineffective. In NSCLC, response to EGFR inhibitors is limited mainly to the rare patients (∼10%) whose tumors harbor somatic, kinase-activated mutants of EGFR (1, 2). Even these patients almost invariably develop resistance to EGFR inhibitors, often through the EGFR “gatekeeper” mutation (T790M; refs. 3, 4) or through upregulation of the HGF receptor c-MET or other receptors (5). Combination therapies present a possible strategy to overcome resistance. In NSCLC, recent investigations suggest promise for combining EGFR inhibitors with chemoradiation (6), the multikinase inhibitor sorafenib (7), or a c-MET inhibitor (8). Scheduling multiple drugs such that initial therapy reprograms cells to respond to another drug is another possible strategy. In one recent example, triple-negative breast cancer cells and NSCLC cells were dramatically sensitized to doxorubicin by pretreatment with the EGFR inhibitor erlotinib (9).

Epithelial–mesenchymal transition (EMT) is another pathway through which cancers of epithelial origin become chemoresistant. EMT is a developmental process whereby epithelial cells lose cell–cell adhesions to become more motile and invasive. Cells undergoing EMT lose expression of epithelial markers (e.g., E-cadherin) and gain expression of mesenchymal markers (e.g., vimentin and fibronectin) through differential expression and activation of transcription factors including Twist, ZEB1, and Snail (10, 11). EMT is frequently hijacked in metastatic progression, and mesenchymal dedifferentiation has been associated with resistance to EGFR inhibitors, chemotherapy, and other targeted drugs in cancers of the lung (12–14), bladder (15), head and neck (16, 17), pancreas (18), and breast (19). In NSCLC, in vitro acquired resistance to the EGFR inhibitor erlotinib can result from the selection of a mesenchymal subpopulation (20), and restoring E-cadherin expression in mesenchymal-like NSCLC cells potentiates sensitivity to EGFR inhibitors (21). In addition, growing evidence for AXL-mediated EGFR inhibitor resistance has been tied to EMT (22). Thus, developing treatments that elicit a mesenchymal-epithelial transition (MET) could be a useful approach for expanding the efficacy of EGFR inhibitors.

Several studies have demonstrated a requirement for extracellular signal-regulated kinase (ERK)1/2 or mitogen-activated protein kinase (MAPK) 3/1 pathway activity in EMT induced by TGF-β in nontransformed cells (23–25). ERK2, but not ERK1, activity also induces EMT in nontransformed mammary epithelial cells (26) and has been implicated as mediating oncogenic KRAS-induced invasion in pancreatic cancer cells (27). Interestingly, ERK2 amplification was recently identified as a mechanism leading to acquired resistance to EGFR inhibitors in NSCLC (28).

Here, we sought to determine ERK's role in governing EMT in NSCLC. In a panel of NSCLC cell lines, inhibition of MEK1/2 (MAPKK1/2) prevented TGF-β–induced EMT and promoted epithelial cellular characteristics when administered alone. Conversely, augmented ERK activation, through KRAS12V expression or ERK2 amplification, promoted mesenchymal characteristics. Furthermore, chronic MEK inhibition for times long enough to observe changes in epithelial and mesenchymal marker expression augmented cellular sensitivity to the EGFR inhibitor gefitinib in cell lines with de novo or acquired resistance to EGFR inhibitors. These changes were reversible and accompanied by shifts in expression of stem cell-like markers CD24 and CD44. These results suggest the potential utility of drug scheduling strategies first targeting ERK to promote epithelial characteristics before targeting EGFR or other oncogenic signaling nodes.

Cell culture

H1666 cells were obtained from the American Type Culture Collection. H322, gefitinib-resistant PC9 (clone GR4, referred to as GR henceforth), and WZ4002-resistant PC9 cells (clone WZR12, referred to as WZR henceforth) were provided by Dr. Pasi Jänne (Dana–Farber Cancer Institute, Boston, MA). Parental PC9 cells were provided by Dr. Douglas Lauffenburger (Massachusetts Institute of Technology, Cambridge, MA). Because PC9 cells came from different labs, we confirmed similar expression of important proteins and response to gefitinib for parental stocks from both labs. H358 cells were provided by Dr. Russ Carstens (University of Pennsylvania, Philadelphia, PA). PC9 (all variants), H322, and H358 cells were maintained in RPMI-1640 supplemented with 10% FBS, 100 U/mL penicillin, 100 μg/mL streptomycin, and 1 mmol/L L-glutamine. H1666 cells were maintained in ACL4 (29). Cell culture reagents were from Life Technologies. Cell lines were validated for anticipated responsiveness to gefitinib and were cultured for less than 2 months from low-passage frozen stocks.

Chronic MEK inhibition

H1666, PC9 GR, and PC9 WZR cells were maintained in 10, 5, and 20 μmol/L U0126, respectively, with controls maintained in dimethyl sulfoxide (DMSO). Responses to 1-hour or 2-day treatments with a range of U0126 concentrations were evaluated for each cell line to identify U0126 concentrations yielding significantly inhibited ERK phosphorylation (Supplementary Fig. S1) but minimal background cell death. For culture experiments, media were changed every 2 days or cells were passed as necessary. When passing, cells were subcultured into inhibitor-free media to promote adhesion. U0126 or DMSO was replaced the following day. When appropriate, cells were lysed or gefitinib was added 24 hours later. Time points in figures with chronic MEK inhibition reflect the total time of U0126 exposure before lysing or gefitinib addition. To probe the reversibility of U0126 effects, cells were split from U0126 cultures after 7 days and maintained in DMSO thereafter. All time courses were performed from freshly thawed cells multiple times to demonstrate reproducibility. Naïve cell death response to U0126 and gefitinib was quantified at multiple points during time courses, including the beginning and end, to verify that the baseline response was not changing.

Wound-healing assay

Confluent cell monolayers in 6-well plates were scratched with a pipet tip, and media were immediately changed. Phase contrast images were taken with a Zeiss Axiovert 40 CFL microscope (×10 objective) every 1 to 3 hours for 11 hours or less. Scratch areas were quantified using ImageJ, and closure rates were calculated from linear fits of areas versus time and reported as percentage of total image area closed per hour normalized to the conditions indicated in figures. Where inhibitors were used, cells were plated at subconfluence and treated at appropriate times with media containing inhibitor. Media and inhibitor were changed every 2 days until wells reached confluence.

Transwell migration assay

Untreated PC9 GR and WZR cells or H1666 cells treated with U0126 or DMSO for 4 days were trypsinized and resuspended in media containing 0.1% FBS and U0126 or DMSO for H1666. Of note, 20,000 untreated or DMSO-treated cells or 50,000 U0126-treated cells (adjusted for U0126-mediated changes in adhesion) were added to 8 μm Transwell membranes (Corning), which were placed in 24-well plate wells containing complete media with DMSO or U0126. After 20 hours, cells on the upper surfaces were removed with a cotton swab. Cells on the lower surfaces were fixed in 4% paraformaldehyde for 30 minutes and washed with PBS, and nuclei were stained with Hoescht-33342. Membranes were mounted and imaged (×10 objective), and nuclei were counted using ImageJ. Counts were normalized to the number of adhered cells from parallel wells for each condition.

Flow cytometry

For cell death assays, cells were plated in 6-well dishes from their various culture conditions and treated with inhibitors. Floating and adherent cells were collected 48 hours later, resuspended in PBS containing TO-PRO3 (Life Technologies), and analyzed within 1 hour. For CD24 and CD44 measurements, cells were collected as above, washed with 0.1% bovine serum albumin in PBS (PBSA), blocked for 10 minutes in PBSA, and incubated for 1 hour with 3 μL each of fluorescein isothiocyanate–conjugated antihuman CD44 antibody (BD Pharmingen, #555478) and Alexa-647 conjugated antihuman CD24 antibody (BioLegend, #311109) in 200 μL. Labeled cells were washed again and resuspended in PBS. Cytometry was performed on a BD Biosciences FACSCalibur cytometer, and data were analyzed using FlowJo.

Antibodies and other reagents

pERK T202/Y204 (#4377) and ERK (#4695) antibodies were from Cell Signaling Technology. E-cadherin (sc-8426), vimentin (sc-373717), fibronectin (sc-8422), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; sc-32233) antibodies were from Santa Cruz Biotechnology. Stocks of U0126 and gefitinib (LC Labs) were prepared in DMSO. Recombinant human EGF and TGF-β were from Peprotech. Infrared dye- and Alexa Fluor-conjugated secondary antibodies were from Rockland Immunochemicals and Invitrogen, respectively.

Additional methods

Western blotting, immunofluorescence, KRAS12V expression, experiments involving CI-1040, chronic MEK inhibition in H322, H358, and SKRB3 cells, and the EGFR internalization assay were performed as described in Supplementary Materials and Methods.

NSCLC cell lines undergo MEK-dependent TGF-β–induced EMT

H1666, H322, and H358 cells were cultured in complete media with TGF-β and EGF with or without U0126 for 4 days, with a media change after 2 days (Fig. 1). In response to TGF-β and EGF, E-cadherin expression decreased and vimentin expression increased in all three cell lines, and fibronectin expression increased in H1666 and H358 cells (Fig. 1A). U0126 cotreatment inhibited changes in E-cadherin and vimentin, but did not prevent fibronectin induction. The same trends were found in H1666 cells using the alternative MEK inhibitor CI-1040 (Supplementary Fig. S2). The conditions used in Fig. 1A were explored in H1666 cells by immunofluorescence (Fig. 1B). Treatment with TGF-β and EGF promoted an elongated cellular morphology and the appearance of F-actin fibers, and decreased the E-cadherin intensity. U0126 addition prevented E-cadherin loss and promoted E-cadherin localization at cell–cell junctions.

Figure 1.

MEK inhibition prevents TGF-β–induced EMT in NSCLC cell lines. A, H1666, H322, and H358 cells were treated for 4 days with 10 ng/mL TGF-β + 50 ng/mL EGF, TGF-β + EGF and 20 μmol/L U0126, or DMSO (control). Whole-cell lysates were analyzed by Western blot analysis with antibodies against indicated proteins. Images are representative of at least three independent experiments. B, H1666 cells on glass coverslips were treated as in A, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates from each of two independent experiments. Scale bar, 20 μm.

Figure 1.

MEK inhibition prevents TGF-β–induced EMT in NSCLC cell lines. A, H1666, H322, and H358 cells were treated for 4 days with 10 ng/mL TGF-β + 50 ng/mL EGF, TGF-β + EGF and 20 μmol/L U0126, or DMSO (control). Whole-cell lysates were analyzed by Western blot analysis with antibodies against indicated proteins. Images are representative of at least three independent experiments. B, H1666 cells on glass coverslips were treated as in A, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates from each of two independent experiments. Scale bar, 20 μm.

Close modal

MEK inhibition promotes epithelial phenotypes in NSCLC cells

We next explored the ability of MEK inhibition alone to promote epithelial characteristics in H1666 cells. As seen when combined with TGF-β and EGF, U0126 promoted E-cadherin localization at cell–cell junctions (Fig. 2A). MEK inhibition also antagonized H1666 wound healing in a treatment time-dependent manner (Fig. 2B and Supplementary Fig. S3). Although U0126 inhibited ERK phosphorylation within 1 hour of addition (Supplementary Fig. S1), U0126 treatment for the duration of the scratch assay (10 hours) did not significantly affect the wound-closure rate (“at scratch” condition vs. DMSO in Fig. 2B). The wound-closure rate decreased significantly with 1 day of U0126 pretreatment and was nearly zero with 4 days of pretreatment. Western blot analyses of lysates created in parallel revealed that E-cadherin expression increased and vimentin expression decreased with increasing the U0126 exposure time, consistent with the time-dependent changes in the wound-closure rate (Fig. 2C and Supplementary Fig. S3). U0126-mediated changes in wound-healing migration were further confirmed by assessing migration across Transwell inserts (Fig. 2D).

Figure 2.

MEK inhibition promotes epithelial characteristics in H1666 cells. A, H1666 cells plated on glass coverslips were treated with 20 μmol/L U0126 for 4 days, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates from each of two independent experiments. Scale bar, 20 μm. B, wound-closure rates were measured for H1666 cells treated with DMSO (control) or 20 μmol/L U0126 at the time of scratch or for 1 or 4 days before scratch. Rates normalized to the control condition for triplicate wells from two different platings (n = 6) are reported as averages ± SEM. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. C, lysates prepared in parallel to B were analyzed by Western blot analysis using antibodies against indicated proteins. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3). D, Transwell migration experiments were performed for H1666 cells pretreated with DMSO or 20 μmol/L U0126 for 4 days. Counts normalized to DMSO control for duplicate wells from three separate experiments (n = 6) are reported as averages ± SEM. * and **, P < 0.05 and P < 0.005, respectively.

Figure 2.

MEK inhibition promotes epithelial characteristics in H1666 cells. A, H1666 cells plated on glass coverslips were treated with 20 μmol/L U0126 for 4 days, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates from each of two independent experiments. Scale bar, 20 μm. B, wound-closure rates were measured for H1666 cells treated with DMSO (control) or 20 μmol/L U0126 at the time of scratch or for 1 or 4 days before scratch. Rates normalized to the control condition for triplicate wells from two different platings (n = 6) are reported as averages ± SEM. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. C, lysates prepared in parallel to B were analyzed by Western blot analysis using antibodies against indicated proteins. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3). D, Transwell migration experiments were performed for H1666 cells pretreated with DMSO or 20 μmol/L U0126 for 4 days. Counts normalized to DMSO control for duplicate wells from three separate experiments (n = 6) are reported as averages ± SEM. * and **, P < 0.05 and P < 0.005, respectively.

Close modal

Chronic MEK inhibition sensitizes NSCLC cells to EGFR inhibition on a time scale consistent with changes in epithelial and mesenchymal markers

Because epithelial characteristics have been connected to EGFR dependence in NSCLC (14, 21), we explored the ability of MEK inhibition to increase sensitivity to gefitinib (Fig. 3). As described in Materials and Methods, H1666 cells were maintained in 10 μmol/L U0126 or DMSO (control) for up to 3 weeks. Throughout the 3-week period, cells were evaluated for death response to gefitinib. Without U0126 pretreatment, 2 days of EGFR and MEK coinhibition led to only 7% cell death in H1666, but this more than doubled with 3 days of U0126 pretreatment (Fig. 3A and Supplementary Fig. S4A). This enhancement further increased with additional U0126 pretreatment time, reaching a maximum at 11 days. Increased cellular sensitivity to gefitinib was accompanied by increased E-cadherin expression (Fig. 3B and Supplementary Fig. S4B). Beyond 11 days, the synergistic effect began to decrease, which was accompanied by increased fibronectin expression and decreased E-cadherin expression. Similar effects were observed in H1666 cells using CI-1040 (Supplementary Fig. S4C).

Figure 3.

Chronic MEK inhibition sensitizes NSCLC cells to EGFR inhibition. A, H1666 cells were cultured in 10 μmol/L U0126 or DMSO (control) for up to 3 weeks and were evaluated for cell death response to 10 μmol/L gefitinib for various U0126 exposure times. Cell death was measured by flow cytometry for TO-PRO3 permeability 48 hours after gefitinib addition. Averaged data are shown ± SEM (n = 3); significance is relative to any other condition from the same day. B, lysates prepared in parallel to A, before gefitinib addition, were analyzed by Western blot analysis with antibodies against the indicated proteins. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3). C, experiments probing the reversibility of U0126-mediated effects were performed as described in Materials and Methods. For cells removed from (U0126 removal) or maintained in U0126, response to the same drug cotreatment was assessed by TO-PRO3 permeability at the indicated times. Data are represented as averages ± SEM (n = 3). D and E, reversibility of U0126 effects was assessed by Western blot analysis (D), using antibodies against indicated proteins, and by flow cytometry for CD44 and CD24 staining (E). Blot images are representative of three replicates. Flow cytometry data are represented as averages ± SEM (n = 3). In all panels, times indicated reflect the total time of U0126 exposure before lysis, gefitinib addition, or staining; * and **, P < 0.05 and P < 0.005, respectively.

Figure 3.

Chronic MEK inhibition sensitizes NSCLC cells to EGFR inhibition. A, H1666 cells were cultured in 10 μmol/L U0126 or DMSO (control) for up to 3 weeks and were evaluated for cell death response to 10 μmol/L gefitinib for various U0126 exposure times. Cell death was measured by flow cytometry for TO-PRO3 permeability 48 hours after gefitinib addition. Averaged data are shown ± SEM (n = 3); significance is relative to any other condition from the same day. B, lysates prepared in parallel to A, before gefitinib addition, were analyzed by Western blot analysis with antibodies against the indicated proteins. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3). C, experiments probing the reversibility of U0126-mediated effects were performed as described in Materials and Methods. For cells removed from (U0126 removal) or maintained in U0126, response to the same drug cotreatment was assessed by TO-PRO3 permeability at the indicated times. Data are represented as averages ± SEM (n = 3). D and E, reversibility of U0126 effects was assessed by Western blot analysis (D), using antibodies against indicated proteins, and by flow cytometry for CD44 and CD24 staining (E). Blot images are representative of three replicates. Flow cytometry data are represented as averages ± SEM (n = 3). In all panels, times indicated reflect the total time of U0126 exposure before lysis, gefitinib addition, or staining; * and **, P < 0.05 and P < 0.005, respectively.

Close modal

H358 and H322 cells and the breast cancer cell line SKBR3 were also evaluated for response to gefitinib following chronic MEK inhibition (Supplementary Fig. S5). H358 cells harbor a KRAS mutation and are ERK addicted (30). Not surprisingly, therefore, H358 response to gefitinib was not augmented by U0126 pretreatment. However, H322 and SKBR3 cells were sensitized to EGFR inhibition with 5 days of U0126 exposure, suggesting fairly broad applicability of this strategy.

We also probed the reversibility of the effects of chronic MEK inhibition in H1666 cells (Fig. 3C–E and Supplementary Fig. S6). After 7 days, a fraction of cells was split from the U0126 culture and maintained in media containing DMSO (control). This “U0126 removal” culture was subsequently plated in parallel with the cells maintained in U0126. After 7 days of U0126 exposure, removal from U0126 for 4 days completely reversed changes in cellular sensitivity to gefitinib and epithelial and mesenchymal marker expression (Fig. 3C and D and Supplementary Fig. S6A).

Stem cell-like subpopulations with intrinsic therapeutic resistance are found within tumors (20, 31) and are typically identified as CD44high/CD24low (20) or E-cadherinlow (32). CD44high/CD24low enrichment from heterogeneous cultures promotes mesenchymal behaviors including resistance to EGFR inhibitors (20). Using flow cytometry, we measured the shift in the CD44high/CD24low subpopulation with U0126 addition and subsequent removal (Fig. 3E and Supplementary Fig. S6B). U0126-treated cells had fewer CD44high/CD24low cells and more CD44low/CD24high cells than controls. As with epithelial/mesenchymal marker expression, U0126 removal for 4 days reversed the effect on the CD44low/CD24high population.

KRAS12V-mediated ERK activation promotes EMT

To further probe the connection between ERK activity and EMT, we expressed KRAS12V in H1666 cells. This produced an anticipated increase in ERK phosphorylation and increased vimentin expression (Fig. 4A). Gefitinib and U0126 promoted an epithelial shift in control cells, as determined by Western blot analysis and wound-closure measurements (Fig. 4A and B). KRAS12V promoted maintenance of ERK phosphorylation in response to gefitinib and largely prevented gefitinib-mediated changes in marker expression and wound closure (Fig. 4A and B). Importantly, the effects of KRAS12V were MEK dependent, as demonstrated by the effects of U0126 (Fig. 4).

Figure 4.

KRAS12V expression promotes mesenchymal characteristics in H1666 cells. A, H1666 cells transduced with KRAS12V or an empty vector (EV) control treated for 3 days with DMSO, 2 μmol/L gefitinib, or 20 μmol/L U0126 were lysed and analyzed by Western blot with antibodies against indicated proteins. Signals normalized to DMSO-treated EV lysates are reported as averages ± SEM (n = 3). B, wound-closure rates were measured for EV- and KRAS12V-transduced cells treated as in A. Rates normalized to the EV DMSO control condition are reported as averages ± SEM for triplicate wells from two different platings (n = 6). Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. * and **, P < 0.05 and P < 0.005, respectively.

Figure 4.

KRAS12V expression promotes mesenchymal characteristics in H1666 cells. A, H1666 cells transduced with KRAS12V or an empty vector (EV) control treated for 3 days with DMSO, 2 μmol/L gefitinib, or 20 μmol/L U0126 were lysed and analyzed by Western blot with antibodies against indicated proteins. Signals normalized to DMSO-treated EV lysates are reported as averages ± SEM (n = 3). B, wound-closure rates were measured for EV- and KRAS12V-transduced cells treated as in A. Rates normalized to the EV DMSO control condition are reported as averages ± SEM for triplicate wells from two different platings (n = 6). Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. * and **, P < 0.05 and P < 0.005, respectively.

Close modal

MEK inhibition antagonizes mesenchymal phenotypes and acquired resistance to EGFR inhibition in an EGFR-mutant–expressing cell line

We tested the effects of chronic MEK inhibition in a panel of cell lines derived from PC9 cells, which express EGFRdelE746_A750 and are gefitinib sensitive. In parental PC9 cells, MEK inhibition prevented TGF-β–mediated EMT and drove changes in epithelial/mesenchymal marker expression and wound healing consistent with the effects observed in H1666 cells (Fig. 5 and Supplementary Fig. S7).

Figure 5.

ERK activity determines epithelial/mesenchymal characteristics in an NSCLC cell line with an EGFR-activating mutation. A, lysates of parental PC9 cells treated for 4 days with 10 ng/mL TGF-β + 50 ng/mL EGF, TGF-β + EGF and 20 μmol/L U0126, or DMSO (control) were analyzed by Western blot analysis with antibodies against indicated proteins. Images are representative of three independent experiments. B, PC9 cells on glass coverslips were treated as in A, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates for each of two independent experiments. Scale bar, 20 μm. C, wound-closure rates were measured for PC9 cells treated with DMSO (control) or 20 μmol/L U0126 at the time of scratch or for 1 or 4 days before scratch. Rates normalized to the control condition for triplicate wells from two different platings (n = 6) are reported as averages ± SEM. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. D, lysates prepared in parallel to C were analyzed by Western blot analysis using antibodies against indicated proteins. Images are representative of three independent experiments. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3); * and **, P < 0.05 and P < 0.005, respectively, compared with control.

Figure 5.

ERK activity determines epithelial/mesenchymal characteristics in an NSCLC cell line with an EGFR-activating mutation. A, lysates of parental PC9 cells treated for 4 days with 10 ng/mL TGF-β + 50 ng/mL EGF, TGF-β + EGF and 20 μmol/L U0126, or DMSO (control) were analyzed by Western blot analysis with antibodies against indicated proteins. Images are representative of three independent experiments. B, PC9 cells on glass coverslips were treated as in A, fixed, and stained for E-cadherin, F-actin, and DNA. Images are representative of three replicates for each of two independent experiments. Scale bar, 20 μm. C, wound-closure rates were measured for PC9 cells treated with DMSO (control) or 20 μmol/L U0126 at the time of scratch or for 1 or 4 days before scratch. Rates normalized to the control condition for triplicate wells from two different platings (n = 6) are reported as averages ± SEM. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. D, lysates prepared in parallel to C were analyzed by Western blot analysis using antibodies against indicated proteins. Images are representative of three independent experiments. Signals normalized to respective DMSO controls are reported as averages ± SEM (n = 3); * and **, P < 0.05 and P < 0.005, respectively, compared with control.

Close modal

A previous study derived PC9 cells that acquired gefitinib resistance through EGFRT790M mutation (28). Another cell line was then derived from these cells with secondary resistance to the irreversible EGFR inhibitor WZ4002, which potently inhibits EGFRT790M (28, 33). WZ4002 resistance arose through ERK2 amplification. We used these GR and WZR clones to examine the effects of ERK2 activity, which has been tied to EMT in nontransformed cells (26). WZR cells displayed a more mesenchymal marker expression pattern than parental or GR cells, with increased fibronectin and vimentin expression and decreased E-cadherin expression (Fig. 6A). WZR cells were also more migratory than GR cells as measured by wound closure and Transwell migration (Fig. 6B and C).

Figure 6.

ERK2 amplification promotes mesenchymal characteristics in PC9 cells with acquired resistance to an irreversible EGFR inhibitor. A, lysates prepared from parental, GR, and WZR PC9 cells grown in complete media were analyzed by Western blot analysis using antibodies against indicated proteins. Images are representative of three independent experiments. Densitometry data are shown normalized to PC9 parental signals and are reported as averages ± SEM (n = 3); **, P < 0.01. B, wound-closure rates were measured for GR and WZR cells. Rates normalized to GR closure rate are reported as averages ± SEM for triplicate wells from each of two different platings (n = 6); *, P < 0.05. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. C, Transwell migration measurements were made for GR and WZR cells. Bars represent averages ± SEM for duplicate wells from three separate experiments (n = 6) normalized to GR cells.

Figure 6.

ERK2 amplification promotes mesenchymal characteristics in PC9 cells with acquired resistance to an irreversible EGFR inhibitor. A, lysates prepared from parental, GR, and WZR PC9 cells grown in complete media were analyzed by Western blot analysis using antibodies against indicated proteins. Images are representative of three independent experiments. Densitometry data are shown normalized to PC9 parental signals and are reported as averages ± SEM (n = 3); **, P < 0.01. B, wound-closure rates were measured for GR and WZR cells. Rates normalized to GR closure rate are reported as averages ± SEM for triplicate wells from each of two different platings (n = 6); *, P < 0.05. Representative phase contrast images are shown, with tracings added to identify open scratch areas. Scale bar, 200 μm. C, Transwell migration measurements were made for GR and WZR cells. Bars represent averages ± SEM for duplicate wells from three separate experiments (n = 6) normalized to GR cells.

Close modal

The effects of chronic MEK inhibition were tested in GR and WZR cells, using a higher U0126 concentration for WZR because of the ERK2 amplification. Death response to cotreatment was enhanced by approximately two-fold within 3 days of U0126 pretreatment in both cell lines (Fig. 7A and Supplementary Fig. S8A and S8B). As in H1666 cells, sufficiently long U0126 exposure resulted in an eventual decrease in augmented response to EGFR inhibition. Increased sensitivity to gefitinib correlated with reduced vimentin expression and slight increases in E-cadherin expression, and the eventual decrease in augmentation was accompanied by increased fibronectin expression in WZR cells (fibronectin was not detectable in GR cells; Fig. 7B and Supplementary Fig. S8C).

Figure 7.

MEK inhibition sensitizes cells with acquired resistance to EGFR inhibitors. A, GR and WZR PC9 cells were cultured in 5 μmol/L and 20 μmol/L U0126, respectively, or DMSO (control) for up to 9 days, as described in Materials and Methods, and were evaluated for cell death response to 5 μmol/L gefitinib for various U0126 exposure times. Cell death was measured by flow cytometry for TO-PRO3 permeability 48 hours after gefitinib addition. Time points represent total time exposed to U0126 before gefitinib addition or lysis. Data are represented as averages ± SEM (n = 3); significance is shown relative to any other condition from the same day. B, GR and WZR cell lysates prepared in parallel to A were analyzed by Western blot analysis using antibodies against indicated proteins. Average signals normalized to respective DMSO controls are reported ± SEM (n = 3). * and **, P < 0.05 and P <0.005, respectively.

Figure 7.

MEK inhibition sensitizes cells with acquired resistance to EGFR inhibitors. A, GR and WZR PC9 cells were cultured in 5 μmol/L and 20 μmol/L U0126, respectively, or DMSO (control) for up to 9 days, as described in Materials and Methods, and were evaluated for cell death response to 5 μmol/L gefitinib for various U0126 exposure times. Cell death was measured by flow cytometry for TO-PRO3 permeability 48 hours after gefitinib addition. Time points represent total time exposed to U0126 before gefitinib addition or lysis. Data are represented as averages ± SEM (n = 3); significance is shown relative to any other condition from the same day. B, GR and WZR cell lysates prepared in parallel to A were analyzed by Western blot analysis using antibodies against indicated proteins. Average signals normalized to respective DMSO controls are reported ± SEM (n = 3). * and **, P < 0.05 and P <0.005, respectively.

Close modal

We characterized the role of ERK in regulating EMT in NSCLC cells. MEK inhibition prevented TGF-β–induced EMT in NSCLC cell lines expressing wild-type (WT) EGFR and KRAS (H1666 and H322), WT EGFR and mutant KRAS (H358), or mutant EGFR (PC9). This suggests a general role for ERK in promoting mesenchymal characteristics across typical NSCLC cell types. MEK inhibition also antagonized mesenchymal-associated characteristics including migration, resistance to EGFR inhibition, and CD44high/CD24low expression.

The role of ERK in EMT has been explored in other cell types. ERK is an established determinant of EMT in nontransformed cells (23, 34–36), with ERK2 specifically implicated as an EMT driver (26, 27, 37). This is consistent with findings in breast cancer (38), where MEK inhibition reverses miR-21–mediated EMT, and with studies of phosphatases regulating ERK (i.e., MKP3, PTPN14, and SHP2; refs. 39–41), which also suggest a connection between ERK activity and EMT.

A number of factors regulate ERK activity in NSCLC cells. We previously demonstrated a connection of impaired mutant EGFR endocytosis with a diminished functional role of SHP2 and impaired ERK activity (29, 42). More recently, in collaboration with others, we identified a coupling between ERK2 amplification and increased EGFR endocytosis in the WZ4002-resistant PC9 cells used here (28). Interestingly, inhibiting EGFR endocytosis in WZ4002-resistant cells reduced ERK phosphorylation and promoted an epithelial marker expression pattern (Supplementary Fig. S9), drawing a potential connection between EGFR endocytosis and EMT. More recently, Sprouty2 has been shown to promote ERK activity in NSCLC cells with or without EGFR mutations (43).

Of course, KRAS mutations may also promote ERK activity in NSCLC. In H1666 cells, KRAS12V-driven ERK activity promoted mesenchymal marker expression and resistance to gefitinib-mediated MET changes. These findings are generally consistent with studies connecting oncogenic HRAS or KRAS mutants with EMT in mammary epithelia (26), pancreatic cancer (27, 30), and lung cancer (30). The MEK dependence of our observations with KRAS12V may support exploration of MEK and ERK inhibitors in NSCLCs with KRAS mutation.

In addition to promoting epithelial marker expression, MEK inhibition promoted response to EGFR inhibition. Some studies have suggested the efficacy of EGFR and MEK coinhibition in gastric cancer (44) and pancreatic cancer cells (45), and current clinical trials are testing erlotinib combined with MEK inhibitors in NSCLC. In light of this, it is worth noting that the effects we observed with MEK inhibition occurred on a time scale that was much longer than that for ERK inhibition and more consistent with changes in epithelial and mesenchymal marker expression. This may suggest considering a clinical scheduling approach wherein a MEK inhibitor is initially used alone to promote an epithelial phenotype, followed by addition of an EGFR inhibitor. An analogous staggered drug scheduling approach was effective in triple-negative breast cancer cells, which were sensitized to doxorubicin by erlotinib pretreatment in vitro and in mouse tumor xenografts (9). Applying this basic concept to future clinical trial design may allow for lower MEK inhibitor doses to curb toxicities (46) and promote response to EGFR inhibitors. Given that the time windows for augmented response to gefitinib and the useful concentrations of U0126 were variable among cell lines, it will of course be important first to examine this strategy in preclinical animal models to assess general efficacy in an in vivo setting and optimize timing and dosing.

We note as well that some recent data suggest that MET is required for efficient colonization of distant metastases (47, 48). Thus, in evaluating different potential strategies for driving MET, the potential to drive proliferation of metastases will have to be weighed against the potential ability to kill tumor cells. ERK may be an especially attractive target in this regard. In at least one study, ERK inhibition through microRNA-mediated reduction of SHP2 (a positive regulator of ERK signaling) reduced metastases in an in vivo breast cancer model (49), which was due to SHP2′s simultaneous regulation of proliferation, invasion, and transcription factor expression. Thus, promoting MET through ERK inhibition, which should additionally inhibit cellular proliferation, may mitigate risks associated with proliferation of micrometastases.

In all three cell lines we tested for effects of chronic MEK inhibition, enhanced gefitinib response eventually decreased. Though initially unresponsive to U0126, U0126-cultured cells displayed noticeable cell death at mid-range time points (partially reflected by cell death measurements for U0126-only samples in Figs. 3 and 7). This could result in selection for U0126-resistant cells over time. Because the fraction of CD44high/CD24low cells in U0126-treated cultures remained low at time points when resistance was observed (Fig. 3C and E), the potentially inherently resistant population was apparently not CD44high/CD24low. Alternatively, resistance could arise from adaptation of the entire cell population, for example, through increased fibronectin-mediated signaling, which has been linked to AKT activation and docetaxel resistance in ovarian and breast cancer cells (50). Indeed, in all cases, the eventual drop in augmented response to gefitinib was accompanied by increased fibronectin expression. Future work should consider effectors of fibronectin-mediated signaling as signaling nodes whose inhibition may augment EGFR inhibitor response.

Although our data suggest potential relevance of our findings to de novo and acquired EGFR inhibitor resistance, responsiveness to chronic MEK inhibition was not uniform among cell lines, which could result from any of the numerous differences in initial conditions among NSCLC cell lines. For example, although H1666 cells express WT EGFR and KRAS, they also express the low-activity G446V BRAF mutant (51), which could influence their death response to MEK inhibition. In the future, therefore, investigating the cellular initial conditions that may determine responsiveness to this approach and identifying alternative signaling nodes whose inhibition may result in more stable maintenance of an epithelial cell type will also be important.

No potential conflicts of interest were disclosed.

Conception and design: J.M. Buonato, M.J. Lazzara

Development of methodology: J.M. Buonato, M.J. Lazzara

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.M. Buonato

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.M. Buonato

Writing, review, and/or revision of the manuscript: J.M. Buonato, M.J. Lazzara

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.J. Lazzara

Study supervision: M.J. Lazzara

The authors thank Drs. Pasi Jänne, Douglas Lauffenburger, Deepak Nihalani, and Russ Carstens for generously providing reagents, Dr. Devraj Basu for technical discussions, and Calixte Monast, Chris Furcht, and Alice Walsh for technical assistance and guidance.

This work was supported, in part, by the NIH under Ruth L. Kirschstein National Research Service Award 2T32HL007954 from the NIH–NHLBI. This material is also based upon work supported by the National Science Foundation Graduate Research Fellowship under grant no. DGE-0822.

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.
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
.
2.
Sordella
R
,
Bell
DW
,
Haber
DA
,
Settleman
J
. 
Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways
.
Science
2004
;
305
:
1163
7
.
3.
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
.
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 Med
2005
;
2
:
e73
.
5.
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
.
6.
Blumenschein
GR
 Jr
,
Paulus
R
,
Curran
WJ
,
Robert
F
,
Fossella
F
,
Werner-Wasik
M
, et al
Phase II study of cetuximab in combination with chemoradiation in patients with stage IIIA/B non-small-cell lung cancer: RTOG 0324
.
J Clin Oncol
2011
;
29
:
2312
8
.
7.
Spigel
DR
,
Burris
HA
 III
,
Greco
FA
,
Shipley
DL
,
Friedman
EK
,
Waterhouse
DM
, et al
Randomized, double-blind, placebo-controlled, phase II trial of sorafenib and erlotinib or erlotinib alone in previously treated advanced non-small-cell lung cancer
.
J Clin Oncol
2011
;
29
:
2582
9
.
8.
Xu
L
,
Kikuchi
E
,
Xu
C
,
Ebi
H
,
Ercan
D
,
Cheng
KA
, et al
Combined EGFR/MET or EGFR/HSP90 inhibition is effective in the treatment of lung cancers codriven by mutant EGFR containing T790M and MET
.
Cancer Res
2012
;
72
:
3302
11
.
9.
Lee
MJ
,
Ye
AS
,
Gardino
AK
,
Heijink
AM
,
Sorger
PK
,
MacBeath
G
, et al
Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks
.
Cell
2012
;
149
:
780
94
.
10.
Kalluri
R
,
Weinberg
RA
. 
The basics of epithelial-mesenchymal transition
.
J Clin Invest
2009
;
119
:
1420
8
.
11.
Zavadil
J
,
Bottinger
EP
. 
TGF-beta and epithelial-to-mesenchymal transitions
.
Oncogene
2005
;
24
:
5764
74
.
12.
Garofalo
M
,
Romano
G
,
Di Leva
G
,
Nuovo
G
,
Jeon
YJ
,
Ngankeu
A
, et al
EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers
.
Nat Med
2011
;
18
:
74
82
.
13.
Thomson
S
,
Petti
F
,
Sujka-Kwok
I
,
Epstein
D
,
Haley
JD
. 
Kinase switching in mesenchymal-like non-small cell lung cancer lines contributes to EGFR inhibitor resistance through pathway redundancy
.
Clin Exp Metastasis
2008
;
25
:
843
54
.
14.
Yauch
RL
,
Januario
T
,
Eberhard
DA
,
Cavet
G
,
Zhu
W
,
Fu
L
, et al
Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients
.
Clin Cancer Res
2005
;
11
:
8686
98
.
15.
Shrader
M
,
Pino
MS
,
Brown
G
,
Black
P
,
Adam
L
,
Bar-Eli
M
, et al
Molecular correlates of gefitinib responsiveness in human bladder cancer cells
.
Mol Cancer Ther
2007
;
6
:
277
85
.
16.
Frederick
BA
,
Helfrich
BA
,
Coldren
CD
,
Zheng
D
,
Chan
D
,
Bunn
PA
 Jr
, et al
Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma
.
Mol Cancer Ther
2007
;
6
:
1683
91
.
17.
Maseki
S
,
Ijichi
K
,
Tanaka
H
,
Fujii
M
,
Hasegawa
Y
,
Ogawa
T
, et al
Acquisition of EMT phenotype in the gefitinib-resistant cells of a head and neck squamous cell carcinoma cell line through Akt/GSK-3beta/snail signalling pathway
.
Br J Cancer
2012
;
106
:
1196
204
.
18.
Yin
T
,
Wang
C
,
Liu
T
,
Zhao
G
,
Zha
Y
,
Yang
M
. 
Expression of snail in pancreatic cancer promotes metastasis and chemoresistance
.
J Surg Res
2007
;
141
:
196
203
.
19.
Li
X
,
Lewis
MT
,
Huang
J
,
Gutierrez
C
,
Osborne
CK
,
Wu
MF
, et al
Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy
.
J Natl Cancer Inst
2008
;
100
:
672
9
.
20.
Yao
Z
,
Fenoglio
S
,
Gao
DC
,
Camiolo
M
,
Stiles
B
,
Lindsted
T
, et al
TGF-beta IL-6 axis mediates selective and adaptive mechanisms of resistance to molecular targeted therapy in lung cancer
.
Proc Natl Acad Sci U S A
2010
;
107
:
15535
40
.
21.
Witta
SE
,
Gemmill
RM
,
Hirsch
FR
,
Coldren
CD
,
Hedman
K
,
Ravdel
L
, et al
Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines
.
Cancer Res
2006
;
66
:
944
50
.
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.
Grande
M
,
Franzen
A
,
Karlsson
JO
,
Ericson
LE
,
Heldin
NE
,
Nilsson
M
. 
Transforming growth factor-beta and epidermal growth factor synergistically stimulate epithelial to mesenchymal transition (EMT) through a MEK-dependent mechanism in primary cultured pig thyrocytes
.
J Cell Sci
2002
;
115
:
4227
36
.
24.
Xie
L
,
Law
BK
,
Chytil
AM
,
Brown
KA
,
Aakre
ME
,
Moses
HL
. 
Activation of the Erk pathway is required for TGF-beta1-induced EMT in vitro
.
Neoplasia
2004
;
6
:
603
10
.
25.
Zavadil
J
,
Bitzer
M
,
Liang
D
,
Yang
YC
,
Massimi
A
,
Kneitz
S
, et al
Genetic programs of epithelial cell plasticity directed by transforming growth factor-beta
.
Proc Natl Acad Sci U S A
2001
;
98
:
6686
91
.
26.
Shin
S
,
Dimitri
CA
,
Yoon
SO
,
Dowdle
W
,
Blenis
J
. 
ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events
.
Mol Cell
2010
;
38
:
114
27
.
27.
Botta
GP
,
Reginato
MJ
,
Reichert
M
,
Rustgi
AK
,
Lelkes
PI
. 
Constitutive K-RasG12D activation of ERK2 specifically regulates 3D invasion of human pancreatic cancer cells via MMP-1
.
Mol Cancer Res
2011
;
10
:
183
96
.
28.
Ercan
D
,
Xu
C
,
Yanagita
M
,
Monast
CS
,
Pratilas
CA
,
Montero
J
, et al
Reactivation of ERK Signaling Causes Resistance to EGFR Kinase Inhibitors
.
Cancer Discov
2012
;
2
:
934
47
.
29.
Lazzara
MJ
,
Lane
K
,
Chan
R
,
Jasper
PJ
,
Yaffe
MB
,
Sorger
PK
, et al
Impaired SHP2-mediated extracellular signal-regulated kinase activation contributes to gefitinib sensitivity of lung cancer cells with epidermal growth factor receptor-activating mutations
.
Cancer Res
2010
;
70
:
3843
50
.
30.
Singh
A
,
Greninger
P
,
Rhodes
D
,
Koopman
L
,
Violette
S
,
Bardeesy
N
, et al
A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival
.
Cancer Cell
2009
;
15
:
489
500
.
31.
Basu
D
,
Nguyen
TT
,
Montone
KT
,
Zhang
G
,
Wang
LP
,
Diehl
JA
, et al
Evidence for mesenchymal-like sub-populations within squamous cell carcinomas possessing chemoresistance and phenotypic plasticity
.
Oncogene
2010
;
29
:
4170
82
.
32.
Basu
D
,
Montone
KT
,
Wang
LP
,
Gimotty
PA
,
Hammond
R
,
Diehl
JA
, et al
Detecting and targeting mesenchymal-like subpopulations within squamous cell carcinomas
.
Cell Cycle
2011
;
10
:
2008
16
.
33.
Zhou
W
,
Ercan
D
,
Chen
L
,
Yun
CH
,
Li
D
,
Capelletti
M
, et al
Novel mutant-selective EGFR kinase inhibitors against EGFR T790M
.
Nature
2009
;
462
:
1070
4
.
34.
Janda
E
,
Lehmann
K
,
Killisch
I
,
Jechlinger
M
,
Herzig
M
,
Downward
J
, et al
Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways
.
J Cell Biol
2002
;
156
:
299
313
.
35.
Lehmann
K
,
Janda
E
,
Pierreux
CE
,
Rytomaa
M
,
Schulze
A
,
McMahon
M
, et al
Raf induces TGFbeta production while blocking its apoptotic but not invasive responses: a mechanism leading to increased malignancy in epithelial cells
.
Genes Dev
2000
;
14
:
2610
22
.
36.
Peinado
H
,
Quintanilla
M
,
Cano
A
. 
Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions
.
J Biol Chem
2003
;
278
:
21113
23
.
37.
von Thun
A
,
Birtwistle
M
,
Kalna
G
,
Grindlay
J
,
Strachan
D
,
Kolch
W
, et al
ERK2 drives tumour cell migration in three-dimensional microenvironments by suppressing expression of Rab17 and liprin-beta2
.
J Cell Sci
2012
;
125
:
1465
77
.
38.
Han
M
,
Liu
M
,
Wang
Y
,
Chen
X
,
Xu
J
,
Sun
Y
, et al
Antagonism of miR-21 reverses epithelial-mesenchymal transition and cancer stem cell phenotype through AKT/ERK1/2 inactivation by targeting PTEN
.
PLoS ONE
2012
;
7
:
e39520
.
39.
Liu
X
,
Yang
N
,
Figel
SA
,
Wilson
KE
,
Morrison
CD
,
Gelman
IH
, et al
PTPN14 interacts with and negatively regulates the oncogenic function of YAP
.
Oncogene
2013
;
32
:
1266
73
.
40.
Wong
VC
,
Chen
H
,
Ko
JM
,
Chan
KW
,
Chan
YP
,
Law
S
, et al
Tumor suppressor dual-specificity phosphatase 6 (DUSP6) impairs cell invasion and epithelial-mesenchymal transition (EMT)-associated phenotype
.
Int J Cancer
2011
;
130
:
83
95
.
41.
Zhou
XD
,
Agazie
YM
. 
Inhibition of SHP2 leads to mesenchymal to epithelial transition in breast cancer cells
.
Cell Death Differ
2008
;
15
:
988
96
.
42.
Furcht
CM
,
Munoz Rojas
AR
,
Nihalani
D
,
Lazzara
MJ
. 
Diminished functional role and altered localization of SHP2 in non-small cell lung cancer cells with EGFR-activating mutations
.
Oncogene
2013
;
32
:
2346
55
.
43.
Walsh
AM
,
Lazzara
MJ
. 
Regulation of EGFR trafficking and cell signaling by Sprouty2 and MIG6 in lung cancer cells
.
J Cell Sci
2013
;
126
:
4339
48
.
44.
Yoon
YK
,
Kim
HP
,
Han
SW
,
Hur
HS
,
Oh do
Y
,
Im
SA
, et al
Combination of EGFR and MEK1/2 inhibitor shows synergistic effects by suppressing EGFR/HER3-dependent AKT activation in human gastric cancer cells
.
Mol Cancer Ther
2009
;
8
:
2526
36
.
45.
Diep
CH
,
Munoz
RM
,
Choudhary
A
,
Von Hoff
DD
,
Han
H
. 
Synergistic effect between erlotinib and MEK inhibitors in KRAS wild-type human pancreatic cancer cells
.
Clin Cancer Res
2011
;
17
:
2744
56
.
46.
Wang
D
,
Boerner
SA
,
Winkler
JD
,
LoRusso
PM
. 
Clinical experience of MEK inhibitors in cancer therapy
.
Biochim Biophys Acta
2007
;
1773
:
1248
55
.
47.
Celia-Terrassa
T
,
Meca-Cortes
O
,
Mateo
F
,
de Paz
AM
,
Rubio
N
,
Arnal-Estape
A
, et al
Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells
.
J Clin Invest
2012
;
122
:
1849
68
.
48.
Tsai
JH
,
Donaher
JL
,
Murphy
DA
,
Chau
S
,
Yang
J
. 
Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis
.
Cancer Cell
2012
;
22
:
725
36
.
49.
Aceto
N
,
Sausgruber
N
,
Brinkhaus
H
,
Gaidatzis
D
,
Martiny-Baron
G
,
Mazzarol
G
, et al
Tyrosine phosphatase SHP2 promotes breast cancer progression and maintains tumor-initiating cells via activation of key transcription factors and a positive feedback signaling loop
.
Nat Med
2012
;
18
:
529
37
.
50.
Xing
H
,
Weng
D
,
Chen
G
,
Tao
W
,
Zhu
T
,
Yang
X
, et al
Activation of fibronectin/PI-3K/Akt2 leads to chemoresistance to docetaxel by regulating survivin protein expression in ovarian and breast cancer cells
.
Cancer Lett
2008
;
261
:
108
19
.
51.
Pratilas
CA
,
Hanrahan
AJ
,
Halilovic
E
,
Persaud
Y
,
Soh
J
,
Chitale
D
, et al
Genetic predictors of MEK dependence in non-small cell lung cancer
.
Cancer Res
2008
;
68
:
9375
83
.