The molecular mechanism by which epidermal growth factor receptor–tyrosine kinase inhibitors (EGFR-TKI) induce apoptosis in non–small cell-lung cancer (NSCLC) cells that are positive for activating mutations of the EGFR remains unclear. In this study, we report the effects of the EGFR-TKI gefitinib on expression of the antiapoptotic protein survivin that have functional consequences in EGFR mutation–positive NSCLC cells. Immunoblot analysis revealed that gefitinib downregulated survivin expression, likely through inhibition of the PI3K-AKT signaling pathway, in NSCLC cells positive for EGFR mutation. Stable overexpression of survivin attenuated gefitinib-induced apoptosis and also inhibited the antitumor effect of gefitinib in human tumor xenografts. Furthermore, the combination of survivin overexpression with inhibition of the gefitinib-induced upregulation of the proapoptotic protein BIM attenuated gefitinib-induced apoptosis to a greater extent than either approach alone. Our results indicate that downregulation of survivin plays a pivotal role in gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells. Furthermore, they suggest that simultaneous interruption of the PI3K-AKT-survivin and MEK-ERK-BIM signaling pathways is responsible for EGFR-TKI–induced apoptotic death in these cells. Cancer Res; 70(24); 10402–10. ©2010 AACR.

Survivin is a member of the inhibitor of apoptosis (IAP) family of proteins and has been shown to inhibit caspases and to prevent caspase-mediated cell death (1–3). Survivin is abundant in many types of cancer cells but not in the corresponding normal cells (4, 5). In nonmalignant proliferating cells, the expression of survivin is regulated in a cell cycle-dependent manner (6, 7). The upregulation of survivin expression in tumors does not seem to be dependent solely on the cell cycle, however, given that it occurs in tumor cells that are not actively cycling (4, 8, 9). Indeed, growth factors have been found to regulate survivin expression in endothelial cells and neuroblastoma cells (10, 11). Although expression of survivin has been demonstrated in non–small cell-lung cancer (NSCLC; refs. 12–14), the mechanism by which such expression is regulated in NSCLC cells has remained unknown.

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is abnormally amplified or activated in a variety of tumors including NSCLC, and it has therefore been identified as an important target in cancer treatment (15–17). Inhibitors of the tyrosine kinase activity of EGFR (EGFR-TKI), which compete with ATP for binding to the tyrosine kinase pocket of the receptor, have been extensively studied in patients with NSCLC (18, 19). Several prospective clinical trials have revealed marked antitumor activity of EGFR-TKIs in NSCLC patients with EGFR mutations. The therapeutic benefit of these drugs is much greater than that historically observed with standard cytotoxic chemotherapy for advanced NSCLC. NSCLC cells with EGFR mutations manifest activation of the PI3K (phosphatidylinositol 3-kinase)–AKT and MEK–ERK (extracellular signal-regulated kinase) signaling pathways under the control of EGFR, and exposure of such cells to EGFR-TKIs blocks signaling by both pathways and induces apoptosis (20–22). The precise molecular mechanism by which EGFR-TKIs induce apoptosis has remained unclear, however. We have therefore now examined the effect of the EGFR-TKI gefitinib on survivin expression as well as further investigated the mechanism of gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells.

Cell culture and reagents

The human NSCLC cell lines PC9, HCC827, NCI-H1975 (H1975), A549, and H1299 were obtained from American Type Culture Collection. The NSCLC line PC9/ZD was obtained as described previously (23). All cells were cultured under a humidified atmosphere of 5% CO2 at 37°C in RPMI 1640 medium (Sigma) supplemented with 10% fetal bovine serum. Gefitinib was obtained from Kemprotec, U0126 and LY294002 were from Cell Signaling Technology and BEZ235 and AZD6244 were from ShangHai Biochempartner.

Immunoblot analysis

Cells were washed twice with ice-cold PBS and then lysed in a solution containing 20 mmol/L Tris-HCl (pH 7.5), 150 mmol/L NaCl, 1 mmol/L EDTA, 1% Triton X-100, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L phenylmethylsulfonyl fluoride, and leupeptin (1 μg/mL). The protein concentration of the cell lysates was determined with the use of the Bradford reagent (Bio-Rad), and equal amounts of protein were subjected to SDS-PAGE on a 7.5% gel. The separated proteins were transferred to a nitrocellulose membrane, which was then exposed to 5% nonfat dried milk in PBS for 1 hour at room temperature before incubation overnight at 4°C with primary antibodies. Rabbit polyclonal antibodies to human phosphorylated EGFR (pY1068), to XIAP, to phosphorylated and total AKT, to phosphorylated and total ERK, to poly(ADP-ribose) polymerase (PARP), to caspase-3, and to BIM were obtained from Cell Signaling Technology; those to survivin were from Santa Cruz Biotechnology; those to cIAP-1 were from R&D Systems; and those to β-actin were from Sigma. Mouse monoclonal antibodies to EGFR were obtained from Invitrogen. All antibodies were used at a 1:1,000 dilution, with the exception of those to β-actin (1:200). The nitrocellulose membrane was then washed with PBS containing 0.05% Tween 20 before incubation for 1 hour at room temperature with horseradish peroxidase-conjugated goat antibodies to rabbit (Sigma) or mouse (Santa Cruz Biotechnology) immunoglobulin G. Immune complexes were finally detected with chemiluminescence reagents (Perkin-Elmer Life Science).

Gene silencing

Cells were plated at 50% to 60% confluence in 6-well plates or 25-cm2 flasks and then incubated for 24 hours before transient transfection for the indicated times with small interfering RNAs (siRNA) mixed with the Lipofectamine reagent (Invitrogen). The siRNAs specific for AKT (AKT-1, 5′-CCAGGUAUUUUGAUGAGGA-3′; AKT-2, 5′-CAACCGCCAUCCAGACUGU-3′), survivin (survivin-1, 5′-GAAGCAGUUUGAAGAAUUA-3′; survivin-2, 5′-AGAAGCAGUUU GAAGAAUU-3′), or BIM (BIM-1, 5′-GGAGGGUAUUUUUGAAUAA-3′) mRNAs as well as corresponding scrambled (control) siRNAs were obtained from Nippon EGT.

Annexin V binding assay

The binding of Annexin V to cells was measured with the use of an Annexin-V-FLUOS Staining Kit (Roche). Cells were harvested by exposure to trypsin-EDTA, washed with PBS, and centrifuged at 200 × g for 5 minutes. The cell pellets were resuspended in 100 μL of Annexin-V-FLUOS labeling solution, incubated for 10 to 15 minutes at 15°C to 25°C, and then analyzed for fluorescence with a flow cytometer (FACSCalibur) and Cell Quest software (Becton Dickinson).

Cell cycle analysis

Cells were harvested, washed with PBS, fixed with 70% methanol, washed again with PBS, and stained with propidium iodide (0.05 mg/mL) in a solution containing 0.1% Triton X-100, 0.1 mmol/L EDTA, and RNase A (0.05 mg/mL). The stained cells were then analyzed for DNA content with a flow cytometer and Modfit software (Verity Software House).

Establishment of cells stably overexpressing survivin

A full-length cDNA fragment encoding human survivin was obtained from HCC827 cells by reverse transcription and PCR with the primers survivin-forward (5′-GCGGCCGCGGCGGCATGGGTGCCCCGACGTTG-3′) and survivin-reverse (5′-GGATCCTCAATCCATGGCAGCCAGCTGCTCG-3′). The amplification product was verified by sequencing after its cloning into the pCR-Blunt II-TOPO vector (Invitrogen). The survivin cDNA was excised from pCR-Blunt II-TOPO and transferred to the pQCXIH retroviral vector (Clontech). Retroviruses encoding survivin were then produced and used to infect PC9 and HCC827 cells as described (24). Cells stably expressing survivin were then isolated by selection with hygromycin at 300 μg/mL (Invivogen).

Growth inhibition assay in vivo

All animal studies were performed in accordance with the Recommendations for Handling of Laboratory Animals for Biomedical Research compiled by the Committee on Safety and Ethical Handling Regulations for Laboratory Animal Experiments, Kinki University (Osaka, Japan). The ethical procedures followed conformed to the guidelines of the United Kingdom Coordinating Committee on Cancer Prevention Research. Tumors cells (5 × 106) were injected subcutaneously into the axilla of 5- to 6-week-old female athymic nude mice (BALB/c nu/nu; CLEA Japan). Treatment was initiated when tumors in each group of 6 mice achieved an average volume of 200 to 400 mm3. Treatment groups consisted of vehicle control and gefitinib (10 or 25 mg/kg). Gefitinib was administered by oral gavage daily for 4 weeks, with control animals receiving a 0.5% (w/v) aqueous solution of hydroxypropylmethylcellulose as vehicle. Tumor volume was determined from caliper measurements of tumor length (L) and width (W) according to the formula LW2/2. Both tumor size and body weight were measured twice per week.

Statistical analysis

Quantitative data are presented as means ± SE from 3 independent experiments or for 6 animals per group unless indicated otherwise. The significance of differences in the percentage of Annexin V-positive cells was evaluated with the unpaired 2-tailed Student's t test. P < 0.05 was considered statistically significant.

Gefitinib downregulates survivin expression in EGFR mutation-positive NSCLC cell lines

We first examined the effects of the EGFR-TKI gefitinib on the expression of IAP family members in a subset of NSCLC cell lines (PC9, HCC827, PC9/ZD, H1975, A549, and H1299) by immunoblot analysis (Fig. 1). PC9 and HCC827 cells harbor an EGFR allele with an activating mutation, whereas A549 and H1299 cells express wild-type EGFR and PC9/ZD and H1975 cells harbor an EGFR allele with both an activating mutation and a mutation (T790M) that confers resistance to EGFR-TKIs. In PC9 and HCC827 cells, gefitinib induced the dephosphorylation of EGFR and reduced the abundance of survivin in a concentration-dependent manner. In contrast, in cells expressing wild-type EGFR or harboring the T790M resistance mutation, gefitinib did not affect the phosphorylation level of EGFR or the expression of survivin. The expression of other IAP family members, including XIAP and cIAP-1, was not substantially affected by gefitinib in any of the cell lines examined. These data thus showed that gefitinib downregulated survivin expression in NSCLC cells with an activating mutation of EGFR.

Figure 1.

Effects of gefitinib on the expression of IAP family proteins in human NSCLC cells. PC9, HCC827, PC9/ZD, H1975, A549, or H1299 cells were incubated in complete medium and in the presence of the indicated concentrations of gefitinib for 24 hours. Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of EGFR, AKT, or ERK, to survivin, to XIAP, to cIAP-1, or to β-actin (loading control). Data are representative of 3 independent experiments.

Figure 1.

Effects of gefitinib on the expression of IAP family proteins in human NSCLC cells. PC9, HCC827, PC9/ZD, H1975, A549, or H1299 cells were incubated in complete medium and in the presence of the indicated concentrations of gefitinib for 24 hours. Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of EGFR, AKT, or ERK, to survivin, to XIAP, to cIAP-1, or to β-actin (loading control). Data are representative of 3 independent experiments.

Close modal

Inhibition of the PI3K-AKT pathway results in survivin downregulation in EGFR mutation–positive cells

To identify the signaling pathway (or pathways) responsible for downregulation of survivin by gefitinib, we examined the effects of specific inhibitors of MEK (U0126 and AZD6244) and PI3K (LY294002 and BEZ235) in EGFR mutation–positive NSCLC cells (PC9 and HCC827). Each of the PI3K inhibitors reduced the abundance of survivin, whereas the MEK inhibitors had no such effect (Fig. 2A), suggesting that the regulation of survivin expression is mediated by PI3K rather than by MEK in EGFR mutation-positive NSCLC cells. Given that the protein kinase AKT is an important downstream target of PI3K, we examined whether the PI3K-dependent survivin expression is also dependent on AKT. Depletion of AKT by transfection of cells with 2 different siRNAs specific for AKT mRNA (AKT-1 and AKT-2 siRNA) resulted in downregulation of survivin expression in both PC9 and HCC827 cells (Fig. 2B). These results thus suggested that gefitinib might regulate survivin expression through inhibition of the PI3K-AKT signaling pathway in EGFR mutation–positive NSCLC cells.

Figure 2.

Effects of inhibition of MEK or PI3K signaling pathways on survivin expression in EGFR mutation–positive NSCLC cells. A, PC9 or HCC827 cells were incubated in the absence (control, 0.1% dimethyl sulfoxide) or the presence of gefitinib (1 μmol/L), LY294002 (20 μmol/L), BEZ235 (0.2 μmol/L), U0126 (20 μmol/L), or AZD6244 (0.2 μmol/L) for 24 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of AKT or ERK, to survivin, or to β-actin. B, Cells were transfected (or not) with 2 different AKT (AKT-1 or AKT-2) or scrambled (control) siRNAs for 48 hours, lysed, and subjected to immunoblot analysis with antibodies to AKT, to survivin, or to β-actin. All data are representative of 3 independent experiments.

Figure 2.

Effects of inhibition of MEK or PI3K signaling pathways on survivin expression in EGFR mutation–positive NSCLC cells. A, PC9 or HCC827 cells were incubated in the absence (control, 0.1% dimethyl sulfoxide) or the presence of gefitinib (1 μmol/L), LY294002 (20 μmol/L), BEZ235 (0.2 μmol/L), U0126 (20 μmol/L), or AZD6244 (0.2 μmol/L) for 24 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of AKT or ERK, to survivin, or to β-actin. B, Cells were transfected (or not) with 2 different AKT (AKT-1 or AKT-2) or scrambled (control) siRNAs for 48 hours, lysed, and subjected to immunoblot analysis with antibodies to AKT, to survivin, or to β-actin. All data are representative of 3 independent experiments.

Close modal

Knockdown of survivin expression induces apoptosis in EGFR mutation–positive cells

To investigate whether downregulation of survivin by gefitinib is related to gefitinib-induced apoptosis, we transfected PC9 or HCC827 cells with 2 independent siRNA specific for survivin mRNA (survivin-1 and survivin-2 siRNAs). Depletion of survivin resulted in generation of the cleaved forms of both caspase-3 and PARP in both cell lines (Fig. 3A). Staining with Annexin V also revealed that the proportion of apoptotic cells was markedly increased by transfection with the survivin siRNAs (Fig. 3B). In addition, depletion of survivin resulted in an increase in the size of the sub-G1 (apoptotic) cell population, as revealed by flow cytometry (Fig. 3C). These data suggested that downregulation of survivin induces apoptosis in EGFR mutation–positive NSCLC cells.

Figure 3.

Effect of survivin depletion on apoptosis in EGFR mutation–positive NSCLC cells. A, PC9 or HCC827 cells were transfected (or not) with 2 different survivin (survivin-1 or survivin-2) or scrambled (control) siRNAs for 48 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to survivin, to PARP, to caspase-3, or to β-actin. Bands corresponding to the cleaved (cl) forms of caspase-3 and PARP are indicated. Data are representative of 3 independent experiments. B, cells were transfected with survivin or scrambled siRNAs for 72 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 versus the corresponding value for cells transfected with the scrambled siRNA. C, cells were transfected with survivin or scrambled siRNAs for 48 hours, fixed, stained with propidium iodide, and analyzed for cell cycle distribution by flow cytometry. Data are means of triplicates from representative experiments that were repeated 3 times.

Figure 3.

Effect of survivin depletion on apoptosis in EGFR mutation–positive NSCLC cells. A, PC9 or HCC827 cells were transfected (or not) with 2 different survivin (survivin-1 or survivin-2) or scrambled (control) siRNAs for 48 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to survivin, to PARP, to caspase-3, or to β-actin. Bands corresponding to the cleaved (cl) forms of caspase-3 and PARP are indicated. Data are representative of 3 independent experiments. B, cells were transfected with survivin or scrambled siRNAs for 72 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 versus the corresponding value for cells transfected with the scrambled siRNA. C, cells were transfected with survivin or scrambled siRNAs for 48 hours, fixed, stained with propidium iodide, and analyzed for cell cycle distribution by flow cytometry. Data are means of triplicates from representative experiments that were repeated 3 times.

Close modal

Overexpression of survivin inhibits gefitinib-induced apoptosis in EGFR mutation–positive cells in vitro

To examine further the role of survivin in gefitinib-induced apoptosis, we established PC9 and HCC827 sublines (PC9S7, PC9S8, HCC827S6, and HCC827S7) that stably overexpress survivin as a result of retroviral infection. The abundance of survivin in these sublines was substantially greater than that in cells infected with the empty virus (PC9-Mock and HCC827-Mock; Fig. 4A). In addition, gefitinib markedly reduced the level of survivin expression in PC9-Mock and HCC827-Mock cells but not in the corresponding sublines overexpressing survivin (Fig. 4B). Immunoblot analysis of the cleaved forms of caspase-3 and PARP (Fig. 4B) as well as staining with Annexin V (Fig. 4C) also revealed that overexpression of survivin resulted in marked inhibition of gefitinib-induced apoptosis. Examination of the effect of gefitinib on cell cycle distribution revealed that gefitinib increased the proportion of cells in G0–G1 phase and reduced that in S phase at 24 hours in a manner independent of survivin overexpression (Fig. 4D). The survivin-overexpressing sublines, however, showed a smaller time-dependent increase in the size of the sub-G1 cell population than did cells infected with the empty virus. These results thus further indicated that downregulation of survivin by gefitinib contributes to the proapoptotic action of this drug in EGFR mutation–positive NSCLC cells.

Figure 4.

Effect of survivin overexpression on gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells in vitro. A, parental (P) PC9 or HCC827 cells or corresponding sublines either stably overexpressing survivin (PC9S7, PC9S8, HCC827S6, and HCC827S7) or infected with the empty retrovirus (PC9-Mock and HCC827-Mock) were cultured overnight in complete medium, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to survivin or to β-actin. B, PC9 or HCC827 isogenic cell lines were incubated in the presence of the indicated concentrations of gefitinib for 48 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of EGFR, AKT, or ERK, to survivin, to PARP, to caspase-3, or to β-actin. Data in A and B are representative of 3 independent experiments. C, PC9 or HCC827 isogenic cell lines were incubated with gefitinib (0.1 μmol/L) for the indicated times, after which the proportion of apoptotic cells was determined by staining with Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 versus the corresponding value for cells infected with the empty retrovirus. D, PC9 or HCC827 isogenic cell lines were incubated with gefitinib (0.1 μmol/L) for the indicated times and then analyzed for cell cycle distribution by flow cytometry. Data are means of triplicates from representative experiments that were repeated 3 times.

Figure 4.

Effect of survivin overexpression on gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells in vitro. A, parental (P) PC9 or HCC827 cells or corresponding sublines either stably overexpressing survivin (PC9S7, PC9S8, HCC827S6, and HCC827S7) or infected with the empty retrovirus (PC9-Mock and HCC827-Mock) were cultured overnight in complete medium, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to survivin or to β-actin. B, PC9 or HCC827 isogenic cell lines were incubated in the presence of the indicated concentrations of gefitinib for 48 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of EGFR, AKT, or ERK, to survivin, to PARP, to caspase-3, or to β-actin. Data in A and B are representative of 3 independent experiments. C, PC9 or HCC827 isogenic cell lines were incubated with gefitinib (0.1 μmol/L) for the indicated times, after which the proportion of apoptotic cells was determined by staining with Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 versus the corresponding value for cells infected with the empty retrovirus. D, PC9 or HCC827 isogenic cell lines were incubated with gefitinib (0.1 μmol/L) for the indicated times and then analyzed for cell cycle distribution by flow cytometry. Data are means of triplicates from representative experiments that were repeated 3 times.

Close modal

Overexpression of survivin inhibits the antitumor effect of gefitinib on EGFR mutation–positive cells in vivo

To investigate whether the antitumor effect of gefitinib on EGFR mutation–positive NSCLC cells might be affected by survivin overexpression in vivo, we injected HCC827-Mock cells or cells of the survivin-overexpressing subline HCC827S7 into nude mice for elicitation of the formation of solid tumors. When the tumors became palpable (200–400 mm3), mice were divided into 3 groups and treated with vehicle (control) or gefitinib at a daily dose of 10 or 25 mg/kg by oral gavage for 4 weeks. Gefitinib treatment at either dose eradicated tumors in mice injected with HCC827-Mock cells (Fig. 5A and C). In contrast, tumors in mice injected with survivin-overexpressing cells were not eradicated by gefitinib even at the dose of 25 mg/kg per day, although tumor growth was partially inhibited by gefitinib in a dose-dependent manner (Fig. 5B and C). These results showed that survivin overexpression inhibits the antitumor effect of gefitinib on EGFR mutation–positive NSCLC cells in vivo.

Figure 5.

Effect of gefitinib on the growth of EGFR mutation–positive NSCLC cells overexpressing survivin in vivo. A and B, nude mice with tumor xenografts established by subcutaneous injection of HCC827-Mock or HCC827S7 cells, respectively, were treated daily for 4 weeks with vehicle (control) or gefitinib (10 or 25 mg/kg). Tumor volume was determined at the indicated times after the onset of treatment. Data are means ± SE of values from 6 mice per group. C, representative mice showing tumors at the end of the 4-week treatment period.

Figure 5.

Effect of gefitinib on the growth of EGFR mutation–positive NSCLC cells overexpressing survivin in vivo. A and B, nude mice with tumor xenografts established by subcutaneous injection of HCC827-Mock or HCC827S7 cells, respectively, were treated daily for 4 weeks with vehicle (control) or gefitinib (10 or 25 mg/kg). Tumor volume was determined at the indicated times after the onset of treatment. Data are means ± SE of values from 6 mice per group. C, representative mice showing tumors at the end of the 4-week treatment period.

Close modal

Effect of attenuation of BIM induction on gefitinib-induced apoptosis in EGFR mutation–positive cells overexpressing survivin

Survivin overexpression did not completely eliminate gefitinib-induced apoptosis in PC9 and HCC827 cells, suggesting that other signaling pathways might contribute to this process. Induction of the proapoptotic BH3-only protein BIM has been found to be important for EGFR-TKI–induced apoptosis in EGFR mutation–positive lung cancers, and inhibition of the EGFR-MEK-ERK signaling pathway is required for BIM induction (25–27). We therefore examined whether survivin overexpression in combination with specific inhibition of BIM induction results in an additive antiapoptotic effect in EGFR mutation–positive NSCLC cells. We transiently transfected survivin-overexpressing sublines of PC9 or HCC827 cells with an siRNA specific for BIM mRNA. Transfection with the BIM siRNA specifically inhibited the induction of BIM expression by gefitinib in both mock-infected and survivin-overexpressing sublines (Fig. 6A). Staining with Annexin V further revealed that the combination of survivin overexpression and attenuation of BIM induction resulted in a greater level of inhibition of gefitinib-induced apoptosis than that observed with either approach alone (Fig. 6B). These data were confirmed with a second BIM siRNA to rule out off-target effects (Supplementary Fig. 1). These results thus suggested that both survivin downregulation and BIM induction contribute independently to gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells.

Figure 6.

Effect of the combination of survivin overexpression and inhibition of BIM induction on gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells. A, cells stably overexpressing survivin (PC9S7 and HCC827S6) or infected with the empty retrovirus (PC9-Mock and HCC827-Mock) were transfected with BIM (BIM-1) or scrambled siRNAs for 24 hours and then incubated for 24 hours in complete medium with or without gefitinib (0.1 μmol/L). Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to survivin, to BIM, or to β-actin. Data are representative of 3 independent experiments. B, cells transfected as in (A) were incubated for 48 hours in the absence or presence of gefitinib (0.1 μmol/L) and then evaluated for the proportion of apoptotic cells by staining with Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 for the indicated comparisons.

Figure 6.

Effect of the combination of survivin overexpression and inhibition of BIM induction on gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells. A, cells stably overexpressing survivin (PC9S7 and HCC827S6) or infected with the empty retrovirus (PC9-Mock and HCC827-Mock) were transfected with BIM (BIM-1) or scrambled siRNAs for 24 hours and then incubated for 24 hours in complete medium with or without gefitinib (0.1 μmol/L). Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to survivin, to BIM, or to β-actin. Data are representative of 3 independent experiments. B, cells transfected as in (A) were incubated for 48 hours in the absence or presence of gefitinib (0.1 μmol/L) and then evaluated for the proportion of apoptotic cells by staining with Annexin V and propidium iodide followed by flow cytometry. Data are means ± SE from 3 independent experiments. *, P < 0.05 for the indicated comparisons.

Close modal

EGFR-TKIs induce marked clinical responses in patients with NSCLC positive for activating mutations of EGFR (1–3). In vitro experiments have shown that EGFR-TKIs induce a substantial level of apoptosis in NSCLC cell lines expressing mutant EGFRs (4). However, the key downstream mediators of EGFR-TKI–induced apoptosis in EGFR mutation–positive cells have remained unidentified. We have now found that gefitinib downregulated survivin expression in EGFR mutation-positive NSCLC cells but not in NSCLC cells expressing wild-type EGFR or EGFR with the T790M resistance mutation. With the use of specific PI3K inhibitors and siRNAs specific for AKT mRNA, we further showed that the downregulation of survivin expression by gefitinib is likely mediated through inhibition of PI3K-AKT signaling. Human epidermal growth factor receptor 2 (HER2)–targeting agents such as lapatinib and trastuzumab were previously found to induce downregulation of survivin through inhibition of the PI3K-AKT pathway in breast cancer cells positive for HER2 amplification (28, 29). Given that downregulation of survivin through inhibition of the PI3K-AKT pathway was induced by EGFR-TKIs in EGFR mutation–positive NSCLC cells and by HER2-targeting agents in breast cancer cells positive for HER2 amplification, the expression of survivin is likely dependent on PI3K-AKT signaling that operates downstream of receptor tyrosine kinases and is essential for cell survival. This hypothesis is further supported by the observation that transfection of EGFR mutation–positive NSCLC cells with an siRNA specific for EGFR mRNA resulted in marked inhibition of survivin expression, whereas transfection of cells expressing wild-type EGFR had no such effect (Supplementary Fig. 2). The PI3K-AKT pathway has been implicated in the regulation of survivin expression by cytokines, growth factors, and chemotherapeutic drugs (8, 10, 30). Although no direct correlation has been established between downregulation of survivin and inhibition of EGFR signaling, these previous findings support the notion that inhibition of the EGFR-PI3K-AKT pathway contributes to downregulation of survivin expression by EGFR-TKIs in EGFR mutation–positive NSCLC cells.

Survivin has been implicated in resistance of cancer cells to apoptosis, although the effect of survivin expression on gefitinib-induced apoptosis in EGFR mutation–positive NSCLC cells has not previously been examined. We have now shown that survivin overexpression inhibited gefitinib-induced apoptosis in such cells. Inhibition of the PI3K-AKT and MEK-ERK pathways was previously found to account for much of the proapoptotic activity of EGFR-TKIs in EGFR mutation–positive NSCLC cells (31). We further found that overexpression of survivin resulted in inhibition of apoptosis induced by a combination of PI3K and MEK inhibitors in such cells (Supplementary Fig. 3). Increased AKT activity as a result either of the loss of PTEN or of expression of a constitutively active form of AKT was previously found to be associated with a reduced sensitivity to EGFR-TKIs in EGFR mutation–positive NSCLC cells (32). However, the principal molecular target underlying the response to inhibition of PI3K-AKT signaling by EGFR-TKIs has remained to be elucidated. In the present study, we show that the sensitivity of EGFR mutation–positive NSCLC cells to EGFR-TKIs depends, at least in part, on survivin downregulation through inhibition of the PI3K-AKT pathway. In our xenograft model, we showed that survivin overexpression inhibited the antitumor effect of gefitinib on EGFR mutation–positive NSCLC cells. The extent of the clinical benefit of EGFR-TKIs varies among NSCLC patients harboring activating EGFR mutations, and the efficacy of these drugs is limited by either de novo resistance or resistance acquired after the onset of therapy (33). Although several mechanisms of acquired resistance have been described, it remains of clinical concern that molecular markers for prediction of de novo resistance to these drugs have not been well delineated (23, 34–38). It will therefore be of interest to determine whether increased survivin expression in tumors is clinically useful as a negative predictive marker of sensitivity to EGFR-TKIs in patients with EGFR mutation–positive NSCLC.

Our observations revealed that survivin overexpression did not completely abolish gefitinib-induced apoptosis, suggesting that another proapoptotic regulator activated after EGFR inhibition might contribute to EGFR-TKI–induced apoptotic cell death. Previous studies have shown that gefitinib induces BIM expression via inhibition of the MEK-ERK pathway and that BIM induction plays a key role in EGFR-TKI–induced apoptosis in EGFR mutation–positive NSCLC cells (25–27). We have now shown that inhibition of both survivin downregulation and BIM induction attenuated gefitinib-induced apoptosis to a greater extent than did inhibition of either process alone. The recent preclinical study showing that the combination of a PI3K inhibitor and a MEK inhibitor, but neither agent alone, induced substantial growth inhibition in EGFR mutation–positive NSCLC cells (31) supports the notion that both the PI3K-AKT-survivin and MEK-ERK-BIM pathways contribute independently to gefitinib-induced apoptosis in such cells (Fig. 7).

Figure 7.

Proposed model for the intracellular signaling underlying EGFR-TKI–induced apoptosis in EGFR mutation–positive NSCLC cells.

Figure 7.

Proposed model for the intracellular signaling underlying EGFR-TKI–induced apoptosis in EGFR mutation–positive NSCLC cells.

Close modal

In conclusion, we have shown that the EGFR-TKI gefitinib downregulated survivin expression, likely through inhibition of PI3K-AKT signaling, and that this effect plays a key role in gefitinib-induced apoptosis. Moreover, we found that survivin downregulation and BIM induction are independently required for EGFR-TKI–induced apoptosis. Our results thus show that simultaneous upstream interruption of the PI3K-AKT-survivin and MEK-ERK-BIM pathways mediates EGFR-TKI–induced apoptosis.

No potential conflicts or interest were disclosed.

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.
Blanc-Brude
OP
,
Mesri
M
,
Wall
NR
,
Plescia
J
,
Dohi
T
,
Altieri
DC
. 
Therapeutic targeting of the survivin pathway in cancer: initiation of mitochondrial apoptosis and suppression of tumor-associated angiogenesis
.
Clin Cancer Res
2003
;
9
:
2683
92
.
2.
Dohi
T
,
Okada
K
,
Xia
F
, et al
An IAP-IAP complex inhibits apoptosis
.
J Biol Chem
2004
;
279
:
34087
90
.
3.
Li
F
,
Ambrosini
G
,
Chu
EY
, et al
Control of apoptosis and mitotic spindle checkpoint by survivin
.
Nature
1998
;
396
:
580
4
.
4.
Ambrosini
G
,
Adida
C
,
Altieri
DC
. 
A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma
.
Nat Med
1997
;
3
:
917
21
.
5.
Ambrosini
G
,
Adida
C
,
Sirugo
G
,
Altieri
DC
. 
Induction of apoptosis and inhibition of cell proliferation by survivin gene targeting
.
J Biol Chem
1998
;
273
:
11177
82
.
6.
Kallio
MJ
,
Nieminen
M
,
Eriksson
JE
. 
Human inhibitor of apoptosis protein (IAP) survivin participates in regulation of chromosome segregation and mitotic exit
.
FASEB J
2001
;
15
:
2721
3
.
7.
Li
F
,
Altieri
DC
. 
The cancer antiapoptosis mouse survivin gene: characterization of locus and transcriptional requirements of basal and cell cycle-dependent expression
.
Cancer Res
1999
;
59
:
3143
51
.
8.
Carter
BZ
,
Milella
M
,
Altieri
DC
,
Andreeff
M
. 
Cytokine-regulated expression of survivin in myeloid leukemia
.
Blood
2001
;
97
:
2784
90
.
9.
Fang
ZH
,
Dong
CL
,
Chen
Z
, et al
Transcriptional regulation of survivin by c-Myc in BCR/ABL-transformed cells: implications in anti-leukaemic strategy
.
J Cell Mol Med
2009
;
13
:
2039
52
.
10.
Beierle
EA
,
Nagaram
A
,
Dai
W
,
Iyengar
M
,
Chen
MK
. 
VEGF-mediated survivin expression in neuroblastoma cells
.
J Surg Res
2005
;
127
:
21
8
.
11.
Tran
J
,
Master
Z
,
Yu
JL
,
Rak
J
,
Dumont
DJ
,
Kerbel
RS
. 
A role for survivin in chemoresistance of endothelial cells mediated by VEGF
.
Proc Natl Acad Sci U S A
2002
;
99
:
4349
54
.
12.
Fan
J
,
Wang
L
,
Jiang
GN
,
He
WX
,
Ding
JA
. 
The role of survivin on overall survival of non-small cell lung cancer, a meta-analysis of published literatures
.
Lung Cancer
2008
;
61
:
91
6
.
13.
Huang
CL
,
Liu
D
,
Nakano
J
, et al
E2F1 overexpression correlates with thymidylate synthase and survivin gene expressions and tumor proliferation in non small-cell lung cancer
.
Clin Cancer Res
2007
;
13
:
6938
46
.
14.
Krepela
E
,
Dankova
P
,
Moravcikova
E
, et al
Increased expression of inhibitor of apoptosis proteins, survivin and XIAP, in non-small cell lung carcinoma
.
Int J Oncol
2009
;
35
:
1449
62
.
15.
Mendelsohn
J
,
Baselga
J
. 
The EGF receptor family as targets for cancer therapy
.
Oncogene
2000
;
19
:
6550
65
.
16.
Schlessinger
J
. 
Cell signaling by receptor tyrosine kinases
.
Cell
2000
;
103
:
211
25
.
17.
Hynes
NE
,
Lane
HA
. 
ERBB receptors and cancer: the complexity of targeted inhibitors
.
Nat Rev Cancer
2005
;
5
:
341
54
.
18.
Paez
JG
,
Janne
PA
,
Lee
JC
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.
19.
Shepherd
FA
,
Pereira
J Rodrigues
,
Ciuleanu
T
, et al
Erlotinib in previously treated non-small-cell lung cancer
.
N Engl J Med
2005
;
353
:
123
32
.
20.
Sordella
R
,
Bell
DW
,
Haber
DA
,
Settleman
J
. 
Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways
.
Science
2004
;
305
:
1163
7
.
21.
Tracy
S
,
Mukohara
T
,
Hansen
M
,
Meyerson
M
,
Johnson
BE
,
Janne
PA
. 
Gefitinib induces apoptosis in the EGFRL858R non-small-cell lung cancer cell line H3255
.
Cancer Res
2004
;
64
:
7241
4
.
22.
Ling
YH
,
Lin
R
,
Perez-Soler
R
. 
Erlotinib induces mitochondrial-mediated apoptosis in human H3255 non-small-cell lung cancer cells with epidermal growth factor receptorL858R mutation through mitochondrial oxidative phosphorylation-dependent activation of BAX and BAK
.
Mol Pharmacol
2008
;
74
:
793
806
.
23.
Koizumi
F
,
Shimoyama
T
,
Taguchi
F
,
Saijo
N
,
Nishio
K
. 
Establishment of a human non-small cell lung cancer cell line resistant to gefitinib
.
Int J Cancer
2005
;
116
:
36
44
.
24.
Tanaka
K
,
Arao
T
,
Maegawa
M
, et al
SRPX2 is overexpressed in gastric cancer and promotes cellular migration and adhesion
.
Int J Cancer
2009
;
124
:
1072
80
.
25.
Costa
DB
,
Halmos
B
,
Kumar
A
, et al
BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations
.
PLoS Med
2007
;
4
:
1669
79
;
discussion 80
.
26.
Cragg
MS
,
Kuroda
J
,
Puthalakath
H
,
Huang
DC
,
Strasser
A
. 
Gefitinib-induced killing of NSCLC cell lines expressing mutant EGFR requires BIM and can be enhanced by BH3 mimetics
.
PLoS Med
2007
;
4
:
1681
89
;
discussion 90
.
27.
Gong
Y
,
Somwar
R
,
Politi
K
, et al
Induction of BIM is essential for apoptosis triggered by EGFR kinase inhibitors in mutant EGFR-dependent lung adenocarcinomas
.
PLoS Med
2007
;
4
:
1655
68
.
28.
Asanuma
H
,
Torigoe
T
,
Kamiguchi
K
, et al
Survivin expression is regulated by coexpression of human epidermal growth factor receptor 2 and epidermal growth factor receptor via phosphatidylinositol 3-kinase/AKT signaling pathway in breast cancer cells
.
Cancer Res
2005
;
65
:
11018
25
.
29.
Xia
W
,
Bisi
J
,
Strum
J
, et al
Regulation of survivin by ErbB2 signaling: therapeutic implications for ErbB2-overexpressing breast cancers
.
Cancer Res
2006
;
66
:
1640
7
.
30.
Zhao
P
,
Meng
Q
,
Liu
LZ
,
You
YP
,
Liu
N
,
Jiang
BH
. 
Regulation of survivin by PI3K/Akt/p70S6K1 pathway
.
Biochem Biophys Res Commun
2010
;
395
:
219
24
.
31.
Faber
AC
,
Li
D
,
Song
Y
, et al
Differential induction of apoptosis in HER2 and EGFR addicted cancers following PI3K inhibition
.
Proc Natl Acad Sci U S A
2009
;
106
:
19503
8
.
32.
Sos
ML
,
Koker
M
,
Weir
BA
, 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
.
33.
Okamoto
I
. 
Epidermal growth factor receptor in relation to tumor development: EGFR-targeted anticancer therapy
.
FEBS J
2009
;
277
:
309
15
.
34.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
35.
Ercan
D
,
Zejnullahu
K
,
Yonesaka
K
, et al
Amplification of EGFR T790M causes resistance to an irreversible EGFR inhibitor
.
Oncogene
2010
;
29
:
2346
56
.
36.
Pao
W
,
Miller
VA
,
Politi
KA
, 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
:
225
35
.
37.
Yano
S
,
Wang
W
,
Li
Q
, et al
Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor-activating mutations
.
Cancer Res
2008
;
68
:
9479
87
.
38.
Takeda
M
,
Okamoto
I
,
Fujita
Y
, et al
De novo resistance to epidermal growth factor receptor-tyrosine kinase inhibitors in EGFR mutation-positive patients with non-small cell lung cancer
.
J Thorac Oncol
2010
;
5
:
399
400
.