Inhibition of RPTOR overcomes resistance to EGFR inhibition in triple-negative breast cancer cells

  • Authors:
    • Kyu Sic You
    • Yong Weon Yi
    • Sahng-June Kwak
    • Yeon-Sun Seong
  • View Affiliations

  • Published online on: January 15, 2018     https://doi.org/10.3892/ijo.2018.4244
  • Pages: 828-840
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Triple-negative breast cancer (TNBC) cells frequently exhibit activated growth factor signaling and resistance to inhibitors for epidermal growth factor receptor (EGFR), despite the overexpression of EGFR protein, and this is associated with a malignant behavior and a poor prognosis. In this study, to elucidate the underlying mechanisms of resistance to EGFR inhibitor and identify inhibitors that exert a synergistic effect with EGFR inhibition, we examined the inhibitory effects of selected protein kinase inhibitors (PKIs) in combination with gefitinib on the viability of a mesenchymal stem-like (MSL) subtype TNBC cell line. MK‑2206, an AKT inhibitor, and a group of mammalian target of rapamycin (mTOR) inhibitors were found to exert synergistic lethal effects in combination with gefitinib in MDA‑MB‑231 cells. The combination of gefitinib/MK‑2206 exerted a prominent synergistic lethal effect in an MTT cell viability assay and a growth inhibitory effect in a long-term colony-forming assay in 2 MSL subtype TNBC cell lines (MDA‑MB‑231 and HS578T) and one basal-like (BL) subtype TNBC cell line (MDA‑MB-468). Gefitinib/MK‑2206 treatment synergistically decreased the mTOR signaling target substrates along with the downregulation of ribosomal protein S6 (RPS6), a marker of cell proliferation and target substrate of the AKT-mTOR signaling pathway. In addition, gefitinib markedly reduced the viability of MDA‑MD‑231 and HS578T cells when regulatory-associated protein of mTOR (RPTOR) was suppressed by siRNA-based knockdown (KD). These results thus suggest that RPTOR mediates, at least partially, the resistance to EGFR inhibition in TNBC cells. Therefore, targeting the mTOR complex 1 (mTORC1) pathway may be a potential strategy for the treatment of EGFR-resistant TNBC.

Introduction

Three signaling receptors are considered as important markers for the grouping of human breast cancer types and the subsequent planning of systemic therapy for breast cancer: Estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) (1). Triple-negative breast cancer (TNBC), which comprises 15–20% of breast cancers (26), is characterized by the immunohistochemical absence of both ER and PR, as well as a lack of the amplification of HER2. Due to the absence of these proteins, the treatment of TNBC with agents targeting these proteins is inadequate. Although conventional adjuvant chemotherapies employed, such as taxanes, anthracyclines, capecitabines and/or platins show a desirable response with a higher rate of pathological complete response (pCR) in initial treatment (7); drug resistance to these agents and distant metastasis commonly occur and are associated with an overall poor prognosis (810).

The human epidermal growth factor receptor (HER) family consists of 4 structurally related receptor tyrosine kinases: HER1 [epidermal growth factor receptor (EGFR)], HER2, HER3, and HER4 (1113). Since EGFR is a well-established oncogenic factor and therapeutic target in certain types of human cancer, a number of humanized monoclonal antibodies and small molecule kinase inhibitors have been approved by the US Food and Drug Administration (FDA) for the treatment of several human cancers (12,13). Unlike other types of hormone therapy-sensitive breast cancer, the overexpression of EGFR is more frequently found in TNBC (1417) and is closely associated with the aggressiveness and drug resistance of malignant cancers, including breast cancer (11,13). Up to 70–80% of metastatic breast cancers have been shown to overexpress EGFR but not to overexpress HER2, the basis for HER2-targeted therapy (18,19). The activation of EGFR transmits signals of cell proliferation, cell survival, drug resistance and metastasis via various signaling cascades, including RAS-mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)-AKT and SRC-signal transducer and activator of transcription (STAT) pathways (20). Although numerous EGFR target drugs, including protein kinase inhibitors (PKIs) and monoclonal antibodies, are easily available, the majority of studies on the therapeutic potential of EGFR targeted therapy have focused on lung cancer, glioblastoma, colon cancer and head and neck cancers (21 and refs therein). In addition, despite the frequent overexpression of EGFR in TNBC, the study of its potential role or therapeutic benefits in TNBC remains still limited and is still under evaluation (11,21,22).

Previously, we identified a potential therapeutic benefit of blocking EGFR and PI3K with a gefitinib/PI-103 combination treatment in basal-like (BL) subtypes of TNBC in vitro (23). However, a mesenchymal stem-like (MSL) subtype of TNBC cells is relatively resistant to this combination (23), probably due to the presence of alternative intracellular signaling pathways that ensure cell survival and proliferation even when EGFR is blocked (2429). We have also reported that the additional blocking of MET coupled with EGFR inhibition effectively exerts anticancer effects in TNBC cells of the MSL subtype with a decrease in p-/total RPS6 levels (30). RPS6 is one of the downstream target of mTOR kinase signaling. In addition, the PI3K-AKT-mTOR signaling pathway, which is frequently deregulated in TNBC, is indispensable for cancer cell survival and is associated with drug resistance (31). In this study, to identify PKIs that induce synthetic lethality in combination with an EGFR inhibitor, we performed an MTT screening in MDA-MB-231 cells with PKIs, including target agents against PI3K, AKT, mTOR and SRC pathways in combination with gefitinib. As a result, we identified and further characterized an AKT inhibitor, MK-2206, as a synthetic lethal agent when used in combination with gefitinib in TNBC cells of the MSL subtype and demonstrated that the antitumor effect was associated with the downregulation of the mammalian target of rapamycin complex 1 (mTORC1)/regulatory-associated protein of mTOR (RPTOR) signaling coupled with a downregulation in the levels of ribosomal protein S6 (RPS6).

Materials and methods

Cell culture and reagents

The MDA-MB-231, HS578T and MDA-MB-468 cell lines were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA) and maintained in Dulbecco's modified Eagle's medium (DMEM; HyClone, Logan, UT, USA) containing 10% heat-inactivated fetal bovine serum (FBS; Corning, Manassas, VA, USA) and 100 units/ml penicillin/streptomycin (Gibco, Grand Island, NY, USA). Cell growth was monitored by trypan blue dye (Sigma-Aldrich St. Louis, MO, USA) exclusion cell counting using the Luna Automated Cell Counter (Logos Biosystems, Gyeonggi-do, Korea). The PKIs used in these experiments were purchased from the following sources: AZD0530, deforolimus, GDC-0941, GSK1059615, IC-87114, KU-0063794, MK-2206, Perifosine, PIK-90, PIK-75, PI-103 TG100-115, TGX221 and WYE-354 were all obtained from SelleckChem (Houston, TX, USA); (−)-Deguelin was purchased from Enzo Life Sciences, Inc. (Farmingdale, NY, USA); BEZ235, dasatinib, everolimus, FK506, pimecrolimus, staurosporine, temsirolimus and ZSTK474 were all obtained from LC Laboratories (Woburn, MA, USA); and LY294002, rapamycin and wortmannin were obtained from Sigma-Aldrich. Each compound was dissolved in dimethyl sulfoxide (DMSO) (Sigma-Aldrich) to create a stock solution and stored at −20°C in small aliquots.

Screening of PKIs

Synthetic lethal screening was performed as previously described (30). In brief, the MDA-MB-231 cells, plated at 1,000 cells/well in 96-well plates 24 h prior to testing, were treated with increasing concentrations of gefitinib and PKIs in duplicate in a 6×5 matrix. Cell viability was determined at 72 h after treatment by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay with 4 mg/ml MTT solution as previously described (25,33). The synergism was determined by calculating the classification index (CI) as previously described (30,32) as follows: (viability of gefitinib) × (viability of PKI)/(viability of the gefitinib and PKI combination). Supra-additivity was defined as CI >1; additivity was defined as CI=1; and subadditivity was defined as CI <1 (32,33). The numbers of combination points with CI values >1.3 that were generated from individual drug concentration points were summated and further assigned as a quantitative index of synergism. The average of CIs >1.3 was used as the qualitative index of synergism.

Clonogenic cell survival assay

The cells were subcultured in 6-well plates at appropriate densities: 1,000 cells/well for the HS578T cells and 2,000 cells/well for the MDA-MB-231 and MDA-MB-468 cells. At 1 day after subculture, the cells were treated with 9 µM gefitinib, 9 µM MK-2206, or a combination of these drugs for 24 h, and then washed and supplemented with fresh normal growth media in the absence of the drugs. The cells were cultured for an additional 10–14 days following treatment with replacement of normal growth media twice per week. The colonies were stained as previously described (30,34). After washing with distilled water (DW), the colonies were imaged with a scanner. The relative amount and number of colonies were quantified as follows: The crystal violet stain for the staining of the colonies was dissolved in a solubilizing buffer [1:1 mixture (v/v) of 0.1 M sodium phosphate buffer (pH 4.5) and ethanol], and the absorbance of the dissolved crystal violet was measured by using a 680 micro-plate reader (Bio-Rad, Hercules, CA, USA).

Western blot analysis and antibodies

For PKI treatment, the HS578T and MDA-MB231 cells were plated at 2×105/60-mm dish at 1 day prior to treatment. The cells were then treated with gefitinib and MK-2206 for 24 or 2 h in the presence of normal serum-containing media. For siRNA transfection, the HS578T and MDA-MB231 cells were plated at 5×104 cells/60-mm dish the day prior to transfection. Following treatment with the drug or siRNA transfection, the cells were lysed with Pierce™ RIPA buffer (Thermo Fisher Scientific, Waltham, MA, USA) containing protease phosphatase inhibitor cocktail (Thermo Fisher Scientific), and the protein concentration was determined with a BCA protein assay kit (Thermo Fisher Scientific). The following antibodies were used in this study: mTOR (#4517), phosphorylated (p-)mTOR (S2448; #2971), extracellular signal-regulated kinase (ERK)1/2 (#9102), p-ERK1/2 (T202/Y204; #4370), SRC (#2108), p-SRC (Y416; #2101), eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1; #9452), p-4E-BP1 (T37/46; #2855), RPS6 (#2217), p-RPS6 (#4856), proline-rich AKT substrate of 40 kDa (PRAS40; #2691), p-PRAS40 (S235/236; #13175), AKT (#9272), p-AKT (S473; #4060), glycogen synthase kinase (GSK)-3β (#9315), p-GSK-3β (S9); #9315), RPTOR (#2280), rapamycin-insensitive companion of mTOR (RICTOR) (#2140), poly(ADP-ribose) polymerase-1 (PARP1; #9542), cleaved PARP (Asp214; #5625), cleaved caspase-3 (Asp175; #9661) and peroxidase-conjugated secondary antibodies (anti-rabbit IgG, #7074; anti-mouse IgG, #7076) were all obtained from Cell Signaling Technology (Danvers, MA, USA); X-linked inhibitor of apoptosis protein antibody (XIAP; #610716) was from BD Sciences (San Jose, CA, USA); β-actin antibody (A330-491A) was from Bethyl Laboratories, Inc. (Montgomery, TX, USA); and β-tubulin antibody (T7816) was from Sigma-Aldrich.

Transfection of siRNA

For the siRNA-mediated knockdown of target gene expression, AccuTarget™ premade siRNA was purchased from Bioneer (Seoul, Korea) for the following target genes and control siRNA: Human RPTOR (gene ID: 57521; #1079283), human RICTOR (gene ID: 253260; #1129356) and control siRNA (SN-1003). The transfection of siRNA was carried out using Lipofectamine 2000 (Invitrogen, Waltham, MA, USA) as previously described (30). In brief, the HS578T (1×105 cells/well) or MDA-MB-231 (5×104 cells/well) cells in 6-well plates were transfected with 100 pmoles of siRNA mixed and pre-incubated with Lipofectamine 2000 in serum-free DMEM media. Following 4 h of incubation, the transfection media was supplemented with an equal volume of DMEM containing 20% FBS and 200 units/ml penicillin/streptomycin to restore the normal growth condition, and the cells were further incubated for 3 days. During the incubation, 9 µM gefitinib or DMSO (vehicle) were added 1 day after the media supplement or the last 2 h of the 3rd day of the incubation period for the cell proliferation assay and western blot analyses.

Cell counting-based cell viability assay and cell proliferation assay

The HS578T and MDA-MB231 cells were plated at 1×105 cells in a 60-mm dish at 1 day prior to treatment. At the indicated time of siRNA transfection or drug treatment (0, 1, 2, 3 and 4 days), cells were detached with trypsin/EDTA (Gibco) and suspended with 0.5 ml of DMEM containing 10% FBS. Cell proliferation or number of viable cells were determined by the trypan blue exclusion method using the Luna Automated Cell Counter (Logos Biosystems) immediately after staining with trypan blue dye (Sigma-Aldrich) to avoid cell death caused by prolonged incubation (34).

Cell cycle analysis

The MDA-MB231 cells were plated at 1×105 cells in a 60-mm dish at 1 day prior to treatment. Following treatment with the drugs, both attached and floating cells were harvested and fixed with 70% ethanol at −20°C for >4 h. The nuclei were then stained with propidium iodide (Sigma-Aldrich). The chromosomal DNA content was measured using a FACSCalibur flow cytometer (BD Sciences, Franklin Lakes, NJ, USA) according to the manufacturer's instructions. The data were analyzed using CellQuest Pro (BD Sciences) and the ModFit LT program (Verity Software House, Topsham, ME, USA).

Statistical analysis

Triplicate experiments were repeated 3 times. Most of the data are presented as the means ± standard deviation (SD). One-way analysis of variance (ANOVA) with a post hoc Tukey's honest significant difference (HSD) test was used to analyze the significance of the differences between groups. Conventionally, values of P<0.05 and P<0.01 were considered to indicate statistically significant and highly statistically significant differences, respectively.

Results

Identification of a set of small molecule PKIs with synergistic effects in combination with gefitinib

In our previous studies, the combination of an EGFR inhibitor (EGFRi) and PI3K/AKT inhibitor or MET inhibitors was found to decrease the viability and proliferation of TNBC cells of the BL subtype or MSL subtype, respectively (23,30). In this study, to further identify PKIs that are effective in the presence of an EGFR inhibitor in TNBC cells of the MSL subtype, a set of known PKIs (Table I) was screened with gefitinib in the MDA-MB231 cell line, and inhibitors of the PI3K/AKT and mTOR signaling pathways were identified (Fig. 1A). For example, GSK1059615 (a PI3K/mTOR inhibitor), MK-2206 (an AKT inhibitor) and rapamycin (an mTOR inhibitor) exerted synergistic effects with gefitinib at various concentrations (Fig. 1A and B). Although MK-2206/gefitinib treatment exhibited fewer points of synergy than that of treatment with rapamycin, it exerted the highest average CI, reflecting strong synergy. The majority of the mTOR pathway inhibitors exerted a synergistic effect at 4–10 drug combination points (Fig. 1A); the overall lethal effects of rapamycin or other mTOR inhibitors in combination with gefitinib were not as potent as those of MK-2206 in terms of the average CI value (Fig. 1).

Table I

List of protein kinase inhibitors used in this study.

Table I

List of protein kinase inhibitors used in this study.

InhibitorOther nameKnown targets [IC50 value (nM)](Refs.)
AZD0530SaracatinibSRC (2.7), LCK (<4), YES (4), EGFR (L861Q) (4), LYN (5), EGFR (L858R) (5), FYN (10), FGR (10), BLK (11), ABL (30), EGFR (66), KIT (200)(60)
BEZ235NVP-BEZ235PI3Kα (4/2), PI3Kβ (75), PI3Kδ (7), PI3Kγ (5), mTOR (20.7/2), DNAPK (5), ATM (7), ATR (21)(61,62)
DasatinibBMS354825, SprycelBCR-ABL (0.8), SRC (0.5), LCK (0.4), YES (0.5), BLK (8), KIT (5), PDGFRβ (28), p38 (100), EGFR (180), EPHB2 (4.4), TXK (<0.3)(63,64)
DeforolimusRidaforolimus AP23573 MK-8669mTOR (0.2)(65)
(−) DeguelinAKT signaling, Hsp90(66)
EverolimusSDZ-RAD, Certican, RAD001mTOR (0.63)(67)
FK-506Tacrolimus, Fujimycin, PrografFKBP12(68)
GDC-0941PI3Kα (3), PI3Kβ (33), PI3Kδ (3), PI3Kγ (75), PI3KC2β (670), mTOR (580)(69)
GSK1059615PK3Kα (0.4), PI3Kβ (0.6), PI3Kγ (5), PI3Kδ (2), mTOR (12)(70)
IC-87114D-030PI3Kδ (130)(71)
KU-0063794mTOR1 (10), mTOR2 (10)(72)
LY294002PI3K (1400), PI3Kα (500) PI3Kδ (570), PI3Kβ (970)(73)
MK-2206AKT1 (5), AKT2 (12), AKT3 (65)(74)
PerifosineKRX-0401AKT translocation inhibitor(75)
PI-103DNAPK (2), PI3Kα (8), PI3Kβ (88), PI3Kδ (48), PI3Kγ (150), mTORC1 (20), mTORC2 (83)(71)
PIK-75DNAPK (2), PI3Kα (5.8), PI3Kγ (76)(71)
PIK-90PI3KC2a (47), PI3KC2b (64), DNAPK (13), PI3Kα (11), PI3Kβ (350), PI3Kδ (58), PI3Kγ (18)(71)
PimecrolimusElidel, SDZ-ASM-981FKBP12(76)
RapamycinRAPA, SirolimusmTOR (0.1)(77)
TemsirolimusCCI779, ToriselmTOR (1.76)(78)
TG100–115PI3Kγ (83), PI3Kσ (235)(79)
TGX221PI3Kβ (10), PI3Kδ (65)(79)
WortmanninKY12420PI3K (0.3), AKT1 (35), AKT2 (30), AKT3 (60), PKCζ (24), p38α (6), p38β (2)(80)
WYE-354mTOR (5)(81)
ZSTK474PI3Kα, PI3Kβ (17), PI3Kγ (53), PI3Kδ (6)(82)
Gefitinib/MK-2206 combination decreases the viability and proliferation of TNBC cells

Among these PKIs, an allosteric AKT inhibitor, MK-2206, was further determined to exert potent synergistic anti-proliferative effects in 2 cell lines of the MSL subtype (MDA-MB-231 and HS578T cells) and 1 cell line of the BL subtype (MDA-MB-468 cells). As was expected, the MDA-MB-468 cells exhibited greater sensitivity following combination treatment with synergistic effect, as well as following treatment with gefitinib or MK-2206 alone (Fig. 2A). Potent synergistic effects were observed in the 2 cell lines of the MSL subtype. In the MDA-MB-231 cells, gefitinib and MK-2206 alone had little or no effect on cell viability; however, the combination of these inhibitors exerted marked synergistic effects (Fig. 2A). The effects of treatment with the gefitinib/MK-2206 combination were further assessed using a long-term colony formation assay. Cells plated in 6-well plates were treated as indicated for 24 h and further cultivated in normal growth media for 14 days. All the TNBC cells tested were minimally affected by treatment with gefitinib or MK-2206 alone. However, the long-term survivals were significantly reduced by the combination treatment (Fig. 2B). The effects of the gefitinib/MK-2206 combination on cell proliferation were also assessed by viable cell counting over time. As shown in Fig. 2C, the gefitinib/MK-2206 combination markedly suppressed the proliferation of the HS578T and MDA-MB-231 cells.

The gefitinib/MK-2206 combination reduces the protein level of RPS6 in TNBC cells of the MSL subtype

To determine the intracellular signaling pathway responsible for the effects of the gefitinib/MK-2206 combination, a series of western blot analyses were carried out. First, the cells were treated with increasing concentrations of gefitinib and MK-2206 in an equimolar ratio for 24 h, and the levels of p-mTOR (S2448), p-SRC (Y416) and p-RPS6 (S235/236) were examined. Of note, both the phosphorylated and total RPS6 levels were decreased in a concentration-dependent manner both in the cells treated with MK-2206 alone and in those that were treated with the gefitinib/MK-2206 combination (Fig. 3A). However, the decrease in the phosphorylated and total RPS6 levels was more prominent with the combination treatment. Previously, we found a similar synergistic decrease in the RPS6 levels following combination treatment with gefitinib and SU11274, a MET inhibitor, in the same TNBC cell line (30). As the overexpression of EGFR in breast cancer is often accompanied by the co-overexpression of SRC, providing synergistic tumorigenic effects between EGFR and SRC (35), TNBC cells of the MSL type exhibit greater sensitivity to dasatinib, an SRC and ABL kinase inhibitor (5,36). Therefore, in this study, we also examined the changes in SRC expression following treatment with gefitinib/MK-2206. The level of p-SRC (Y416) (37) was not affected under these conditions (Fig. 3A). However, the level of p-mTOR (S2448) (38) was more markedly decreased by the gefitinib/MK-2206 combination than with treatment with either agent alone (Fig. 3A).

The effect of the gefitinib/MK-2206 combination was further analyzed with a fixed concentration of both PKIs. Unlike p-ERK1/2 (T202/Y204), which exhibited no apparent change in the MDA-MB-231 cells and even increased in the HS578T cells, the expression of p-AKT (S473) and p-PRAS40 (T246) (39), a downstream substrate of AKT, disappeared in the cells treated with MK-2206 and the gefitinib/MK-2206 combination (Fig. 3B). In addition, the level of p-mTOR was decreased by the combination treatment. The phosphorylation of 4E-BP1, which is known to be mediated by mTORC1 (39,40), at the threonine 37 and 46 residues, was apparently diminished by gefitinib/MK-2206 combination treatment.

To identify the immediate intracellular signaling changes that occur prior to the onset of autonomous feedback regulation, we further assessed the immediate-early phosphorylation status of the AKT and mTOR signaling molecules. As shown in Fig. 3C, the level of p-ERK1/2 (T202/Y204) was not affected, even in the HS578T and MDA-MB-231 cells treated with the gefitinib/MK-2206 combination, suggesting that ERK1/2 may be uncoupled from EGFR in these cells. By contrast, p-AKT (S473) expression disappeared both in the cells treated with MK-2206 alone and in those treated with the gefitinib/MK-2206 combination (Fig. 3C). Accordingly, the phosphorylation of AKT substrates such as PRAS40 (T246) and GSK-3β (S9) was reduced in MK-2206-treated cells and further decreased in gefitinib/MK-2206 combination-treated cells within 2 h. However, the phosphorylation status of mTOR (S2448) differed between the HS578T and MDA-MB-231 cells. The level of p-mTOR was synergistically decreased in the HS578T cells, whereas the change in p-mTOR expression in the MDA-MB-231 cells was not as evident. Under these conditions, the level of p-4E-BP1 (T37) was synergistically decreased by the gefitinib/MK-2206 combination treatment. Of note, the levels of phosphorylated and total RPS6 were decreased as early as 2 h after treatment in both cell lines, and the gefitinib/MK-2206 combination further decreased the levels of phosphorylated/total RPS6 in both the HS578T and MDA-MB231 cells. A similar early decrease in p-RPS6 and RPS6 expression within 2 h was also previously observed with MET inhibitor/gefitinib combination treatment in the same TNBC cell lines (30).

The gefitinib/MK-2206 combination decreases XIAP expression without affecting the levels of apoptotic markers

As the gefitinib/MK-2206 combination effectively inhibited cell proliferation and clonogenicity, we analyzed cell cycle progression following treatment with the drug. The MDA-MB-231 cells were treated as indicated for 72 h and were then subjected to flow cytometric analysis. As shown in Fig. 4A, no apparent cell cycle arrest or accumulation of apoptotic cells was observed in the MDA-MB-231 cells. We also used western blot analysis to examine PARP cleavage and XIAP expression following treatment of the HS578T and MDA-MB-231 cells with the drugs for 24 or 72 h. As shown in Fig. 4B, the expression of full-length PARP was decreased by treatment with the drugs in both cell lines. However, the cleaved product of PARP was not observed. The level of XIAP was decreased in the cells treated with the gefitinib/MK-2206 combination for 72 h. Additional western blot analyses were performed using antibodies for the cleaved forms of caspase-3 and PARP, and staurosporine-treated samples were simulatnously tested as positive controls. Staurosporine induced the cleavage of both caspase-3 and PARP in the TNBC cells tested. On the contrary, no evident cleavage of capase-3 and PARP was observed in the TNBC cells treated with gefitinib, MK-2206 or their combination (Fig. 4C).

Gefitinib with RPTOR knockdown decreases the level of p-/total RPS6 and the viability of TNBC cells of the MSL subtype

In addition to MK-2206, several mTOR pathway inhibitors and a subset of PI3K/AKT pathway inhibitors consistently exhibited synergism with gefitinib in the initial PKI screening (Fig. 1). Based on these findings, we speculated that the resistance to EGFR inhibition and the effects of the gefitinib/MK-2206 combination in TNBC cells of the MSL subtype are to be attributed to the mTOR pathway. mTOR is a protein kinase in two distinct protein complexes, mTORC1 and mTORC2. Therefore, to further elucidate which mTOR pathway contributes to the resistance to EGFR inhibition, the mTORC1 and mTORC2 pathways were selectively blocked by the siRNA-based knockdown (KD) of RPTOR or RICTOR, respectively, in both the MDA-MB231 and HS578T cell lines. RPTOR KD decreased the level of phosphorylated/total RPS6, and the effect was potentiated by the addition of gefitinib in both cell lines (Fig. 5A). However, RICTOR KD with gefitinib treatment did not affect the level of RPS6 protein. As RPTOR KD potentiated the de-phosphorylation of RPS6, we examined the effects of the gefitinib/RPTOR KD combination on cell proliferation. As shown in Fig. 5B, the percentage cell viability was significantly decreased to approximately 50% inhibited by the combination of RPTOR KD and gefitinib treatment. To determine the synergistic inhibitory effects on cell proliferation, the CI values (33) of each group after RPTOR KD and/or gefitinib treatment were calculated. The CI values from 5 independent cell proliferation experiments indicated that this combination was supra-additive, with mean CI values of 1.38±0.31 and 1.34±0.25 in the HS578T and MDA-MB-231 cells, respectively.

Discussion

Due to the lack of appropriate therapies and a poor prognosis of patients with TNBC, the identification of a 'druggable' target is an unmet requirement for the development of an effective TNBC therapy. As numerous types of cancer, including breast cancer have limited driver oncogene activation or tumor suppressor gene mutations (41), proper control of the key surrogate/redundant pathway crosstalk involved in EGFR resistance can provide appropriate therapeutic strategies against the particular type of breast cancer with EGFR activation/overexpression (2224,2628). Among the TNBC subgroups classified according to Lehmann et al, the BL and MSL subtype exhibits activated EGFR signaling (5). In addition to the activation of the epithelial-mesenchymal transition, the MSL-type TNBC exhibits relatively greater resistance to EGFR inhibitors than the BL subtype, regardless of the over-expression status of EGFR (23,30).

In this study, we screened a series of known PKIs in the presence of gefitinib in typical EGFR inhibitor-resistant TNBC cells of the MSL subtype (MDA-MB-231 cells) and identified that MK-2206, an AKT inhibitor, exerts a potent lethal effect with gefitinib combination treatment. In addition, a group of inhibitors against mTOR kinase, including rapamycin, temsirolimus, WYE-354 and GSK1059615 also synergistically inhibited cell viability with gefitinib at multiple points of combination than those of MK2206. However, to investigate resistance to gefitinib in TNBC cells of the MSL-subtype, the investigation of the effects of the MK-2206/gefitinib combination, which exhibited potent lethal synergistic effects (average CI value, 4.21; Fig. 1A), would be more feasible to dissect the downstream signaling cascade to unveil the mechanism of resistance to EGFR inhibitors. MK-2206 treatment alone, which effectively blocked AKT activation and phosphorylation of its downstream substrates, had minimal effect in TNBC cells, indicating that EGFR signaling is indispensable for TNBC cell viability. In addition, gefitinib alone had little effect on the TNBC cells of the MSL subtype tested, whereas the gefitinib/MK-2206 combination was synthetic lethal. The synergistic effects of the gefitinib/MK-2206 combination were also demonstrated by a long-term clonogenic assay following a 24-h treatment with this combination. These results suggest that the AKT-mTOR pathway may contribute to resistance to EGFR inhibitors in MSL TNBC cells.

The decreased phosphorylation of both mTOR and 4E-BP1 (Thr 37/46) suggested that the nodal signaling pathways of the MK-2206/gefitinib combination converge on mTOR signaling. As a downstream signal transducer of the PI3K/AKT pathway, mTORC1, composed of mTOR, RPTOR, PRAS40, mammalian lethal with SEC13 protein 8 (mLST8) and DEP-domain-containing mTOR-interacting protein (DEPTOR), mediates protein synthesis for cell proliferation and cell cycle progression by promoting mRNA translocation as a serine-threonine kinase (42,43). A closely related, but structurally heterologous complex, mTORC2 [composed of mTOR, RICTOR, GβL, mammalian stress-activated protein kinase interacting protein-1 (mSIN1), Protor 1/2 and DEPTOR] mediates AKT activation and cytoskeletal actin remodeling (43,44). mTOR pathway activation is more frequent in TNBC than non-TNBC and is associated with a poor prognosis in early-stage TNBC (45,46). In addition, the activated PI3K mutation and loss of PTEN function, along with the recent identification of mTOR activation in TNBC, rationalizes the use of mTOR-targeted therapy (31,47). The safety and efficacy of an oral mTOR inhibitor, everolimus, was previously evaluated in phase II clinical studies of patients with metastatic or recurrent breast cancer (48). In a recent report of phase II clinical trials, the use of everolimus in combination with paclitaxel and cisplatin neoadjuvant therapy in TNBC patients revealed more adverse effects without a clinically significant improvement (49). Other independent phase II clinical trials of everolimus combined with conventional chemotherapeutics, such as paclitaxel with anthracyclines also revealed no significant benefit in TNBC patients (50). To date, several phase I/II clinical trials targeting EGFR and mTOR pathways in an advanced setting of breast cancers, including TNBC have been carried out or are still undergoing: Erlotinib and everolimus treatment in metastatic breast cancer (NCT00574366, completed); lapatinib and everolimus treatment in TNBC (NCT01272141, terminated); letrozole, lapatinib and everolimus treatment in advanced endocrine resistant breast cancer (NCT01499160, terminated); temsirolimus, cisplatin and erlotinib treatment in TNBC (NCT00998036, completed); lapatinib and erlotinib treatment in HER2-positive metastatic breast cancer (NCT01283789, active); and everolimus, lapatinib and capecitabine treatment in HER2-positive breast cancer with CNS metastasis (NCT01783756, active) (31,51). While the effectiveness of the combination of EGFR inhibitors and rapalogs with the appearance of apoptotic cells has been reported in some TNBC cell lines, the mechanisms responsible for these synergistic effects have not been reported (52). The phosphorylation of eukaryotic translation initiation factor 4B (eIF4B) in TNBC is a possible fragile point in EGFRi/rapalog synergy (53). As a component of mTORC1, RPTOR is a stoichiometric adaptor molecule for mTOR kinase (54). In the present study, the suppression of mTORC1 via siRNA-mediated RPTOR KD with gefitinib decreased the level of both phosphorylated and total RPS6 protein and inhibited cell viability. These results suggest that mTORC1 provides, at least partially, resistance to EGFR inhibitors in TNBC cells of the MSL subtype, and targeting mTORC1 is a potential alternative therapeutic strategy to overcome resistance to EGFR inhibitors in a subset of TNBCs. Previously, we identified RPS6 dephosphorylation/degradation as a readout pathway of the gefitinib/MET inhibitor combination treatment, and RPS6 KD eventually reduced TNBC cell proliferation (30). A high level of phosphorylated RPS6 is closely associated with active mTOR and is a poor prognostic marker of various cancers including non-small cell lung cancer (NSCLC), hepatocellular carcinoma, sarcoma and pancreatic neuroendocrine tumors (5558). Importantly, the inhibition of mTOR reduces the level of p-RPS6 in renal carcinoma cells (59).

In this study, we demonstrated the necessity for mTORC1-targeted therapy in EGFRi-resistant TNBC cells and that the dephosphorylation/degradation of RPS6 is closely associated with the mTORC1 signaling pathway along with the survival of a subgroup of TNBC cells. Although we demonstrated an association between resistance to gefitinib and mTORC1 activation, other possible crosstalk mechanisms need to be explored to overcome resistance to EGFR inhibitors and for the successful intervention and control of TNBC. In addition, whether the regulation of RPS6 protein is under the control of a cancer-specific growth signaling pathway remains unclear. Further investigations are warranted in order to dissect the RPS6 de-phosphorylation and/or the degradation pathway and to identify the cancer-specific altered homeostasis of the RPS6 pathway to further develop therapeutic targets in malignant cancers, including TNBCs.

Acknowledgments

This study was supported by a grant (NRF-2015R1D1A1A 01057893 to Y.-S.S.) funded by the National Research Foundation of Korea.

Notes

[1] Competing interests

The authors declare that they have no competing interests.

References

1 

Mohamed A, Krajewski K, Cakar B and Ma CX: Targeted therapy for breast cancer. Am J Pathol. 183:1096–1112. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Brenton JD, Carey LA, Ahmed AA and Caldas C: Molecular classification and molecular forecasting of breast cancer: Ready for clinical application? J Clin Oncol. 23:7350–7360. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Morris GJ, Naidu S, Topham AK, Guiles F, Xu Y, McCue P, Schwartz GF, Park PK, Rosenberg AL, Brill K, et al: Differences in breast carcinoma characteristics in newly diagnosed African-American and Caucasian patients: A single-institution compilation compared with the National Cancer Institute's Surveillance, Epidemiology, and End Results database. Cancer. 110:876–884. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Podo F, Buydens LM, Degani H, Hilhorst R, Klipp E, Gribbestad IS, Van Huffel S, van Laarhoven HW, Luts J, Monleon D, et al FEMME Consortium: Triple-negative breast cancer: Present challenges and new perspectives. Mol Oncol. 4:209–229. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y and Pietenpol JA: Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 121:2750–2767. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Lehmann BD and Pietenpol JA: Identification and use of biomarkers in treatment strategies for triple-negative breast cancer subtypes. J Pathol. 232:142–150. 2014. View Article : Google Scholar :

7 

Liedtke C, Mazouni C, Hess KR, André F, Tordai A, Mejia JA, Symmans WF, Gonzalez-Angulo AM, Hennessy B, Green M, et al: Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 26:1275–1281. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Kassam F, Enright K, Dent R, Dranitsaris G, Myers J, Flynn C, Fralick M, Kumar R and Clemons M: Survival outcomes for patients with metastatic triple-negative breast cancer: Implications for clinical practice and trial design. Clin Breast Cancer. 9:29–33. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, Chae YK, Cristofanilli M, Gradishar WJ and Giles FJ: Targeting Epidermal Growth Factor Receptor in triple negative breast cancer: New discoveries and practical insights for drug development. Cancer Treat Rev. 53:111–119. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P and Narod SA: Triple-negative breast cancer: Clinical features and patterns of recurrence. Clin Cancer Res. 13:4429–4434. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Eccles SA: The epidermal growth factor receptor/Erb-B/HER family in normal and malignant breast biology. Int J Dev Biol. 55:685–696. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Wheeler DL, Dunn EF and Harari PM: Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat Rev Clin Oncol. 7:493–507. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Yarden Y and Pines G: The ERBB network: At last, cancer therapy meets systems biology. Nat Rev Cancer. 12:553–563. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Reis-Filho JS and Tutt AN: Triple negative tumours: A critical review. Histopathology. 52:108–118. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT and Perou CM: Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol. 19:264–271. 2006. View Article : Google Scholar

16 

Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, et al: Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 10:5367–5374. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Nakai K, Hung MC and Yamaguchi H: A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am J Cancer Res. 6:1609–1623. 2016.PubMed/NCBI

18 

Reis-Filho JS, Milanezi F, Carvalho S, Simpson PT, Steele D, Savage K, Lambros MB, Pereira EM, Nesland JM, Lakhani SR, et al: Metaplastic breast carcinomas exhibit EGFR, but not HER2, gene amplification and overexpression: Immunohistochemical and chromogenic in situ hybridization analysis. Breast Cancer Res. 7:R1028–R1035. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Reis-Filho JS, Pinheiro C, Lambros MB, Milanezi F, Carvalho S, Savage K, Simpson PT, Jones C, Swift S, Mackay A, et al: EGFR amplification and lack of activating mutations in metaplastic breast carcinomas. J Pathol. 209:445–453. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Yarden Y and Sliwkowski MX: Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2:127–137. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Burness ML, Grushko TA and Olopade OI: Epidermal growth factor receptor in triple-negative and basal-like breast cancer: Promising clinical target or only a marker? Cancer J. 16:23–32. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Alvarez RH, Valero V and Hortobagyi GN: Emerging targeted therapies for breast cancer. J Clin Oncol. 28:3366–3379. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Yi YW, Hong W, Kang HJ, Kim HJ, Zhao W, Wang A, Seong YS and Bae I: Inhibition of the PI3K/AKT pathway potentiates cytotoxicity of EGFR kinase inhibitors in triple-negative breast cancer cells. J Cell Mol Med. 17:648–656. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Jin Q and Esteva FJ: Cross-talk between the ErbB/HER family and the type I insulin-like growth factor receptor signaling pathway in breast cancer. J Mammary Gland Biol Neoplasia. 13:485–498. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Karamouzis MV, Konstantinopoulos PA and Papavassiliou AG: Targeting MET as a strategy to overcome crosstalk-related resistance to EGFR inhibitors. Lancet Oncol. 10:709–717. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Nahta R, Yu D, Hung MC, Hortobagyi GN and Esteva FJ: Mechanisms of disease: Understanding resistance to HER2-targeted therapy in human breast cancer. Nat Clin Pract Oncol. 3:269–280. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Yamaguchi H, Chang SS, Hsu JL and Hung MC: Signaling crosstalk in the resistance to HER family receptor targeted therapy. Oncogene. 33:1073–1081. 2014. View Article : Google Scholar

29 

Baselga J: Targeting tyrosine kinases in cancer: The second wave. Science. 312:1175–1178. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Yi YW, You K, Bae EJ, Kwak SJ, Seong YS and Bae I: Dual inhibition of EGFR and MET induces synthetic lethality in triple-negative breast cancer cells through downregulation of ribosomal protein S6. Int J Oncol. 47:122–132. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Massihnia D, Galvano A, Fanale D, Perez A, Castiglia M, Incorvaia L, Listì A, Rizzo S, Cicero G, Bazan V, et al: Triple negative breast cancer: Shedding light onto the role of pi3k/akt/mtor pathway. Oncotarget. 7:60712–60722. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Goldstein D, Bushmeyer SM, Witt PL, Jordan VC and Borden EC: Effects of type I and II interferons on cultured human breast cells: Interaction with estrogen receptors and with tamoxifen. Cancer Res. 49:2698–2702. 1989.PubMed/NCBI

33 

Duong HQ, You KS, Oh S, Kwak SJ and Seong YS: Silencing of NRF2 reduces the expression of ALDH1A1 and ALDH3A1 and sensitizes to 5-FU in pancreatic cancer cells. Antioxidants. 6:62017. View Article : Google Scholar

34 

Kim SI, Kim HJ, Lee HJ, Lee K, Hong D, Lim H, Cho K, Jung N and Yi YW: Application of a non-hazardous vital dye for cell counting with automated cell counters. Anal Biochem. 492:8–12. 2016. View Article : Google Scholar

35 

Biscardi JS, Ishizawar RC, Silva CM and Parsons SJ: Tyrosine kinase signalling in breast cancer: Epidermal growth factor receptor and c-Src interactions in breast cancer. Breast Cancer Res. 2:203–210. 2000. View Article : Google Scholar

36 

Finn RS, Dering J, Ginther C, Wilson CA, Glaspy P, Tchekmedyian N and Slamon DJ: Dasatinib, an orally active small molecule inhibitor of both the src and abl kinases, selectively inhibits growth of basal-type/'triple-negative' breast cancer cell lines growing in vitro. Breast Cancer Res Treat. 105:319–326. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Feder D and Bishop JM: Purification and enzymatic characterization of pp60c-src from human platelets. J Biol Chem. 265:8205–8211. 1990.PubMed/NCBI

38 

Sekulić A, Hudson CC, Homme JL, Yin P, Otterness DM, Karnitz LM and Abraham RT: A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res. 60:3504–3513. 2000.

39 

Kovacina KS, Park GY, Bae SS, Guzzetta AW, Schaefer E, Birnbaum MJ and Roth RA: Identification of a proline-rich Akt substrate as a 14-3-3 binding partner. J Biol Chem. 278:10189–10194. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Gingras AC, Gygi SP, Raught B, Polakiewicz RD, Abraham RT, Hoekstra MF, Aebersold R and Sonenberg N: Regulation of 4E-BP1 phosphorylation: A novel two-step mechanism. Genes Dev. 13:1422–1437. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr and Kinzler KW: Cancer genome landscapes. Science. 339:1546–1558. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Wullschleger S, Loewith R and Hall MN: TOR signaling in growth and metabolism. Cell. 124:471–484. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Laplante M and Sabatini DM: mTOR signaling at a glance. J Cell Sci. 122:3589–3594. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Sabatini DM: mTOR and cancer: Insights into a complex relationship. Nat Rev Cancer. 6:729–734. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Ueng SH, Chen SC, Chang YS, Hsueh S, Lin YC, Chien HP, Lo YF, Shen SC and Hsueh C: Phosphorylated mTOR expression correlates with poor outcome in early-stage triple negative breast carcinomas. Int J Clin Exp Pathol. 5:806–813. 2012.PubMed/NCBI

46 

Walsh S, Flanagan L, Quinn C, Evoy D, McDermott EW, Pierce A and Duffy MJ: mTOR in breast cancer: Differential expression in triple-negative and non-triple-negative tumors. Breast. 21:178–182. 2012. View Article : Google Scholar

47 

Montero JC, Esparís-Ogando A, Re-Louhau MF, Seoane S, Abad M, Calero R, Ocaña A and Pandiella A: Active kinase profiling, genetic and pharmacological data define mTOR as an important common target in triple-negative breast cancer. Oncogene. 33:148–156. 2014. View Article : Google Scholar

48 

Ellard SL, Clemons M, Gelmon KA, Norris B, Kennecke H, Chia S, Pritchard K, Eisen A, Vandenberg T, Taylor M, et al: Randomized phase II study comparing two schedules of everolimus in patients with recurrent/metastatic breast cancer: NCIC Clinical Trials Group IND.163. J Clin Oncol. 27:4536–4541. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Jovanović B, Mayer IA, Mayer EL, Abramson VG, Bardia A, Sanders ME, Kuba MG, Estrada MV, Beeler JS, Shaver TM, et al: A Randomized phase II neoadjuvant study of cisplatin, paclitaxel with or without everolimus in patients with stage II/III triple-negative breast cancer (TNBC): Responses and long-term outcome correlated with increased frequency of DNA damage response gene mutations, TNBC subtype, AR status, and Ki67. Clin Cancer Res. 23:4035–4045. 2017. View Article : Google Scholar

50 

Gonzalez-Angulo AM, Akcakanat A, Liu S, Green MC, Murray JL, Chen H, Palla SL, Koenig KB, Brewster AM, Valero V, et al: Open-label randomized clinical trial of standard neoadjuvant chemotherapy with paclitaxel followed by FEC versus the combination of paclitaxel and everolimus followed by FEC in women with triple receptor-negative breast cancer. Ann Oncol. 25:1122–1127. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Oualla K, El-Zawahry HM, Arun B, Reuben JM, Woodward WA, Gamal El-Din H, Lim B, Mellas N, Ueno NT and Fouad TM: Novel therapeutic strategies in the treatment of triple-negative breast cancer. Ther Adv Med Oncol. 9:493–511. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Liu T, Yacoub R, Taliaferro-Smith LD, Sun SY, Graham TR, Dolan R, Lobo C, Tighiouart M, Yang L, Adams A, et al: Combinatorial effects of lapatinib and rapamycin in triple-negative breast cancer cells. Mol Cancer Ther. 10:1460–1469. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Madden JM, Mueller KL, Bollig-Fischer A, Stemmer P, Mattingly RR and Boerner JL: Abrogating phosphorylation of eIF4B is required for EGFR and mTOR inhibitor synergy in triple-negative breast cancer. Breast Cancer Res Treat. 147:283–293. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H, Tempst P and Sabatini DM: mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell. 110:163–175. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Chen B, Tan Z, Gao J, Wu W, Liu L, Jin W, Cao Y, Zhao S, Zhang W, Qiu Z, et al: Hyperphosphorylation of ribosomal protein S6 predicts unfavorable clinical survival in non-small cell lung cancer. J Exp Clin Cancer Res. 34:1262015. View Article : Google Scholar : PubMed/NCBI

56 

Iwenofu OH, Lackman RD, Staddon AP, Goodwin DG, Haupt HM and Brooks JS: Phospho-S6 ribosomal protein: A potential new predictive sarcoma marker for targeted mTOR therapy. Mod Pathol. 21:231–237. 2008. View Article : Google Scholar

57 

Komori Y, Yada K, Ohta M, Uchida H, Iwashita Y, Fukuzawa K, Kashima K, Yokoyama S, Inomata M and Kitano S: Mammalian target of rapamycin signaling activation patterns in pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci. 21:288–295. 2014. View Article : Google Scholar

58 

Masuda M, Chen WY, Miyanaga A, Nakamura Y, Kawasaki K, Sakuma T, Ono M, Chen CL, Honda K and Yamada T: Alternative mammalian target of rapamycin (mTOR) signal activation in sorafenib-resistant hepatocellular carcinoma cells revealed by array-based pathway profiling. Mol Cell Proteomics. 13:1429–1438. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Knoll M, Macher-Goeppinger S, Kopitz J, Duensing S, Pahernik S, Hohenfellner M, Schirmacher P and Roth W: The ribosomal protein S6 in renal cell carcinoma: Functional relevance and potential as biomarker. Oncotarget. 7:418–432. 2016. View Article : Google Scholar :

60 

Green TP, Fennell M, Whittaker R, Curwen J, Jacobs V, Allen J, Logie A, Hargreaves J, Hickinson DM, Wilkinson RW, et al: Preclinical anticancer activity of the potent, oral Src inhibitor AZD0530. Mol Oncol. 3:248–261. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Maira SM, Stauffer F, Brueggen J, Furet P, Schnell CC, Brachmann S, Chène P, De Pover A, Schoemaker K, et al: Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Mol Cancer Ther. 7:1851–1863. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Toledo LI, Murga M, Zur R, Soria R, Rodriguez A, Martinez S, Oyarzabal J, Pastor J, Bischoff JR and Fernandez-Capetillo O: A cell-based screen identifies ATR inhibitors with synthetic lethal properties for cancer-associated mutations. Nat Struct Mol Biol. 18:721–727. 2011. View Article : Google Scholar : PubMed/NCBI

63 

O'Hare T, Walters DK, Stoffregen EP, Jia T, Manley PW, Mestan J, Cowan-Jacob SW, Lee FY, Heinrich MC, Deininger MW, et al: In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer Res. 65:4500–4505. 2005. View Article : Google Scholar : PubMed/NCBI

64 

Shah NP, Lee FY, Luo R, Jiang Y, Donker M and Akin C: Dasatinib (BMS-354825) inhibits KITD816V, an imatinib-resistant activating mutation that triggers neoplastic growth in most patients with systemic mastocytosis. Blood. 108:286–291. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Rivera VM, Squillace RM, Miller D, Berk L, Wardwell SD, Ning Y, Pollock R, Narasimhan NI, Iuliucci JD, Wang F, et al: Ridaforolimus (AP23573; MK-8669), a potent mTOR inhibitor, has broad antitumor activity and can be optimally administered using intermittent dosing regimens. Mol Cancer Ther. 10:1059–1071. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Chun KH, Kosmeder JW II, Sun S, Pezzuto JM, Lotan R, Hong WK and Lee HY: Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells. J Natl Cancer Inst. 95:291–302. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Schuler W, Sedrani R, Cottens S, Häberlin B, Schulz M, Schuurman HJ, Zenke G, Zerwes HG and Schreier MH: SDZ RAD, a new rapamycin derivative: Pharmacological properties in vitro and in vivo. Transplantation. 64:36–42. 1997. View Article : Google Scholar : PubMed/NCBI

68 

Flanagan WM, Corthésy B, Bram RJ and Crabtree GR: Nuclear association of a T-cell transcription factor blocked by FK-506 and cyclosporin A. Nature. 352:803–807. 1991. View Article : Google Scholar : PubMed/NCBI

69 

Folkes AJ, Ahmadi K, Alderton WK, Alix S, Baker SJ, Box G, Chuckowree IS, Clarke PA, Depledge P, Eccles SA, et al: The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J Med Chem. 51:5522–5532. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Carnero A: Novel inhibitors of the PI3K family. Expert Opin Investig Drugs. 18:1265–1277. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Knight ZA, Gonzalez B, Feldman ME, Zunder ER, Goldenberg DD, Williams O, Loewith R, Stokoe D, Balla A, Toth B, et al: A pharmacological map of the PI3-K family defines a role for p110alpha in insulin signaling. Cell. 125:733–747. 2006. View Article : Google Scholar : PubMed/NCBI

72 

García-Martínez JM, Moran J, Clarke RG, Gray A, Cosulich SC, Chresta CM and Alessi DR: Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR). Biochem J. 421:29–42. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Vlahos CJ, Matter WF, Hui KY and Brown RF: A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem. 269:5241–5248. 1994.PubMed/NCBI

74 

Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K, Ueno Y, Hatch H, Majumder PK, Pan BS, et al: MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 9:1956–1967. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Gills JJ and Dennis PA: Perifosine: Update on a novel Akt inhibitor. Curr Oncol Rep. 11:102–110. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Nghiem P, Pearson G and Langley RG: Tacrolimus and pimecrolimus: From clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis. J Am Acad Dermatol. 46:228–241. 2002. View Article : Google Scholar : PubMed/NCBI

77 

Edwards SR and Wandless TJ: The rapamycin-binding domain of the protein kinase mammalian target of rapamycin is a destabilizing domain. J Biol Chem. 282:13395–13401. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Shor B, Zhang WG, Toral-Barza L, Lucas J, Abraham RT, Gibbons JJ and Yu K: A new pharmacologic action of CCI-779 involves FKBP12-independent inhibition of mTOR kinase activity and profound repression of global protein synthesis. Cancer Res. 68:2934–2943. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Marone R, Cmiljanovic V, Giese B and Wymann MP: Targeting phosphoinositide 3-kinase: Moving towards therapy. Biochim Biophys Acta. 1784:159–185. 2008. View Article : Google Scholar

80 

Somwar R, Niu W, Kim DY, Sweeney G, Randhawa VK, Huang C, Ramlal T and Klip A: Differential effects of phosphatidylinositol 3-kinase inhibition on intracellular signals regulating GLUT4 translocation and glucose transport. J Biol Chem. 276:46079–46087. 2001. View Article : Google Scholar : PubMed/NCBI

81 

Yu K, Toral-Barza L, Shi C, Zhang WG, Lucas J, Shor B, Kim J, Verheijen J, Curran K, Malwitz DJ, et al: Biochemical, cellular, and in vivo activity of novel ATP-competitive and selective inhibitors of the mammalian target of rapamycin. Cancer Res. 69:6232–6240. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Yaguchi S, Fukui Y, Koshimizu I, Yoshimi H, Matsuno T, Gouda H, Hirono S, Yamazaki K and Yamori T: Antitumor activity of ZSTK474, a new phosphatidylinositol 3-kinase inhibitor. J Natl Cancer Inst. 98:545–556. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 52 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
You KS, Yi YW, Kwak S and Seong Y: Inhibition of RPTOR overcomes resistance to EGFR inhibition in triple-negative breast cancer cells. Int J Oncol 52: 828-840, 2018
APA
You, K.S., Yi, Y.W., Kwak, S., & Seong, Y. (2018). Inhibition of RPTOR overcomes resistance to EGFR inhibition in triple-negative breast cancer cells. International Journal of Oncology, 52, 828-840. https://doi.org/10.3892/ijo.2018.4244
MLA
You, K. S., Yi, Y. W., Kwak, S., Seong, Y."Inhibition of RPTOR overcomes resistance to EGFR inhibition in triple-negative breast cancer cells". International Journal of Oncology 52.3 (2018): 828-840.
Chicago
You, K. S., Yi, Y. W., Kwak, S., Seong, Y."Inhibition of RPTOR overcomes resistance to EGFR inhibition in triple-negative breast cancer cells". International Journal of Oncology 52, no. 3 (2018): 828-840. https://doi.org/10.3892/ijo.2018.4244