Abstract
Background
Recent therapeutic strategies for KRAS-mutated cancers that inhibit the MAPK pathway have attracted considerable attention. The RAF/MEK clamp avutometinib (VS-6766/CH5126766/RO5126766/CKI27) is promising for patients with KRAS-mutated cancers. Although avutometinib monotherapy has shown clinical activity in patients with KRAS-mutated cancers, effective combination strategies will be important to develop.
Methods
Using a phosphorylation kinase array kit, we explored the feedback mechanism of avutometinib in KRAS-mutated NSCLC cells, and investigated the efficacy of combining avutometinib with inhibitors of the feedback signal using in vitro and in vivo experiments. Moreover, we searched for a biomarker for the efficacy of combination therapy through an in vitro study and analysis using the The Cancer Genome Atlas Programme dataset.
Results
Focal adhesion kinase (FAK) phosphorylation/activation was increased after avutometinib treatment and synergy between avutometinib and FAK inhibitor, defactinib, was observed in KRAS-mutated NSCLC cells with an epithelial rather than mesenchymal phenotype. Combination therapy with avutometinib and defactinib induced apoptosis with upregulation of Bim in cancer cells with an epithelial phenotype in an in vitro and in vivo study.
Conclusions
These results demonstrate that the epithelial-mesenchymal transition status may be a promising biomarker for the efficacy of combination therapy with avutometinib and defactinib in KRAS-mutated NSCLC.
Subject terms: Non-small-cell lung cancer, Tumour biomarkers
Introduction
The development of molecular-targeted therapies has dramatically improved the prognosis of cancers with driver mutations. KRAS mutations are commonly observed in several cancers, including non-small-cell lung cancer (NSCLC), colon cancer, and pancreatic cancer. The frequency of KRAS mutations in NSCLC varies by race, with the mutations found in 20–30% of lung adenocarcinomas in Caucasians and 8–10% in Asians [1–3]. Although EGFR, ALK, and BRAF inhibitors have been successfully developed, targeted therapies for KRAS-mutated cancers have only recently been approved.
Two KRAS G12C inhibitors, sotorasib and adagrasib, are currently approved by the Food and Drug Administration to treat patients with NSCLC harbouring KRAS G12C mutation [4, 5]. A phase III clinical trial revealed that sotorasib provided significantly better progression-free survival (PFS) than docetaxel in previously treated patients with KRAS G12C-mutated NSCLC [6]. Therefore, sotorasib has been approved for treating KRAS G12C-mutated NSCLC in the United States, Japan, and other countries. However, the objective response rate to sotorasib and adagrasib in patients with KRAS G12C-mutated NSCLC is 28.1% [6] and 42.9% [5], respectively. Although several other KRAS inhibitors are in development in addition to sotorasib and adagrasib and are expected to have further clinical applications, alternative strategies for KRAS-mutated cancers are attracting attention.
Considerable efforts have been dedicated to exploring alternative strategies for the treatment of KRAS-mutated cancers, with a focus on inhibiting downstream signalling within the mitogen-activated protein kinase (MAPK; RAF/MEK/ERK) pathway [7]. Among the several therapeutic strategies for KRAS-mutated cancers, MEK inhibition is a potential treatment. However, several clinical trials have shown that MEK inhibitor monotherapy does not provide significant efficacy in patients with KRAS-mutated NSCLC [8–10]. The limited efficacy observed for MEK inhibitors may involve adaptive feedback mechanisms through the reactivation of MAPK signalling caused by the blockade of ERK signalling [11–13]. On the other hand, both a first-in-class MEK inhibitor trametinib (Mekinist) and a RAF/MEK clamp avutometinib (VS-6766/CH5126766/RO5126766/CKI27) were discovered by our original screening system [14]. Importantly, avutometinib is considered to be more effective in RAS-mutated malignancies than other MEK inhibitors including trametinib, because avutometinib can strongly inhibit CRAF, one of the main downstream signals of RAS, as well as MEK [11, 15, 16]. We therefore used avutometinib in the present study of KRAS-mutated malignancies.
A phase I trial demonstrated that 30% of patients with KRAS-mutated NSCLC treated with avutometinib had an objective response; all these patients had a low frequency of adverse events that lasted longer than 6 months [17]. While avutometinib monotherapy displayed better efficacy than other MEK inhibitors in KRAS-mutated NSCLC, the development of effective combination strategies remains crucial. The combination of avutometinib and the FAK inhibitor defactinib has demonstrated clinical efficacy in both KRAS-mutated NSCLC and low-grade serous ovarian cancer (LGSOC) [18–20].
In this study, we aimed to elucidate the molecular mechanism of resistance induced by avutometinib in KRAS-mutated NSCLC cells and propose an effective therapeutic combination strategy for patients with KRAS-mutated NSCLC.
Methods
Cell cultures and reagents
Nine human NSCLC cell lines with KRAS mutations were used. The cell lines NCI-H358, NCI-H2122, NCI-H441, A549, SW1573, Calu-1, and Calu-6 were purchased from the American Type Culture Collection (Manassas, VA, USA), while the NCI-H23 and HOP-62 cell lines were obtained from the NCI-60 cancer cell line panel of the National Cancer Institute Developmental Therapeutics Programme. We obtained information on the type of point mutations in the KRAS oncogene from the Catalogue of Somatic Mutations in Cancer (COSMIC) database (https://cancer.sanger.ac.uk/cosmic/).
All cell lines were cultured in Roswell Park Memorial Institute (RPMI) 1640 medium supplemented with 10% foetal bovine serum (FBS), 2 mmol/L glutamine, 50 U/mL penicillin, and 100 µg/mL streptomycin in a 5% CO2 incubator at 37 °C. All cells were passaged for less than 3 months before being renewed with frozen early passage stocks.
Avutometinib was provided by Chugai Pharmaceuticals (Tokyo, Japan). Defactinib was obtained from Selleck Chemicals (Houston, TX, USA). For in vitro experiments, the inhibitors were dissolved in dimethyl sulfoxide (DMSO) and stored at −20 °C. Recombinant human transforming growth factor (TGF)-β1 protein was purchased from R&D Systems (Abingdon, UK).
RTK phosphorylation antibody array
Cell lysates (500 μg/mL) were prepared from NCI-H358 cells treated with DMSO or avutometinib at the indicated concentrations for 72 h. We analysed the phosphorylation status of receptor tyrosine kinases (RTK) in the relevant pathways using RayBio® C-Series Human RTK Phosphorylation Antibody Array C1 (RayBiotech Inc., Norcross, GA, USA) following the manufacturer’s instructions. Arrays were imaged and quantified using the ChemiDoc Imaging System (Bio-Rad, Hercules, CA, USA).
Antibodies and western blotting
Cells treated with each agent at the indicated concentrations were lysed in RIPA buffer (50 mmol/L Tris-HCl, pH 8.0; 150 mmol/L NaCl; 1% NP-40; 0.5% deoxycholic acid; 0.1% SDS; 1 mmol/L dithiothreitol; and 0.5 mmol/L phenylmethylsulfonyl fluoride) supplemented with phosphatase inhibitors (#07574; Nacalai Tesque, Kyoto, Japan) for 30 min at 4 °C and centrifuged. The total protein concentration was measured using a NanoDrop Lite spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). Protein aliquots of 10 µg each were separated using SDS-PAGE. The proteins were subsequently transferred to Immobilon-P membranes (Millipore; Merck KGaA, Darmstadt, Germany), which were then blocked in Tris-buffered saline (TBS) containing 5% skim milk for 1 h at room temperature. After washing three times with TBS with tween-20 (TBS-T), the membranes were incubated overnight at 4 °C with primary antibodies against pFAK (Tyr397, #8556), FAK (#3285), pAKT (Ser473, #4060), AKT (#4691), pERK (#4370), ERK (#4696), Bim (#2933), pFoxO3a (Ser253, #9466), FoxO3a (#2497), Cleaved PARP (#5625), Cleaved Caspase 3 (#9661), pIGF-1R (#3021), IGF-1R (#9750), E-Cadherin (#3195 S), Vimentin (#5741 S) and ZEB1 (#3396 S) (1:1000 dilution; Cell Signalling Technology, Danvers, MA, USA), β-actin (#A5441) (1:1000 dilution; Sigma-Aldrich, Burlington, MA, USA), and cMyc (#C2012) (1:1000 dilution; Abcam, Cambridge MA, UK).
After being washed three times with TBS-T, the membranes were incubated for 1 h at room temperature with HRP-conjugated species-specific secondary antibodies. Immunoreactive bands were visualised using an ECL chemiluminescent detection reagent (Immobilon Western Chemiluminescent HRP Substrate; Merck‐Millipore, Darmstadt, Germany), imaged, and quantified using the ChemiDoc Imaging System. All immunoblotting experiments were performed independently at least three times.
Cell viability assay
Cell viability was evaluated using the Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan). Cells (2–3 × 103 cells/well) were seeded in 96-well plates and incubated with the indicated concentrations of various compounds for 72 h. The CCK-8 solution was added to each well, and the medium’s absorbance (450 nm) was determined using SpectraMax® iD3 (Molecular Devices, LLC., San Jose, CA, USA) after another 4 h of incubation. The percentage growth was determined relative to the DMSO treatment. Experiments were repeated at least three times with triplicate samples.
Colony formation assay
The cells were seeded at a density of 5 × 103 cells/well in 6-well plates. After incubation for 24 h, the cells were treated with each agent at the indicated concentrations for 15 days, and the drugs were replenished every 72 h. The plates were stained with 0.1% crystal violet, fixed in formalin, and visually examined. Representative plates from three independent experiments are shown.
Transfection experiments
Duplexed Silencer Select siRNAs against MYC (s9129) and focal adhesion kinase (FAK) (#1: PTK2HSS108800 and #2: PTK2HSS183870) were purchased from Invitrogen (Carlsbad, CA, USA). Cells (1–2 × 105 cells/well) were seeded into 6-well plates. After incubation for 24 h, the cells were transfected using Lipofectamine RNAiMAX (Invitrogen) with these siRNAs (20 nmol/L) or Stealth RNAi as a negative control (Invitrogen) as a scrambled control, according to the manufacturer’s instructions. After incubation for 6 h, the medium containing Lipofectamine was replaced with the medium. Each sample was tested in triplicate in three independent experiments.
Quantitative real-time RT-PCR
The total cellular RNA was extracted from the NCI-H358 and NCI-H2122 cells treated with DMSO or avutometinib (150 nmol/L) for 24 h using Sepasol-RNA I Super G (Nacalai Tesque). Complementary DNA (cDNA) was synthesised from total RNA using ReverTra Ace® qPCR RT Master Mix kit (TOYOBO, Osaka, Japan) according to the manufacturer’s instructions. Quantitative RT-PCR was performed using a QuantStudio 3 Real-Time PCR System (Thermo Fisher Scientific) with cDNA and TaqMan probes (Applied Biosystems) for IGF-1 (Hs01547656_m1) and GAPDH (Hs02758991_g1). The expression level of every gene was determined from standard curves to obtain a relative IGF-1/GAPDH ratio using the 2−ΔΔCT method. Each sample was tested in triplicate in three independent experiments.
Gene set variation analysis (GSVA) of cell lines using the epithelial and mesenchymal gene signature set
RNA-seq gene expression data for the nine KRAS-mutated NSCLC cell lines were retrieved from the Cell Model Passports database (https://cellmodelpassports.Sanger.Ac.uk). These data were log2-transformed from TMM-normalised count data. We calculated the epithelial and mesenchymal scores using GSVA [21]. Scoring was performed using the R package, GSVA. For each cell line (one expression data point), we computed two scores using epithelial (288 genes) and mesenchymal (188 genes) gene lists [22]. Briefly, GSVA estimates a cumulative density function for each gene using all samples, ranks genes across samples, and calculates a score between −1 and 1 for each set of genes using the Kolmogorov–Smirnov random walk statistic. The difference between the two scores was calculated and used as the epithelial-mesenchymal transition (EMT) score. Cell lines with positive (high) EMT scores were mostly mesenchymal, whereas those with negative (low) EMT scores were mostly epithelial. We conducted a hierarchical cluster analysis of the expression of signature genes in all cell lines. The distance between clusters was measured using the Ward method and visualised as a heatmap using Python and Python libraries (Pandas, Matplotlib, and Seaborn).
Quantification of cell cycle and apoptosis
Cells (1–2 × 105 cells/well) were seeded into 6-well plates. After incubation for 24 h, the cells treated with each agent for 48 or 72 h were harvested via trypsinization. After washing twice with phosphate buffered saline (PBS), the cells were suspended in PBS containing 0.1% Triton X-100 and 50 g/mL propidium iodide to stain the nuclei. The cells were analysed using a BD Accuri C6 Flow Cytometer (BD Biosciences, San Jose, CA, USA), and the number of cells in the G0-G1, S, G2-M, and sub G1 phases was quantified.
Scratch assay
NCI-H358 and NCI-H2122 cells were seeded into a 6-well plate at a concentration of 1 × 105 cells/mL and cultured for 24 h at 37 °C. After nearly confluent cell monolayers were formed, a linear scratch wound was generated on each plate using a plastic pipette tip. Images of the scratched wound before treatment and 24 h after treatment were photographed to evaluate the relative migration of the cells. Representative images from three independent experiments are shown.
Cell-line-derived xenograft mouse study
Suspensions of NCI-H358 cells (4 × 106 cells/mouse) were injected subcutaneously into the right flank of 6-week-old female BALB/c nu/nu mice obtained from Charles River Laboratories (Yokohama, Japan) and maintained under pathogen-free conditions. Tumours were measured twice a week using calipers, and tumour volumes were calculated using the following formula: 0.5 × lengths × width2. The mice were randomly divided into four groups (n = 6 in each group) when the tumour volume reached approximately 70–120 mm3: 1) vehicle, 2) defactinib (10.0 mg/kg), 3) avutometinib (0.3 mg/kg), and 4) avutometinib + defactinib. Avutometinib and defactinib was dissolved in 20% hydroxypropyl-β-cyclodextrin/saline. Drugs were administered once daily via oral gavage up to day 28, and the mice’s body weight and general condition were monitored daily. Approval was obtained from the Institutional Review Board of the Kyoto Prefectural University of Medicine for the mice study (approval no. M2021-38) following the ARRIVE guidelines.
Quantification of immunohistochemistry staining
Formalin-fixed, paraffin-embedded tissue blocks were cut into serial 4-µm thick sections, deparaffinized, and immunostained with Ki-67 (Clone MIB-1; Dako, Agilent, Santa Clara, CA, USA). Apoptosis was quantified via the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labelling (TUNEL) method using a Vectastain ABC Kit (Vector Laboratories, Burlingame, CA, USA), according to the manufacturer’s instructions. Hematoxylin and eosin staining was performed to identify tumour cells. Five randomly selected areas (0.1 mm2/section) containing the most positively stained cells in each section were selected using light microscopy at 400 × magnification. The percentage of cells positive for each immunohistochemical stain was assessed using the ImageJ software.
Statistical analysis
The statistical significance of differences was analysed using student’s t test and one-way ANOVA. The combination index (CI) values were calculated using CompuSyn® software (version 2.0; Biosoft, Cambridge, UK) [21] with cell viability representing the effect when exposed to the drugs at 72 h; CI < 0.7, 0.7 ≤ CI ≤ 1.2, and CI > 1.2 were judged as having synergistic, additive, and antagonistic effects, respectively. All statistical analyses were performed using GraphPad Prism Ver. 9.0 (GraphPad Software, Inc., San Diego, CA, USA). A two-sided p < 0.05 was considered statistically significant.
Results
Dual RAF/MEK inhibitor avutometinib activates FAK and AKT and reactivates ERK
When NCI-H358 cells were treated with avutometinib at concentrations above 1.5 µmol/L for 72 h, ~45% of the cells survived (Fig. S1). To elucidate the mechanism underlying the inhibitory effect of avutometinib, we used an RTK phosphorylation array kit to evaluate the differences in the phosphorylation states of molecules in NCI-H358 cells treated with or without avutometinib for 72 h. The phosphorylation of FAK (pFAK) increased after 72 h of treatment with avutometinib (Fig. 1a). This result suggests that the FAK pathway may function as a bypass signalling pathway against RAF/MEK inhibition by avutometinib. Kinetic analysis revealed that ERK phosphorylation was remarkably inhibited by avutometinib at 3 h but reactivated at 72 h in NCI-H2122 and NCI-H358 cells (Fig. 1b). Conversely, phosphorylation of FAK and AKT was steadily increased after 72 h of avutometinib treatment in these two cell lines (Fig. 1b).
Fig. 1. Avutometinib activates FAK in KRAS-mutated NSCLC cells.
a The results of human RTK phosphorylation array. NCI-H358 cells are treated with or without avutometinib (150 nmol/L) for 72 h. b NCI-H358 and NCI-H2122 cells are treated with avutometinib (150 nmol/L) for indicated times, and each protein is detected in cell lysates using western blotting. c The indicated cells are treated with nonspecific control (siSCR) or siRNA specific to FAK (siFAK) for 72 h in the presence or absence of avutometinib (150 nmol/L), and cell viability is determined using CCK-8 assay. The cell viability rate of cells treated with siFAK in the presence of avutometinib is compared with that of cells treated with siSCR. p values are calculated using one-way ANOVA. *: p < 0.001. d SiSCR or siFAK are introduced into the indicated cells. After 24 h, the cells are incubated with or without avutometinib (150 nmol/L) for 72 h and lysed, and the indicated proteins are detected using western blotting. e The indicated cells are incubated with or without avutometinib (150 nmol/L) for 24 h, and quantitative RT-PCR is used to assess the expression of IGF-1 at the mRNA level in indicated cells. p values are calculated using student’s t-tests. f The indicated cells are treated with avutometinib (150 nmol/L) for indicated times, and each protein is detected in cell lysates using western blotting.
Next, we used siRNAs to evaluate the effects of FAK knockdown on the viability of NCI-H358 and NCI-H2122 cells. The knockdown of FAK expression alone did not affect cell viability. Meanwhile, the knockdown of FAK expression following treatment with avutometinib significantly decreased the viability of both cancer cell lines (Fig. 1c). These results suggest that FAK contributes to the survival of several types of NSCLC cells with KRAS mutations when the MAPK pathway is inhibited by avutometinib. Furthermore, FAK knockdown decreased the avutometinib-induced phosphorylation of AKT but not of ERK (Fig. 1d).
Avutometinib also significantly increased the IGF-1 mRNA levels in NCI-H358 and NCI-H2122 cells (Fig. 1e). Avutometinib treatment decreased c-MYC expression and enhanced IGF-1R phosphorylation over time (Fig. 1f). The knockdown of MYC expression increased the levels of IGF-1 mRNA and the phosphorylation of IGF-1R, FAK, and AKT in both NCI-H358 and NCI-H2122 cells (Fig. S2A, B).
EMT status correlates with synergistic effects of defactinib in combination with avutometinib
To characterise the EMT status in the nine KRAS-mutated lung cancer cell lines, we performed western blot analysis for E-cadherin and vimentin. Additionally, we acquired information on the type of point mutations in the KRAS oncogene status from the COSMIC database (Fig. 2a). We investigated the effects of avutometinib monotherapy on cell viability on KRAS-mutated NSCLC cells (Fig. S3A). There was no significant difference in the efficacy of avutometinib monotherapy in cell lines with epithelial and mesenchymal phenotypes (p = 0.456) or between KRAS G12C and non-G12C point mutations (p = 0.926) (Figs. 2a and S3B–D).
Fig. 2. EMT status correlates with synergistic effects of defactinib in combination with avutometinib.
a The KRAS-mutated NSCLC cell lines NCI-H358, NCI-H2122, NCI-H441, A549, SW1573, Calu-1, NCI-H23, Calu-6, and HOP-62 are lysed, and the indicated proteins are detected using western blotting. The mutation status of KRAS in these cell lines obtained from the database are shown. b The combination index with avutometinib and defactinib in the indicated cell lines is calculated using CompuSyn®. c Based on the result of (a) and (b), cells are further divided into groups with high and low expression of phosphorylation of FAK, KRAS G12C, and non-G12C point mutations; comparative data are shown (p = 0.392, p = 0.879, respectively; p values are calculated using student’s t-tests). d Hierarchical clustering and heatmap of EMT signature gene expression levels in the indicated cell lines from the Cell Model Passports dataset. e Distribution of the 9 KRAS-mutated NSCLC cell lines in the epithelial-mesenchymal score space following the GSVA scoring. f The correlation coefficients between the calculated EMT scores and the combination index are examined. The white boxes and black circles in (e) and (f) correspond to epithelial and mesenchymal-type cell lines, respectively, as shown in (a). EMT epithelial-mesenchymal transition, NSCLC Non-small cell lung cancer, GSVA Gene set variation analysis.
Moreover, we calculated the CI values of the FAK inhibitor defactinib in combination with avutometinib using CompuSyn. Among the nine KRAS-mutated NSCLC cell lines, NCI-H358, NCI-H2122, and NCI-H441 had values < 0.7. Meanwhile, the other cell lines had values between 0.7 and 1.2 (Fig. 2b). We found a strong combined effect of avutometinib and defactinib in cancer cells with an epithelial phenotype, but not in those with a mesenchymal phenotype, regardless of the presence or absence of phosphorylated FAK expression, or KRAS point mutation (G12C or not) (Fig. 2c). Therefore, the EMT status might be a biomarker for combination therapy with avutometinib and defactinib.
Next, we evaluated the EMT status in the nine KRAS-mutated NSCLC cell lines based on the mRNA expression levels of the 416 signature EMT genes. RNA-seq data were obtained from the Cell Model Passports database. Hierarchical cluster analysis revealed that the transcriptomes of cell lines were broadly classified into two groups, each corresponding to epithelial or mesenchymal cell lines, based on western blotting results (Fig. 2d). To examine the degree of EMT in these cells, we calculated an epithelial and a mesenchymal score that summarised the overall transcriptional activities of epithelial and mesenchymal genes for each cell line. These scores were based on GSVA using a list of 228 epithelial genes and 188 mesenchymal genes as signature gene sets [22]. A positive/high value for each score indicates more epithelial or mesenchymal cells. The transcriptional state of the cell lines was unevenly distributed in the epithelial and mesenchymal regions in the epithelial and mesenchymal score spaces (Fig. 2e). Therefore, the western blot result-based classification of epithelial or mesenchymal cell lines was validated via a transcriptome analysis of 416 genes related to EMT.
Furthermore, the relationship between the CI values and degree of EMT in these cell lines was examined. EMT scores were calculated as a single score representing the degree of EMT and evaluated for correlation with the CI value, as described in the Methods section. Cell lines with positive/high EMT scores are highly mesenchymal, whereas those with negative/low scores are highly epithelial. A strong positive correlation was observed between CI value and EMT score (R2 = 0.7937; Fig. 2f). Therefore, cell lines with CI values above 0.7 were confirmed to be in a stronger mesenchymal state. These results indicate that the EMT status, not the FAK phosphorylation status or KRAS point mutation type, is a predictive biomarker for the efficacy of combination therapy with avutometinib and defactinib.
Defactinib sensitises epithelial phenotype cancer cells to avutometinib
We evaluated the combined effects of avutometinib and defactinib on KRAS-mutated NSCLC cells. Although avutometinib alone did not adequately reduce cell viability in both the epithelial and mesenchymal phenotypes of KRAS-mutated NSCLC cells (Fig. S3A), the combined use of avutometinib and defactinib significantly reduced the viability of epithelial cells (Figs. 2b and S4A). Moreover, continuous co-treatment for 15 days decreased the viability of epithelial-phenotype cells, but not of mesenchymal-phenotype cells (Fig. S4B).
Next, we evaluated the effects of these drugs on the expression of various proteins using western blotting. Combination treatment for 6 h inhibited the activation of FAK and AKT induced by avutometinib alone in epithelial-phenotype cells but not in mesenchymal-phenotype cells (Fig. 3a). Subsequently, we examined the alterations in the cell cycle and sub G1 populations with a combination of avutometinib and defactinib. The sub G1 population was induced by the combination of epithelial phenotype cells but not in mesenchymal phenotype cells (Fig. 3b). Conversely, the cell cycle was not obviously affected by the combination (Fig. S5). Regarding cell migration, no additional effects of defactinib were observed in cells with an epithelial phenotype (Fig. S6). Given the increase in sub G1 with combination therapy, we evaluated the apoptosis-associated Bim protein. The combination treatment for 24 h increased Bim protein levels more than avutometinib alone in NCI-H358 and NCI-H441 cells (Fig. 3c). We then assessed the effects of the combination treatment on the Bim-associated protein, FoxO3a, and found a decrease in the phosphorylation of FoxO3a induced by avutometinib alone in epithelial cells (Fig. 3c). The combination treatment for 72 h inhibited the activation of FAK and AKT induced by avutometinib alone in epithelial cells (Fig. 3d). Consistent with the flow cytometry results, combination treatment with avutometinib and defactinib for 72 h increased the cleavage of caspase 3 and PARP, markers of apoptotic cells, compared with avutometinib alone in epithelial phenotype cells (Fig. 3d). Hence, the use of defactinib in combination with avutometinib may induce apoptosis in epithelial-phenotype KRAS-mutated NSCLC cells via the FoxO3a-Bim pathway, regardless of the type of KRAS point mutation.
Fig. 3. Defactinib sensitises epithelial phenotype KRAS-mutated non-small cell lung cancer cells to avutometinib.
Cells are cultured with avutometinib (150 nmol/L) in the presence or absence of defactinib (1 mol/L) for 6 h (a), 24 h (c), and 72 h (d). The cells are lysed, and the indicated proteins detected using western blotting. b The sub G1 population of the indicated cells treated with avutometinib (15 nmol/L) in the presence or absence of defactinib (1 mol/L) is analysed via flow cytometry using FACS Accuri. The sub G1 rate of the combination therapy is compared to that of each single treatment in the indicated cells. p values are calculated using one-way ANOVA.
EMT induction reduces the activity of the combination treatment of avutometinib and defactinib
EMT plays an important role in cancer progression and is induced by TGF-β [23]. We evaluated the effects of combination therapy with avutometinib and defactinib by inducing the mesenchymal phenotype. First, NCI-H358 cells were incubated with TGF-β1 for 2 weeks to induce EMT. EMT-induced cells (H358-TGF-β cells) showed a spindle-cell shape, loss of polarity, and decreased intercellular adhesion compared to parental cells (Fig. 4a). Moreover, E-cadherin expression was decreased, while those of ZEB1 and vimentin increased in H358-TGF-β cells compared to parental cells (Fig. 4b). Regarding the cell growth and cell cycle, TGF-β-induced EMT slowed down cell growth and increased the percentage of cells in the G0-G1 phase (Fig. 4c, d).
Fig. 4. Induction of epithelial-mesenchymal transition reduces the effectiveness of the combination treatment of avutometinib and defactinib.
a Representative microscopic images of NCI-H358 cells and H358-TGF-β cells are shown. b The cells are lysed, and the indicated proteins detected using western blotting. c Cell growth is determined using CCK-8 assay. d Cell cycle is defined as collection reduces via flow cytometry using FACS Accuri. e The combination index with avutometinib and defactinib in cells is calculated using CompuSyn®. The combination index of NCI-H358 is compared with that of H358-TGF-β. p values are calculated using student’s t-tests. f The cells are treated with avutometinib (150 nmol/L) in the presence or absence of defactinib (1 μmol/L) for 6 h. The cells are lysed, and the indicated proteins detected using western blotting.
Next, we calculated the CI values for avutometinib and defactinib in NCI-H358 and H358-TGF-β cells. CI values in H358-TGF-β cells were significantly higher than those in parental NCI-H358 cells (p = 0.012; Fig. 4e). Thus, EMT appeared to decrease the effects of avutometinib and defactinib combination therapy. When treated with avutometinib alone, FAK was activated in NCI-H358 cells but not in H358-TGF-β cells (Fig. 4f). Combination therapy with avutometinib and defactinib for 6 h inhibited the phosphorylation of AKT induced by avutometinib alone in NCI-H358 cells, but not in H358-TGF-β cells (Fig. 4f). Moreover, steady-state levels of AKT phosphorylation were higher in H358-TGF-β cells than in NCI-H358 cells. However, there was no difference in the inhibitory effect of avutometinib on phosphorylated ERK between NCI-H358 and H358-TGF-β cells (Fig. 4f). These results suggested that the additive effect of defactinib on epithelial-type KRAS-mutated cell lines treated with avutometinib was due to its contribution to the FAK-AKT-mediated feedback mechanism.
Combination therapy of avutometinib and defactinib maintains tumour regression in vivo
In vivo experiments were conducted to evaluate the effects of avutometinib and defactinib on NCI-H358 cells. Mice were treated with avutometinib alone, defactinib alone, or a combination of these therapies via daily oral gavage up to day 28. Treatment with avutometinib alone and combination therapy with avutometinib plus defactinib caused tumour regression. After treatment cessation, tumours treated with avutometinib alone exhibited rapid regrowth, while those subjected to combination therapy maintained a sustained tumour-suppressive effect. Tumour regression was observed in two mice receiving combination therapy. (Fig. S7A). Defactinib monotherapy only suppressed tumour growth without regression (Fig. 5a). No apparent adverse effects, including weight loss, were observed during the treatment period (Fig. S7B). The size and weight of the resected tumours were measured at the end of the experiment (day 49) to evaluate the effects of avutometinib and defactinib. Tumour-suppressive effect was observed with avutometinib alone and with combination therapy (Fig. 5b). The mean tumour weight in the combination therapy group was significantly lower than that in the avutometinib-treated group (p = 0.029; Fig. 5c).
Fig. 5. Combination of defactinib with avutometinib inhibits the growth of NCI-H358 tumours in vivo.
a NCI-H358 cell-line-derived xenograft (CDX) model mice, after randomisation, are treated daily by oral administration for 28 days with vehicle, defactinib (10.0 mg/kg), avutometinib (0.3 mg/kg), or a combination with defactinib (10.0 mg/kg) and avutometinib (0.3 mg/kg) (n = 6 per group). Tumour volumes are measured over time from the start of treatment, and the results are shown (mean ± SEM). b Images of NCI-H358 CDX tumours at the end of the experiment are shown. c NCI-H358 CDX tumours weights in each group at the end of the experiment are compared. p values are calculated using one-way ANOVA. d The NCI-H358 CDX model mice are treated with avutometinib (0.3 mg/kg) with or without defactinib (10.0 mg/kg) for 4 days. After collection, the tumours are lysed, and the indicated proteins detected using western blotting. e Representative images of hematoxylin & eosin staining and immunohistochemical staining with antibodies specific for human Ki-67 and TUNEL of tumours collected after 4 days of the treatment with each drug on NCI-H358 CDX model are shown. f The percentage of the Ki-67- and TUNEL-positive cells in each group is calculated from the average of the five evaluated areas. These are compared in each group (mean ± SD). p values are calculated using one-way ANOVA.
To further investigate the molecular changes induced by avutometinib and defactinib, we excised tumours from mice treated with these agents for 4 days and performed western blotting and immunohistochemical staining. Similar to the in vitro assays, the in vivo experiments showed that the combination of defactinib and avutometinib inhibited the avutometinib-activated phosphorylation of FAK and AKT, and Bim expression was higher in the combination group than in the other groups (Fig. 5d). The number of Ki-67-positive proliferative tumour cells was significantly lower in the combination group than in the avutometinib alone group (p < 0.001; Fig. 5e, f). Conversely, the number of TUNEL-positive tumour cells, i.e., apoptotic cells, was significantly higher in the combination group than in the avutometinib alone group (p < 0.001; Fig. 5e, f). Thus, these in vivo analyses showed that combination therapy with avutometinib and defactinib inhibited the AKT and MAPK pathways and induced apoptosis, resulting in the regression of the epithelial phenotype in KRAS-mutated NSCLC tumours (Fig. S8).
Discussion
FAK is a cytoplasmic non-receptor protein kinase belonging to the FAK family and was first identified in 1992 in BALB/c 3T3 cells using a homology-based cDNA cloning approach [24, 25]. FAK is activated by various transmembrane receptors such as integrins, growth factors, and cytokine receptors [26], and then the functional role of FAK is involved in cell proliferation, migration, and survival [27]. The FAK inhibitor, defactinib, appears to have more activity in KRAS-mutated NSCLC cells when compared to those with wild-type KRAS [28]. Meanwhile, defactinib monotherapy was not sufficiently effective in patients with KRAS-mutated NSCLC [29]. Therefore, it is necessary to identify drugs that could be used in combination with defactinib for KRAS-mutated NSCLC patients.
FAK is overexpressed in various malignant tumours, including lung cancer, and promotes progression, metastasis, and drug resistance [30, 31]. The activation of FAK signalling in EGFR-mutated NSCLC cells induces acquired resistance to an EGFR tyrosine kinase inhibitor (EGFR-TKI), and combination therapy with EGFR-TKI and FAK inhibitor has shown activity for these cells [32, 33]. We found that combination therapy with avutometinib and defactinib was active against KRAS-mutated NSCLC cells with epithelial phenotype, in which FAK was activated by avutometinib.
Several signalling pathways, such as the TGF-β signalling pathway, the Wnt/β-catenin signalling pathway, and the integrin signalling pathway, play important roles in EMT induction [34]. When EMT is induced, Snail, ZEB, and Twist repress epithelial gene expression and induce mesenchymal genes’ expression such as EXT1 [35, 36]. Several reports have demonstrated that, although RTKs are activated as a physiological negative feedback loop upon treatment with MEK inhibitors in KRAS-mutated NSCLC cells, the feedback loop is caused by different bypass signals in the epithelial and mesenchymal phenotypes. Epithelial cells activate IGF-1R and ERBB3 via c-MYC, whereas mesenchymal phenotype cells activate FRS2 via SPRY4 [37, 38]. When KRAS-mutated cancer cells with a mesenchymal phenotype are treated with KRAS inhibitors, dephosphorylated YAP translocates to the nucleus and binds to TEAD. This induces transcription of downstream genes such as MRAS, leading to resistance to KRAS inhibitors [39]. Combining TEAD inhibitors may overcome this KRAS inhibitor resistance [40]. In KRAS-mutated pancreatic ductal cancer cells, KRAS inhibition decreased c-Myc expression, upregulated the expression of a gene set associated with the IGF-1 pathway in gene array-based transcriptome analysis, and increased IGF-1R phosphorylation [41]. The FERM domain of FAK directly binds to the β-subunit of IGF-1R, housing the intracellular tyrosine kinase domain, thereby activating FAK in human cells [42, 43]. Therefore, our results show for the first time that combination therapy with avutometinib and defactinib is active against KRAS-mutated NSCLC cells with an epithelial phenotype via the cMYC-IGF-1R-FAK pathway (Fig. S8). The activation of AKT by long-term exposure to TGF-β (Fig. 4f) was presumed to be due to the induction of EMT.
Previous reports indicated that in KRAS-mutated cancer cells, trametinib, a MEK inhibitor, caused an increase in the total protein level of Bim, which is a proapoptotic BH-3-only protein [38, 44]. Consistent with a previous report on other MEK inhibitors, we showed that avutometinib induced Bim expression in KRAS-mutated NSCLC cells with an epithelial phenotype (Fig. 3c). Additionally, the use of defactinib in combination with avutometinib further upregulated Bim expression in vitro and in vivo (Figs. 3c and 5d). To our knowledge, there are no published reports on the upregulation of BIM following treatment with FAK inhibitors. In the combination group, tumour regrowth was not observed even 3 weeks after the cessation of administration, and the tumour-suppressive effect was sustained (Fig. 5a). This delay in tumour regrowth may be due to the ability of both agents to induce apoptosis more strongly.
The suppression of the AKT pathway activates downstream FoxO3a, resulting in the restoration of Bim transcriptional induction by FoxO3a [45, 46]. In our study, a decrease in phosphorylated FoxO3a was also observed when defactinib was used in combination with avutometinib (Fig. 3c). This suggests that Bim expression may have increased further through this pathway. Therefore, owing to the additional induction of Bim expression, combination therapy with avutometinib and defactinib was found to be strongly effective in KRAS-mutated NSCLC cells with the epithelial phenotype by inducing apoptosis (Fig. S8).
Several clinical trials have been conducted using MEK inhibitors in combination with other inhibitors for patients with KRAS-mutated NSCLC. Combination therapy with the pan-HER family tyrosine kinase inhibitor afatinib, which can inhibit ERBB3 activation, and selumetinib is a promising strategy focusing on the feedback mechanism. However, its efficacy is limited owing to its side effects [47]. In contrast, combination therapy with avutometinib and defactinib was generally well-tolerated in a phase I trial (NCT03875820) [18, 48]. Therefore, this combination therapy is promising and is currently being tested in clinical trials for patients with several cancers, including KRAS-mutated NSCLC and LGSOC. In this phase I/II trial, the objective response rate to combination therapy was 15.0% in patients with KRAS-mutated NSCLC and 44.8% in those with LGSOC [18, 19, 48, 49]. Additionally, two ongoing phase II trials aim to evaluate the effectiveness of avutometinib and defactinib combination therapy in patients with KRAS-mutated NSCLC and in those with LGSOC [18, 20, 50], and a useful biomarker is urgently needed for this combination therapy in patients with KRAS-mutated NSCLC. As shown in Fig. 2e, EMT status may serve as a practical biomarker.
In summary, the FAK inhibitor defactinib suppressed the bypass signal activated by treatment with the RAF/MEK clamp avutometinib in KRAS-mutated NSCLC cells with an epithelial phenotype. Moreover, combination therapy effectively induced the intrinsic apoptosis-mediated FoxO3a and Bim pathways. Further clinical investigations are needed to validate the usefulness of EMT status as a biomarker for combination therapy.
Supplementary information
Acknowledgements
We thank Editage (www.editage.com) for help with English language editing and M. Sukeno and M. Morita for technical support with the experiments.
Author contributions
Conception and design: AY and MH. Development of the methodology: AY, MH and T Yaoi. Acquisition of data (providing animals, acquiring and managing patients, and providing facilities): AY and T Yaoi. Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): AY, MH, T Yaoi and HO. Writing – Original Draft: AY, MH, and T Yaoi. Writing – Review and revision of manuscript: AY, MH, T Yaoi, HO, KI, T Yamada, KT, and TS. Administrative, technical, or material support (i.e., reporting or organising data and constructing databases): AY, T Yaoi, KI, T Yamada, and KT. Study supervision: TS.
Funding
This research was supported by a grant-in-aid from the Public Promoting Association Asano Foundation for Studies on Medicine (Yoshimura, Japan).
Data availability
The data that support the findings of this study are available from the authors upon reasonable request.
Competing interests
TY received research grants from Ono Pharmaceutical, Janssen, AstraZeneca, and Takeda Pharmaceutical and a personal fee from Eli Lilly. KT received research grants from Chugai Pharmaceutical and Ono Pharmaceutical and personal fees from AstraZeneca, Chugai Pharmaceutical, MSD-Merck, Eli Lilly, Boehringer-Ingelheim, and Daiichi-Sankyo. TS received research grants from Otsuka Pharmaceutical, Taiho Pharmaceutical, and Oncolys BioPharma and a patent fee from JT Pharmaceutical. The remaining authors declare no potential conflicts of interest.
Ethics approval
Approval was obtained from the Institutional Review Board of the Kyoto Prefectural University of Medicine for the mice study (approval no. M2021-38) following the ARRIVE guidelines.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
The online version contains supplementary material available at 10.1038/s41416-024-02727-2.
References
- 1.Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K, et al. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res. 2015;4:156–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.El Osta B, Behera M, Kim S, Berry LD, Sica G, Pillai RN, et al. Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: the lung cancer mutation consortium experience. J Thorac Oncol. 2019;14:876–89. 10.1016/j.jtho.2019.01.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Prior IA, Hood FE, Hartley JL. The frequency of ras mutations in cancer. Cancer Res. 2020;80:2969–74. 10.1158/0008-5472.CAN-19-3682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383:1207–17. 10.1056/NEJMoa1917239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jänne PA, Riely GJ, Gadgeel SM, Heist RS, Ou SI, Pacheco JM, et al. Adagrasib in non-small-cell lung cancer harboring a KRAS(G12C) mutation. N Engl J Med. 2022;387:120–31. 10.1056/NEJMoa2204619 [DOI] [PubMed] [Google Scholar]
- 6.de Langen AJ, Johnson ML, Mazieres J, Dingemans AC, Mountzios G, Pless M, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRAS(G12C) mutation: a randomised, open-label, phase 3 trial. Lancet. 2023;401:733–46. 10.1016/S0140-6736(23)00221-0 [DOI] [PubMed] [Google Scholar]
- 7.Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011;11:761–74. 10.1038/nrc3106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Carter CA, Rajan A, Keen C, Szabo E, Khozin S, Thomas A, et al. Selumetinib with and without erlotinib in KRAS mutant and KRAS wild-type advanced nonsmall-cell lung cancer. Ann Oncol. 2016;27:693–9. 10.1093/annonc/mdw008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Infante JR, Fecher LA, Falchook GS, Nallapareddy S, Gordon MS, Becerra C, et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:773–81. 10.1016/S1470-2045(12)70270-X [DOI] [PubMed] [Google Scholar]
- 10.Blumenschein GR Jr, Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)†. Ann Oncol. 2015;26:894–901. 10.1093/annonc/mdv072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lito P, Saborowski A, Yue J, Solomon M, Joseph E, Gadal S, et al. Disruption of CRAF-mediated MEK activation is required for effective MEK inhibition in KRAS mutant tumors. Cancer Cell. 2014;25:697–710. 10.1016/j.ccr.2014.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer. 2015;15:577–92. 10.1038/nrc4000 [DOI] [PubMed] [Google Scholar]
- 13.Lake D, Corrêa SA, Müller J. Negative feedback regulation of the ERK1/2 MAPK pathway. Cell Mol Life Sci. 2016;73:4397–413. 10.1007/s00018-016-2297-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sakai T. “RB-reactivator screening” as a novel cell-based assay for discoveries of molecular targeting agents including the first-in-class MEK inhibitor trametinib (trade name: Mekinist). Pharmacol Ther. 2022;236:108234. 10.1016/j.pharmthera.2022.108234 [DOI] [PubMed] [Google Scholar]
- 15.Ishii N, Harada N, Joseph EW, Ohara K, Miura T, Sakamoto H, et al. Enhanced inhibition of ERK signaling by a novel allosteric MEK inhibitor, CH5126766, that suppresses feedback reactivation of RAF activity. Cancer Res. 2013;73:4050–60. 10.1158/0008-5472.CAN-12-3937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wada M, Horinaka M, Yamazaki T, Katoh N, Sakai T. The dual RAF/MEK inhibitor CH5126766/RO5126766 may be a potential therapy for RAS-mutated tumor cells. PLoS One. 2014;9:e113217. 10.1371/journal.pone.0113217 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Guo C, Chénard-Poirier M, Roda D, de Miguel M, Harris SJ, Candilejo IM, et al. Intermittent schedules of the oral RAF-MEK inhibitor CH5126766/VS-6766 in patients with RAS/RAF-mutant solid tumours and multiple myeloma: a single-centre, open-label, phase 1 dose-escalation and basket dose-expansion study. Lancet Oncol. 2020;21:1478–88. 10.1016/S1470-2045(20)30464-2 [DOI] [PubMed] [Google Scholar]
- 18.Krebs MG, Shinde R, Rahman RA, Grochot R, Little M, King J, et al. A phase I trial of the combination of the dual RAF-MEK inhibitor VS-6766 and the FAK inhibitor defactinib: evaluation of efficacy in KRAS mutated NSCLC. Cancer Res. 2021;81:CT2019. suppl (July 01, 2021). 10.1158/1538-7445.AM2021-CT019 [DOI] [Google Scholar]
- 19.Banerjee, SN, Ring, KL, Nieuwenhuysen, EV, Fabbro, M, Aghajanian, C, Oaknin, A et al. Initial efficacy and safety results from ENGOT-ov60/GOG-3052/RAMP 201: a phase 2 study of avutometinib (VS-6766) ± defactinib in recurrent low-grade serous ovarian cancer (LGSOC). J Clin Oncol. 41, suppl (June 01, 2023) #5515 (2023).
- 20.Reuss, JE, Gandhi, SG, Spigel, DR, Janne, PA, Paz-Ares, LG, Gadgeel, SM et al. RAMP 202: a phase 2 study of avutometinib (VS-6766) ± defactinib, in patients with advanced KRAS G12V mutant non-small cell lung cancer (NSCLC). J Clin Oncol. 41, suppl (June 01, 2023) #9100 (2023).
- 21.Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7. 10.1186/1471-2105-14-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tan TZ, Miow QH, Miki Y, Noda T, Mori S, Huang RY, et al. Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol Med. 2014;6:1279–3. 10.15252/emmm.201404208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 2009;19:156–72. 10.1038/cr.2009.5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hanks SK, Calalb MB, Harper MC, Patel SK. Focal adhesion protein-tyrosine kinase phosphorylated in response to cell attachment to fibronectin. Proc Natl Acad Sci USA. 1992;89:8487–91. 10.1073/pnas.89.18.8487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Schaller MD, Borgman CA, Cobb BS, Vines RR, Reynolds AB, Parsons JT. pp125FAK a structurally distinctive protein-tyrosine kinase associated with focal adhesions. Proc Natl Acad Sci USA. 1992;89:5192–6. 10.1073/pnas.89.11.5192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18:516–23. 10.1016/j.ceb.2006.08.011 [DOI] [PubMed] [Google Scholar]
- 27.Hauck CR, Hsia DA, Schlaepfer DD. The focal adhesion kinase–a regulator of cell migration and invasion. IUBMB Life. 2002;53:115–9. 10.1080/15216540211470 [DOI] [PubMed] [Google Scholar]
- 28.Tang KJ, Constanzo JD, Venkateswaran N, Melegari M, Ilcheva M, Morales JC, et al. Focal adhesion kinase regulates the DNA damage response and its inhibition radiosensitizes mutant KRAS lung cancer. Clin Cancer Res. 2016;22:5851–63. 10.1158/1078-0432.CCR-15-2603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gerber DE, Camidge DR, Morgensztern D, Cetnar J, Kelly RJ, Ramalingam SS, et al. Phase 2 study of the focal adhesion kinase inhibitor defactinib (VS-6063) in previously treated advanced KRAS mutant non-small cell lung cancer. Lung Cancer. 2020;139:60–67. 10.1016/j.lungcan.2019.10.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tai YL, Chen LC, Shen TL. Emerging roles of focal adhesion kinase in cancer. Biomed Res Int. 2015;2015:690690. 10.1155/2015/690690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yoon H, Dehart JP, Murphy JM, Lim ST. Understanding the roles of FAK in cancer: inhibitors, genetic models, and new insights. J Histochem Cytochem. 2015;63:114–28. 10.1369/0022155414561498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fu Y, Zhang Y, Lei Z, Liu T, Cai T, Wang A, et al. Abnormally activated OPN/integrin αVβ3/FAK signalling is responsible for EGFR-TKI resistance in EGFR mutant non-small-cell lung cancer. J Hematol Oncol. 2020;13:169. 10.1186/s13045-020-01009-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ichihara E, Westover D, Meador CB, Yan Y, Bauer JA, Lu P, et al. SFK/FAK signaling attenuates osimertinib efficacy in both drug-sensitive and drug-resistant models of EGFR-mutant lung cancer. Cancer Res. 2017;77:2990–3000. 10.1158/0008-5472.CAN-16-2300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 2014;7:re8. 10.1126/scisignal.2005189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sleeman JP, Thiery JP. SnapShot: The epithelial-mesenchymal transition. Cell. 2011;145:162.e161. 10.1016/j.cell.2011.03.029 [DOI] [PubMed] [Google Scholar]
- 36.Solaimuthu B, Khatib A, Tanna M, Karmi A, Hayashi A, Abu Rmaileh A, et al. The exostosin glycosyltransferase 1/STAT3 axis is a driver of breast cancer aggressiveness. Proc Natl Acad Sci USA. 2024;121:e2316733121. 10.1073/pnas.2316733121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kitai H, Ebi H, Tomida S, Floros KV, Kotani H, Adachi Y, et al. Epithelial-to-mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS-mutant lung cancer. Cancer Discov. 2016;6:754–69. 10.1158/2159-8290.CD-15-1377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sun C, Hobor S, Bertotti A, Zecchin D, Huang S, Galimi F, et al. Intrinsic resistance to MEK inhibition in KRAS mutant lung and colon cancer through transcriptional induction of ERBB3. Cell Rep. 2014;7:86–93. 10.1016/j.celrep.2014.02.045 [DOI] [PubMed] [Google Scholar]
- 39.Adachi Y, Kimura R, Hirade K, Yanase S, Nishioka Y, Kasuga N, et al. Scribble mis-localization induces adaptive resistance to KRAS G12C inhibitors through feedback activation of MAPK signaling mediated by YAP-induced MRAS. Nat Cancer. 2023;4:829–43. 10.1038/s43018-023-00575-2 [DOI] [PubMed] [Google Scholar]
- 40.Edwards AC, Stalnecker CA, Jean Morales A, Taylor KE, Klomp JE, Klomp JA, et al. TEAD inhibition overcomes YAP1/TAZ-driven primary and acquired resistance to KRASG12C inhibitors. Cancer Res. 2023;83:4112–29. 10.1158/0008-5472.CAN-23-2994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rajbhandari N, Lin WC, Wehde BL, Triplett AA, Wagner KU. Autocrine IGF1 signaling mediates pancreatic tumor cell dormancy in the absence of oncogenic drivers. Cell Rep. 2017;18:2243–55. 10.1016/j.celrep.2017.02.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zheng D, Kurenova E, Ucar D, Golubovskaya V, Magis A, Ostrov D, et al. Targeting of the protein interaction site between FAK and IGF-1R. Biochem Biophys Res Commun. 2009;388:301–5. 10.1016/j.bbrc.2009.07.156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Andersson S, D’Arcy P, Larsson O, Sehat B. Focal adhesion kinase (FAK) activates and stabilizes IGF-1 receptor. Biochem Biophys Res Commun. 2009;387:36–41. 10.1016/j.bbrc.2009.06.088 [DOI] [PubMed] [Google Scholar]
- 44.Yamada T, Amann JM, Tanimoto A, Taniguchi H, Shukuya T, Timmers C, et al. Histone deacetylase inhibition enhances the antitumor activity of a MEK inhibitor in lung cancer cells harboring RAS mutations. Mol Cancer Ther. 2018;17:17–25. 10.1158/1535-7163.MCT-17-0146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.De Bruyne E, Bos TJ, Schuit F, Van Valckenborgh E, Menu E, Thorrez L, et al. IGF-1 suppresses Bim expression in multiple myeloma via epigenetic and posttranslational mechanisms. Blood. 2010;115:2430–40. 10.1182/blood-2009-07-232801 [DOI] [PubMed] [Google Scholar]
- 46.Sunters A, Fernández de Mattos S, Stahl M, Brosens JJ, Zoumpoulidou G, Saunders CA, et al. FoxO3a transcriptional regulation of Bim controls apoptosis in paclitaxel-treated breast cancer cell lines. J Biol Chem. 2003;278:49795–805. 10.1074/jbc.M309523200 [DOI] [PubMed] [Google Scholar]
- 47.van Brummelen EMJ, Huijberts S, van Herpen C, Desar I, Opdam F, van Geel R, et al. Phase I Study of afatinib and selumetinib in patients with KRAS-mutated colorectal, non-small cell lung, and pancreatic cancer. Oncologist. 2021;26:290–e545. 10.1002/onco.13631 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Stewart, AR, Pickard, L, Paranjape, E, Perez, VS, Chowdhury, S, Lustgarten, S et al. Mechanistic evaluation of VS-6766 (dual RAF/MEK inhibitor) and defactinib (FAK inhibitor) in low-grade serous ovarian cancer models with correlations to clinical response. Cancer Res.82, suppl (June 15, 2022) #3476 (2022).
- 49.Banerjee, S, Grochot, R, Shinde, R, Lima, J, Krebs, M, Rahman, R et al. Phase I study of the combination of the dual RAF/MEK inhibitor VS-6766 and the FAK inhibitor defactinib: results of efficacy in low-grade serous ovarian cancer. Ann. Oncol.32, suppl (Sep 01, 2021) #2799 (2021).
- 50.Capelletto E, Bironzo P, Denis L, Koustenis A, Bungaro M, Novello S. Single agent VS-6766 or VS-6766 plus defactinib in KRAS-mutant non-small-cell lung cancer: the RAMP-202 phase II trial. Future Oncol. 2022;18:1907–15. 10.2217/fon-2021-1582 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support the findings of this study are available from the authors upon reasonable request.