Purpose: Non–small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide due to the limited availability of effective therapeutic options. For instance, there are no effective strategies for NSCLCs that harbor mutant KRAS, the most commonly mutated oncogene in NSCLC. Thus, our purpose was to make progress toward the generation of a novel therapeutic strategy for NSCLC.

Experimental Design: We characterized the effects of suppressing focal adhesion kinase (FAK) by RNA interference (RNAi), CRISPR/CAS9 gene editing or pharmacologic approaches in NSCLC cells and in tumor xenografts. In addition, we tested the effects of suppressing FAK in association with ionizing radiation (IR), a standard-of-care treatment modality.

Results: FAK is a critical requirement of mutant KRAS NSCLC cells. With functional experiments, we also found that, in mutant KRAS NSCLC cells, FAK inhibition resulted in persistent DNA damage and susceptibility to exposure to IR. Accordingly, administration of IR to FAK-null tumor xenografts causes a profound antitumor effect in vivo.

Conclusions: FAK is a novel regulator of DNA damage repair in mutant KRAS NSCLC and its pharmacologic inhibition leads to radiosensitizing effects that could be beneficial in cancer therapy. Our results provide a framework for the rationale clinical testing of FAK inhibitors in NSCLC patients. Clin Cancer Res; 22(23); 5851–63. ©2016 AACR.

Translational Relevance

There is a dearth of therapeutic options for mutant KRAS non–small cell lung cancer (NSCLC), a disease associated with an aggressive clinical course and resistance to therapy. We report that focal adhesion kinase (FAK) represents a vulnerability of mutant KRAS NSCLC. Suppression or pharmacologic inhibition of FAK causes DNA damage and radiosensitizing effects that promote the therapeutic effect of ionizing radiations both in cultured cells and in lung cancer xenografts. Several FAK inhibitors have entered clinical testing, but it is still undefined in which patient population they could be effective and whether any biomarker exists to identify patients likely to respond to therapy. Thus, our findings provide a framework for their clinical development in the context of radiotherapy, a common form of therapy used for lung cancer.

Non–small cell lung cancer (NSCLC) is associated with an aggressive course and poor prognosis. These features are due to the fact that this cancer type is often diagnosed at a stage not amenable to surgical resection. In addition, NSCLC is either endowed with or acquires resistance to available medical treatments. Accordingly, NSCLC is a leading cause of cancer-related deaths worldwide (1). In NSCLC several known oncogenes promote tumorigenesis and predict response to therapy. For instance, approximately 40% of NSCLC harbor mutations of KRAS, EGFR, ALK translocation, or amplification of c-MET (2, 3). While there are specific inhibitors of EGFR, ALK, or MET that cause clinically meaningful, but short-lived antitumor responses, mutant KRAS remains undruggable (4, 5). Thus, therapeutic options are still limited for patients with oncogenic KRAS NSCLC.

KRAS belongs to a family of small guanosine triphosphatases (GTPase). Tumor-associated mutations lock KRAS in a constitutively active state (i.e., oncogenic KRAS; refs. 6, 7). When bound to GTP, KRAS activates several critical cell proliferation and survival signals, which include the PI3K/mTOR, MEK1/2/ERK1/2, RHOA-Focal adhesion kinase (FAK), and TBK1 signaling networks (6, 8–10).

Oncogenic KRAS is not only sufficient to induce lung cancer, but also required for its maintenance in transgenic mice and in human lung cancer cells (11–14). However, progression to high-grade lung adenocarcinoma requires co-occurring mutations, such as the loss of TP53, CDKN2AB, Ataxia Telangiectasia Mutated (ATM), or LKB1, which allow bypass of tumor suppressive responses induced by inappropriate proliferative stimuli, DNA damage, or energetic stress (2, 3, 8, 11–13, 15–17). The loss of tumor suppressors implicated in DNA damage repair is relevant because oncogenic KRAS also stimulates the production of reactive oxygen species (ROS), which promote DNA damage and genomic instability (18, 19).

Because attempts to develop direct inhibitors of oncogenic KRAS have been unsuccessful (4), intense efforts have been spent on targeting critical components of its downstream signaling networks in preclinical models. Several PI3K, mTOR, MEK1/2, and FAK inhibitors (FAKi) are undergoing clinical testing, but they have not been approved for therapy of lung cancer (20–23). Thus, there is still an urgent need for the development of therapies that target oncogenic KRAS tumors.

Protein tyrosine kinase 2 (PTK2), also known as FAK, is a nonreceptor tyrosine kinase and a major mediator of integrin signaling. Upon autophosphorylation at Tyr397, FAK interacts with SRC protein kinase family members, initiating several signaling cascades that regulate cytoskeleton remodeling, cell migration and resistance to anoikis (24). FAK is amplified or overexpressed in several cancer types, including ovarian, colon, breast, and lung cancers (25, 26). Importantly, FAK inhibition is detrimental to breast and lung cancer cells: in this context, disruption of FAK is associated with alterations in the cytoskeleton or induction of senescence and activation of DNA damage pathways, respectively (8, 25). Furthermore, in lung cancer, mutant KRAS is a positive regulator of FAK (8). However, the mechanisms underlying senescence induction following FAK inhibition and the functional consequences of this event in cancer cells remain unexplored.

In this study, we characterized the effects of FAK suppression in a large panel of NSCLC cells representative of frequent cancer associated mutations. We found that FAK inhibition invariably impairs the viability of mutant KRAS NSCLC cells. In this genetic context, suppression or inhibition of FAK was accompanied by DNA damage. In addition, we demonstrate that FAK suppression synergizes with radiotherapy both in vivo and in vitro. We propose that combination therapy with FAK inhibition and ionizing radiation (IR) may lead to important clinical benefits in the treatment of NSCLC with oncogenic KRAS.

Cell cultures and reagents

Human NSCLC cell lines and human bronchoalveolar cells were provided by Dr. John Minna (UT Southwestern Medical Center, Dallas, TX) and cultured as described (27, 28). A549 and H460 cells expressing vector control or LKB1 were previously described (17). All cells were mycoplasma free and were identified by DNA fingerprinting. Supplementary Table S1 shows the mutations that they harbor according to COSMIC: Catalogue of Somatic Mutations in Cancer (Cosmic; cancer.sanger.ac.uk). FAKi PF-562,271 and VS-4718 were obtained from Selleckchem and Verastem, Inc., respectively (29, 30). Inhibitors were added to mid-log phase cell cultures at the indicated concentrations. All other chemicals were purchased from Sigma-Aldrich.

RNA interference

Stable FAK mRNA knockdown was performed with lentiviral vectors containing shRNAs against FAK obtained from the RNAi Consortium (TRC) following procedure described previously (8). Inducible expression of FAK shRNA was performed with the GEPIR vector (31). See also Supplementary Materials and Methods.

Gene editing with CRISPR/CAS9

We followed established procedures to ablate FAK (exon 4) using the following vectors: pCW57.1 (Addgene plasmid 50661) and pLX-sgRNA (Addgene plasmid 50662; ref. 32). We selected several single clones and assessed FAK editing by direct sequencing and by lack of FAK protein by Western blot (WB) analysis.

Cell viability and proliferation curves

Cells (0.5×104 to 1.5×104) were plated in triplicate (24-well plates) 14 to 16 hours prior to exposure to pharmacologic inhibitors. At the indicated time points or 72 hours later in cell viability assays, the cells were fixed with 10% formalin and stained with 0.1% crystal violet (8).

Immunoblotting and antibodies

WB analyses were performed as previously described (33). We used the following antibodies: FAK, Tyr397 phospho-FAK, Akt, Ser473 phospho-Akt, S6, Ser235/236 phospho-S6, Ser139 γ-H2AX (Cell Signaling Technology), H2AX (Bethyl), β-Tubulin and GAPDH (Santa Cruz Biotechnology), Vinculin and LKB1 (Cell Signaling Technology).

Colony formation assays

For colony formation assays in plastic, we plated 500 to 1,500 cells in 60-mm tissue culture dishes and scored colonies of >50 normal-appearing cells after 12 to 30 days. For soft-agar colony assays, we seeded 2,000 cells/well on semisolid agar medium in a 6-well plate in triplicate and after 14 to 21 days, colonies larger than 50 μm were counted using an inverted microscope (34).

Flow cytometry

Cells were allowed to adhere overnight. When indicated, cells were treated with PF-562,271 and/or IR (2 Gy). Analysis of the cell cycle and percentage of cells stained with propidium iodide (PI; Sigma-Aldrich) and γ-H2AX (Millipore) were performed following a standard procedure with a FC500 Beckman Coulter flow cytometer using the WinMDI V2.8 software (35, 36).

Plasmids, transfections, and retroviral transductions

pMXs-Puro-GFP-Fak was obtained from Addgene (FAK-Plasmid #38194). Retroviral transductions were performed as described (37).

Expression profiling

Gene expression profiles were obtained from exponentially growing HBEC cells transduced with pMXs-Puro-GFP or pMXs-Puro-GFP-Fak. Microarray results have been deposited in NCBI's Gene Expression Omnibus and are accessible through GEO series accession number GSE72470. See also Supplementary Data.

Clonogenic survival assays after exposure to IRs

Clonogenic assays were performed as described previously (38). Surviving fractions (SF) were derived using the number of colonies formed after treatment, divided by the number of cells seeded multiplied by plating efficiency. Cells were plated in triplicate onto 60-mm dishes 14 to 16 hours prior to irradiation. We added PF-562,271 at indicated concentrations to the cells four hours before irradiation. Drug-containing medium was replaced with drug-free medium after 48 hours. Cells were irradiated with a Mark I cesium irradiator (1.31 Gy/min). We scored colonies of >50 normal-appearing cells 12 to 30 days after treatment and graphed the SF versus dose of IR (Gy). Do (relative dose of IR required for 37% lethality on a log-phase kill curve), Dq (inherent DNA repair capacity: dose (Gy) required to eliminate the survival curve shoulder) and dose enhancement ratios (DERs at LD50 and LD20) were calculated as described (39).

Immunofluorescence

Immunofluorescence was performed as described previously using γ-H2AX (Millipore), TP53BP1 (Bethyl), and FAK (Abicam ab40794) antibodies (8).

Mouse studies

Xenograft experiments using T2.2 FAK wild-type (FAK+) and T2.2 FAK-null (FAK) H460 NSCLC cells were performed by subcutaneous inoculation of cells into 6-week-old female athymic nude mice. Mice with xenograft tumors of 300 mm3 (7 mice/group) were treated with IR. Mice were irradiated with five 4-Gy fractions every other day for 10 days using an X-RAD 320 irradiator (Precision X-Ray, Inc.) to deliver local irradiation to the flank or thigh of lead-shielded mice. Tumor volumes were calculated every other day using the formula: (length × width2)/2. All studies were performed according to the guidelines of the UT Southwestern Institutional Animal Care and Use Committee.

Statistical analyses

All data presented are the average ± standard deviations of experiments repeated three or more times. Significance was determined using two-tailed unpaired Student t tests or one-way ANOVA. Curve fitting for the radiosensitization experiments was performed using the linear-quadratic formula which has two components of cell killing: one is proportional to the dose of IR (aD) and the other is proportional to the square of the dose of IR (bD2): exp (aD+bD2) (38, 40).

Pharmacologic inhibition or silencing of FAK impairs the viability of lung cancer cells

The goal of our study was to shed light on the function of FAK in lung cancer cells and to leverage this knowledge to identify novel therapeutic opportunities. Pharmacologic inhibition of FAK with the small molecule inhibitor (FAKi) PF-562,271 led to a striking inhibition of cell viability in a panel of NSCLC cell lines (detailed genotype information is provided in Supplementary Table S1). We noted that the IC50 of mutant KRAS NSCLC cell lines ranged between 2 and 4 μmol/L, while in wild-type KRAS cell lines the IC50 was 8 μmol/L or higher (Fig. 1A; Supplementary Table S2). When used at a concentration between 2 and 4 μmol/L, PF-562,271 led to approximately a 50% reduction of auto-phosphorylation of FAK at Y397 (P-FAK, the active form of FAK), both in wild-type and in mutant KRAS NSCLC cells (Fig. 1B; Supplementary Fig. S1A). We obtained a similar inhibition of cell viability and P-FAK when treating NSCLC cells with VS-4718, a FAKi structurally distinct from PF-562,271 (Supplementary Fig. S1B–S1D).

Figure 1.

Suppression of FAK leads to loss of viability in mutant KRAS NSCLC cells. A, Viability assay of NSCLC cell lines treated with FAKi PF-562,271 at the indicated concentrations. B, WB of H460-mutant KRAS NSCLC cells treated with the indicated concentrations of PF-562,271. C, histogram showing viability of NSCLC cells expressing the indicated shRNAs. The mutation status of the cell lines is indicated. D, WB of H460 cells expressing the indicated shRNAs. E, WB of CRISPR/CAS9-edited H460 cell clones T2.2 and T2.7; FAK+ and FAK indicate wild-type and null status, respectively. F and G, Proliferation assay of H460 T.2.2 and T2.7 clones. FAK status is indicated. A representative picture of a tissue culture well is provided. Error bars are indicated.

Figure 1.

Suppression of FAK leads to loss of viability in mutant KRAS NSCLC cells. A, Viability assay of NSCLC cell lines treated with FAKi PF-562,271 at the indicated concentrations. B, WB of H460-mutant KRAS NSCLC cells treated with the indicated concentrations of PF-562,271. C, histogram showing viability of NSCLC cells expressing the indicated shRNAs. The mutation status of the cell lines is indicated. D, WB of H460 cells expressing the indicated shRNAs. E, WB of CRISPR/CAS9-edited H460 cell clones T2.2 and T2.7; FAK+ and FAK indicate wild-type and null status, respectively. F and G, Proliferation assay of H460 T.2.2 and T2.7 clones. FAK status is indicated. A representative picture of a tissue culture well is provided. Error bars are indicated.

Close modal

Of note, pharmacokinetic studies have demonstrated that PF-562,271 and VS-4718 (formerly known as PND-1186) reach a concentration of 1 to 2 μmol/L in vivo (29, 41). Notably, PF-562,271 inhibits its target, causing antitumor effects in vivo in a mutant KRAS lung cancer model (8).

Integrin engagement with the extracellular matrix has been implicated in the activation of FAK; thus, we tested whether culturing NSCLC cells on collagen coated plates would affect their vulnerability to FAKi. However, culturing NSCLC cells on collagen-coated plates did not affect their sensitivities to FAKi (Supplementary Fig. S1E–S1G). Taken together, our findings suggest that pharmacologic inhibition of FAK may be of therapeutic value in mutant KRAS NSCLC.

To validate, with an alternative technique, the results obtained with FAKi, we inactivated FAK genetically in a panel of 21 NSCLC cells harboring mutant KRAS or wild-type KRAS, mutant EGFR, the EML4–ALK fusion gene or MET amplification and loss/mutations of the TP53, CDKN2A/B, or LKB1 tumor suppressors (detailed genotype information is provided in Supplementary Table S1). We found that silencing FAK invariably leads to loss of viability in mutant KRAS lung cancer cells (Fig. 1C). Notably, the degree of FAK silencing was comparable between wild-type and mutant KRAS NSCLC cells (Fig. 1C–D; Supplementary Fig. S1H). In contrast, the effect of FAK silencing on cell viability was not consistent in lung cancer cells carrying genotypes other than mutant KRAS. For instance, FAK silencing strikingly reduced the viability of H1975 (mutant EGFR) and H1993 (MET amplified) cells, but not of H1650 and HCC827 (mutant EGFR) or H920 cells (MET amplified). We also noticed that the vulnerability to FAK silencing was comparable between mutant KRAS cells that carry p53, CDKN2a, or LKB1 mutations (Fig. 1C; Supplementary Table S1). This result confirms and extends our previous observations in a smaller sample size of mutant KRAS NSCLC cells, providing further support to the notion that FAK is a therapeutic target in mutant KRAS NSCLC (8).

It is well known that small molecule inhibitors of protein tyrosine kinases as well as RNAi-mediated gene silencing, may lead to off-target effects. Thus, we ablated FAK by CRISPR/CAS9 gene editing in H460-mutant KRAS lung NSCLC cells, as a representative example of the NSCLC used in our studies. We carried out our analysis in two independent FAK-null (T2.2 FAK and T2.7 FAK) and wild-type FAK (T2.2 FAK+ and T2.7 FAK+) H460 clones (Fig. 1E). We determined that, also in this setting, loss of FAK dramatically affects the proliferative capacity of H460 cells (Fig. 1F and G).

FAK is required for the oncogenic properties of mutant KRAS NSCLC cells in vitro

Next, we determined that FAK silencing impairs the ability of mutant KRAS H460, but not of wild-type KRAS H522 NSCLC cells, which we chose as representative examples, to grow in an anchorage-independent manner, a hallmark of oncogenic transformation and tumor aggressiveness (ref. 42; Fig. 2A). These cell lines are widely used in the literature for similar experiments. Furthermore, we determined that CRISPR/CAS9-mediated ablation of FAK significantly reduces the clonogenic ability of H460 cells when cultured on plastic dishes and in soft agar (Fig. 2B and C).

Figure 2.

Suppression of FAK impairs the clonogenic ability of mutant KRAS NSCLC cells. A, Representative soft-agar colony formation assay of H460 and H522 cells transduced with the indicated shRNAs. The histogram shows a quantification of three experiments performed in triplicate. B, Representative focus assay of CRISPR/CAS9-edited H460 T2.2 and T2.7 clones. FAK status is indicated. The histogram shows a quantification of three independent experiments performed in triplicate. C,Representative soft-agar colony formation assay of CRISPR/CAS9-edited H460 cells. The histogram shows a quantification of three independent experiments performed in triplicate. D, Representative focus assay of H460 cells expressing the doxy-dependent FAK shRNA1 grown in the absence (−) or presence (+) of doxy, which was added 48 hours after plating. The histogram shows a quantification of three independent experiments performed in triplicate. *, P < 0.05; **, P < 0.01; NS = nonstatistically significant.

Figure 2.

Suppression of FAK impairs the clonogenic ability of mutant KRAS NSCLC cells. A, Representative soft-agar colony formation assay of H460 and H522 cells transduced with the indicated shRNAs. The histogram shows a quantification of three experiments performed in triplicate. B, Representative focus assay of CRISPR/CAS9-edited H460 T2.2 and T2.7 clones. FAK status is indicated. The histogram shows a quantification of three independent experiments performed in triplicate. C,Representative soft-agar colony formation assay of CRISPR/CAS9-edited H460 cells. The histogram shows a quantification of three independent experiments performed in triplicate. D, Representative focus assay of H460 cells expressing the doxy-dependent FAK shRNA1 grown in the absence (−) or presence (+) of doxy, which was added 48 hours after plating. The histogram shows a quantification of three independent experiments performed in triplicate. *, P < 0.05; **, P < 0.01; NS = nonstatistically significant.

Close modal

It is also noteworthy that the acute silencing of FAK with a retrovirus that expresses a doxycycline (doxy)-inducible FAK shRNA, which mimics the action of a pharmacologic agent, significantly impairs the clonogenic ability of H460 cells when grown on plastic (Fig. 2D).

Taken together, these data support the conclusion that FAK is required for the transforming ability of oncogenic KRAS, which could be targeted for therapeutic purposes in lung cancer.

Suppression of FAK leads to a growth arrest in the G2 phase of the cell cycle

Flow cytometry revealed that pharmacologic inhibition of FAK leads to a significant increase of the percentage of the G2 phase of the cell cycle in mutant KRAS NSCLC cells, but not in wild-type KRAS H522 NSCLC cells (Fig. 3A and B). These data suggest that the effects of FAK suppression are mediated by a mechanism that utilizes a G2 cell-cycle checkpoint.

Figure 3.

Suppression of FAK promotes a DNA damage response. A and B, Cell-cycle analysis of H460 and H522 cells treated as indicated. The percentage of cells in each phase of the cell cycle is indicated. Note a significant increase in the percentage of G2 cells after treatment with FAKi. C, Heat maps of the genes enriched in a genome-wide expression profiling experiment, illustrating the changes in gene expression of HBEC cells expressing either pMXs-Puro-GFP or pMXs-Puro-GFP-Fak. Expression level shown is representative of ± log (2.5) of each replicate (n = 3 samples/condition). Red signal denotes higher expression relative to the mean expression level within the group and blue signal denotes lower expression relative to the mean expression level within the group. The histogram shows Gene Ontology analysis of differentially expressed genes in enrichment analysis. D, WB analysis of H460 and H522 cells expressing the indicated shRNAs. Note upregulation of γ-H2AX in H460 cells with FAK knockdown. E, WB analysis of NSCLC cells treated with FAKi as indicated. Note upregulation of γ-H2AX in mutant, but not in wild-type, KRAS NSCLC cells treated with FAKi. F, WB analysis of H460 T2.2 FAK+ and FAK cells; note upregulation of γ-H2AX in FAK-null cells. G and H, WB analysis of H460 or A549 cells (both LKB1 mutant) stably transduced with either control or with LKB1 expressing retroviral vectors (H460 or A549 + control and H460 or A549 + LKB1, respectively). Cells were treated as indicated. Note that PF-562,271 cause upregulation of γ-H2AX to a degree comparable to IR in all cell lines.

Figure 3.

Suppression of FAK promotes a DNA damage response. A and B, Cell-cycle analysis of H460 and H522 cells treated as indicated. The percentage of cells in each phase of the cell cycle is indicated. Note a significant increase in the percentage of G2 cells after treatment with FAKi. C, Heat maps of the genes enriched in a genome-wide expression profiling experiment, illustrating the changes in gene expression of HBEC cells expressing either pMXs-Puro-GFP or pMXs-Puro-GFP-Fak. Expression level shown is representative of ± log (2.5) of each replicate (n = 3 samples/condition). Red signal denotes higher expression relative to the mean expression level within the group and blue signal denotes lower expression relative to the mean expression level within the group. The histogram shows Gene Ontology analysis of differentially expressed genes in enrichment analysis. D, WB analysis of H460 and H522 cells expressing the indicated shRNAs. Note upregulation of γ-H2AX in H460 cells with FAK knockdown. E, WB analysis of NSCLC cells treated with FAKi as indicated. Note upregulation of γ-H2AX in mutant, but not in wild-type, KRAS NSCLC cells treated with FAKi. F, WB analysis of H460 T2.2 FAK+ and FAK cells; note upregulation of γ-H2AX in FAK-null cells. G and H, WB analysis of H460 or A549 cells (both LKB1 mutant) stably transduced with either control or with LKB1 expressing retroviral vectors (H460 or A549 + control and H460 or A549 + LKB1, respectively). Cells were treated as indicated. Note that PF-562,271 cause upregulation of γ-H2AX to a degree comparable to IR in all cell lines.

Close modal

Suppression of FAK leads to activation of the DNA damage response in mutant KRAS NSCLC cells

To characterize the effect of FAK suppression in NSCLC cells, we assessed the status of several targets of KRAS (6). We found that pharmacologic inhibition of FAK (FAKi) or FAK gene silencing did not affect p-AKT, p-ERK, and p-S6 levels (Supplementary Fig. S2A–S2E). This observation suggests that inhibitory effects on these signaling pathways do not cause the effect of FAKi on cell proliferation and viability.

To gain insight into the cellular networks affected by FAK, we performed gene expression analysis of primary human bronchoalveolar epithelial cells (HBEC) immortalized by expression of hTERT and CDK4. These cells have been extensively studied to determine the effect of oncogenic mutations in respiratory epithelial cells (28). In addition, using this cellular system we previously demonstrated that mutant KRAS activates FAK (8). Therefore, we reasoned that these cells are an experimental system to assess the cellular networks regulated by FAK. To this end, we compared the transcriptome of HBEC cells expressing pMXs-Puro-GFP or pMXs-Puro-GFP-Fak. Database for Annotation, Visualization and Integrated Discovery-based (DAVID) functional enrichment analysis and Gene Set Enrichment Analysis (GSEA) between experimental groups revealed a significant upregulation of genes that regulate G2–M DNA damage checkpoint, TGFβ signaling, and PKA signaling (Fig. 3C).

To address the relationship of FAK with DNA damage repair networks, we tested whether loss of FAK affects activation of γ-H2AX, an occurrence that was also previously reported to occur in breast cancer cells (25). Indeed, we found that silencing, pharmacologic inhibition, or ablation of FAK leads to upregulation of γ-H2AX in mutant KRAS NSCLC A549 and H460 cells but not in wild-type KRAS NSCLC H522 and H596 cells. Notably, there were no major differences in baseline γ-H2AX among the cell lines used for these experiments (Fig. 3D–F; Supplementary Fig. S2F). LKB1, which is lost in a significant percentage of mutant KRAS lung cancer, has been implicated in the regulation of the DNA damage response (43). To test whether this is the case also in lung cancer cells, we treated with FAKi PF-562,271 or IRs H460 (LKB1 mutant), A549 cells (LKB1 mutant) and their counterparts where LKB1 was reintroduced by retroviral transduction, H358 (wild-type LKB1) and HCC4017 (wild-type LKB1) cells (17). We found that PF-562,271 and 4 Gy of IR cause a comparable degree of upregulation of γ-H2AX. Furthermore, LKB1 did not influence the upregulation of γ-H2AX (Fig 3G and H; and Supplementary Fig. S3A–S3C). Taken together, our data support the conclusion that loss of FAK leads to a DNA damage response in mutant KRAS NSCLC cells.

FAK silencing or pharmacologic inhibition sensitizes oncogenic KRAS NSCLC cells to the effects of IRs

Our results suggest that FAK is required for promoting DNA damage repair in oncogenic KRAS NSCLC cells. Therefore, we tested whether pharmacologic inhibition of FAK sensitized NSCLC cells expressing oncogenic KRAS to the antiproliferative effects of IR. We performed clonogenic survival assays with H460, H358, H522, and H596 cells as representative examples of mutant and wild-type KRAS NSCLC cells, respectively, exposed to increasing doses of IR (1–6 Gy). We chose H358 cells for this experiment because they are LKB1 wild-type, while H460 and A549 are LBK1 mutant, to rule out the contribution of LKB1 loss to radiosensitization. We administered 1 μmol/L PF-562,271, because we found that this concentration inhibits P-FAK in a comparable manner in the cells we used for this study (data not shown), 4 hours before exposure to IR. We limited the incubation time to 48 hours not to affect plating efficiency.

We found that pharmacologic inhibition of FAK with PF-562,271 to irradiated mutant KRAS H460 and H358 NSCLC cells resulted in profound changes in Dq as well as significant, but less dramatic, decreases in Do (Fig. 4A and B). Thus, we concluded that FAKi reduces inherent DNA repair capacity (Dq) as indicated by significant changes in the shoulder of the survival curves. In contrast, exposure of wild-type KRAS NSCLC cells (H522 or H596; Fig. 4C and D) to PF-562,271 had no significant effect on IR-induced lethality as noted by the survival curve and estimations of Dq and Do. Most importantly, the presence of FAKi increased dose enhancement ratios (DER) with values ranging between 2.1 and 1.9 at LD50 levels, and between 1.7 and 1.4 at LD20 levels, respectively (Supplementary Table S3).

Figure 4.

FAK blockade sensitizes mutant KRAS NSCLC cells to the effects of IRs. A–D, Clonogenic survival assays of NSCLC cells treated as indicated. E and F, Clonogenic survival assays of H460 and H522 cells stably expressing a doxy-dependent FAK shRNA (shRNA1). Doxy turns on the shRNA. Doxy-treated cells are indicated. Colony number was calculated from three replicate plates of three independent experiments; bars, SD. Gy = Grey.

Figure 4.

FAK blockade sensitizes mutant KRAS NSCLC cells to the effects of IRs. A–D, Clonogenic survival assays of NSCLC cells treated as indicated. E and F, Clonogenic survival assays of H460 and H522 cells stably expressing a doxy-dependent FAK shRNA (shRNA1). Doxy turns on the shRNA. Doxy-treated cells are indicated. Colony number was calculated from three replicate plates of three independent experiments; bars, SD. Gy = Grey.

Close modal

Notably, the effect of pharmacologic inhibition of FAK was comparable to the dual PI3K/mTOR inhibitor BEZ-235, which is a known radiosensitizing agent (Fig. 4A; refs. 20, 44).

To confirm these findings with an independent approach, we silenced FAK. Because prolonged silencing of FAK severely impairs clonogenic growth of mutant KRAS NSCLC cell lines, we used the GEPIR retrovirus to express FAK shRNA1 in a doxycycline-regulated manner, obtaining results equivalent to FAKi (Fig. 4E and F). We found that inducible silencing of FAK had effects similar to PF-562,271 treatment in both mutant and wild-type KRAS NSCLC cells.

These data indicate that silencing or pharmacologic inhibition of FAK inhibits the DNA repair and/or augment DNA damage created by IR.

Radiosensitization induced by FAK blockade or loss is accompanied by persistence of DNA damage foci

Our data suggest that inhibition of FAK facilitates the cytotoxic effects of IR by promoting increased or unresolved DNA damage. Thus, we examined whether PF-562,271 affects induction and repair of DNA breaks after IR exposure in H460 cells. We determined that resolution of γ-H2AX foci, a well-known marker of DNA double-strand break (DSB) damage and repair (when foci decrease), occurred rapidly after treatment with IR (2 Gy). In contrast, treatment with the FAK inhibitor PF-562,271 in combination with IR (2 Gy) led to a striking persistence of γ-H2AX foci at 24 hours after IR administration compared with exposure with IR alone. The effect of PF-562,271 in regard to inducing the persistence of γ-H2AX foci was comparable to the effects of the dual PI3K/mTOR inhibitor BEZ-235, a known radiosensitizer (Fig. 5A; refs. 20, 44). Inhibition of FAK in the absence of IR induces γ-H2AX foci slightly above the background level in this assay (Fig. 5A; Supplementary Fig. S4A). We obtained equivalent results in H358 and HCC4017 (LKB1 wild-type) cells (Supplementary Fig. S4B–S4E).

Figure 5.

Radiosensitization induced by FAK blockade is accompanied by persistence of DNA damage foci. A, Detection by immunofluorescence of γ-H2AX foci in H460 cells treated with DMSO, PF-562,271, or BEZ-235 followed by 2 Gy of IR. Foci were detected at the indicated time points. Note striking increase in the number of foci 24 hours after treatment with PF-562,271 and BEZ-235 (a known radiosensitizing drug) in combination with IR. Bar, 25 μm. B, Detection by immunofluorescence of γ-H2AX foci in T2.2 FAK+ and FAK H460 cells after treatment with 2 Gy of IR. Foci were detected at the indicated time points. Note striking increase in the number of foci at 9 and 24 hours after IR. Bar, 25 μm. C, Cell cycle analysis of H460 cells treated as indicated. The percentage of cells in each phase of the cell cycle is indicated. Note a significant increase in the percentage of G2 cells after combination treatment with FAKi and IR. D, WB of H460 cells treated with PF-562,271 as indicated. E, WB of T2.2 FAK+ and FAK H460 cells after IR.

Figure 5.

Radiosensitization induced by FAK blockade is accompanied by persistence of DNA damage foci. A, Detection by immunofluorescence of γ-H2AX foci in H460 cells treated with DMSO, PF-562,271, or BEZ-235 followed by 2 Gy of IR. Foci were detected at the indicated time points. Note striking increase in the number of foci 24 hours after treatment with PF-562,271 and BEZ-235 (a known radiosensitizing drug) in combination with IR. Bar, 25 μm. B, Detection by immunofluorescence of γ-H2AX foci in T2.2 FAK+ and FAK H460 cells after treatment with 2 Gy of IR. Foci were detected at the indicated time points. Note striking increase in the number of foci at 9 and 24 hours after IR. Bar, 25 μm. C, Cell cycle analysis of H460 cells treated as indicated. The percentage of cells in each phase of the cell cycle is indicated. Note a significant increase in the percentage of G2 cells after combination treatment with FAKi and IR. D, WB of H460 cells treated with PF-562,271 as indicated. E, WB of T2.2 FAK+ and FAK H460 cells after IR.

Close modal

We also obtained equivalent results when we determined the induction and resolution of γ-H2AX and TP53BP1 foci, as readout of DNA damage, in CRISPR/CAS9 H460 T2.2 FAK+ and H460 T2.2 FAK cells (Fig. 5B; Supplementary Fig. S5A).

Next, we confirmed by flow cytometry that inhibition of FAK with PF-562,271 or in FAK null T2.2 H460 cells affects the percentage of γ-H2AX–positive cells after exposure to IR at every time point tested (Supplementary Fig. S5B and S5C). We found that mutant KRAS H460 cells treated with FAKi display a significant increase of the percentage of cells in the G2 phase of the cell cycle, which was further increased 4 and 24 hours after combination treatment with FAKi and IR as compared to H460 cells treated with IR only (Fig. 5C).

Western blot analysis confirmed that PF-562,271 increases γ-H2AX and in combination of IR leads to further upregulation and persistence of γ-H2AX in H460 cells (Fig. 5D). Furthermore, we found that IR activates P-FAK, which persists for at least 8 hours after IR (Fig. 5D). In addition, cell fractionation experiments revealed that a portion of FAK resides in the cell nucleus where DNA repair takes place (Supplementary Fig. S5D). FAK-null H460 cells not only display an increased basal level of γ-H2AX and its further upregulation after IR treatment, as compared to wild-type FAK H460 cells, but also persistent activation of CHK2 after exposure to IR (Fig. 5E). We detected similar findings with respect to γ-H2AX in H460 cells that underwent FAK inhibition or CRISPR/CAS9-mediated ablation, with only minor differences in the kinetics of resolution of γ-H2AX activation at later time points (Fig. 5D and E; and Supplementary Figs. S4A, S5A–C, and S5E). In this regard, there were small incongruences between the intensities of the signals obtained by IF, WB, and flow cytometry, which may be due to the fact that WB detects total cellular γ-H2AX while IF detects γ-H2AX accumulated in foci, which are also more readily detected by flow cytometry. Because γ-H2AX is a marker of DNA damage when it is accumulated in nuclear foci, we conclude that the presence of nuclear foci is representative of ongoing DNA damage.

Taken together, these data suggest that inhibition/suppression of FAK results in persistent DNA damage in NSCLC cells because of inhibition of DNA repair or augmentation of damage by cell-cycle checkpoint abrogation, which occur without affecting the activation and recruitment to sites of DNA damage of the DNA damage–sensing machinery. These observations also suggest that IR therapy could be exploited to sensitize cancer cells to therapy with FAK inhibitors.

Combination of FAK inhibition and radiotherapy is an effective antitumor strategy in vivo

We tested the antitumor effects of FAK inactivation mediated by CRISP/CAS9 editing in combination with IR in H460 NSCLC xenografts, a well-established model of aggressive NSCLC and a representative example of the cells we used in tissue culture experiments.

We generated four cohorts of seven athymic nude mice bearing xenografts of H460 T2.2 FAK+ and H460 T2.2 FAK cells of 300-mm3 average size. Mice were either mock treated or treated with five 4-Gy fractions every other day for 10 days. We delivered local irradiation to the flank or thigh of lead-shielded mice using a fractionated dose to limit overall tissue toxicity and mimic the administration modality used in the clinic (45). Notably, this xenograft volume and IR dose is comparable with previous studies involving H460 xenografts (20, 46).

As expected, FAK ablation impaired xenograft growth compared to the parental H460 cells (Fig. 6A). The median survivals of mock-treated mice carrying H460 T2.2 FAK+ and FAK cells were 19 days and 24 days, respectively (P = 0.0467; Fig. 6B). IR treatment of H460 T2.2 FAK cells resulted in a greater than 75% reduction in xenograft volume as compared with H460 T2.2 FAK+ cells 30 days after the first dose of IR (P < 0.001; Fig. 6C and D). All but one of the irradiated mice carrying xenografts of H460 T2.2 FAK cells were alive 43 days after the initiation of IR treatment; in contrast, every mouse carrying xenografts of H460 T2.2 FAK+ cells was sacrificed between days 34 and 43 after radiation due to excessive tumor burden (Fig. 6E).

Figure 6.

Loss of FAK is radiosensitizing in a xenograft tumor model. A, Xenograft growth of H460 T2.2 FAK+ and FAK cells in nude mice treated as indicated. The graph shows xenograft volumes. Points represent the mean of tumor volume (mm3) at each time point. B, Kaplan–Meyer curve of xenografts of H460 T2.2 FAK+ and FAK cells. P = 0.0467. C, Xenograft growth of H460 T2.2 FAK+ and FAK cells in nude mice treated with IR as indicated. D, Tumor burden in representative mice carrying xenografts of the indicated genotype 20 days after initiation of IR treatment. E, Kaplan–Meyer curve of xenografts of H460 T2.2 FAK+ and FAK cells treated with IR as indicated. P = 0.0006. Number of mice = 7/group; mice were sacrificed when tumor volume reached 2,000 mm3; bars, SE; P value is indicated.

Figure 6.

Loss of FAK is radiosensitizing in a xenograft tumor model. A, Xenograft growth of H460 T2.2 FAK+ and FAK cells in nude mice treated as indicated. The graph shows xenograft volumes. Points represent the mean of tumor volume (mm3) at each time point. B, Kaplan–Meyer curve of xenografts of H460 T2.2 FAK+ and FAK cells. P = 0.0467. C, Xenograft growth of H460 T2.2 FAK+ and FAK cells in nude mice treated with IR as indicated. D, Tumor burden in representative mice carrying xenografts of the indicated genotype 20 days after initiation of IR treatment. E, Kaplan–Meyer curve of xenografts of H460 T2.2 FAK+ and FAK cells treated with IR as indicated. P = 0.0006. Number of mice = 7/group; mice were sacrificed when tumor volume reached 2,000 mm3; bars, SE; P value is indicated.

Close modal

H460 T2.2 FAK cells resumed their growth about 40 days after the first dose of radiation was administered. Xenograft growth eventually reached the maximum size allowed in these experiments, and mice had to be sacrificed. By IF analysis of frozen sections we determined that T2.2 FAK cells remained negative for FAK at the experimental endpoint (Supplementary Fig. S5F). This finding suggests that FAK-independent mechanisms may mediate resistance to FAK inhibition. It is possible that other tyrosine kinases or other prosurvival networks may mediate resistance to FAK inhibition. IR treatment was well tolerated in xenograft bearing nude mice and we did not observe any drop in body weight or other signs of toxicity in both groups (data not shown).

These results indicate that the ablation of FAK leads to significant radiosensitizing effects, which in turn led to significant antitumor effects in vivo.

NSCLC remains a significant clinical challenge due to the fact that few medical treatments are effective in this disease. It is well known that NSCLC displays either primary or acquired resistance to chemotherapy and targeted therapy. The limitations of current therapies are evident in mutant KRAS NSCLC. For instance, direct inhibitors of oncogenic KRAS lack the specificity needed for their deployment in vivo (47–49). Furthermore, inhibition of the canonical KRAS signaling pathways MEK1/2, PI3K, and mTORC1/2 have not shown benefits in lung cancer patients that justify their FDA approval for clinical use (4). As a consequence, there has been an intense interest in the identification of novel therapeutic targets for mutant KRAS NSCLC.

The data presented in this article lead to several important conclusions: not only is FAK a targetable vulnerability of mutant KRAS lung cancer, but its inhibition also leads to significant radiosensitizing effects that can be exploited in a combination therapy regimen. This finding is of significance because of the availability of several FAKi in clinical development.

We previously reported a mutant KRAS–RHOA–FAK signaling axis in NSCLC and that both RHOA and FAK are requirements of high-grade NSCLC (8). Our analysis in a large panel of lung cancer cells confirms and extends our prior findings obtained with RNAi in a smaller sample set or NSCLC cells and in a genetically engineered mouse model of lung cancer (8). Our new set of data is of significance because we used specific FAKi PF-562,271 (the parental compound of Vs6063, also known as defactinib) and VS-4718. Both defactinib and VS-4718 are being evaluated in clinical trials in lung cancer and mesothelioma. Furthermore, we used shRNAs and gene editing, which are complementary and nonoverlapping methods to genetically inactivate FAK. Thus, we reason that by using these complementary approaches we adequately addressed the concern that the interpretation of RNAi experiments may be hampered by off-target effects (50). Notably, the results we obtained are internally consistent, indicating that the deleterious effects on cell proliferation, clonogenic capacity, and delayed xenograft grow in vivo are caused by FAK inhibition.

Even though we tested NSCLC cells carrying oncogenotypes other than mutant KRAS, we did not find antiproliferative responses consistently in wild-type KRAS NSCLC cells. Thus, we conclude that the dependency on FAK is a specific feature of mutant KRAS NSCLC cells. Accordingly, mutant KRAS status could be used as a biomarker for the enrolment of patients in clinical trials using FAK inhibitors.

Our data indicate that FAK is implicated in the response to DNA damage in mutant KRAS NSCLC cells. Pharmacologic inhibition of FAK protein, FAK gene silencing, or ablation results in activation and maintenance of a DNA damage response as demonstrated by the presence of γ-H2AX in Western blots and the recruitment of γ-H2AX and TP53BP1 to DNA damage foci. In this regard, it is also noteworthy that it was reported that FAK loss in murine breast cancer cells and primary fibroblast is associated with induction of replicative senescence, even though the mechanism underlying this outcome was not defined (25). In addition, FAK inhibition was implicated in mediating radiosensitization in several other settings, for instance, melanoma and head and neck cancers (51, 52).

In xenograft experiments with FAK-null lung cancer cells, we noted that the impairment of the growth of FAK-null xenografts declined over time. The observation that FAK-null xenografts stain negative for FAK at the experimental endpoint suggests that FAK-independent mechanisms mediate resistance to FAK inhibition. It is possible that other tyrosine kinases or other prosurvival networks may mediate this phenomenon. The identification of mechanisms that mediate resistance to FAK inhibition is the subject of ongoing investigations in our lab.

Our report also provides the first demonstration that the FAK blockade is an effective strategy to sensitize mutant KRAS NSCLC cancer cells to the deleterious effects of IR both in clonogenic assays and in vivo. In this regard, it is also possible that IR, by potentiating the therapeutic effects of FAKi, may delay or prevent the emergence of drug resistance.

These findings suggest that FAK blockade/ablation in combination with IR both inhibits inherent DNA repair [i.e., decreased shoulder (Dq) values] in oncogenic KRAS-driven NSCLC, as well as augments DSBs (noted by decreased Do values, representing the number of hits needed to cause death, high-dose enhancement ratios), consistent with enhanced formation of foci due to DNA DSBs. These interpretations are consistent with the observation that the DNA damage foci induced by IR persist in FAK-deficient cells. In this setting, decreased clonogenic capacity may be caused by growth arrest or impaired cell proliferation, inhibition of prosurvival stress responses or mitotic catastrophe. Indeed, we found that inhibition or loss of FAK leads to a G2 cell-cycle arrest, which is further increased by IR treatment. Thus, it is likely that the effects of FAK inhibition/suppression are mediated by loss of G2 checkpoint control that, in turn, leads to an inherent loss of DNA repair capacity. Alternatively, these effects could be caused by inhibition of a DSB repair protein (such as ATM, ATR, or DNA-PKcs) that affects DSB repair and cell-cycle checkpoint control. In this respect, it is noteworthy that PF-562,271 exposure phenocopies the effects of the dual PI3K/mTOR inhibitor BEZ-235, which was reported to be an inhibitor of ATM and DNA-PK (20, 44). In the future, it will be of interest to differentiate between these two possibilities.

We noted for the first time that IR activates FAK and that FAK depletion promotes the hyperactivation of checkpoint kinase CHK2, a known downstream target of ATM/ATR and G2–M effectors of cell-cycle arrest or apoptosis in response to DNA damage.

In this respect, it is noteworthy that FAK was reported to interact in the nucleus with PIAS1, a SUMO E3 ligase that interacts with BRCA1 in the DNA damage response (53–55). Thus, it is tempting to speculate that the requirement for FAK in mutant KRAS NSCLC cells is due to the fact that in basal growth conditions FAK promotes the repair of the DNA damage caused by oncogenic KRAS activation and that the requirement for FAK is further magnified in cells exposed to IR. In addition, p53, CDKN2A, and ATM tumor suppressors, which are well-known players in the DNA damage response and are also frequently comutated with mutant KRAS, may collaborate with FAK either directly or indirectly in mediating DNA damage repair (2, 3, 56, 57). The results of the experiments with H460 and A549 cells with restored LKB1 lead to the conclusion that loss of LKB1, another tumor suppressor implicated in DNA damage response, does not determine the dependency on FAK, at least in this context (43).

The FAKi defactinib and VS-4718 are being tested in phase I/II clinical trials, including lung cancer (58). Therefore, our work provides preclinical data useful for the design of therapeutic protocols using this novel class of drugs. We propose that mutant KRAS should represent a biomarker for the enrollment of patients in clinical trials using FAKi in NSCLC. Furthermore, we suggest that the clinical testing of combined therapy using FAKi and IR should be prioritized. In the future, it will be of interest to determine the mechanistic underpinning of the role of FAK in DNA damage repair to optimize the use of FAKi in cancer patients, identify biomarkers that predict clinical response and test FAKi in other tumor types that harbor mutant KRAS.

J.V. Heymach reports receiving other commercial research support from AstraZeneca, Bayer, and GlaxoSmithKline; and is a consultant/advisory board member for Ariad, AstraZeneca, Boerhinger Ingelheim, Exelixis, Genentech, GlaxoSmithKline, Lilly, Novartis, and Synta. No potential conflicts of interest were disclosed by the other authors.

Conception and design: K.-J. Tang, J.D. Constanzo, N. Venkateswaran, P.P. Scaglioni

Development of methodology: K.-J. Tang, J.D. Constanzo, N. Venkateswaran, M. Melegari, J.C. Morales, P.P. Scaglioni

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K.-J. Tang, J.D. Constanzo, N. Venkateswaran, M. Ilcheva, F. Skoulidis, J.V. Heymach, P.P. Scaglioni

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.-J. Tang, J.D. Constanzo, N. Venkateswaran, M. Ilcheva, J.V. Heymach, P.P. Scaglioni

Writing, review, and/or revision of the manuscript: K.-J. Tang, J.D. Constanzo, N. Venkateswaran, M. Ilcheva, F. Skoulidis, J.V. Heymach, D.A. Boothman, P.P. Scaglioni

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.-J. Tang, N. Venkateswaran, M. Melegari, D.A. Boothman, P.P. Scaglioni

Study supervision: M. Melegari, D.A. Boothman, P.P. Scaglioni

The authors thank Dr. John Minna, Luc Girard, and Amit Das at the UT Southwestern Medical Center for providing unpublished data regarding oncogenic mutations of NSCLC cell lines. They thank Mahesh Padval and Jonathan Pachter at Verastem for providing VS-4718 and for helpful discussions.

This study was financially supported by CDMRP LCRP grant #LC110229, American Cancer Society Scholar Award 13-068-01-TBG, UT Southwestern Friends of the Comprehensive Cancer Center, Texas 4000 (PPS), #2012J5100031 Science and Technology Program of Guangzhou, China (K.-J. Tang), NCI #1F31CA180689-01 and NCI T32CA124334 (JDC), NCI R01 CA102972 (D.A. Boothman), Lung Cancer Moonshot Program; V Foundation Grant; Ford Petrin Donation; CCSG Program (J.V. Heymach), The University of Texas Southwestern Medical Center and The University of Texas MD Anderson Cancer Center Lung SPORE grant 5 P50 CA070907, NIH Cancer Center Grant CA016672 and 2P30 CA142543-06.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2015
.
CA Cancer J Clin
2015
;
65
:
5
29
.
2.
Cancer Genome Atlas Research
N
. 
Comprehensive molecular profiling of lung adenocarcinoma
.
Nature
2014
;
511
:
543
50
.
3.
Cancer Genome Atlas Research
N
. 
Comprehensive genomic characterization of squamous cell lung cancers
.
Nature
2012
;
489
:
519
25
.
4.
Gysin
S
,
Salt
M
,
Young
A
,
McCormick
F
. 
Therapeutic strategies for targeting ras proteins
.
Genes Cancer
2011
;
2
:
359
72
.
5.
Cagle
PT
,
Chirieac
LR
. 
Advances in treatment of lung cancer with targeted therapy
.
Arch Pathol Lab Med
2012
;
136
:
504
9
.
6.
Pylayeva-Gupta
Y
,
Grabocka
E
,
Bar-Sagi
D
. 
RAS oncogenes: weaving a tumorigenic web
.
Nat Rev Cancer
2011
;
11
:
761
74
.
7.
Rodenhuis
S
,
Slebos
RJ
,
Boot
AJ
,
Evers
SG
,
Mooi
WJ
,
Wagenaar
SS
, et al
Incidence and possible clinical significance of K-ras oncogene activation in adenocarcinoma of the human lung
.
Cancer Res
1988
;
48
:
5738
41
.
8.
Konstantinidou
G
,
Ramadori
G
,
Torti
F
,
Kangasniemi
K
,
Ramirez
RE
,
Cai
Y
, et al
RHOA-FAK is a required signaling axis for the maintenance of KRAS-driven lung adenocarcinomas
.
Cancer Discov
2013
;
3
:
444
57
.
9.
Barbie
DA
,
Tamayo
P
,
Boehm
JS
,
Kim
SY
,
Moody
SE
,
Dunn
IF
, et al
Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1
.
Nature
2009
;
462
:
108
12
.
10.
Chien
Y
,
Kim
S
,
Bumeister
R
,
Loo
YM
,
Kwon
SW
,
Johnson
CL
, et al
RalB GTPase-mediated activation of the IkappaB family kinase TBK1 couples innate immune signaling to tumor cell survival
.
Cell
2006
;
127
:
157
70
.
11.
Fisher
GH
,
Wellen
SL
,
Klimstra
D
,
Lenczowski
JM
,
Tichelaar
JW
,
Lizak
MJ
, et al
Induction and apoptotic regression of lung adenocarcinomas by regulation of a K-Ras transgene in the presence and absence of tumor suppressor genes
.
Genes Dev
2001
;
15
:
3249
62
.
12.
Jackson
EL
,
Willis
N
,
Mercer
K
,
Bronson
RT
,
Crowley
D
,
Montoya
R
, et al
Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras
.
Genes Dev
2001
;
15
:
3243
8
.
13.
Johnson
L
,
Mercer
K
,
Greenbaum
D
,
Bronson
RT
,
Crowley
D
,
Tuveson
DA
, et al
Somatic activation of the K-ras oncogene causes early onset lung cancer in mice
.
Nature
2001
;
410
:
1111
6
.
14.
Singh
A
,
Greninger
P
,
Rhodes
D
,
Koopman
L
,
Violette
S
,
Bardeesy
N
, et al
A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival
.
Cancer Cell
2009
;
15
:
489
500
.
15.
Schuster
K
,
Venkateswaran
N
,
Rabellino
A
,
Girard
L
,
Pena-Llopis
S
,
Scaglioni
PP
. 
Nullifying the CDKN2AB locus promotes mutant K-ras lung tumorigenesis
.
Mol Cancer Res
2014
;
12
:
912
23
.
16.
Ji
H
,
Ramsey
MR
,
Hayes
DN
,
Fan
C
,
McNamara
K
,
Kozlowski
P
, et al
LKB1 modulates lung cancer differentiation and metastasis
.
Nature
2007
;
448
:
807
10
.
17.
Skoulidis
F
,
Byers
LA
,
Diao
L
,
Papadimitrakopoulou
VA
,
Tong
P
,
Izzo
J
, et al
Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities
.
Cancer Discov
2015
;
5
:
860
77
.
18.
Weinberg
F
,
Hamanaka
R
,
Wheaton
WW
,
Weinberg
S
,
Joseph
J
,
Lopez
M
, et al
Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity
.
Proc Natl Acad Sci U S A
2010
;
107
:
8788
93
.
19.
Abulaiti
A
,
Fikaris
AJ
,
Tsygankova
OM
,
Meinkoth
JL
. 
Ras induces chromosome instability and abrogation of the DNA damage response
.
Cancer Res
2006
;
66
:
10505
12
.
20.
Konstantinidou
G
,
Bey
EA
,
Rabellino
A
,
Schuster
K
,
Maira
MS
,
Gazdar
AF
, et al
Dual phosphoinositide 3-kinase/mammalian target of rapamycin blockade is an effective radiosensitizing strategy for the treatment of non-small cell lung cancer harboring K-RAS mutations
.
Cancer Res
2009
;
69
:
7644
52
.
21.
Engelman
JA
,
Chen
L
,
Tan
X
,
Crosby
K
,
Guimaraes
AR
,
Upadhyay
R
, et al
Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers
.
Nat Med
2008
;
14
:
1351
6
.
22.
Greshock
J
,
Bachman
KE
,
Degenhardt
YY
,
Jing
J
,
Wen
YH
,
Eastman
S
, et al
Molecular target class is predictive of in vitro response profile
.
Cancer Res
2010
;
70
:
3677
86
.
23.
Pratilas
CA
,
Solit
DB
. 
Targeting the mitogen-activated protein kinase pathway: physiological feedback and drug response
.
Clin Cancer Res
2010
;
16
:
3329
34
.
24.
Sulzmaier
FJ
,
Jean
C
,
Schlaepfer
DD
. 
FAK in cancer: mechanistic findings and clinical applications
.
Nat Rev Cancer
2014
;
14
:
598
610
.
25.
Pylayeva
Y
,
Gillen
KM
,
Gerald
W
,
Beggs
HE
,
Reichardt
LF
,
Giancotti
FG
. 
Ras- and PI3K-dependent breast tumorigenesis in mice and humans requires focal adhesion kinase signaling
.
J Clin Invest
2009
;
119
:
252
66
.
26.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.
27.
Phelps
RM
,
Johnson
BE
,
Ihde
DC
,
Gazdar
AF
,
Carbone
DP
,
McClintock
PR
, et al
NCI-Navy Medical Oncology Branch cell line data base
.
J Cell Biochem Suppl
1996
;
24
:
32
91
.
28.
Ramirez
RD
,
Sheridan
S
,
Girard
L
,
Sato
M
,
Kim
Y
,
Pollack
J
, et al
Immortalization of human bronchial epithelial cells in the absence of viral oncoproteins
.
Cancer Res
2004
;
64
:
9027
34
.
29.
Roberts
WG
,
Ung
E
,
Whalen
P
,
Cooper
B
,
Hulford
C
,
Autry
C
, et al
Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271
.
Cancer Res
2008
;
68
:
1935
44
.
30.
Tanjoni
I
,
Walsh
C
,
Uryu
S
,
Tomar
A
,
Nam
JO
,
Mielgo
A
, et al
PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments
.
Cancer Biol Ther
2010
;
9
:
764
77
.
31.
Fellmann
C
,
Hoffmann
T
,
Sridhar
V
,
Hopfgartner
B
,
Muhar
M
,
Roth
M
, et al
An optimized microRNA backbone for effective single-copy RNAi
.
Cell Rep
2013
;
5
:
1704
13
.
32.
Wang
T
,
Wei
JJ
,
Sabatini
DM
,
Lander
ES
. 
Genetic screens in human cells using the CRISPR-Cas9 system
.
Science
2014
;
343
:
80
4
.
33.
Scaglioni
PP
,
Yung
TM
,
Cai
LF
,
Erdjument-Bromage
H
,
Kaufman
AJ
,
Singh
B
, et al
A CK2-dependent mechanism for degradation of the PML tumor suppressor
.
Cell
2006
;
126
:
269
83
.
34.
Sun
H
,
Taneja
R
. 
Analysis of transformation and tumorigenicity using mouse embryonic fibroblast cells
.
Methods Mol Biol
2007
;
383
:
303
10
.
35.
Nicoletti
I
,
Migliorati
G
,
Pagliacci
MC
,
Grignani
F
,
Riccardi
C
. 
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry
.
J Immunol Methods
1991
;
139
:
271
9
.
36.
Huang
X
,
Darzynkiewicz
Z
. 
Cytometric assessment of histone H2AX phosphorylation: a reporter of DNA damage
.
Methods Mol Biol
2006
;
314
:
73
80
.
37.
Scaglioni
PP
,
Rabellino
A
,
Yung
TM
,
Bernardi
R
,
Choi
S
,
Konstantinidou
G
, et al
Translation-dependent mechanisms lead to PML upregulation and mediate oncogenic K-RAS-induced cellular senescence
.
EMBO Mol Med
2012
;
4
:
594
602
.
38.
Franken
NA
,
Rodermond
HM
,
Stap
J
,
Haveman
J
,
van Bree
C
. 
Clonogenic assay of cells in vitro
.
Nat Protoc
2006
;
1
:
2315
9
.
39.
Boothman
DA
,
Greer
S
,
Pardee
AB
. 
Potentiation of halogenated pyrimidine radiosensitizers in human carcinoma cells by beta-lapachone (3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione), a novel DNA repair inhibitor
.
Cancer Res
1987
;
47
:
5361
6
.
40.
Valenzuela
MT
,
Guerrero
R
,
Nunez
MI
,
Ruiz De Almodovar
JM
,
Sarker
M
,
de Murcia
G
, et al
PARP-1 modifies the effectiveness of p53-mediated DNA damage response
.
Oncogene
2002
;
21
:
1108
16
.
41.
Walsh
C
,
Tanjoni
I
,
Uryu
S
,
Tomar
A
,
Nam
JO
,
Luo
H
, et al
Oral delivery of PND-1186 FAK inhibitor decreases tumor growth and spontaneous breast to lung metastasis in pre-clinical models
.
Cancer Biol Ther
2010
;
9
:
778
90
.
42.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
43.
Gupta
R
,
Liu
AY
,
Glazer
PM
,
Wajapeyee
N
. 
LKB1 preserves genome integrity by stimulating BRCA1 expression
.
Nucleic Acids Res
2015
;
43
:
259
71
.
44.
Mukherjee
B
,
Tomimatsu
N
,
Amancherla
K
,
Camacho
CV
,
Pichamoorthy
N
,
Burma
S
. 
The dual PI3K/mTOR inhibitor NVP-BEZ235 is a potent inhibitor of ATM- and DNA-PKCs-mediated DNA damage responses
.
Neoplasia
2012
;
14
:
34
43
.
45.
Fairchild
A
,
Harris
K
,
Barnes
E
,
Wong
R
,
Lutz
S
,
Bezjak
A
, et al
Palliative thoracic radiotherapy for lung cancer: a systematic review
.
J Clin Oncol
2008
;
26
:
4001
11
.
46.
Iwasa
T
,
Okamoto
I
,
Suzuki
M
,
Nakahara
T
,
Yamanaka
K
,
Hatashita
E
, et al
Radiosensitizing effect of YM155, a novel small-molecule survivin suppressant, in non-small cell lung cancer cell lines
.
Clin Cancer Res
2008
;
14
:
6496
504
.
47.
Ostrem
JM
,
Peters
U
,
Sos
ML
,
Wells
JA
,
Shokat
KM
. 
K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions
.
Nature
2013
;
503
:
548
51
.
48.
Patgiri
A
,
Yadav
KK
,
Arora
PS
,
Bar-Sagi
D
. 
An orthosteric inhibitor of the Ras-Sos interaction
.
Nat Chem Biol
2011
;
7
:
585
7
.
49.
Zimmermann
G
,
Papke
B
,
Ismail
S
,
Vartak
N
,
Chandra
A
,
Hoffmann
M
, et al
Small molecule inhibition of the KRAS-PDEdelta interaction impairs oncogenic KRAS signalling
.
Nature
2013
;
497
:
638
42
.
50.
Kaelin
WG
 Jr
. 
Molecular biology. Use and abuse of RNAi to study mammalian gene function
.
Science
2012
;
337
:
421
2
.
51.
Tavora
B
,
Reynolds
LE
,
Batista
S
,
Demircioglu
F
,
Fernandez
I
,
Lechertier
T
, et al
Endothelial-cell FAK targeting sensitizes tumours to DNA-damaging therapy
.
Nature
2014
;
514
:
112
6
.
52.
Eke
I
,
Deuse
Y
,
Hehlgans
S
,
Gurtner
K
,
Krause
M
,
Baumann
M
, et al
beta(1)Integrin/FAK/cortactin signaling is essential for human head and neck cancer resistance to radiotherapy
.
J Clin Invest
2012
;
122
:
1529
40
.
53.
Galanty
Y
,
Belotserkovskaya
R
,
Coates
J
,
Polo
S
,
Miller
KM
,
Jackson
SP
. 
Mammalian SUMO E3-ligases PIAS1 and PIAS4 promote responses to DNA double-strand breaks
.
Nature
2009
;
462
:
935
9
.
54.
Morris
JR
,
Boutell
C
,
Keppler
M
,
Densham
R
,
Weekes
D
,
Alamshah
A
, et al
The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress
.
Nature
2009
;
462
:
886
90
.
55.
Kadare
G
,
Toutant
M
,
Formstecher
E
,
Corvol
JC
,
Carnaud
M
,
Boutterin
MC
, et al
PIAS1-mediated sumoylation of focal adhesion kinase activates its autophosphorylation
.
The J Biol Chem
2003
;
278
:
47434
40
.
56.
Bakkenist
CJ
,
Kastan
MB
. 
DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation
.
Nature
2003
;
421
:
499
506
.
57.
Kastan
MB
,
Onyekwere
O
,
Sidransky
D
,
Vogelstein
B
,
Craig
RW
. 
Participation of p53 protein in the cellular response to DNA damage
.
Cancer Res
1991
;
51
:
6304
11
.
58.
Golubovskaya
VM
. 
Targeting FAK in human cancer: from finding to first clinical trials
.
Front Biosci
2014
;
19
:
687
706
.