Purpose: The MEK inhibitor trametinib radiosensitizes KRAS-mutant non–small cell lung cancer (NSCLC) and is being tested clinically with chemoradiation. However, variability in response to trametinib suggests that additional pathways are involved. The mechanism of resistance to trametinib radiosensitization is still unknown.

Experimental Design: We used a panel of KRAS-mutant NSCLC cells and tested the radiosensitization effects of trametinib by clonogenic survival assay. Then, we investigated the mechanisms underlying the resistance to the combination therapy through several knockout and overexpression systems. Finally, we validated our findings in syngeneic mouse models in a treatment setting that mimicked the standard of care in the clinic.

Results: Radiosensitization by trametinib was effective only in KRAS-LKB1–mutated cells with wild-type (WT) p53, and we found that restoring LKB1 expression in those cells blocked that sensitization. Trametinib and radiotherapy both induced senescence in a p53-dependent manner, but in WT LKB1 cells, the combination also activated the AMPK-autophagy pathway to rescue damaged cells from senescence. LKB1-knockout or autophagy inhibition in WT LKB1 cells potentiated trametinib radiosensitization. In syngeneic animal models of Kras-mutant lung tumors, Lkb1-knockout tumors were resistant to trametinib and chemoradiation given separately, but the combination greatly controlled tumor growth and prolonged survival.

Conclusions: The LKB1 mutation in KRAS-mutant NSCLC conferred enhanced radiosensitization in combination with trametinib. The WT LKB1 could activate autophagy through AMPK pathway to induce resistance to the combination of trametinib and radiation. The KRAS-LKB1 mutation could potentially be a biomarker to select patients for trametinib and radiotherapy combination therapy. Clin Cancer Res; 24(22); 5744–56. ©2018 AACR.

Translational Relevance

We found trametinib only sensitized the KRAS-LKB1 subtype of non–small cell lung cancer to radiotherapy and that restoring LKB1 expression in those cells blocked that sensitization. The autophagy activated by the LKB1-AMPK pathway induced the resistance in LKB1 WT cells. Knockout of LKB1 or autophagy inhibition potentiated the cells to trametinib radiosensitization. Our study strongly suggests a tumor suppressor like LKB1 can also drive resistance to cancer therapy, which in turn suggests that the genetic context be considered when combining therapeutic modalities. We believe our discoveries have two important indications: (1) it emphasize the complexity of tumor signaling depending on the genetic background, especially when multiple treatment modalities are being considered, and (2) it provides compelling evidence to show KRAS-LKB1 mutation could be a biomarker for vulnerability to trametinib and chemoradiation combination which warrants further validation for personalized patients care or trial design.

Lung cancer is the leading cause of death from cancer in the United States (1) and is commonly managed with various combinations of surgery, chemotherapy, and radiotherapy. The 5-year overall survival rate for patients with lung cancer of any stage is 17%; for advanced disease, that rate is only 4% (1), indicating the need for improved therapeutic strategies. About 85% of lung cancers are non–small cell lung cancer (NSCLC), and KRAS is the most often mutated oncogene in NSCLC (2). KRAS-mutant NSCLC is a genetically and functionally complex disease with several subtypes, mainly defined by frequent comutations in p53 and LKB1 (STK11; ref. 3). p53 plays important roles in genome stability, DNA damage response, senescence, apoptosis, and cell-cycle regulation (4). The p53 signaling pathway is activated by several kinds of stress signals. The protein level of p53 is strictly regulated by MDM2, an E3 ubiquitin ligase. In cells without stress, MDM2 monoubiquitinates p53. Once cells are under stress, the interaction between p53 and MDM2 will be disrupted, stabilizing the p53 protein to trigger downstream signals (5). Liver Kinase B1 (LKB1), also named Serine/threonine kinase 11 (STK11), is a tumor-suppressor gene that is mutated in 15% to 30% of NSCLCs (6). A germline mutation in LKB1 is responsible for Peutz–Jeghers syndrome (7). LKB1 directly regulates the AMPK-mTOR pathway to control cell growth and metabolism, rendering LKB1-mutant cells resistant to metabolic stress (8). Mutations in LKB1 in patients with lung cancer are strongly associated with KRAS mutations and a history of smoking (9). Several studies have illustrated the relationship between LKB1 mutation and response to therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy (10–14).

In KRAS-mutant NSCLC, p53 and LKB1 mutations define the two major subtypes of lung cancer having distinct gene expression profiles and responses to therapies (3). In one study of genetically modified mice, Kras-Lkb1–mutated cells produced squamous tumors, whereas Kras or Kras-p53–mutated cells produced adenocarcinomas (15). Chen and colleagues showed that the effect of docetaxel could be enhanced by the MEK inhibitor selumetinib in Kras and Kras-p53 mouse lung cancer models but not in the Kras-Lkb1 model (16). Herter-Sprie and colleagues induced single lung tumor nodules in genetically modified mice implanted with Kras, Kras-p53, or Kras-Lkb1-mutated cells by injecting Adeno-Cre and treating the tumors by radiotherapy. They found that radiotherapy effectively controlled tumor growth in the Kras and Kras-p53 tumor models, but Kras-Lkb1 tumors had grown rapidly at 4 weeks after treatment (14). Collectively, these findings suggest that KRAS-LKB1 subtype may be more resistant to conventional therapies; therefore, specific therapeutic strategies should be developed.

Radiotherapy is a part of standard treatments for lung cancer (17). However, KRAS-mutant lung cancers can be radioresistant (18–20). The ability to sensitize tumor cells with molecular targeted drugs can increase the probability of tumor control in response to similar or even lower radiotherapy doses. In a previous high-content drug screen for radiosensitizers, we identified several MEK inhibitors as being radiosensitizers in KRAS-mutant lung cancer (21). Further preclinical studies indicated that one of those MEK inhibitors, trametinib, sensitized KRAS-mutant NSCLC cell lines to radiotherapy both in vitro and in vivo (21). Trametinib is an MEK1/2 inhibitor that has been approved for the treatment of BRAF V600E–mutant metastatic melanoma and is currently in phase I clinical trials in combination with chemoradiation for stage III NSCLC (NCT01912625). However, the effects of trametinib vary considerably among different cell lines, suggesting that additional mechanisms contribute to the radiosensitization effect. We undertook this study to identify a specific subgroup of KRAS-mutant NSCLC that can be radiosensitized by trametinib and to investigate mechanisms mediating resistance to trametinib sensitization, with the ultimate goal of generating novel strategies to overcome such resistance.

Cell lines

All of the human NSCLC cell lines were obtained from the American Type Culture Collection or provided by Dr. John Minna (University of Texas Southwestern Medical Center). Mouse lung adenocarcinoma cell lines were obtained from Dr. Tyler Jacks (Massachusetts Institute of Technology) and Dr. Jonathan Kurie (MD Anderson Cancer Center). All cell lines were maintained in RPMI1640 + glutamine medium supplemented with 10% FBS in a 37°C incubator with 5% carbon dioxide. Cell lines were authenticated every 6 months of use at the Characterized Cell Line Core Facility at MD Anderson Cancer Center with the short tandem repeat method. All cell lines were regularly tested for mycoplasma.

Reagents and irradiator

Trametinib, AICAR (Acadesine), and hydroxychloroquine sulfate (HCQ) were purchased from Selleck Chemicals. Cells and mice were irradiated with a Shepherd mark I-68A Cesium irradiator. Dosimetry and experimental set-up were done as previously described (22).

CRISPR knockout of p53 and Lkb1

The lentiCRISPRv2 plasmid was provided by Dr. Feng Zhang (Massachusetts Institute of Technology) through Addgene (#52961). Guide DNA sequences were synthesized by Integrated DNA Technologies and cloned to the plasmid according to the methods described elsewhere (23). Oligo sequences are listed in Supplementary Table S1. The constructed lentiCRISPRv2, pRSV-Rev, pMDLg/pRRE, and pMD2G plasmids were transfected to 293FT cells by TransIT-LT1 (Mirus) to generate lentivirus. The 293FT cell medium was changed at 24 hours after transfection, and at 48 hours, the supernatant was used to infect the target cells. Single-cell–derived stable clones were obtained after 2 to 3 weeks of puromycin selection (2–5 μg/μL). Sanger sequencing of the targeted DNA region and Western blotting of targeted proteins were done to validate successful knockouts.

siRNA knockdown

The siRNA knockdown experiments were done as described previously (24). siRNAs were transfected into cells at 24 hours before treatment with radiotherapy, trametinib, or both, and cell lysates were collected at 24 hours after treatment. Western blotting was done to test the knockdown efficiency. The siRNAs used in the study were SignalSilence Atg5 siRNA I (Cell Signaling Technology; #6345), SignalSilence Control siRNA (unconjugated; Cell Signaling Technology; #6568), and AMPK α1 siRNA (m) (Santa Cruz Biotechnology; sc-29674).

LKB1 overexpression

Whole-length LKB1 was cloned by PCR from human cDNA and then ligated to SFB (S-protein/Flag/SBP)–tagged plasmid driven by a CMV promoter. Cells were transfected by using the TransIT-LT1 (Mirus) according to the manufacturer's instructions. At 48 hours after transfection, the cells were treated with trametinib followed 4 hours later by radiotherapy or single treatment alone. Transfection efficiency was tested by Western blotting.

Clonogenic survival assay

The clonogenic survival assay was carried out as we previously described (22, 24). Briefly, cells were trypsinized to single-cell suspension and then seeded in 6-well plates 16 hours before treatment. Each treatment condition had three to six replicates. After treating by vehicle (dimethylsulfoxide) or trametinib (10–60 nmol/L) for 4 to 5 hours, cells were treated with 0, 2, 4, or 6 Gy of radiation. Drugs were washed out 24 hours later, and the cells maintained for 2 to 3 weeks. Clonogenic survival after chemoradiation was assayed as reported previously (22). Briefly, the chemotherapy agents (7.02 nmol/L paclitaxel and 48.46 nmol/L carboplatin) were mixed and used to treat cells together with trametinib followed 4 hours later with radiotherapy. Medium was changed after 24 hours. After the colonies were fixed and stained by 0.1% crystal violet in 20% methanol, the colonies consisting of at least 50 cells were counted. Data were analyzed by using SigmaPlot 10.0 (Systat Software Inc.) or GraphPad Prism 7.0 (GraphPad Software, Inc.). The dose enhancement ratio 0.5 (DER0.5) was calculated as follows: DER0.5 = (Radiation dose correlated with 50% survival fraction in DMSO-treated cells)/(Radiation dose correlated with 50% survival fraction in drug-treated cells). A drug or drug combination that generated a DER0.5 of ≥1.2 was considered a radiosensitizer.

Cell proliferation assay

LKR13-scrambled and LKR13-Lkb1-KO cells were seeded in 6-well plates at a density of 5 × 104 cells per well. Cells were trypsinized and counted with a hemocytometer after 1, 2, and 3 days of culture. Three wells of cells were counted at each time point for each cell type. The relative cell numbers were calculated and plotted by using GraphPad Prism 7.0 (GraphPad Software, Inc.).

Senescence detecting assays

For the beta-gal histology staining, a kit for staining senescent cells was purchased from Sigma (CS0030). Briefly, cells were plated in 6-well plates 1 day before treatment. The next day, cells were treated with trametinib with or without radiotherapy 4 to 5 hours later. After a 72-hour incubation, cells were fixed and stained according to the manufacturer's protocol. Five to six pictures of stained cells at random fields were taken with an inverted microscope, and then the blue-stained cells (senescence-positive) and nonstaining cells in each picture were counted on a computer. The positive percentages were calculated and presented by using GraphPad Prism 7.0 (GraphPad Software, Inc.). For the senescence detection by flow cytometry, a Senescence Detection Kit was purchased from BioVision (K991). Cells were treated for 48 hours before being tested following the manufacturer's protocol. Senescent-positive cells were detected on a BD C6 flow cytometer in the FL1 channel. Results were analyzed by FlowJo software (V10).

Reactive oxygen species detecting assay

The CellROX Green Reagent was purchased from ThermoFisher (C10444). Cells were seeded in 6-well plates 1 day prior to the treatment. Next morning, cells were treated by trametinib (30 nmol/L) followed by radiotherapy (6 Gy) 4 to 5 hours later. Six hours after radiotherapy, cells were incubated with 5 μmol/L CellROX Green Reagent for 30 minutes and then trypsinized and washed by PBS. Cells were analyzed by flow cytometer according to the manufacturer's protocol.

Western blots

Western blotting was done as previously described (22). Antibodies were purchased from Cell Signaling Technology and Abcam. For Western blot of tumor protein lysates, tumors were snap frozen by liquid nitrogen and then homogenized by a Brinkmann Polytron Homogenizer (PT 10-35) and lysed in RIPA buffer (Cell Signaling Technology) supplemented with Protease Inhibitor Cocktail (ThermoFisher; 78430). Antibodies used in this study were LC3B (CST, 3868 for human; Abcam, 51520 for mouse), GAPDH (CST, 2118), HSP90 (CST, 4874), Actin (Sigma, A2228), LKB1 (CST, 3047), p53 (CST, 2524 for mouse; Santa Cruz Biotechnology, 47698 for human), phospho-p53-Ser15 (CST, 9284), p21 (Abcam, 109199), AMPK (CST, 5832), phospho-AMPK-Thr172 (CST, 2535), ULK1 (CST, 8054), phospho-ULK1-Ser555 (CST, 5869), ERK1/2 (CST, 4695), phospho-ERK1/2 (CST, 4370), ATG5 (CST, 8540), MDM2 (CST, 86934), and phospho-MDM2-Ser166 (CST, 3521). Quantification of band intensity was carried out by the volume tools within the Image Lab software (Bio-Rad).

Animal models

All experiments involving animals were approved by the Institutional Animal Care and Use Committee of The University of Texas MD Anderson Cancer Center. All animal experiments were carried out in accordance with the institutional guidelines and the approved protocol. Male 129S background mice were purchased from the Jackson Laboratory. When the animals were 8 to 10 weeks old, 5 × 105 LKR13 or LKR13-Lkb1 KO cells were s.c. injected into the right thighs of the mice. When the tumors reached 10 mm in diameter, the mice were randomly assigned to the following treatment groups: control; chemoradiation; trametinib; chemoradiation + trametinib; HCQ; and HCQ + chemoradiation + trametinib. Chemotherapy (10 mg/kg paclitaxel and 30 mg/kg carboplatin) was administered on day 1 by i.p. injection. Radiotherapy was given as 2 Gy once daily for 5 days, with a jig used to confine the mice and expose the tumor to radiotherapy as previously reported (22). Trametinib was administered daily at a dose of 2 mg/kg for 5 days by oral gavage. The trametinib was dissolved in DMSO and diluted 1:9 in 1% carboxymethylcellulose (medium viscosity) and 0.4% Tween-80 (Sigma). HCQ was dissolved in PBS and administered at a daily dose of 60 mg/kg for 5 days by i.p. injection. Tumor diameters were measured 2 to 3 times each week, and tumor volumes were calculated as 0.5 × length × width × width.

Assessment of lung metastasis

Three animals from each treatment group were sacrificed at day 7 of the experiment. Lungs were fixed in 10% formalin overnight and embedded in paraffin. Three sections representing different locations from each sample were stained by hematoxylin and eosin (H&E). Total number of metastatic nodules from each section was counted under a microscope.

Trametinib radiosensitized the KL subtype, but not the KP subtype, of NSCLC cell lines

Because comutations in KRAS can affect cells' response to therapies, we tested a panel of human KRAS-TP53–mutated (KP) and KRAS-LKB1–mutated (KL) NSCLC cell lines. DER0.5 greater than 1.2 was considered sensitization. Clonogenic survival assays indicated that trametinib radiosensitized KL cells, but not KP cells (Fig. 1A–C).

Figure 1.

The MEK inhibitor trametinib radiosensitized KRAS-LKB1–mutant NSCLC cells. A, Clonogenic survival assays of KRAS-LKB1 (KL) NSCLC cells with or without trametinib (30 nmol/L). B, Clonogenic survival assays of KRAS-p53 (KP) NSCLC cells with or without trametinib. C, Statistical comparison (t test) of trametinib DER in KL and KP cells. A DER0.5 > 1.2 was used as the cut point to determine sensitization. Error bars show SEM.

Figure 1.

The MEK inhibitor trametinib radiosensitized KRAS-LKB1–mutant NSCLC cells. A, Clonogenic survival assays of KRAS-LKB1 (KL) NSCLC cells with or without trametinib (30 nmol/L). B, Clonogenic survival assays of KRAS-p53 (KP) NSCLC cells with or without trametinib. C, Statistical comparison (t test) of trametinib DER in KL and KP cells. A DER0.5 > 1.2 was used as the cut point to determine sensitization. Error bars show SEM.

Close modal

Trametinib and radiotherapy induced p53-dependent senescence in KRAS-LKB1–mutated NSCLC cell lines

We observed that for the sensitive KL cell line H460, combination trametinib plus radiotherapy produced enlarged, flat cells, which was not observed in the KP cell line H441 (Supplementary Fig. S1A and S1B). Because this morphologic change often represents senescence, we assessed the senescence marker beta-galactosidase and found that trametinib and radiotherapy were able to induce strong beta-galactosidase activity in H460 KL cells but not in H441 KP cells (Fig. 2A and B). The senescence marker p21 was also upregulated in the A549 and H460 KL cell lines at 48 hours after treatment, but was not upregulated in H441 KP cells. Moreover, treatment with combined radiotherapy plus trametinib led to higher p21 levels in A549 and H460 cells than did either treatment alone, suggesting that the combination robustly induced senescence (Fig. 2C). Next, to determine if p53 was required for this effect, we generated p53-knockout (KO) A549 and H460 cell lines by CRISPR-Cas9 targeting. The KO cells did not express p53 and could not upregulate p21 after irradiation (Fig. 2D). Clonogenic survival assays showed that the p53 KO cells were not radiosensitized by trametinib (Fig. 2E) and did not undergo senescence (Supplementary Fig. S1C). We also examined the H23 cell line harboring natural KRAS-p53-LKB1 triple mutation and found it could not be radiosensitized by trametinib either (Supplementary Fig. S1D). Next, we measured levels of MDM2 in A549 and H460 cells under various treatment conditions. As expected (25), MDM2 expression was upregulated by radiotherapy. Trametinib did not affect MDM2 expression but robustly inhibited MDM2 phosphorylation at Ser-166, which has previously been shown to increase the ubiquitin ligase activity of MDM2 (26). Both the activity and levels of p53 were acutely induced by radiotherapy, as indicated by the levels of p53 phosphorylation at Ser-15 and total p53 levels. As a result of these converging pathways on p53 levels and activity, p21 was strongly activated only by the combination of radiotherapy and trametinib (Fig. 2F). Together, the results suggest that trametinib inhibited phosphorylation of MDM2 to stabilize p53 after radiotherapy and induced p53-dependent senescence in KRAS-LKB1–mutant NSCLC cells.

Figure 2.

Trametinib and radiotherapy-induced senescence in KL cells in a p53-dependent manner. A and B, Senescence-associated (SA)-beta gal staining results of H460 (KL) and H441 (KP) cells. T test was used to compare the difference between groups. Error bars show SEM. C, Western blot of senescence marker, p21, after trametinib (30 nmol/L) and radiotherapy (2 Gy) treatments in A549, H460, and H441 cells. D, Western blot of p53 and p21 in p53-KO and scrambled control cells treated or not treated with 6 Gy of ionizing radiation. Tram, trametinib. E, Clonogenic survival assays of p53-KO A549 and H460 cells. F, Western blot of MDM2 Ser166 level changes in A549 at different times after treatments.

Figure 2.

Trametinib and radiotherapy-induced senescence in KL cells in a p53-dependent manner. A and B, Senescence-associated (SA)-beta gal staining results of H460 (KL) and H441 (KP) cells. T test was used to compare the difference between groups. Error bars show SEM. C, Western blot of senescence marker, p21, after trametinib (30 nmol/L) and radiotherapy (2 Gy) treatments in A549, H460, and H441 cells. D, Western blot of p53 and p21 in p53-KO and scrambled control cells treated or not treated with 6 Gy of ionizing radiation. Tram, trametinib. E, Clonogenic survival assays of p53-KO A549 and H460 cells. F, Western blot of MDM2 Ser166 level changes in A549 at different times after treatments.

Close modal

LKB1 abrogated the radiosensitizing effect of trametinib

To better understand the functions of comutated TP53 and LKB1 in KRAS-mutant lung cancer, we used a Kras-MUT, p53-WT, and Lkb1-WT mouse lung cancer cell line, LKR13, to generate p53 and Lkb1 KO cells (Supplementary Fig. S2). We first tested the response of the KP and KL cells to radiotherapy and trametinib separately. The p53 and Lkb1 KO in LKR13 cells induced radioresistance (Fig. 3A). Interestingly, however, KO of Lkb1 also increased resistance to trametinib (Fig. 3B). Next, in testing the responses to trametinib-induced radiosensitization, we found that trametinib radiosensitized the Lkb1 KO cells in a dose-dependent manner, but not the scrambled control or the p53 KO cells (Fig. 3C). To further explore the role of LKB1 in trametinib-induced radiosensitization, we overexpressed LKB1 in A549 and H460 cells (both KL). Clonogenic survival assays showed that the A549-LKB1 and H460-LKB1 cell lines were resistant to trametinib radiosensitization (Fig. 3D and E). Overexpression of LKB1 in A549 cells also reduced senescence after trametinib plus radiotherapy (Fig. 3F; Supplementary Fig. S3A). This is also observed in the LKR13 cells, as the combination of trametinib and radiotherapy induced senescence only in Lkb1-KO cells, but not in the scrambled control (Supplementary Fig. S3B and S3C). Thus, LKB1 deletion seems to be required for the trametinib-induced radiosensitization.

Figure 3.

LKB1 loss of function was required for trametinib-induced radiosensitization. A and B, Trametinib IC50 and survival fraction at 2 Gy of LKR13-scrambled control, p53-KO, and Lkb1-KO clones. Three stables clones from each KO and scrambled control were tested. C, Clonogenic survival assays of LKR13-KO cells with different concentrations of Trametinib. D and E, Clonogenic survival assays of A549 and H460 cells with LKB1 overexpression treated by Trametinib (30 nmol/L) and radiotherapy. F, SA-beta gal staining results of A549 control and LKB1-overexpressing cells.

Figure 3.

LKB1 loss of function was required for trametinib-induced radiosensitization. A and B, Trametinib IC50 and survival fraction at 2 Gy of LKR13-scrambled control, p53-KO, and Lkb1-KO clones. Three stables clones from each KO and scrambled control were tested. C, Clonogenic survival assays of LKR13-KO cells with different concentrations of Trametinib. D and E, Clonogenic survival assays of A549 and H460 cells with LKB1 overexpression treated by Trametinib (30 nmol/L) and radiotherapy. F, SA-beta gal staining results of A549 control and LKB1-overexpressing cells.

Close modal

Resistance to trametinib radiosensitization may operate via the AMPK pathway through LKB1

We next investigated the mechanism of LKB1-induced resistance to sensitization by trametinib. One of the pathways directly regulated by LKB1 is AMPK (27), which is known to regulate senescence (28–31). Western blotting of LKB1-overexpressing A549 and H460 cells and LKR13 p53- or LKB1-KO cells at 24 hours after combination treatment showed that AMPK was robustly activated in the cells with WT LKB1 (Fig. 4A and B; Supplementary Fig. S4) and was activated to a lesser extent by either treatment alone. AMPK activation was completely absent in the A549 and LKR13 cells without LKB1, suggesting that the activation of AMPK by radiotherapy and trametinib could be LKB1-dependent. To further investigate the roles of AMPK in trametinib-induced radiosensitization, we treated A549 cells with an AMPK activator (AICAR) simultaneously with trametinib and radiotherapy. AICAR can lead to the direct activation of AMPK in A549 cells despite the loss of LKB1 (Fig. 4C). In clonogenic survival assays, activation of AMPK abrogated trametinib-induced radiosensitization (Fig. 4D). Next, we knocked down AMPK in LKR13 cells using siRNAs (Fig. 4E) and found that the LKR13-AMPK-KD cells were radiosensitized by trametinib (DER0.5 = 1.38 vs. DER0.5 = 0.95 in scrambled siRNA control) despite their also having WT LKB1 (Fig. 4F), suggesting that inactivation of LKB1-mediated AMPK pathway is required for the trametinib-induced radiosensitization.

Figure 4.

LKB1-induced activation of AMPK blocked trametinib-induced radiosensitization. A and B, Western blot of phosphor-AMPK and phosphor-ERK in A549-LKB1 cells and LKR13 p53- or LKB1-KO cells after trametinib (30 nmol/L) with radiotherapy (2 Gy for A549, 4 Gy for LKR13). C, Western blot of phosphor-AMPK in A549 cells at different times after treatment with the AMPK activator AICAR. D, Clonogenic survival assays of A549 cells treated with trametinib (30 nmol/L), AICAR (0.5 μmol/L), and trametinib + AICAR. E, Western blot of AMPK in LKR13 cells transfected with AMPK siRNA. F, Clonogenic survival assays of LKR13 and LKR13-AMPK-KD cells treated by trametinib.

Figure 4.

LKB1-induced activation of AMPK blocked trametinib-induced radiosensitization. A and B, Western blot of phosphor-AMPK and phosphor-ERK in A549-LKB1 cells and LKR13 p53- or LKB1-KO cells after trametinib (30 nmol/L) with radiotherapy (2 Gy for A549, 4 Gy for LKR13). C, Western blot of phosphor-AMPK in A549 cells at different times after treatment with the AMPK activator AICAR. D, Clonogenic survival assays of A549 cells treated with trametinib (30 nmol/L), AICAR (0.5 μmol/L), and trametinib + AICAR. E, Western blot of AMPK in LKR13 cells transfected with AMPK siRNA. F, Clonogenic survival assays of LKR13 and LKR13-AMPK-KD cells treated by trametinib.

Close modal

Blocking autophagy reversed resistance to trametinib-induced radiosensitization

AMPK is known to regulate autophagy (32, 33), a self-degradation program in starving and damaged cells (34) that may protect damaged cells from senescence (35–37). We next examined if AMPK activation resulted in upregulation of autophagy. Autophagy activity was indeed increased in LKB1-overexpressing A549 and H460 cells, as evidenced by the increased LC3B II/I ratio (Fig. 5A and B; Supplementary Fig. S5A). In LKR13-Lkb1-KO cells, the level of LC3B-II was much lower compared with the scrambled control after treatment of trametinib alone or trametinib and radiotherapy. Another marker of autophagy activation, phospho-ULK1-Ser555, which could be directly phosphorylated by AMPK (38), was strongly upregulated by trametinib and further intensified by radiotherapy in the LKR13-scrambled control cells. However, in the Lkb1-KO cells, phospho-ULK1-Ser555 was lower compared with scrambled control at baseline level and was not upregulated by treatment, most likely as the consequence of AMPK inactivation in the absence of LKB1 (Supplementary Fig. S5B). Because autophagy may rescue cells from senescence, we next tested if inhibition of autophagy with HCQ could increase senescence in LKB1 WT cells and found that A549-LKB1 cells underwent senescence after being treated with the combination of HCQ, trametinib, and radiotherapy (Fig. 5C). Furthermore, the A549-LKB1 and LKR13 cells were radiosensitized by HCQ + trametinib but not by trametinib alone (Fig. 5D; Supplementary Fig. S5C). Knockdown of ATG5, which is a critical mediator of autophagy (39, 40), in A549-LKB1 cells (Fig. 5E) converts its resistant phenotype in trametinib-induced radiosensitization (Fig. 5F) and increased senescence after the combined treatment of trametinib and radiotherapy (Supplementary Fig. S5D). Because autophagy is capable of reducing reactive oxygen species (ROS) in stressed cancer cells (41), we next examined if autophagy deficiency in Lkb1-KO cells results in elevated ROS levels. Indeed, the LKR13-Lkb1-KO cells had higher baseline ROS levels, but trametinib treatment did not increase ROS generation in both KO and scrambled control cells (Supplementary Fig. S5E). In sum, our results showed that autophagy induced by the LKB1-AMPK activation may induce the resistance to trametinib and radiotherapy combination treatment.

Figure 5.

Autophagy inhibition partially potentiated LKB1 WT cells to trametinib-induced radiosensitization. A and B, Western blot of autophagy marker LC3B in A549 cells treated by trametinib (30 nmol/L) and radiotherapy (2 Gy). Error bars show SD. C, SA-beta gal staining results of A549-LKB1 cells treated by trametinib (30 nmol/L), radiotherapy (2 Gy), and HCQ (50 μmol/L). Cells were treated by HCQ and/or trametinib 4 hours prior to radiotherapy. Drugs were washed out 24 hours after radiotherapy. Cells were incubated for additional 48 hours before staining. D, Clonogenic survival assay of A549-LKB1 cells treated with trametinib (30 nmol/L) and HCQ (50 μmol/L). E, Western blots of ATG5 in A549-LKB1 cells after transfection with ATG5 siRNA. F, Clonogenic survival assay of A549-LKB1-ATG5-KD and scrambled KD cells.

Figure 5.

Autophagy inhibition partially potentiated LKB1 WT cells to trametinib-induced radiosensitization. A and B, Western blot of autophagy marker LC3B in A549 cells treated by trametinib (30 nmol/L) and radiotherapy (2 Gy). Error bars show SD. C, SA-beta gal staining results of A549-LKB1 cells treated by trametinib (30 nmol/L), radiotherapy (2 Gy), and HCQ (50 μmol/L). Cells were treated by HCQ and/or trametinib 4 hours prior to radiotherapy. Drugs were washed out 24 hours after radiotherapy. Cells were incubated for additional 48 hours before staining. D, Clonogenic survival assay of A549-LKB1 cells treated with trametinib (30 nmol/L) and HCQ (50 μmol/L). E, Western blots of ATG5 in A549-LKB1 cells after transfection with ATG5 siRNA. F, Clonogenic survival assay of A549-LKB1-ATG5-KD and scrambled KD cells.

Close modal

LKR13-Lkb1-KO tumors were resistant to trametinib or chemoradiation but sensitive to the combination

To validate our findings, we established s.c. tumors by implantation of the LKR13-Lkb1 KO cells or scrambled controls in mice and treated the mice according to the schedule depicted in Fig. 6A. Daily 2 Gy fractionated radiotherapy in combination with paclitaxel and carboplatin was used to reflect the clinical standard of care for lung cancer. To test whether the radiosensitization effect of trametinib was still valid after chemoradiation, we used an MTS assay and found that KO of p53 or Lkb1 did not affect cell sensitivity to carboplatin and paclitaxel doublet chemotherapy (Supplementary Fig. S6A). Clonogenic survival assays showed that trametinib sensitized the LKR13-Lkb1 KO cells to both radiotherapy and chemoradiation (Supplementary Fig. S6B). Tumors formed from implanted Lkb1 KO cells were much more aggressive than those from the scrambled control (Supplementary Fig. S7A), despite their having similar growth rates in vitro (Supplementary Fig. S7B). For the LKR13-Lkb1-KO tumors, neither trametinib nor chemoradiation inhibited tumor growth when given separately, but the combination of trametinib and chemoradiation led to significantly better tumor control (Fig. 6B). For both the control and chemoradiation-treated animals, we observed that the animals had significant morbidity and died rapidly. H&E staining of lung tissue collected at day 7 revealed numerous metastatic nodules in the control group and the chemoradiation group, which were significantly reduced by trametinib, and was further reduced by chemoradiation (P = 0.04; Fig. 6D and E). Although trametinib treatment reduced lung metastasis and prolonged survival compared with the controls (P = 0.026), those mice had to be sacrificed before the end of the experiment because of the excessive primary tumor burden. The combination of chemoradiation and trametinib resulted in tumor stabilization and long-term survival (P < 0.0001; Fig. 6B and C; Supplementary Fig. S8). Tumors collected on day 7 showed significant upregulation of p21 levels in the chemoradiation and trametinib group that were not seen in the other treatment groups (Fig. 6F). For the LKR13-scrambled tumors, both trametinib alone and chemoradiation alone significantly inhibited tumor growth relative to control. The combination of chemoradiation and trametinib appears better than either treatment alone, but this difference was not statistically significant (P = 0.060). The autophagy inhibitor HCQ alone had no effect on tumor growth, but the combination of HCQ, trametinib, and chemoradiation produced better tumor growth inhibition than trametinib and chemoradiation or either treatment alone (P < 0.001; Fig. 6G). Together, the results showed that the combination of trametinib and chemoradiation was more effective than either treatment alone in the Kras-Lkb1–mutant tumors. Our data also suggested that inhibition of autophagy may overcome the LKB1-induced resistance in the LKB1-wild-type tumors.

Figure 6.

Combining trametinib with chemoradiation synergistically inhibited Kras-Lkb1–mutant murine lung tumor growth in vivo. A, Treatment schema for tumor-bearing mice. B, Growth curves of LKR13-Lkb1-KO tumors after treatment of vehicle control, trametinib, chemoradiation, and trametinib plus chemoradiation. Six to 10 mice were randomized to each treatment group. Error bars show SD; t tests were used to compare differences in tumor volume at different times. C, Survival curves of LKR13-Lkb1-KO tumor-bearing mice. Log-rank tests were used to compare difference between curves. D, Number of lung metastasis nodules detected in each group. Error bars show SD; t tests were used to compare the difference. E, Representative staining of lung sections. Arrows point to nodules. F, Western blot analysis of tumor protein lysates collected at day 7. G, Growth curves of LKR13-scrambled control tumors after treatment of vehicle control, Trametinib, HCQ (autophagy inhibitor), chemoradiation, trametinib plus chemoradiation, and HCQ, plus trametinib plus chemoradiation. Six to 10 mice were randomized to each treatment group. Error bars show SD; t tests were used to compare differences in tumor volume at different times.

Figure 6.

Combining trametinib with chemoradiation synergistically inhibited Kras-Lkb1–mutant murine lung tumor growth in vivo. A, Treatment schema for tumor-bearing mice. B, Growth curves of LKR13-Lkb1-KO tumors after treatment of vehicle control, trametinib, chemoradiation, and trametinib plus chemoradiation. Six to 10 mice were randomized to each treatment group. Error bars show SD; t tests were used to compare differences in tumor volume at different times. C, Survival curves of LKR13-Lkb1-KO tumor-bearing mice. Log-rank tests were used to compare difference between curves. D, Number of lung metastasis nodules detected in each group. Error bars show SD; t tests were used to compare the difference. E, Representative staining of lung sections. Arrows point to nodules. F, Western blot analysis of tumor protein lysates collected at day 7. G, Growth curves of LKR13-scrambled control tumors after treatment of vehicle control, Trametinib, HCQ (autophagy inhibitor), chemoradiation, trametinib plus chemoradiation, and HCQ, plus trametinib plus chemoradiation. Six to 10 mice were randomized to each treatment group. Error bars show SD; t tests were used to compare differences in tumor volume at different times.

Close modal

In the current study, we found that the KRAS-LKB1–mutant lung cancer is very sensitive to the combination of trametinib and radiotherapy. The synergy of trametinib and radiotherapy was not seen in KRAS-p53 or KRAS-mutant–alone lung cancer cells. Our data also suggested that the wild-type tumor-suppressor LKB1 could induce resistance to the combination therapy. Specifically, in KRAS-mutant NSCLC, LKB1 induced resistance to this combination through activating the AMPK-autophagy pathway to rescue cells from undergoing senescence. As a result, tumors with both KRAS and LKB1 mutations were very sensitive to this combination because of the absence of LKB1-driven resistance. The combination of trametinib and radiotherapy induced senescence in cells with KRAS and LKB1 mutations, but not in cells with loss-of-function p53 or with WT LKB1. These intriguing findings could reflect the complicated interplay between the MAPK pathway and tumor-suppressor pathways. Our findings suggest that trametinib, when given with radiotherapy, simultaneously affects two tumor-suppressor pathways: it stabilizes p53 by reducing MDM2-Ser166 phosphorylation and by activating AMPK depending on LKB1. The p53 pathway is well known to be activated by radiotherapy (42), but the level of activation is strictly regulated by the negative feedback loop of MDM2. MDM2 phosphorylation at Ser166 reportedly increases the ubiquitin ligase activity of MDM2 (26, 43); thus, trametinib could stabilize the p53 protein after radiotherapy to augment the radiotherapy-induced senescent effects. This radiosensitization mechanism requires functional p53 signaling, suggesting the tumors with KRAS-LKB1-p53 triple mutation may not be radiosensitized by trametinib. In the LKB1 WT tumors, however, trametinib also acted synergistically with radiotherapy to induce robust AMPK activation and eventually upregulated autophagy to rescue cells from senescence. Autophagy is one mechanism that prevents cells from undergoing senescence (35–37). In a recent study of breast cancer, the CDK 4/6 inhibitor palbociclib induced both autophagy and senescence, but combining palbociclib with the autophagy inhibitor HCQ led to inhibited tumor cell growth and increased senescence both in vitro and in vivo (44). In summary, although LKB1 mutation in our study led to resistance to radiotherapy or trametinib given separately, it rendered the KL subtype of NSCLC vulnerable to the combination of these two treatments because of the absence of LKB1-AMPK autophagy–driven resistance. Our proposed working model for these effects is illustrated in Supplementary Fig. S9.

It has been previously shown that murine Kras-Lkb1–mutant lung tumors were more resistant to radiotherapy compared with Kras-p53 or Kras-mutant tumors (14). Our results also demonstrated that Lkb1 KO in LKR13 cell lines conferred resistance to radiotherapy in vitro and chemoradiation in vivo. However, the mechanism by which LKB1 deficiency induces radioresistance is still unknown. The overexpression of LKB1 in A549 and H460 increased radiosensitivity in those cells through the downregulation of DNA damage repair–related genes, such as RAD50 and XRCC1 (45), suggesting LKB1 may be involved in DNA damage repair. Interestingly, MEK inhibition has been reported to attenuate DNA damage repair in lung cancer cells after radiotherapy (46, 47). Whether LKB1 and MAPK pathway interconnections regulate DNA repair needs future investigation.

Certain limitations exist in this study. First, the subcutaneous tumor model may not reflect the microenvironment in the lung. However, attaining intraparenchymal single orthotopic Kras-Lkb1 tumor model using the LKR13 cell line for lung radiotherapy experiments was not feasible, due to the aggressive nature of such double mutant tumors, which led to rapid dissemination. Second, although CRISPR KO of p53 and Lkb1 in the LKR13 cells provided a simple model system for us to assess the relative functions of these two tumor suppressors in the controlled genetic context, they still may not represent their functions in tumor initiation and evolution in vivo. Validation studies using cell lines isolated from genetically modified mouse models are needed in the future. Despite these limitations, we believe our present study has unveiled an interesting complexity of tumor signaling which is dependent on the genetic background, especially when multiple treatment modalities are being considered. Usually, deficiencies in tumor suppressors are linked with tumor initiation and progression. Activation of the tumor suppressor, LKB1, and its downstream AMPK pathway is being increasingly recognized as a novel cancer therapeutic strategy. The AMPK activator metformin has long been used for the treatment of diabetes, but it also has antitumor effects (48–50). However, our study suggests that the tumor-suppressive pathways could also be activated by certain combinations to induce therapy resistance. In the KRAS-LKB1–mutant tumors, although the genetic aberrations could portend to greater resistance to individual conventional therapies, synthetic lethality was conferred when the various therapies are combined. This implies that identifying proper combination of therapies in specific genetic contexts will enable improvements in therapy outcomes. More importantly, our in vivo models also showed that the current standard of care, which is chemoradiation, could be potentially enhanced by trametinib in KRAS-LKB1–mutant tumors. Whether our findings are translatable to the clinic will require prospective validation on future clinical trials.

S.H. Lin is a consultant/advisory board member for AstraZeneca, Inc., and reports receiving commercial research grants from BeyondSpring Pharmaceuticals, Inc., Genentech, Hitachi Chemical Diagnostic. No potential conflicts of interest were disclosed by the other authors.

Conception and design: Y. Wang, N. Li, M. Zhang, M.-C. Hung, S.H. Lin

Development of methodology: Y. Wang, N. Li, C. Xu, S.H. Lin

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Wang, N. Li, R. Ye, Y. Qiao, A. Sharma, S.H. Lin

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Wang, W. Jiang, W. Deng, C. Xu, M.-C. Hung, S.H. Lin

Writing, review, and/or revision of the manuscript: Y. Wang, W. Jiang, C. Xu, A. Sharma, S.H. Lin

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.H. Lin

Study supervision: M.-C. Hung, S.H. Lin

Other (funding): S.H. Lin

This work was supported in part by the Mabuchi Program in Targeted Radiotherapy, Uniting Against Lung Cancer, NIH/NCI Lung Cancer SPORE (5P50CA070907), and Cancer Center Support (Core) Grant CA016672 from the NIH/NCI, to The University of Texas MD Anderson Cancer Center. Y. Wang is supported by a T.C. Hsu Memorial Scholarship from The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences. The authors thank Christine Wogan of the Division of Radiation Oncology at MD Anderson for editorial assistance. The authors thank Jie Zhang of the Center for Radiation Oncology Research at MD Anderson for technical assistance in histology.

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, 2017
.
CA Cancer J Clin
2017
;
67
:
7
30
.
2.
Imielinski
M
,
Berger
AH
,
Hammerman
PS
,
Hernandez
B
,
Pugh
TJ
,
Hodis
E
, et al
Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing
.
Cell
2012
;
150
:
1107
20
.
3.
Skoulidis
F
,
Byers
LA
,
Diao
LX
,
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
.
4.
Bieging
KT
,
Mello
SS
,
Attardi
LD
. 
Unravelling mechanisms of p53-mediated tumour suppression
.
Nat Rev Cancer
2014
;
14
:
359
70
.
5.
Moll
UM
,
Petrenko
O
. 
The MDM2-p53 interaction
.
Mol Cancer Res
2003
;
1
:
1001
8
.
6.
Ding
L
,
Getz
G
,
Wheeler
DA
,
Mardis
ER
,
McLellan
MD
,
Cibulskis
K
, et al
Somatic mutations affect key pathways in lung adenocarcinoma
.
Nature
2008
;
455
:
1069
75
.
7.
Hemminki
A
,
Markie
D
,
Tomlinson
I
,
Avizienyte
E
,
Roth
S
,
Loukola
A
, et al
A serine/threonine kinase gene defective in Peutz-Jeghers syndrome
.
Nature
1998
;
391
:
184
7
.
8.
Alessi
DR
,
Sakamoto
K
,
Bayascas
JR
. 
LKB1-dependent signaling pathways
.
Annu Rev Biochem
2006
;
75
:
137
63
.
9.
Koivunen
JP
,
Kim
J
,
Lee
J
,
Rogers
AM
,
Park
JO
,
Zhao
X
, et al
Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients
.
Br J Cancer
2008
;
99
:
245
52
.
10.
Richer
AL
,
Cala
JM
,
O'Brien
K
,
Carson
VM
,
Inge
LJ
,
Whitsett
TG
. 
WEE1 kinase inhibitor AZD1775 has preclinical efficacy in LKB1-deficient non-small cell lung cancer
.
Cancer Res
2017
;
77
:
4663
72
.
11.
Koyama
S
,
Akbay
EA
,
Li
YY
,
Aref
AR
,
Skoulidis
F
,
Herter-Sprie
GS
, et al
STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment
.
Cancer Res
2016
;
76
:
999
1008
.
12.
Liu
Y
,
Li
Y
,
Wang
X
,
Liu
F
,
Gao
P
,
Quinn
MM
, et al
Gemcitabine and Chk1 inhibitor AZD7762 synergistically suppress the growth of Lkb1-deficient lung adenocarcinoma
.
Cancer Res
2017
;
77
:
5068
76
.
13.
Herter-Sprie
GS
,
Koyama
S
,
Korideck
H
,
Hai
J
,
Deng
J
,
Li
YY
, et al
Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer
.
JCI insight
2016
;
1
:
e87415
.
14.
Herter-Sprie
GS
,
Korideck
H
,
Christensen
CL
,
Herter
JM
,
Rhee
K
,
Berbeco
RI
, et al
Image-guided radiotherapy platform using single nodule conditional lung cancer mouse models
.
Nat Commun
2014
;
5
:
5870
.
15.
Ji
HB
,
Ramsey
MR
,
Hayes
DN
,
Fan
C
,
McNamara
K
,
Kozlowski
P
, et al
LKB1 modulates lung cancer differentiation and metastasis
.
Nature
2007
;
448
:
807
10
.
16.
Chen
Z
,
Cheng
K
,
Walton
Z
,
Wang
YC
,
Ebi
H
,
Shimamura
T
, et al
A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response
.
Nature
2012
;
483
:
613
7
.
17.
Molina
JR
,
Yang
PG
,
Cassivi
SD
,
Schild
SE
,
Adjei
AA
. 
Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship
.
Mayo Clin Proc
2008
;
83
:
584
94
.
18.
Wang
M
,
Han
J
,
Marcar
L
,
Black
J
,
Liu
Q
,
Li
XY
, et al
Radiation resistance in KRAS-mutated lung cancer is enabled by stem-like properties mediated by an osteopontin-EGFR pathway
.
Cancer Res
2017
;
77
:
2018
28
.
19.
Affolter
A
,
Drigotas
M
,
Fruth
K
,
Schmidtmann
I
,
Brochhausen
C
,
Mann
WJ
, et al
Increased radioresistance via G12S K-Ras by compensatory upregulation of MAPK and PI3K pathways in epithelial cancer
.
Head Neck-J Sci Spec
2013
;
35
:
220
8
.
20.
Williams
TM
,
Yang
L
,
Estrada
A
,
Chatterjee
M
,
Robb
R
. 
KRAS oncogenic mutations induce intrinsic resistance to radiation through up-regulation of DNA repair pathways
.
Int J Radiat Oncol
2016
;
96
:
S238
.
21.
Lin
SH
,
Zhang
J
,
Giri
U
,
Stephan
C
,
Sobieski
M
,
Zhong
L
, et al
A high content clonogenic survival drug screen identifies MEK inhibitors as potent radiation sensitizers for KRAS mutant non-small-cell lung cancer
.
J Thorac Oncol
2014
;
9
:
965
73
.
22.
Wang
Y
,
Liu
H
,
Diao
LX
,
Potter
A
,
Zhang
J
,
Qiao
Y
, et al
Hsp90 inhibitor ganetespib sensitizes non-small cell lung cancer to radiation but has variable effects with chemoradiation
.
Clin Cancer Res
2016
;
22
:
5876
86
.
23.
Sanjana
NE
,
Shalem
O
,
Zhang
F
. 
Improved vectors and genome-wide libraries for CRISPR screening
.
Nat Methods
2014
;
11
:
783
4
.
24.
Wang
Y
,
Gudikote
J
,
Giri
U
,
Yan
J
,
Deng
W
,
Ye
R
, et al
RAD50 expression is associated with poor clinical outcomes after radiotherapy for resected non-small cell lung cancer
.
Clin Cancer Res
2018
;
24
:
341
50
.
25.
Perry
ME
. 
Mdm2 in the response to radiation
.
Mol Cancer Res
2004
;
2
:
9
19
.
26.
Malmlof
M
,
Roudier
E
,
Hogberg
J
,
Stenius
U
. 
MEK-ERK-mediated phosphorylation of Mdm2 at Ser-166 in hepatocytes - Mdm2 is activated in response to inhibited Akt signaling
.
J Biol Chem
2007
;
282
:
2288
96
.
27.
Shackelford
DB
,
Shaw
RJ
. 
The LKB1-AMPK pathway: metabolism and growth control in tumour suppression
.
Nat Rev Cancer
2009
;
9
:
563
75
.
28.
Humbert
N
,
Navaratnam
N
,
Augert
A
,
Da Costa
M
,
Martien
S
,
Wang
J
, et al
Regulation of ploidy and senescence by the AMPK-related kinase NUAK1
.
EMBO J
2010
;
29
:
376
86
.
29.
Ido
Y
,
Duranton
A
,
Lan
F
,
Cacicedo
JM
,
Chen
TC
,
Breton
L
, et al
Acute activation of AMP-activated protein kinase prevents H2O2-induced premature senescence in primary human keratinocytes
.
PLoS One
2012
;
7
:
e35092
.
30.
Wang
Y
,
Liang
Y
,
Vanhoutte
PM
. 
SIRT1 and AMPK in regulating mammalian senescence: a critical review and a working model
.
FEBS Lett
2011
;
585
:
986
94
.
31.
Han
X
,
Tai
H
,
Wang
X
,
Wang
Z
,
Zhou
J
,
Wei
X
, et al
AMPK activation protects cells from oxidative stress-induced senescence via autophagic flux restoration and intracellular NAD(+) elevation
.
Aging Cell
2016
;
15
:
416
27
.
32.
Alers
S
,
Loffler
AS
,
Wesselborg
S
,
Stork
B
. 
Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks
.
Mol Cell Biol
2012
;
32
:
2
11
.
33.
Kim
J
,
Kundu
M
,
Viollet
B
,
Guan
KL
. 
AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1
.
Nat Cell Biol
2011
;
13
:
132
41
.
34.
Glick
D
,
Barth
S
,
Macleod
KF
. 
Autophagy: cellular and molecular mechanisms
.
J Pathol
2010
;
221
:
3
12
.
35.
Kang
C
,
Xu
QK
,
Martin
TD
,
Li
MZ
,
Demaria
M
,
Aron
L
, et al
The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4
.
Science
2015
;
349
:
aaa5612
.
36.
Galluzzi
L
,
Pedro
JMBS
,
Kroemer
G
. 
Autophagy mediates tumor suppression via cellular senescence
.
Trends Cell Biol
2016
;
26
:
1
3
.
37.
Garcia-Prat
L
,
Martinez-Vicente
M
,
Perdiguero
E
,
Ortet
L
,
Rodriguez-Ubreva
J
,
Rebollo
E
, et al
Autophagy maintains stemness by preventing senescence
.
Nature
2016
;
529
:
37
.
38.
Roach
PJ
. 
AMPK → ULK1 → autophagy
.
Mol Cell Biol
2011
;
31
:
3082
4
.
39.
Williams
RAM
,
Smith
TK
,
Cull
B
,
Mottram
JC
,
Coombs
GH
. 
ATG5 is essential for ATG8-dependent autophagy and mitochondrial homeostasis in leishmania major
.
PLoS Pathog
2012
;
8
:
e1002695
.
40.
Vuppalapati
KK
,
Bouderlique
T
,
Newton
PT
,
Kaminskyy
VO
,
Wehtje
H
,
Ohlsson
C
, et al
Targeted deletion of autophagy genes Atg5 or Atg7 in the chondrocytes promotes caspase-dependent cell death and leads to mild growth retardation
.
J Bone Miner Res
2015
;
30
:
2249
61
.
41.
Poillet-Perez
L
,
Despouy
G
,
Delage-Mourroux
R
,
Boyer-Guittaut
M
. 
Interplay between ROS and autophagy in cancer cells, from tumor initiation to cancer therapy
.
Redox Biol
2015
;
4
:
184
92
.
42.
Fei
P
,
El-Deiry
WS
. 
P53 and radiation responses
.
Oncogene
2003
;
22
:
5774
83
.
43.
Ogawara
Y
,
Kishishita
S
,
Obata
T
,
Isazawa
Y
,
Suzuki
T
,
Tanaka
K
, et al
Akt enhances Mdm2-mediated ubiquitination and degradation of p53
.
J Biol Chem
2002
;
277
:
21843
50
.
44.
Vijayaraghavan
S
,
Karakas
C
,
Doostan
I
,
Chen
X
,
Bui
T
,
Yi
M
, et al
CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers
.
Nat Commun
2017
;
8
:
15916
.
45.
Yang
C
,
Giri
U
,
Cortez
MA
,
Gudikote
J
,
Fan
Y
,
Diao
L
, et al
Abstract 1589: LKB1 overexpression regulates DNA repair pathway and sensitivity to radiation in LKB1 mutant non-small cell lung cancer cell lines
.
Cancer Res
2013
;
73
:
1589
.
46.
Kriegs
M
,
Kasten-Pisula
U
,
Rieckmann
T
,
Holst
K
,
Saker
J
,
Dahm-Daphi
J
, et al
The epidermal growth factor receptor modulates DNA double-strand break repair by regulating non-homologous end-joining
.
DNA Repair
2010
;
9
:
889
97
.
47.
Wang
M
,
Morsbach
F
,
Sander
D
,
Gheorghiu
L
,
Nanda
A
,
Benes
C
, et al
EGF receptor inhibition radiosensitizes NSCLC cells by inducing senescence in cells sustaining DNA double-strand breaks
.
Cancer Res
2011
;
71
:
6261
9
.
48.
Zi
F
,
Zi
H
,
Li
Y
,
He
J
,
Shi
Q
,
Cai
Z
. 
Metformin and cancer: an existing drug for cancer prevention and therapy
.
Oncol Lett
2018
;
15
:
683
90
.
49.
DeWaal
D
,
Nogueira
V
,
Terry
AR
,
Patra
KC
,
Jeon
SM
,
Guzman
G
, et al
Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin
.
Nat Commun
2018
;
9
:
446
.
50.
Zhang
Y
,
Li
L
,
Wang
L
,
Li
J
,
Fan
Z
,
Yang
L
, et al
Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer
.
Cancer Res
2018
;
78
:
1779
91
.