NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5

  • Authors:
    • Kimberly A. Foster
    • Esther P. Jane
    • Daniel R. Premkumar
    • Alejandro Morales
    • Ian F. Pollack
  • View Affiliations

  • Published online on: June 5, 2015     https://doi.org/10.3892/ijo.2015.3035
  • Pages: 506-516
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

We previously observed that glioma cells are differentially sensitive to TRAIL-induced toxicity. Based on our observation that TRAIL-resistant glioma cell lines typically exhibited high levels of Akt activation, we hypothesized that inhibition of Akt signaling using the PI3 kinase inhibitor NVP-BKM120 could promote TRAIL-induced apoptosis in gliomas. We assessed this combination in established and primary cultured glioma cells. Combination treatment led to significant cellular death when compared to either drug alone, but had no effect in normal human astrocytes, and demonstrated activation of the caspase cascade. This enhanced apoptosis appears dependent upon the loss of mitochondrial membrane potential and the release of Smac/DIABLO, AIF and cytochrome c into the cytosol. The upregulation of Noxa and sequestration of Mcl-1 by Noxa were important factors for cell death. Knockdown of Noxa abrogated apoptosis and suggested dependency on Noxa in combination-induced apoptosis. BKM120 upregulated cell surface expression of death receptor 5 (DR5), but did not increase levels of the other major TRAIL receptor, death receptor 4 (DR4). This study demonstrates that antagonizing apoptosis-resistance pathways, such as the PI3/Akt pathway, in combination with death receptor activation, may induce cell death in TRAIL-resistant glioma.

Introduction

Malignant glioma is an aggressive brain tumor that responds poorly to conventional treatment modalities (13). Gliomas demonstrate multiple modes of resistance and single-agent therapy has proven ineffective in treatment of malignant glioma (4,5). While the underlying basis for resistance to apoptosis is not fully understood (6,7), recent discoveries as to the pathogenesis of glioblastoma have led to the development of rationally-designed, targeted therapies aimed at disrupting key signaling pathways.

Tumor necrosis factor-related apoptosis inducing ligand (TRAIL), a member of the TNF family, is a potential cancer agent due to its tumor-specific induction of apoptosis and human recombinant TRAIL is being tested in clinical trials for various cancer types (812). TRAIL initiates apoptosis by binding one of two main receptors, death receptor 4 and 5 (DR4 and DR5, respectively), which signal via an adaptor intermediate molecule that promotes formation of a death-inducing signaling cascade (DISC), and ultimately activation of caspase-8 and caspase-3 (13). While most human cancer cell lines express death receptors for Apo2L/TRAIL, many cancer types have proven resistant to TRAIL-induced cellular death (1416). We, among others, have shown that many malignant gliomas are also TRAIL resistant, despite expression of TRAIL receptors (1719) and aberrance in the NF-κB, protein kinase C, Bcl-2 and Akt pathways have all been implicated in TRAIL resistance in glioma (1921). In particular, Akt pathway activation, which can occur secondary to PTEN deletion and/or PIK3CA mutations, is common in malignant glioma (22).

NVP-BKM120 is a highly selective pan-class I phosphatidylinositol-3 kinase (PI3K) inhibitor (23), shown to effectively induce apoptosis in tumor cell lines and animal models of cancer at clinically achievable doses (2426), and a preliminary clinical trial suggests this agent is well-tolerated (27). A recent study demonstrated effective, synergistic induction of apoptosis in lung cancer cell lines when cells were co-treated with BKM-120 and TRAIL (28). To date, it has not been reported whether BKM120 can sensitize TRAIL-resistant malignant glioma cell lines to apoptosis. We previously observed that TRAIL-resistant glioma cell lines often exhibited high levels of Akt activation (Fig. 1) and, thus, we hypothesized that Akt pathway inhibition via the PI3 kinase inhibitor BKM120 could work with TRAIL to promote cell death in human glioma.

Herein, we show the combination of BKM120 and TRAIL in glioblastoma is effective in promoting cellular death and demonstrate that the effectiveness of this combination is likely contingent on the upregulation of the pro-apoptotic protein Noxa and the death receptor DR5.

Materials and methods

Cell lines

Two TRAIL-resistant cell lines were utilized (U87 and LNZ308). U87 was obtained from American Type Culture Collection (Manasas, VA, USA) and LNZ308 was kindly provided by Dr Nicolas de Tribolet (Switzerland). We obtained primary GBM cells from Conversant Biologics (Huntsville, AL, USA). Normal human astrocytes (HA) and media were obtained from ScienCell Research Laboratories (Carlsbad, CA, USA). Cell culture conditions of these cell lines were as previously described (29,30).

Reagents and antibodies

Soluble human recombinant SuperKillerTRAIL (referred to as TRAIL in this study) was purchased from Enzo Biochemicals (Enzo Life Sciences, Farmingdale, NY, USA). NVP-BKM120 was purchased from Chemie Tek (Indianapolis, IN, USA). Caspase inhibitors (z-VAD-fmk, z-IETD-fmk, z-DEVD-fmk, and z-LEHD-fmk) were purchased from R&D Systems (Minneapolis, MN, USA). The following antibodies were used: Mcl-1 (#4572), cytochrome c (#4280), cleaved poly-ADP-ribose polymerase (PARP, #9546), cleaved caspase-3 (#9664), cleaved caspase-8 (#9496), cleaved caspase-9 (#9501), Smac/DIABLO (#2954), pAKT (#5106), AKT (#2920), pS6kinase (#5364), pBAD (#9105), BIM (#2819), Bcl-2 (#2872), Bcl-xL (#2764), surviving (#2808), XIAP (#2042), DcR1 (#4756) and β-actin (#4970) were from Cell Signaling Technology (Beverly, MA, USA). Noxa (sc-26917) and apoptosis-inducing factor (AIF; sc-5586) were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). DR4 (#IMG-141A) and DR5 (#IMG-122A) were from Imgenex (San Diego, CA, USA).

Annexin V apoptosis assay

Apoptosis was evaluated using a fluorescent Annexin V/propidium iodide (PI) assay kit (Invitrogen, Carlsbad, CA, USA) as described previously (29,30). Cells were treated with or without inhibitors for various intervals, harvested, and pelleted by centrifugation (1,000 rpm for 5 min); washed in ice-cold phosphate-buffered saline (PBS); and re-suspended in the Annexin V-fluorescein isothiocyanate/PI reagent in the dark for 15 min before flow cytometric analysis using a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA, USA).

DiOC6 labeling and detection of mitochondrial membrane depolarization

Mitochondrial membrane depolarization was measured as described previously (29,30). Nonadherent cells were collected, and attached cells were trypsinized and resuspended in phosphate-buffered saline (PBS). Cells were loaded with 50 nM 3′,3′-dihexyloxacarbo-cyanine iodide (DiOC6; Invitrogen), which accumulates in intact mitochondria, at 37°C for 15 min. Cells were then spun at 3,000 × g, rinsed with PBS, and resuspended. Fluorescence intensity was detected by flow cytometry and analyzed with CellQuest (BD Biosciences) and FlowJo (Tree Star, Inc., Ashland, OR, USA) analysis software.

Immunoprecipitation and western blot analysis

Western blot and immunoprecipitation were performed as previously described (18). Cells were washed in ice-cold PBS and lysed in buffer containing 30 mM HEPES, 10% glycerol, 1% Triton X-100, 100 mM NaCl, 10 mM MgCl2, 5 mM EDTA, 2 mM Na3VO4, 2 mM β-glycerophosphate, 1 mM phenylmethyl-sulfonyl fluoride, 1 mM 4-(2-aminoethyl) benzenesulfonyl fluoride, 0.8 μM aprotinin, 50 μM bestatin, 15 μM E-64, 20 μM leupeptin, and 10 μM pepstatin A for 15 min on ice. Samples were centrifuged at 12,000 × g for 15 min, supernatants were isolated, and protein was quantified using protein assay reagent (Pierce Chemical, Rockford, IL, USA). Equal amounts of protein were separated by SDS-PAGE and electro-transferred onto a nylon membrane (Invitrogen). Nonspecific antibody binding was blocked by incubation of the membranes with 4% bovine serum albumin in Tris-buffered saline (TBS)/Tween-20 (0.1%). The membranes were incubated with primary antibody overnight at 4°C, washed in TBS/Tween-20, and incubated with a 1:2000 dilution of horseradish peroxidase-conjugated secondary antibody in TBS/Tween-20 at room temperature for 1 h. Proteins were visualized by western blot chemiluminescence reagent (Cell Signaling). Where indicated, the membranes were re-probed with antibodies against β-actin to ensure equal loading and transfer of proteins.

For immunoprecipitation, cell extracts were prepared by lysing 5×106 cells on ice for 30 min in CHAPS lysis buffer (10 mM HEPES, pH 7.4, 150 mM NaCl, 1% CHAPS, protease, phosphatase inhibitors). Lysates were clarified by centrifugation, and equal amounts of protein extracts were incubated overnight with primary antibody. Dynabeads Protein G (Invitrogen) was added for 2 h, followed by magnetic separation of the immunoprecipitated fraction; western blot analysis was conducted as outlined already herein. Scanning densitometry was performed using acquisition into Adobe Photoshop (Adobe Systems, Inc., San Jose, CA, USA) followed by image analysis (Un-Scan-It gel, version 6.1; Silk Scientific, Orem, UT, USA).

Subcellular fractionation

Cells were treated with or without inhibitors, and cytosolic proteins were fractionated as described previously (29). Cells were resuspended in lysis buffer containing 0.025% digitonin, sucrose (250 mM), HEPES (20 mM, pH 7.4), magnesium chloride (5 mM), potassium chloride (10 mM), EDTA (1 mM), phenylmethylsulfonyl fluoride (1 mM), 10 μg/ml aprotinin, and 10 μg/ml leupeptin. After 10 min of incubation at 4°C, cells were centrifuged (2 min at 13,000 × g), and the supernatant (cytosolic fraction) was removed and frozen at −80°C for subsequent use.

Transient transfection

Transient transfection was performed as previously described (18). Optimal 29mer-pRS-small hairpin (sh) RNA constructs were obtained from Origene (Rockville, MD). Sequences specific for human Noxa (GGA GGT GCT ACA CAA TGT GGC GTC GGC AC), Mcl-1 (ACC TAG AAG GTG GCA TCA GGA ATG TGC TG) and control sequences (GCA CTA CCA GAG CTA ACT CAG ATA GTA CT) (non-target shRNA) were used for this study. Glioma cells were seeded in six-well plates and allowed to reach 70% confluence. Transfection of targeting or control shRNA was performed using FuGENE6 per the manufacturer’s recommendations (Roche Applied Science, Indianapolis, IN, USA). One microgram of Mcl-1 or Noxa or nontargeting shRNA in 100 μl of Opti-MEM medium was mixed with 2 μl of FuGENE6 and incubated at room temperature for 20 min, followed by the addition of complete medium to make the total volume up to 2 ml. After 48 h, media were changed and cells were incubated with inhibitors or vehicle for 24 h. Assessment of cell viability (Annexin V binding) was carried out as described (18).

Flow cytometry of death receptors

Cells were analyzed for surface expression of DR4 and DR5 by indirect staining with anti-human DR4 and DR5 (Imgenex), followed by FITC-conjugated IgG. Briefly, cells (1×106) were stained with 300 μl PBS containing saturating amounts of anti-DR4 and anti-DR5 overnight at 4°C. After incubation, cells were washed with PBS and subjected to secondary antibody (IgG) for 1 h at room temperature. After washing, expression of death receptors were subjected to flow cytometric analysis using a FACSCalibur flow cytometer (BD Biosciences).

Statistical analysis

Unless otherwise indicated, data are expressed as mean ± SD for at least three separate experiments performed in triplicate. The significance of differences between experimental conditions was determined using a two-tailed Student’s t-test. Differences were considered significant at P-values ≤0.05.

Results

Combination treatment with BKM120 and TRAIL induces apoptosis in TRAIL-resistant glioma lines but not human astrocytes

To quantitatively assess the effects of this combination, LNZ308 and U87 cells were subjected to an Annexin V/PI analysis. Single agent treatment resulted in modest Annexin labeling, while co-treatment enhanced Annexin V/PI positive labeling. To demonstrate that sensitivity to this combination was not restricted to established cell lines, primary cultures were created from patients diagnosed with glioblastoma and were modestly sensitive to BKM120 or TRAIL as single agents, while the combination induced apoptosis (Fig. 2A). Statistical significance was reached when comparing single agent TRAIL treatment to combination treatment for both the established and primary lines. Human astrocytes were subjected to individual agents and co-treatment for 24 h and analyzed by Annexin V/PI assay; neither individual agents nor co-treatment showed significant toxicity in non-neoplastic human astrocytes (Fig. 2B). Having validated the hypothesis that inhibition of Akt signaling via BKM120 can promote TRAIL activity, we examined potential mechanisms involved, including promotion of caspase cleavage and activation of the mitochondrial apoptotic pathway.

Co-treatment of TRAIL-resistant glioma cell lines with BKM120 and TRAIL activates the caspase cascade

Western blot analysis [dose-response (Fig. 3A) and time-course (Fig. 3B)] demonstrates activation of the main effector caspase-3 more robustly with combination treatment than with treatment of TRAIL or BKM120 alone and in a time-dependent manner, as early as 6 h in LNZ308; cleavage of PARP was seen as early as 12 h. The initiator caspase-8 was activated by proteolytic cleavage as shown by intermediate (43-kDa) and active (18-kDa) fragments. The downstream caspase-7 showed early cleavage in LNZ308.

As caspases appeared to have a central role in BKM120 and TRAIL-mediated apoptosis, LNZ308, U87 and primary patient GBM cells were preincubated with caspase-specific inhibitors (z-DEVD-fmk, caspase-3 inhibitor; z-IETD-fmk, caspase-8 inhibitor; z-VAD-fmk, pan-caspase inhibitor) and the effect on cell death was examined with Annexin V/PI assay. Inhibition of caspase-3 and use of a pan-caspase inhibitor decreased co-treatment-induced cellular death in all lines, while caspase-8 inhibition significantly diminished cell death in LNZ308 and the primary patient sample (Fig. 3C).

BKM120 and TRAIL co-treatment induces loss of mitochondrial membrane potential (Δψm) and release of mitochondrial protein into the cytosol

Mitochondria play the central role in the intrinsic pathway of cellular death. Evidence suggests that loss of mitochondrial Δψm and release of soluble proteins, such as cytochrome c, into the cytosol are major events associated with intrinsic pathway apoptosis (30,31). To evaluate if mitochondrial-related cell death played a role in combination-induced apoptosis, we utilized the DiOC6 assay. In both established and primary cells, TRAIL (Fig. 4A) and BKM120 (Fig. 4B) induced minimal loss in mitochondrial Δψm, while co-treatment significantly increased the cellular population with loss of mitochondrial Δψm (Fig. 4C). This result was most robustly seen in LNZ308 and the primary patient samples.

Loss of mitochondrial Δψm is associated with release of mitochondrial intermembrane proteins into the cytosol that are critical in activating initiator caspases. We evaluated the release of Smac/DIABLO, apoptosis-inducing factor (AIF) and cytochrome c and observed accumulation in the cytosolic fraction (Fig. 4D) after combination exposure. The release of Smac/DIABLO and AIF occurred as early as 6 h post-treatment in both lines, while cytochrome c showed expression at 24 h.

BKM120-induced Noxa upregulation and Noxa interaction with Mcl-1 may potentiate TRAIL toxicity

Levels of the Bcl-2 family members Mcl-1 (anti-apoptotic) and Noxa (pro-apoptotic), as well as their interaction, play a critical role in determining TRAIL lethality (18,20,3234). Therefore, we hypothesized that shifting Bcl family protein homeostasis may contribute to BKM120-TRAIL cellular death. Western blot analysis demonstrated that target inhibition occurred with BKM120 exposure, as levels of pAKT (but not total AKT) were diminished in both lines in a time- and dose-dependent manner (Fig. 5A). We examined whether BKM120 exposure could alter the levels of proteins known to be crucial to both the intrinsic and extrinsic apoptotic pathways, including Bcl-2 and XIAP family members.

Figure 5

BKM120-induced Noxa upregulation and Noxa interaction with Mcl-1 may potentiate TRAIL toxicity. (A) Cells were treated with BKM120 at indicated doses for 24 h, as well as exposed to BKM120 2.5 μmol/l for indicated time and examined in a time-dependent manner. Cell extracts were subjected to western blot analysis with indicated antibody. β-actin served as loading control. (B) LNZ308 and U87 cells were treated with indicated concentrations of TRAIL or BKM120 or combination of both for indicated duration. Cell extracts were subjected to western blot analysis with indicated antibody. β-actin served as loading control. (C) LNZ308 and U87 cells were treated with indicated concentrations of TRAIL or BKM120 or combination of both for 24 h. An equal amount of protein (500 μg) was immunoprecipitated (IP) with Mcl-1 antibody and subjected to western blot analysis using the indicated antibodies. IP:Mcl-1 probed with Mcl-1 served as control (IP:Mcl-1; WB:Mcl-1). (D) LNZ308 cells were subjected to non-target (control), Noxa and Mcl-1 shRNA, as described in Materials and methods. Following transfection, cells were incubated in the presence of SAHA and ABT-737 (indicated concentration) for 24 h. Control cells received DMSO (vehicle). At the end of the treatment period, the viable cell numbers were determined by flow cytometric analysis. The mean number of apoptotic cells acquired from two independent experiments is shown. The standard deviation was <5% and error bars are shown. **p<0.05 values were considered statistically significant and represent the decrease in percent of apoptosis following transfection with shNoxa; shMcl-1 transfection showed a trend towards increased apoptosis compared to control (NT shRNA), but was not statistically significant.

We found little effect on expression of major Bcl-2 family proteins Bcl-2 and Bcl-xL or the IAP family member XIAP (Fig. 5A); however, we observed increased levels of Noxa, a pro-apoptotic Bcl-2 family member, and decreased levels of Mcl-1, a well-established pro-survival protein and downstream target of the Akt-signaling pathway, in a time- and dose-dependent manner (Fig. 5A). Levels of survivin, phosphorylated Bad (S112) and BIM remained unchanged (data not shown).

Next we examined expression levels in a time-dependent manner after co-treatment with TRAIL and BKM120 and again found downregulation of Mcl-1 and upregulation of Noxa, with unchanged levels of Bcl-2 and Bcl-xL (Fig. 5B). To determine if this combination alters Mcl-1:Noxa interactions, given that sequestration of the pro-survival Mcl-1 by Noxa could tip the balance to cellular death, we performed immunoprecipitation of Mcl-1 and subjected the product to western blot analysis of Noxa. The combination increased Mcl-1:Noxa association (Fig. 5C). To examine the hypothesis that Mcl-1 downregulation and Noxa upregulation are required for TRAIL-induced apoptosis, we performed RNA interference experiments to knock-down the expression of both proteins. We transiently transfected and treated cells as indicated in Fig. 5D, then analyzed cellular viability with flow cytometry. After co-treatment, a decrease in Noxa levels resulted in a statistically significant decrease in cell toxicity, while Mcl-1 knockdown showed a trend toward increased death after co-treatment.

BKM120 upregulates cell surface expression of DR5

The death receptor-induced apoptotic pathway (also called the extrinsic apoptotic pathway) is initiated by death ligands, most commonly Fas ligand (FasL), tumor necrosis factor (TNF), and TRAIL. It has been shown that Akt inhibition upregulates FasL (35,36) and that PI3K inhibition can sensitize glioblastoma to death receptor-induced apoptosis (3); we hypothesized that Akt inhibition would affect expression of death receptors. In our study, BMK120 treatment produced an increase in the expression of DR5, with evidence of protein expression at 1 μmol/l at 24 h post-treatment. There was no change in the expression levels of DR4 or decoy receptor 1 (DcR1) in either cell line (Fig. 6A). In a time-dependent manner, BKM120 alone and combination treatment produced increased levels of DR5 in both lines as early as 6 h, but no time-dependent change in expression of DR4 or DcR1 was observed. FACS analysis showed that the cell surface expression of DR5 was increased in LNZ308 and U87 cell lines treated with BKM120, without a change in the expression of DR4 (Fig. 6B).

Discussion

Drug resistance remains a major barrier for cancer therapeutics and multiple pathways contribute to cell death resistance in glioma. Currently, there is great interest in aberrant PI3K/Akt signaling in glioma and increased activity of this pathway may be responsible for the dismal prognosis associated with glioblastoma (37). While many human cancer cells express death receptors, various cancer types, including some glioma cell lines, remain resistant to TRAIL. Akt pathway activation, a frequent finding in human glioma, correlates with TRAIL resistance. Studies have shown that inhibition of PI3K/Akt pathway with siRNA or small molecule inhibitors can sensitize cancer cells to TRAIL (3,20,28,38). The efficacy of PI3K/Akt/mTOR pathway inhibitors in combination with TRAIL has been previously demonstrated (28,39,40), while the effect of BKM120 and TRAIL co-treatment has not been examined in glioma.

In our study, both the extrinsic and intrinsic pathways of cellular death are implicated. As monotherapy, neither TRAIL nor BKM120 demonstrated a robust change in the mitochondrial Δψm, while the combination induced loss of mitochondrial Δψm. Moreover, analysis of the cytosolic fraction demonstrated the presence of Smac/DIABLO, AIF and cytochrome c, all critical players in the intrinsic pathway. Regulation of the mitochondrial pathway occurs via an intricate balance between expression levels of pro-apoptotic and pro-survival members of the Bcl-2 family, and levels of two proteins in this family, Mcl-1 and Noxa, appear critical.

It is well-established that Mcl-1, a known downstream regulator of the PI3K/Akt pathway, plays a prominent role in the inhibition of apoptosis, while Noxa is crucial in facilitating apoptosis; cellular death following co-treatment may be more contingent on levels of Noxa than Mcl-1. In this regard, we observed significant upregulation of the pro-apoptotic mediator Noxa associated with sequestration of the anti-apoptotic mediator Mcl-1, without significant changes in other Bcl-2 family members, suggesting the important role of these targets in regulating the responses observed. While it has been reported that TRAIL actually increases levels of the pro-survival protein Mcl-1, likely contributing to TRAIL resistance, studies have shown that a second agent that can reduce Mcl-1 levels could serve as effective co-treatment (14).

After knock-down of Mcl-1, we did not observe a statistically significant increase in the amount of cell death from control. On the other hand, western blot analysis showed an increase in the levels of Noxa after treatment with BKM120 alone and co-treatment. As shown by immunoprecipitation, Noxa sequestered Mcl-1, suggesting that levels of the pro-apoptotic protein Noxa may be more critical to the mechanism of cellular death than Mcl-1. shRNA experiments demonstrated that the loss of expression of Noxa limited the ability of this combination to produce cell death. In short, Noxa upregulation may post-transcriptionally inactivate Mcl-1 and allow for displacement of other pro-apoptotic proteins bound to Mcl-1 (41,42).

Our study showed an increase in total death receptor protein levels and death receptor expression at the cellular membrane, specifically DR5 and not DR4, after treatment with BKM120. Since TRAIL triggers apoptosis by binding DR4 and DR5, the expression of death receptors may be pivotal in determining the apoptotic response to death receptor-mediated cell death (43). Multiple recent studies have shown that the upregulation of death receptors, in particular DR5, by other agents such as proteosome inhibitors (44), amiodorone (45), arsenic trioxide (46), and platinum-based chemotherapies (47), can sensitize TRAIL-resistant glioma cell lines to TRAIL-induced cellular death and the use of sensitizers that can upregulate DR5 expression may allow for the selective death of glioma, sparing normal human astrocytes. Recently, an intimate connection has been shown to exist between diminished Akt pathway activation and an increase in DR5 expression (48). Nonetheless, a study evaluating co-treatment with BKM120 and TRAIL in lung cancer cell lines, at the same doses as used in our study, examined levels of DR4 and DR5 after treatment with BKM120 for 16 h and saw no change in receptor levels (in fact, a decrease in DR4 levels) (28), in contrast to the clear increase in DR5 expression we observed in human glioma. The mechanism by which PI3K/Akt pathway inhibition may lead to DR5 upregulation remains to be elucidated. The effect of PI3K/Akt pathway inhibition on the expression of death receptors deserves further examination and future studies looking at this combination should consider evaluation of extrinsic pathway proteins.

This study demonstrates that BKM120 augments TRAIL-induced apoptosis in proven TRAIL-resistant glioma cell lines. The combination shows efficacy in primary patient samples and no evidence of significant cellular death in normal human astrocytes. While Mcl-1 is downregulated by combination treatment, it appears that increased levels of Noxa are critical to cellular death in glioma. Moreover, treatment of TRAIL-resistant glioma cells with BKM120 increases expression of DR5, but not DR4, in both the intracellular milieu and at the cell surface. Our findings suggest further investigation of this combination in vivo is warranted.

Acknowledgements

This work was supported by National Institutes of Health Grant P01NS40923 (I.F.P.), by the Walter L. Copeland of The Pittsburgh Foundation (K.A.F., D.R.P.), and grants from the Ian’s Friends Foundation (to I.F.P.) in honor of Ian Yagoda, and the Children’s Hospital of Pittsburgh Foundation (to I.F.P.).

References

1 

Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK and DePinho RA: Malignant glioma: Genetics and biology of a grave matter. Genes Dev. 15:1311–1333. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Opel D, Westhoff MA, Bender A, Braun V, Debatin KM and Fulda S: Phosphatidylinositol 3-kinase inhibition broadly sensitizes glioblastoma cells to death receptor- and drug-induced apoptosis. Cancer Res. 68:6271–6280. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Pollack IF, Jakacki RI, Blaney SM, Hancock ML, Kieran MW, Phillips P, Kun LE, Friedman H, Packer R, Banerjee A, et al: Phase I trial of imatinib in children with newly diagnosed brainstem and recurrent malignant gliomas: A Pediatric Brain Tumor Consortium report. Neuro Oncol. 9:145–160. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Prados MD, Chang SM, Butowski N, DeBoer R, Parvataneni R, Carliner H, Kabuubi P, Ayers-Ringler J, Rabbitt J, Page M, et al: Phase II study of erlotinib plus temozolomide during and after radiation therapy in patients with newly diagnosed glioblastoma multiforme or gliosarcoma. J Clin Oncol. 27:579–584. 2009. View Article : Google Scholar :

6 

Omuro AM, Faivre S and Raymond E: Lessons learned in the development of targeted therapy for malignant gliomas. Mol Cancer Ther. 6:1909–1919. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Kelley SK and Ashkenazi A: Targeting death receptors in cancer with Apo2L/TRAIL. Curr Opin Pharmacol. 4:333–339. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Bellail AC, Qi L, Mulligan P, Chhabra V and Hao C: TRAIL agonists on clinical trials for cancer therapy: The promises and the challenges. Rev Recent Clin Trials. 4:34–41. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Hall MA and Cleveland JL: Clearing the TRAIL for cancer therapy. Cancer Cell. 12:4–6. 2007. View Article : Google Scholar : PubMed/NCBI

11 

von Pawel J, Harvey JH, Spigel DR, Dediu M, Reck M, Cebotaru CL, Humphreys RC, Gribbin MJ, Fox NL and Camidge DR: Phase II trial of mapatumumab, a fully human agonist monoclonal antibody to tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1), in combination with paclitaxel and carboplatin in patients with advanced non-small-cell lung cancer. Clin Lung Cancer. 15:188–196.e2. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Forero-Torres A, Infante JR, Waterhouse D, Wong L, Vickers S, Arrowsmith E, He AR, Hart L, Trent D, Wade J, et al: Phase 2, multicenter, open-label study of tigatuzumab (CS-1008), a humanized monoclonal antibody targeting death receptor 5, in combination with gemcitabine in chemotherapy-naive patients with unresectable or metastatic pancreatic cancer. Cancer Med. 2:925–932. 2013. View Article : Google Scholar

13 

Takeda K, Stagg J, Yagita H, Okumura K and Smyth MJ: Targeting death-inducing receptors in cancer therapy. Oncogene. 26:3745–3757. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Zhang L and Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 12:228–237. 2005. View Article : Google Scholar

15 

Russo M, Mupo A, Spagnuolo C and Russo GL: Exploring death receptor pathways as selective targets in cancer therapy. Biochem Pharmacol. 80:674–682. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Yang A, Wilson NS and Ashkenazi A: Proapoptotic DR4 and DR5 signaling in cancer cells: Toward clinical translation. Curr Opin Cell Biol. 22:837–844. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Kim EH, Kim HS, Kim SU, Noh EJ, Lee JS and Choi KS: Sodium butyrate sensitizes human glioma cells to TRAIL-mediated apoptosis through inhibition of Cdc2 and the subsequent down-regulation of survivin and XIAP. Oncogene. 24:6877–6889. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Premkumar DR, Jane EP, Foster KA and Pollack IF: Survivin inhibitor YM-155 sensitizes tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cells to apoptosis through Mcl-1 downregulation and by engaging the mitochondrial death pathway. J Pharmacol Exp Ther. 346:201–210. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Jane EP, Premkumar DR and Pollack IF: Bortezomib sensitizes malignant human glioma cells to TRAIL, mediated by inhibition of the NF-{kappa}B signaling pathway. Mol Cancer Ther. 10:198–208. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Opel D, Naumann I, Schneider M, Bertele D, Debatin KM and Fulda S: Targeting aberrant PI3K/Akt activation by PI103 restores sensitivity to TRAIL-induced apoptosis in neuroblastoma. Clin Cancer Res. 17:3233–3247. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Okhrimenko H, Lu W, Xiang C, Hamburger N, Kazimirsky G and Brodie C: Protein kinase C-epsilon regulates the apoptosis and survival of glioma cells. Cancer Res. 65:7301–7309. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Salmena L, Carracedo A and Pandolfi PP: Tenets of PTEN tumor suppression. Cell. 133:403–414. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Maira SM, Pecchi S, Huang A, Burger M, Knapp M, Sterker D, Schnell C, Guthy D, Nagel T, Wiesmann M, et al: Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol Cancer Ther. 11:317–328. 2012. View Article : Google Scholar

24 

Zheng Y, Yang J, Qian J, Zhang L, Lu Y, Li H, Lin H, Lan Y, Liu Z, He J, et al: Novel phosphatidylinositol 3-kinase inhibitor NVP-BKM120 induces apoptosis in myeloma cells and shows synergistic anti-myeloma activity with dexamethasone. J Mol Med Berl. 90:695–706. 2012. View Article : Google Scholar

25 

Koul D, Fu J, Shen R, LaFortune TA, Wang S, Tiao N, Kim YW, Liu JL, Ramnarian D, Yuan Y, et al: Antitumor activity of NVP-BKM120 - a selective pan class I PI3 kinase inhibitor showed differential forms of cell death based on p53 status of glioma cells. Clin Cancer Res. 18:184–195. 2012. View Article : Google Scholar

26 

Brachmann SM, Kleylein-Sohn J, Gaulis S, Kauffmann A, Blommers MJ, Kazic-Legueux M, Laborde L, Hattenberger M, Stauffer F, Vaxelaire J, et al: Characterization of the mechanism of action of the pan class I PI3K inhibitor NVP-BKM120 across a broad range of concentrations. Mol Cancer Ther. 11:1747–1757. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Bendell JC, Rodon J, Burris HA, de Jonge M, Verweij J, Birle D, Demanse D, De Buck SS, Ru QC, Peters M, et al: Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J Clin Oncol. 30:282–290. 2012. View Article : Google Scholar

28 

Ren H, Zhao L, Li Y, Yue P, Deng X, Owonikoko TK, Chen M, Khuri FR and Sun SY: The PI3 kinase inhibitor NVP-BKM120 induces GSK3/FBXW7-dependent Mcl-1 degradation, contributing to induction of apoptosis and enhancement of TRAIL-induced apoptosis. Cancer Lett. 338:229–238. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Premkumar DR, Jane EP, DiDomenico JD, Vukmer NA, Agostino NR and Pollack IF: ABT-737 synergizes with bortezomib to induce apoptosis, mediated by Bid cleavage, Bax activation, and mitochondrial dysfunction in an Akt-dependent context in malignant human glioma cell lines. J Pharmacol Exp Ther. 341:859–872. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Jane EP, Premkumar DR, DiDomenico JD, Hu B, Cheng SY and Pollack IF: YM-155 potentiates the effect of ABT-737 in malignant human glioma cells via survivin and Mcl-1 down-regulation in an EGFR-dependent context. Mol Cancer Ther. 12:326–338. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Tait SW and Green DR: Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 11:621–632. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Kim SH, Ricci MS and El-Deiry WS: Mcl-1: A gateway to TRAIL sensitization. Cancer Res. 68:2062–2064. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Ricci MS, Kim SH, Ogi K, Plastaras JP, Ling J, Wang W, Jin Z, Liu YY, Dicker DT, Chiao PJ, et al: Reduction of TRAIL-induced Mcl-1 and cIAP2 by c-Myc or sorafenib sensitizes resistant human cancer cells to TRAIL-induced death. Cancer Cell. 12:66–80. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Naumann I, Kappler R, von Schweinitz D, Debatin KM and Fulda S: Bortezomib primes neuroblastoma cells for TRAIL-induced apoptosis by linking the death receptor to the mitochondrial pathway. Clin Cancer Res. 17:3204–3218. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Suhara T, Kim HS, Kirshenbaum LA and Walsh K: Suppression of Akt signaling induces Fas ligand expression: Involvement of caspase and Jun kinase activation in Akt-mediated Fas ligand regulation. Mol Cell Biol. 22:680–691. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Uriarte SM, Joshi-Barve S, Song Z, Sahoo R, Gobejishvili L, Jala VR, Haribabu B, McClain C and Barve S: Akt inhibition upregulates FasL, downregulates c-FLIPs and induces caspase-8-dependent cell death in Jurkat T lymphocytes. Cell Death Differ. 12:233–242. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Chakravarti A, Zhai G, Suzuki Y, Sarkesh S, Black PM, Muzikansky A and Loeffler JS: The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. J Clin Oncol. 22:1926–1933. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Rychahou PG, Murillo CA and Evers BM: Targeted RNA interference of PI3K pathway components sensitizes colon cancer cells to TNF-related apoptosis-inducing ligand (TRAIL). Surgery. 138:391–397. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Kim S, Kang J, Qiao J, Thomas RP, Evers BM and Chung DH: Phosphatidylinositol 3-kinase inhibition down-regulates survivin and facilitates TRAIL-mediated apoptosis in neuroblastomas. J Pediatr Surg. 39:516–521. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Bagci-Onder T, Wakimoto H, Anderegg M, Cameron C and Shah K: A dual PI3K/mTOR inhibitor, PI-103, cooperates with stem cell-delivered TRAIL in experimental glioma models. Cancer Res. 71:154–163. 2011. View Article : Google Scholar

41 

Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, Colman PM, Day CL, Adams JM and Huang DC: Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 17:393–403. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Okumura K, Huang S and Sinicrope FA: Induction of Noxa sensitizes human colorectal cancer cells expressing Mcl-1 to the small-molecule Bcl-2/Bcl-xL inhibitor, ABT-737. Clin Cancer Res. 14:8132–8142. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Kischkel FC, Lawrence DA, Chuntharapai A, Schow P, Kim KJ and Ashkenazi A: Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity. 12:611–620. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Seol DW: p53-Independent up-regulation of a TRAIL receptor DR5 by proteasome inhibitors: A mechanism for proteasome inhibitor-enhanced TRAIL-induced apoptosis. Biochem Biophys Res Commun. 416:222–225. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Kim IY, Kang YJ, Yoon MJ, Kim EH, Kim SU, Kwon TK, Kim IA and Choi KS: Amiodarone sensitizes human glioma cells but not astrocytes to TRAIL-induced apoptosis via CHOP-mediated DR5 upregulation. Neuro Oncol. 13:267–279. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Kim EH, Yoon MJ, Kim SU, Kwon TK, Sohn S and Choi KS: Arsenic trioxide sensitizes human glioma cells, but not normal astrocytes, to TRAIL-induced apoptosis via CCAAT/enhancer-binding protein homologous protein-dependent DR5 up-regulation. Cancer Res. 68:266–275. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Ding L, Yuan C, Wei F, Wang G, Zhang J, Bellail AC, Zhang Z, Olson JJ and Hao C: Cisplatin restores TRAIL apoptotic pathway in glioblastoma-derived stem cells through up-regulation of DR5 and down-regulation of c-FLIP. Cancer Invest. 29:511–520. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Wang J, Xu F, Zhu D, Duan Y, Chen J, Sun X, He X, Li P, Sun W and Feng J: Schistosoma japonicum soluble egg antigens facilitate hepatic stellate cell apoptosis by downregulating Akt expression and upregulating p53 and DR5 expression. PLoS Negl Trop Dis. 8:e31062014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2015
Volume 47 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Foster KA, Jane EP, Premkumar DR, Morales A and Pollack IF: NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5. Int J Oncol 47: 506-516, 2015
APA
Foster, K.A., Jane, E.P., Premkumar, D.R., Morales, A., & Pollack, I.F. (2015). NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5. International Journal of Oncology, 47, 506-516. https://doi.org/10.3892/ijo.2015.3035
MLA
Foster, K. A., Jane, E. P., Premkumar, D. R., Morales, A., Pollack, I. F."NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5". International Journal of Oncology 47.2 (2015): 506-516.
Chicago
Foster, K. A., Jane, E. P., Premkumar, D. R., Morales, A., Pollack, I. F."NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5". International Journal of Oncology 47, no. 2 (2015): 506-516. https://doi.org/10.3892/ijo.2015.3035