Metabolic targeting of EGFRvIII/PDK1 axis in temozolomide resistant glioblastoma

Kiran K Velpula, Maheedhara R Guda, Kamlesh Sahu, Jack Tuszynski, Swapna Asuthkar, Sarah E Bach, Justin D Lathia, Andrew J Tsung, Kiran K Velpula, Maheedhara R Guda, Kamlesh Sahu, Jack Tuszynski, Swapna Asuthkar, Sarah E Bach, Justin D Lathia, Andrew J Tsung

Abstract

Glioblastomas are characterized by amplification of EGFR. Approximately half of tumors with EGFR over-expression also express a constitutively active ligand independent EGFR variant III (EGFRvIII). While current treatments emphasize surgery followed by radiation and chemotherapy with Temozolomide (TMZ), acquired chemoresistance is a universal feature of recurrent GBMs. To mimic the GBM resistant state, we generated an in vitro TMZ resistant model and demonstrated that dichloroacetate (DCA), a metabolic inhibitor of pyruvate dehydrogenase kinase 1 (PDK1), reverses the Warburg effect. Microarray analysis conducted on the TMZ resistant cells with their subsequent treatment with DCA revealed PDK1 as its sole target. DCA treatment also induced mitochondrial membrane potential change and apoptosis as evidenced by JC-1 staining and electron microscopic studies. Computational homology modeling and docking studies confirmed DCA binding to EGFR, EGFRvIII and PDK1 with high affinity. In addition, expression of EGFRvIII was comparable to PDK1 when compared to EGFR in GBM surgical specimens supporting our in silico prediction data. Collectively our current study provides the first in vitro proof of concept that DCA reverses the Warburg effect in the setting of EGFRvIII positivity and TMZ resistance leading to GBM cytotoxicity, implicating cellular tyrosine kinase signaling in cancer cell metabolism.

Keywords: DCA; EGFR; EGFRvIII; glioblastoma; homology modeling.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors declare no conflict of interest exists with this manuscript.

Figures

Figure 1. Detection of EGFR, EGFRvIII and…
Figure 1. Detection of EGFR, EGFRvIII and PDK1 in hGBM clinical specimens
A. Immunohistochemical staining of a primary human GBM with EGFR-, EGFRvIII-, or PDK1-specific antibody (top) on consecutive sections is shown (brown, diaminobenzidine; light blue, nuclear counterstain with DAPI; Bar=100 μm) B. Dual immunohistochemical staining for co-localization was conducted with anti-EGFRvIII and anti-PDK1 antibodies followed by the secondary antibodies conjugated with fluorophores for red (PDK1), green (EGFRvIII) and blue (DAPI) fluorescence, respectively. Representative merged images show the cells expressing PDK1 and EGFRvIII. Bar=100 μm. C. Western blot analysis of EGFR, EGFRvIII and PDK1 protein expression in hGBM specimens D. Immunoprecipitation experiments were conducted on hGBM patient specimen tissue lysates by using PDK1 antibody. Western blotting analysis was performed on these immuno-precipitated samples using EGFRvIII antibody. IgG probing was done to confirm equal loading.
Figure 2. Binding energies and Protein Ligand…
Figure 2. Binding energies and Protein Ligand Interaction fingerprint (PLIF) diagrams
A. Binding Energies and Protein Ligand Interaction fingerprint (PLIF) diagrams showing atoms of DCA interacting with residues of A. EGFR B. EGFRvIII C. PDK1. Binding energies and structure of best pose for D. PDK1-EGFR complex E. PDK1-EGFRvIII complex. F. Top conformations of DCA docked in site close to TYR869 in case of wild type EGFR whereas this site does not anchor DCA conformations if TYR869 is mutated to Phenylalanine.
Figure 3. Development and validation of temozolomide…
Figure 3. Development and validation of temozolomide resistant model
A. Schematic representation depicting generation of U373vIII cells with 150μM TMZ resistance (U373vIIIR). Micrographs in lower panel demonstrate the timeline and development of U373vIIIR cells; Panel B1=confluent cells, B2=150 μM TMZ block, B3= TMZ resistant cells, B4=TMZ resistance cells growing to near confluence B. Immunofluorescence staining for the presence of GFAP was conducted on the U373, U373vIII and U373vIIIR cells followed by the secondary antibodies conjugated with green fluorophore. DAPI was used to stain the nuclei. Representative merged images show the cells expressing GFAP Bar=100 μm C. FACS analysis was done to analyze the cell cycle in U373vIII and U373vIIIR cells when treated with 150μM TMZ D. Western blot analysis of EGFR, EGFRvIII, pEGFR and PDK1 protein expression in U373, U373vIII and U373vIIIR cells. GAPDH was used as a loading control E. Semi-quantitative qRT-PCR analysis for EGFR, EGFRvIII and PDK1 in U373 and its EGFRvIII counterparts F. U373vIII or U373vIIIR cells were cultured with 1mM DCA and cell proliferation was measured by MTT assay G. Cells were stained for apoptosis using TUNEL assay. Results represent the mean ± SD of three experiments performed in triplicate.
Figure 4. Differential expression of key genes…
Figure 4. Differential expression of key genes involved in glucose metabolism; si-PDK1/DCA induce mitochondrial apoptosis and ΔΨm change
A. Heat map demonstrating the differential expression of various key genes involved in GBM glucose metabolism. Red=strong downregulation, Orange=modest downregulation, Yellow=modest upregulation and Green=strong upregulation B. Quantification of HIF1α gene expression with response to 1mM DCA treatment on U373vIII/U373vIIIR cells (n = 4; p = 0.005). The data presented is normalized to loading control GAPDH C. GBM cells were transfected with siRNA for PDK1. After 72h of transfections, both control and treated cells were monitored for ΔΨm change using JC-1 dye and was analyzed by fluorescence microscopy (Red= J aggregation (live cells); Green=JC-1 monomer (dead cells) D. Identification of mitochondrial morphologies by electron microscopy. Bar=200 nm.
Figure 5. Measurement of bioenergetic parameters of…
Figure 5. Measurement of bioenergetic parameters of U373vIII/U373vIIIR cells using Seahorse assays
DCA treatment activates U373vIII A. U373vIIIR B. cells towards energetic phase. DCA treatment is believed to increase the aerobic potential as shown by the difference in stressed OCR between the control (blue) and treated (red) values. The assay was performed in triplicate. Effect of DCA on mitochondrial respiration and phenotype. U373vIII C. and U373vIIIR D. cells were treated with 1mM DCA 24 h. Oxygen consumption rate measured under basal conditions, following the addition of the oligomycin (mitochondrial F1-F0-ATPase inhibitor), FCCP (uncoupler) and rotenone (complex I inhibitor). Oxygen consumption rate (OCR) was measured using the Seahorse™ XFp Extracellular Flux analyzer. Each data point is the average of five independent measurements. Error bars indicate ±S.E.M.
Figure 6. DCA treatment reduced EGFRvIII/EGFRvIIIR- induced…
Figure 6. DCA treatment reduced EGFRvIII/EGFRvIIIR- induced tumor growth in mice
A. Dual immunohistochemical staining for colocalization was conducted on the U373vIIIR cells with anti-EGFRvIII antibody and mitotracker followed by the secondary antibodies conjugated with fluorophores for green (EGFRvIII) and mitotracker (red) fluorescence, respectively. Representative merged images show the cells expressing EGFRvIII in colocalized in the mitochondria (Bar=100mm) B. Whole cell lysates of U373vIIIR and DCA treated cells were subjected to EGFR phosphorylation array and its representative images were presented C. Immuno-histochemical analysis of EGFRvIII and PDK1 expression in U373vIII/ U373vIIIR- induced tumors and their respective DCA treatment with 100gm/kg body weight. Representative H&E staining is seen in the insets of left panel (Bar=100mm) (n = 3) D. Kaplan–Meier survival curves from mice bearing intracranial EGFRvIII/EGFRvIIIR- induced tumors. DCA treatment group had an increased survival compared with the control group with a median survival 3-5 weeks. E. Immuno-fluorescent staining for PDK1 (red), EGFRvIII (green), COX2 (blue), demonstrating intense PDK1-EGFR co-localization (yellow) in U373vIIIR cells.

References

    1. van den Bent MJ, Gao Y, Kerkhof M, Kros JM, Gorlia T, van Zwieten K, Prince J, van Duinen S, Sillevis Smitt PA, Taphoorn M, French PJ. Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro-oncology. 2015;17:935–941.
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. The New England journal of medicine. 2005;352:987–996.
    1. Lee EQ, Kaley TJ, Duda DG, Schiff D, Lassman AB, Wong ET, Mikkelsen T, Purow BW, Muzikansky A, Ancukiewicz M, Huse JT, Ramkissoon S, Drappatz J, et al. A Multicenter, Phase II, Randomized, Noncomparative Clinical Trial of Radiation and Temozolomide with or without Vandetanib in Newly Diagnosed Glioblastoma Patients. Clinical cancer research. 2015;21:3610–3618.
    1. Robins HI, Zhang P, Gilbert MR, Chakravarti A, de Groot JF, Grimm SA, Wang F, Lieberman FS, Krauze A, Trotti AM, Mohile N, Kee AY, Colman H, et al. A randomized phase I/II study of ABT-888 in combination with temozolomide in recurrent temozolomide resistant glioblastoma: an NRG oncology RTOG group study. Journal of neuro-oncology. 2016;126:309–316.
    1. Chong DQ, Toh XY, Ho IA, Sia KC, Newman JP, Yulyana Y, Ng WH, Lai SH, Ho MM, Dinesh N, Tham CK, Lam PY. Combined treatment of Nimotuzumab and rapamycin is effective against temozolomide-resistant human gliomas regardless of the EGFR mutation status. BMC cancer. 2015;15 255-015-1191-3.
    1. Ohka F, Natsume A, Wakabayashi T. Current trends in targeted therapies for glioblastoma multiforme. Neurology research international. 2012;2012:878425.
    1. Dai Z, Pan S, Chen C, Cao L, Li X, Chen X, Su X, Lin S. Down-regulation of succinate dehydrogenase subunit B and up-regulation of pyruvate dehydrogenase kinase 1 predicts poor prognosis in recurrent nasopharyngeal carcinoma. Tumour biology. 2016;37:5145–5152.
    1. Ho N, Coomber BL. Pyruvate dehydrogenase kinase expression and metabolic changes following dichloroacetate exposure in anoxic human colorectal cancer cells. Experimental cell research. 2015;331:73–81.
    1. Velpula KK, Bhasin A, Asuthkar S, Tsung AJ. Combined targeting of PDK1 and EGFR triggers regression of glioblastoma by reversing the Warburg effect. Cancer research. 2013;73:7277–7289.
    1. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell metabolism. 2006;3:177–185.
    1. Wang H, Xu T, Jiang Y, Xu H, Yan Y, Fu D, Chen J. The challenges and the promise of molecular targeted therapy in malignant gliomas. Neoplasia. 2015;17:239–255.
    1. Azuaje F, Tiemann K, Niclou SP. Therapeutic control and resistance of the EGFR-driven signaling network in glioblastoma. Cell communication and signaling. 2015;13 23-015-0098-6.
    1. Velpula KK, Dasari VR, Tsung AJ, Gondi CS, Klopfenstein JD, Mohanam S, Rao JS. Regulation of glioblastoma progression by cord blood stem cells is mediated by downregulation of cyclin D1. PloS one. 2011;6:e18017.
    1. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. The New England journal of medicine. 2005;352:997–1003.
    1. Wang Y, Chen L, Bao Z, Li S, You G, Yan W, Shi Z, Liu Y, Yang P, Zhang W, Han L, Kang C, Jiang T. Inhibition of STAT3 reverses alkylator resistance through modulation of the AKT and beta-catenin signaling pathways. Oncology reports. 2011;26:1173–1180.
    1. Happold C, Roth P, Wick W, Schmidt N, Florea AM, Silginer M, Reifenberger G, Weller M. Distinct molecular mechanisms of acquired resistance to temozolomide in glioblastoma cells. Journal of neurochemistry. 2012;122:444–455.
    1. Terme M, Colussi O, Marcheteau E, Tanchot C, Tartour E, Taieb J. Modulation of immunity by antiangiogenic molecules in cancer. Clinical & developmental immunology. 2012;2012:492920.
    1. Hermisson M, Klumpp A, Wick W, Wischhusen J, Nagel G, Roos W, Kaina B, Weller M. O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells. Journal of neurochemistry. 2006;96:766–776.
    1. Oliva CR, Nozell SE, Diers A, McClugage SG, 3rd, Sarkaria JN, Markert JM, Darley-Usmar VM, Bailey SM, Gillespie GY, Landar A, Griguer CE. Acquisition of temozolomide chemoresistance in gliomas leads to remodeling of mitochondrial electron transport chain. The Journal of biological chemistry. 2010;285:39759–39767.
    1. Wicks RT, Azadi J, Mangraviti A, Zhang I, Hwang L, Joshi A, Bow H, Hutt-Cabezas M, Martin KL, Rudek MA, Zhao M, Brem H, Tyler BM. Local delivery of cancer-cell glycolytic inhibitors in high-grade glioma. Neuro-oncology. 2015;17:70–80.
    1. Li C, Meng G, Su L, Chen A, Xia M, Xu C, Yu D, Jiang A, Wei J. Dichloroacetate blocks aerobic glycolytic adaptation to attenuated measles virus and promotes viral replication leading to enhanced oncolysis in glioblastoma. Oncotarget. 2015;6:1544–1555. doi: 10.18632/oncotarget.2838.
    1. Henze AT, Garvalov BK, Seidel S, Cuesta AM, Ritter M, Filatova A, Foss F, Dopeso H, Essmann CL, Maxwell PH, Reifenberger G, Carmeliet P, Acker-Palmer A, et al. Loss of PHD3 allows tumours to overcome hypoxic growth inhibition and sustain proliferation through EGFR. Nature communications. 2014;5:5582.
    1. Chesnelong C, Chaumeil MM, Blough MD, Al-Najjar M, Stechishin OD, Chan JA, Pieper RO, Ronen SM, Weiss S, Luchman HA, Cairncross JG. Lactate dehydrogenase A silencing in IDH mutant gliomas. Neuro-oncology. 2014;16:686–695.
    1. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, Rak J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nature cell biology. 2008;10:619–624.
    1. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer cell. 2012;21:297–308.
    1. Warburg O. On respiratory impairment in cancer cells. Science. 1956;124:269–270.
    1. Iagaru A, Mosci C, Mittra E, Zaharchuk G, Fischbein N, Harsh G, Li G, Nagpal S, Recht L, Gambhir SS. Glioblastoma Multiforme Recurrence: An Exploratory Study of (18)F FPPRGD2 PET/CT. Radiology. 2015;277:497–506.
    1. Johannessen TC, Bjerkvig R. Molecular mechanisms of temozolomide resistance in glioblastoma multiforme. Expert review of anticancer therapy. 2012;12:635–642.
    1. Spiegl-Kreinecker S, Pirker C, Filipits M, Lotsch D, Buchroithner J, Pichler J, Silye R, Weis S, Micksche M, Fischer J, Berger W. O6-Methylguanine DNA methyltransferase protein expression in tumor cells predicts outcome of temozolomide therapy in glioblastoma patients. Neuro-oncology. 2010;12:28–36.
    1. Park I, Mukherjee J, Ito M, Chaumeil MM, Jalbert LE, Gaensler K, Ronen SM, Nelson SJ, Pieper RO. Changes in pyruvate metabolism detected by magnetic resonance imaging are linked to DNA damage and serve as a sensor of temozolomide response in glioblastoma cells. Cancer research. 2014;74:7115–7124.
    1. Sampson JH, Aldape KD, Archer GE, Coan A, Desjardins A, Friedman AH, Friedman HS, Gilbert MR, Herndon JE, McLendon RE, Mitchell DA, Reardon DA, Sawaya R, et al. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro-oncology. 2011;13:324–333.
    1. Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY, Olson JJ. Exciting new advances in neuro-oncology: the avenue to a cure for malignant glioma. CA Cancer J Clin. 2010;60:166–193.
    1. Paff M, Alexandru-Abrams D, Hsu FP, Bota DA. The evolution of the EGFRvIII (rindopepimut) immunotherapy for glioblastoma multiforme patients. Human vaccines & immunotherapeutics. 2014;10:3322–3331.
    1. Choi BD, Archer GE, Mitchell DA, Heimberger AB, McLendon RE, Bigner DD, Sampson JH. EGFRvIII-targeted vaccination therapy of malignant glioma. Brain pathology. 2009;19:713–723.
    1. Heimberger AB, Suki D, Yang D, Shi W, Aldape K. The natural history of EGFR and EGFRvIII in glioblastoma patients. Journal of translational medicine. 2005;3:38.
    1. Jutten B, Rouschop KM. EGFR signaling and autophagy dependence for growth, survival, and therapy resistance. Cell cycle. 2014;13:42–51.
    1. Masui K, Tanaka K, Akhavan D, Babic I, Gini B, Matsutani T, Iwanami A, Liu F, Villa GR, Gu Y, Campos C, Zhu S, Yang H, et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell metabolism. 2013;18:726–739.
    1. Nam SO, Yotsumoto F, Miyata K, Fukagawa S, Yamada H, Kuroki M, Miyamoto S. Warburg effect regulated by amphiregulin in the development of colorectal cancer. Cancer medicine. 2015;4:575–587.
    1. Stacpoole PW, Nagaraja NV, Hutson AD. Efficacy of dichloroacetate as a lactate-lowering drug. Journal of clinical pharmacology. 2003;43:683–691.
    1. Ghosh JC, Siegelin MD, Vaira V, Faversani A, Tavecchio M, Chae YC, Lisanti S, Rampini P, Giroda M, Caino MC, Seo JH, Kossenkov AV, Michalek RD, et al. Adaptive mitochondrial reprogramming and resistance to PI3K therapy. Journal of the National Cancer Institute. 2015:107. doi: 10.1093/jnci/dju502.
    1. Sutendra G, Michelakis ED. Pyruvate dehydrogenase kinase as a novel therapeutic target in oncology. Frontiers in oncology. 2013;3:38.
    1. Kato M, Li J, Chuang JL, Chuang DT. Distinct structural mechanisms for inhibition of pyruvate dehydrogenase kinase isoforms by AZD7545, dichloroacetate, and radicicol. Structure. 2007;15:992–1004.
    1. Gonzalez-Gomez P, Sanchez P, Mira H. MicroRNAs as regulators of neural stem cell-related pathways in glioblastoma multiforme. Molecular neurobiology. 2011;44:235–249.
    1. Abildgaard C, Dahl C, Basse AL, Ma T, Guldberg P. Bioenergetic modulation with dichloroacetate reduces the growth of melanoma cells and potentiates their response to BRAFV600E inhibition. Journal of translational medicine. 2014;12:247.
    1. Maleszewska M, Kaminska B. Is glioblastoma an epigenetic malignancy? Cancers. 2013;5:1120–1139.
    1. Pfleger J, He M, Abdellatif M. Mitochondrial complex II is a source of the reserve respiratory capacity that is regulated by metabolic sensors and promotes cell survival. Cell death & disease. 2015;6:e1835.
    1. Dasari VR, Velpula KK, Alapati K, Gujrati M, Tsung AJ. Cord blood stem cells inhibit epidermal growth factor receptor translocation to mitochondria in glioblastoma. PloS one. 2012;7:e31884.
    1. Cvrljevic AN, Akhavan D, Wu M, Martinello P, Furnari FB, Johnston AJ, Guo D, Pike L, Cavenee WK, Scott AM, Mischel PS, Hoogenraad NJ, Johns TG. Activation of Src induces mitochondrial localisation of de2-7EGFR (EGFRvIII) in glioma cells: implications for glucose metabolism. Journal of cell science. 2011;124:2938–2950.
    1. Boerner JL, Demory ML, Silva C, Parsons SJ. Phosphorylation of Y845 on the epidermal growth factor receptor mediates binding to the mitochondrial protein cytochrome c oxidase subunit II. Molecular and cellular biology. 2004;24:7059–7071.
    1. Nagane M, Levitzki A, Gazit A, Cavenee WK, Huang HJ. Drug resistance of human glioblastoma cells conferred by a tumor-specific mutant epidermal growth factor receptor through modulation of Bcl-XL and caspase-3-like proteases. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:5724–5729.

Source: PubMed

3
Tilaa