Journal of Experimental & Clinical Cancer Research | |
Dual-targeting of aberrant glucose metabolism in glioblastoma | |
Kerrie L McDonald3  Philip J Hogg1  Peter P Luk1  Sylvia A Chung3  Eric Hau2  Pierre J Dilda1  Stephanie Decollogne1  Han Shen3  | |
[1] Tumour Metabolism Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney 2052, NSW, Australia;Cancer Care Centre, St George Hospital, Kogarah 2217, NSW, Australia;Cure Brain Cancer Neuro-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney 2052, Australia | |
关键词: Apoptosis; Dichloroacetate; PENAO; Metabolism; Cancer; | |
Others : 1133388 DOI : 10.1186/s13046-015-0130-0 |
|
received in 2014-12-11, accepted in 2015-01-28, 发布年份 2015 | |
【 摘 要 】
Background
Glioblastoma (GBM) is the most common and malignant primary brain tumor. In contrast to some other tumor types, aberrant glucose metabolism is an important component of GBM growth and chemoresistance. Recent studies of human orthotopic GBM in mice and in situ demonstrated GBM cells rely on both glycolysis and mitochondrial oxidation for glucose catabolism. These observations suggest that the homeostasis of energy metabolism of GBM cells might be further disturbed by dual-inhibition of glucose metabolism. The present study aimed to evaluate the efficacy and the mechanisms of dual-targeting therapy in GBM cells.
Methods
Representative GBM cells (immortalized GBM cell lines and patient-derived GBM cells) and non-cancerous cells were treated with 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid (PENAO), an in-house designed novel arsenic-based mitochondrial toxin, in combination with dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor. The efficacy of this combinatorial therapy was evaluated by MTS assay, clonogenic surviving assay and apoptotic assays. The underlying mechanisms of this dual-targeting treatment were unraveled by using mitochondrial membrane potential measurements, cytosol/mitochondrial ROS detection, western blotting, extracellular flux assay and mass spectrometry.
Results
As monotherapies, both PENAO and DCA induced proliferation arrest in a panel of GBM cell lines and primary isolates. PENAO inhibited oxygen consumption, induced oxidative stress and depolarized mitochondrial membrane potential, which in turn activated mitochondria-mediated apoptosis. By combining DCA with PENAO, the two drugs worked synergistically to inhibit cell proliferation (but had no significant effect on non-cancerous cells), impair the clonogenicity, and induce mitochondria-mediated apoptosis. An oxidative stress of mitochondrial origin takes a prominent place in the mechanism by which the combination of PENAO and DCA induces cell death. Additionally, PENAO-induced oxidative damage was enhanced by DCA through glycolytic inhibition which in turn diminished acid production induced by PENAO. Moreover, DCA treatment also led to an alteration in the multidrug resistance (MDR) phenotype of GBM cells, thereby leading to an increased cytosolic accumulation of PENAO.
Conclusions
The findings of this study shed a new light with respect to the dual-targeting of glucose metabolism in GBM cells and the innovative combination of PENAO and DCA shows promise in expanding GBM therapies.
【 授权许可】
2015 Shen et al.; licensee BioMed Central.
【 预 览 】
Files | Size | Format | View |
---|---|---|---|
20150304143505973.pdf | 1095KB | download | |
Figure 5. | 30KB | Image | download |
Figure 4. | 20KB | Image | download |
Figure 3. | 35KB | Image | download |
Figure 2. | 34KB | Image | download |
Figure 1. | 51KB | Image | download |
【 图 表 】
Figure 1.
Figure 2.
Figure 3.
Figure 4.
Figure 5.
【 参考文献 】
- [1]Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al.: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005, 352:987-996.
- [2]Warburg O: On the origin of cancer cells. Science 1956, 123:309-314.
- [3]de Groof AJ, te Lindert MM, van Dommelen MM, Wu M, Willemse M, Smift AL, et al.: Increased OXPHOS activity precedes rise in glycolytic rate in H-RasV12/E1A transformed fibroblasts that develop a Warburg phenotype. Mol Cancer 2009, 8:54. BioMed Central Full Text
- [4]El Mjiyad N, Caro-Maldonado A, Ramirez-Peinado S, Munoz-Pinedo C: Sugar-free approaches to cancer cell killing. Oncogene 2011, 30:253-264.
- [5]Ramsay EE, Hogg PJ, Dilda PJ: Mitochondrial metabolism inhibitors for cancer therapy. Pharm Res 2011, 28:2731-2744.
- [6]Pelicano H, Martin DS, Xu RH, Huang P: Glycolysis inhibition for anticancer treatment. Oncogene 2006, 25:4633-4646.
- [7]Pathania D, Millard M, Neamati N: Opportunities in discovery and delivery of anticancer drugs targeting mitochondria and cancer cell metabolism. Adv Drug Deliv Rev 2009, 61:1250-1275.
- [8]Carew JS, Huang P: Mitochondrial defects in cancer. Mol Cancer 2002, 1:9. BioMed Central Full Text
- [9]Chen V, Staub RE, Fong S, Tagliaferri M, Cohen I, Shtivelman E: Bezielle selectively targets mitochondria of cancer cells to inhibit glycolysis and OXPHOS. PLoS One 2012, 7:e30300.
- [10]Gang BP, Dilda PJ, Hogg PJ, Blackburn AC: Targeting of two aspects of metabolism in breast cancer treatment. Cancer Biol Ther 2014, 15(11):1533-41. doi: 10.4161/15384047.2014.955992. ( http://www.ncbi.nlm.nih.gov/pubmed/25482950 )
- [11]Bonnet S, Archer SL, Allalunis-Turner J, Haromy A, Beaulieu C, Thompson R, et al.: A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell 2007, 11:37-51.
- [12]McFate T, Mohyeldin A, Lu H, Thakar J, Henriques J, Halim ND, et al.: Pyruvate dehydrogenase complex activity controls metabolic and malignant phenotype in cancer cells. J Biol Chem 2008, 283:22700-22708.
- [13]Sun RC, Fadia M, Dahlstrom JE, Parish CR, Board PG, Blackburn AC: Reversal of the glycolytic phenotype by dichloroacetate inhibits metastatic breast cancer cell growth in vitro and in vivo. Breast Cancer Res Treat 2010, 120:253-260.
- [14]Chen Y, Cairns R, Papandreou I, Koong A, Denko NC: Oxygen consumption can regulate the growth of tumors, a new perspective on the Warburg effect. PLoS One 2009, 4:e7033.
- [15]Michelakis ED, Sutendra G, Dromparis P, Webster L, Haromy A, Niven E, et al.: Metabolic modulation of glioblastoma with dichloroacetate. Sci Transl Med 2010, 2:31ra34.
- [16]Papandreou I, Goliasova T, Denko NC: Anticancer drugs that target metabolism: is dichloroacetate the new paradigm? Int J Cancer 2011, 128:1001-1008.
- [17]Madhok BM, Yeluri S, Perry SL, Hughes TA, Jayne DG: Dichloroacetate induces apoptosis and cell-cycle arrest in colorectal cancer cells. Br J Cancer 2010, 102:1746-1752.
- [18]Liu H, Hu YP, Savaraj N, Priebe W, Lampidis TJ: Hypersensitization of tumor cells to glycolytic inhibitors. Biochemistry 2001, 40:5542-5547.
- [19]Kurtoglu M, Lampidis TJ: From delocalized lipophilic cations to hypoxia: blocking tumor cell mitochondrial function leads to therapeutic gain with glycolytic inhibitors. Mol Nutr Food Res 2009, 53:68-75.
- [20]Sun RC, Board PG, Blackburn AC: Targeting metabolism with arsenic trioxide and dichloroacetate in breast cancer cells. Mol Cancer 2011, 10:142. BioMed Central Full Text
- [21]Stockwin LH, Yu SX, Borgel S, Hancock C, Wolfe TL, Phillips LR, et al.: Sodium dichloroacetate selectively targets cells with defects in the mitochondrial ETC. Int J Cancer 2010, 127:2510-2519.
- [22]Xiao H, Yan L, Zhang Y, Qi R, Li W, Wang R, et al.: A dual-targeting hybrid platinum(IV) prodrug for enhancing efficacy. Chem Commun 2012, 48:10730-10732.
- [23]Xie J, Wang BS, Yu DH, Lu Q, Ma J, Qi H, et al.: Dichloroacetate shifts the metabolism from glycolysis to glucose oxidation and exhibits synergistic growth inhibition with cisplatin in HeLa cells. Int J Oncol 2011, 38:409-417.
- [24]Haugrud AB, Zhuang Y, Coppock JD, Miskimins WK: Dichloroacetate enhances apoptotic cell death via oxidative damage and attenuates lactate production in metformin-treated breast cancer cells. Breast Cancer Res Treat 2014, 147:539-550.
- [25]Dilda PJ, Decollogne S, Weerakoon L, Norris MD, Haber M, Allen JD, et al.: Optimization of the antitumor efficacy of a synthetic mitochondrial toxin by increasing the residence time in the cytosol. J Med Chem 2009, 52:6209-6216.
- [26]Park D, Chiu J, Perrone GG, Dilda PJ, Hogg PJ: The tumour metabolism inhibitors GSAO and PENAO react with cysteines 57 and 257 of mitochondrial adenine nucleotide translocase. Cancer Cell Int 2012, 12:11. BioMed Central Full Text
- [27]Don AS, Kisker O, Dilda P, Donoghue N, Zhao X, Decollogne S, et al.: A peptide trivalent arsenical inhibits tumor angiogenesis by perturbing mitochondrial function in angiogenic endothelial cells. Cancer Cell 2003, 3:497-509.
- [28]Abuhusain HJ, Matin A, Qiao Q, Shen H, Kain N, Day BW, et al.: A metabolic shift favoring sphingosine 1-phosphate at the expense of ceramide controls glioblastoma angiogenesis. J Biol Chem 2013, 288:37355-37364.
- [29]Halestrap AP: What is the mitochondrial permeability transition pore? J Mol Cell Cardiol 2009, 46:821-831.
- [30]Mah LJ, El-Osta A, Karagiannis TC: gammaH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 2010, 24:679-686.
- [31]Kumar A, Kant S, Singh SM: Antitumor and chemosensitizing action of dichloroacetate implicates modulation of tumor microenvironment: a role of reorganized glucose metabolism, cell survival regulation and macrophage differentiation. Toxicol Appl Pharmacol 2013, 273:196-208.
- [32]Marin-Valencia I, Yang C, Mashimo T, Cho S, Baek H, Yang XL, et al.: Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab 2012, 15:827-837.
- [33]Griguer CE, Oliva CR: Bioenergetics pathways and therapeutic resistance in gliomas: emerging role of mitochondria. Curr Pharm Des 2011, 17:2421-2427.
- [34]Mischel PS: HOT models in flux: mitochondrial glucose oxidation fuels glioblastoma growth. Cell Metab 2012, 15:789-790.
- [35]Cheng G, Zielonka J, Dranka BP, McAllister D, Mackinnon AC Jr, Joseph J, et al.: Mitochondria-targeted drugs synergize with 2-deoxyglucose to trigger breast cancer cell death. Cancer Res 2012, 72:2634-2644.
- [36]Dilip A, Cheng G, Joseph J, Kunnimalaiyaan S, Kalyanaraman B, Kunnimalaiyaan M, et al.: Mitochondria-targeted antioxidant and glycolysis inhibition: synergistic therapy in hepatocellular carcinoma. Anticancer Drugs 2013, 24:881-888.
- [37]Dunbar EM, Coats BS, Shroads AL, Langaee T, Lew A, Forder JR, et al.: Phase 1 trial of dichloroacetate (DCA) in adults with recurrent malignant brain tumors. Invest New Drugs 2014, 32:452-464.
- [38]Wong JY, Huggins GS, Debidda M, Munshi NC, De Vivo I: Dichloroacetate induces apoptosis in endometrial cancer cells. Gynecol Oncol 2008, 109:394-402.
- [39]Duan Y, Zhao X, Ren W, Wang X, Yu KF, Li D, et al.: Antitumor activity of dichloroacetate on C6 glioma cell: in vitro and in vivo evaluation. Onco Targets Ther 2013, 6:189-198.
- [40]Kumar K, Wigfield S, Gee HE, Devlin CM, Singleton D, Li JL, et al.: Dichloroacetate reverses the hypoxic adaptation to bevacizumab and enhances its antitumor effects in mouse xenografts. J Mol Med 2013, 91:749-758.
- [41]Choi YW, Lim IK: Sensitization of metformin-cytotoxicity by dichloroacetate via reprogramming glucose metabolism in cancer cells. Cancer Lett 2014, 346:300-308.
- [42]Dai Y, Xiong X, Huang G, Liu J, Sheng S, Wang H, et al.: Dichloroacetate enhances adriamycin-induced hepatoma cell toxicity in vitro and in vivo by increasing reactive oxygen species levels. PLoS One 2014, 9:e92962.
- [43]Tong J, Xie G, He J, Li J, Pan F, Liang H: Synergistic antitumor effect of dichloroacetate in combination with 5-fluorouracil in colorectal cancer. J Biomed Biotechnol 2011, 2011:740564.
- [44]Lin G, Hill DK, Andrejeva G, Boult JK, Troy H, Fong AC, et al.: Dichloroacetate induces autophagy in colorectal cancer cells and tumours. Br J Cancer 2014, 111:375-385.
- [45]Sinclair WK, Morton RA: X-ray sensitivity during the cell generation cycle of cultured Chinese hamster cells. Radiat Res 1966, 29:450-474.
- [46]Sinclair WK: Cyclic x-ray responses in mammalian cells in vitro. Radiat Res 1968, 33:620-643.
- [47]Pawlik TM, Keyomarsi K: Role of cell cycle in mediating sensitivity to radiotherapy. Int J Radiat Oncol Biol Phys 2004, 59:928-942.
- [48]Shen H, Luk PP, Chung SA, Decollogne S, Dilda PJ, Hogg PJ, et al.: PENAO, a novel mitochondria-targeted agent, has shown potent antitumor effect on glioblastoma in vitro and in vivo. AACR 104th Annual Meeting, Apr 6–10, Washington DC, USA 2013.
- [49]Sanchez-Arago M, Chamorro M, Cuezva JM: Selection of cancer cells with repressed mitochondria triggers colon cancer progression. Carcinogenesis 2010, 31:567-576.
- [50]Ayyanathan K, Kesaraju S, Dawson-Scully K, Weissbach H: Combination of sulindac and dichloroacetate kills cancer cells via oxidative damage. PLoS One 2012, 7:e39949.
- [51]Ishiguro T, Ishiguro R, Ishiguro M, Iwai S: Co-treatment of dichloroacetate, omeprazole and tamoxifen exhibited synergistically antiproliferative effect on malignant tumors: in vivo experiments and a case report. Hepatogastroenterology 2012, 59:994-996.
- [52]Abe T, Mori T, Wakabayashi Y, Nakagawa M, Cole SP, Koike K, et al.: Expression of multidrug resistance protein gene in patients with glioma after chemotherapy. J Neurooncol 1998, 40:11-18.
- [53]Ngo H, Tortorella SM, Ververis K, Karagiannis TC: Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009, 324(5930):1029-33. doi: 10.1126/science.1160809. ( http://www.ncbi.nlm.nih.gov/pubmed/19460998 )
- [54]Keunen O, Johansson M, Oudin A, Sanzey M, Rahim SA, Fack F, et al.: Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma. Proc Natl Acad Sci U S A 2011, 108:3749-3754.