Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter March 24, 2013

Antitumor effects of energy restriction-mimetic agents: thiazolidinediones

  • Hany A. Omar , Samir A. Salama , El-Shaimaa A. Arafa and Jing-Ru Weng EMAIL logo
From the journal Biological Chemistry

Abstract

Distinct metabolic strategies used by cancer cells to gain growth advantages, such as shifting from oxidative phosphorylation to glycolysis, constitute a basis for their selective targeting as a novel approach for cancer therapy. Thiazolidinediones (TZDs) are ligands for the nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) and they are clinically used as oral hypoglycemic agents. Accumulating evidence suggests that the ability of TZDs to suppress cancer cell proliferation through the interplay between apoptosis and autophagy was, at least in part, mediated through PPARγ-independent mechanisms. This review highlights recent advances in the pharmacological exploitation of the PPARγ-independent anticancer effects of TZDs to develop novel agents targeting tumor metabolism, including glucose transporter inhibitors and adenosine monophosphate-activated protein kinase, which have translational potential as cancer therapeutic agents.


Corresponding author: Jing-Ru Weng, Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan

References

Bishayee, A. (2009). Cancer prevention and treatment with resveratrol: from rodent studies to clinical trials. Cancer Prev. Res. 2, 409–418.10.1158/1940-6207.CAPR-08-0160Search in Google Scholar

Blanquicett, C., Roman, J., and Hart, C.M. (2008). Thiazolidinediones as anti-cancer agents. Cancer Ther. 6, 25–34.Search in Google Scholar

Cairns, R.A., Harris, I.S., and Mak, T.W. (2011). Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95.10.1038/nrc2981Search in Google Scholar

Chen, C.H., Huang, P.H., Chu, P.C., Chen, M.C., Chou, C.C., Wang, D., Kulp, S.K., Teng, C.M., Wang, Q., and Chen, C.S. (2011). Energy restriction-mimetic agents induce apoptosis in prostate cancer cells in part through epigenetic activation of KLF6 tumor suppressor gene expression. J. Biol. Chem. 286, 9968–9976.10.1074/jbc.M110.203240Search in Google Scholar

Chen, S.W., Tsan, Y.T., Chen, J.D., Hsieh, H.I., Lee, C.H., Lin, H.H., Wang, J.D., and Chen, P.C. (2013). Use of thiazolidinediones and the risk of colorectal cancer in patients with diabetes: a nationwide, population-based, case-control study. Diabetes Care 36, 369–375.10.2337/dc11-2197Search in Google Scholar

Colmers, I.N., Bowker, S.L., and Johnson, J.A. (2012). Thiazolidinedione use and cancer incidence in type 2 diabetes: a systematic review and meta-analysis. Diabetes Metab. 38, 475–484.10.1016/j.diabet.2012.06.003Search in Google Scholar

De Lena, M., Lorusso, V., Latorre, A., Fanizza, G., Gargano, G., Caporusso, L., Guida, M., Catino, A., Crucitta, E., Sambiasi, D., et al. (2001). Paclitaxel, cisplatin and lonidamine in advanced ovarian cancer. A phase II study. Eur. J. Cancer 37, 364–368.10.1016/S0959-8049(00)00400-7Search in Google Scholar

Egan, D.F., Shackelford, D.B., Mihaylova, M.M., Gelino, S., Kohnz, R.A., Mair, W., Vasquez, D.S., Joshi, A., Gwinn, D.M., Taylor, R., et al. (2011). Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331, 456–461.10.1126/science.1196371Search in Google Scholar PubMed PubMed Central

Guh, J.H., Chang, W.L., Yang, J., Lee, S.L., Wei, S., Wang, D., Kulp, S.K., and Chen, C.S. (2010). Development of novel adenosine monophosphate-activated protein kinase activators. J. Med. Chem. 53, 2552–2561.10.1021/jm901773dSearch in Google Scholar PubMed PubMed Central

Hardie, D.G. (2007). AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat. Rev. Mol. Cell Biol. 8, 774–785.10.1038/nrm2249Search in Google Scholar PubMed

Holley, A.K., Dhar, S.K., and St Clair, D.K. (2012). Curbing cancer’s sweet tooth: Is there a role for MnSOD in regulation of the Warburg effect? Mitochondrion 13, 170–188.10.1016/j.mito.2012.07.104Search in Google Scholar PubMed PubMed Central

Hsu, P.P. and Sabatini, D.M. (2008). Cancer cell metabolism: Warburg and beyond. Cell 134, 703–707.10.1016/j.cell.2008.08.021Search in Google Scholar PubMed

Huang, J.W., Shiau, C.W., Yang, Y.T., Kulp, S.K., Chen, K.F., Brueggemeier, R.W., Shapiro, C.L., and Chen, C.S. (2005). Peroxisome proliferator-activated receptor gamma-independent ablation of cyclin D1 by thiazolidinediones and their derivatives in breast cancer cells. Mol. Pharmacol. 67, 1342–1348.10.1124/mol.104.007732Search in Google Scholar PubMed

Jiang, M., Shappell, S.B., and Hayward, S.W. (2004). Approaches to understanding the importance and clinical implications of peroxisome proliferator-activated receptor γ (PPARγ) signaling in prostate cancer. J. Cell Biochem. 91, 513–527.10.1002/jcb.10770Search in Google Scholar PubMed

Kaelin, W.G. Jr. and Thompson, C.B. (2010). Q&A: Cancer: clues from cell metabolism. Nature 465, 562–564.10.1038/465562aSearch in Google Scholar PubMed

Kahn, B.B., Alquier, T., Carling, D., and Hardie, D.G. (2005). AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 1, 15–25.10.1016/j.cmet.2004.12.003Search in Google Scholar PubMed

Kim, J., Kundu, M., Viollet, B., and Guan, K.L. (2011). AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, 132–141.10.1038/ncb2152Search in Google Scholar PubMed PubMed Central

Lee, K.H., Hsu, E.C., Guh, J.H., Yang, H.C., Wang, D., Kulp, S.K., Shapiro, C.L., and Chen, C.S. (2011). Targeting energy metabolic and oncogenic signaling pathways in triple-negative breast cancer by a novel adenosine monophosphate-activated protein kinase (AMPK) activator. J. Biol. Chem. 286, 39247–39258.10.1074/jbc.M111.264598Search in Google Scholar PubMed PubMed Central

Li, M., Lee, T.W., Yim, A.P., Mok, T.S., and Chen, G.G. (2006). Apoptosis induced by troglitazone is both peroxisome proliferator-activated receptor-γ- and ERK-dependent in human non-small lung cancer cells. J. Cell. Physiol. 209, 428–438.10.1002/jcp.20738Search in Google Scholar PubMed

Lin, H.Y., Kuo, Y.C., Weng, Y.I., Lai, I.L., Huang, T.H., Lin, S.P., Niu, D.M., and Chen, C.S. (2012). Activation of silenced tumor suppressor genes in prostate cancer cells by a novel energy restriction-mimetic agent. Prostate 72, 1767–1768.10.1002/pros.22530Search in Google Scholar PubMed PubMed Central

Lin, J.N., Lin, V.C., Rau, K.M., Shieh, P.C., Kuo, D.H., Shieh, J.C., Chen, W.J., Tsai, S.C., and Way, T.D. (2010). Resveratrol modulates tumor cell proliferation and protein translation via SIRT1-dependent AMPK activation. J. Agric. Food Chem. 58, 1584–1592.10.1021/jf9035782Search in Google Scholar PubMed

Liu, Y. (2006). Fatty acid oxidation is a dominant bioenergetic pathway in prostate cancer. Prostate Cancer Prostatic Dis. 9, 230–234.10.1038/sj.pcan.4500879Search in Google Scholar PubMed

Locasale, J.W. (2012). Metabolic rewiring drives resistance to targeted cancer therapy. Mol. Syst. Biol. 8, 597–598.10.1038/msb.2012.30Search in Google Scholar PubMed PubMed Central

Milane, L., Duan, Z., and Amiji, M. (2011). Therapeutic efficacy and safety of paclitaxel/lonidamine loaded EGFR-targeted nanoparticles for the treatment of multi-drug resistant cancer. PloS One 6, e24075–e24085.10.1371/journal.pone.0024075Search in Google Scholar PubMed PubMed Central

Olefsky, J.M. (2000). Treatment of insulin resistance with peroxisome proliferator-activated receptor gamma agonists. J. Clin. Invest. 106, 467–472.10.1172/JCI10843Search in Google Scholar PubMed PubMed Central

Omar, H.A., Berman-Booty, L., Kulp, S.K., and Chen, C.S. (2010). Energy restriction as an antitumor target. Future Oncol. 6, 1675–1679.10.2217/fon.10.130Search in Google Scholar PubMed

Omar, H.A., Berman-Booty, L., and Weng, J.R. (2012). Energy restriction: stepping stones towards cancer therapy. Future Oncol. 8, 1503–1506.10.2217/fon.12.142Search in Google Scholar PubMed

Omar, H.A., Sargeant, A.M., Weng, J.R., Wang, D., Kulp, S.K., Patel, T., and Chen, C.S. (2009). Targeting of the Akt-nuclear factor-κB signaling network by [1-(4-chloro-3-nitrobenzenesulfonyl)-1H-indol-3-yl]-methanol (OSU-A9), a novel indole-3-carbinol derivative, in a mouse model of hepatocellular carcinoma. Mol. Pharmacol. 76, 957–968.10.1124/mol.109.058180Search in Google Scholar PubMed PubMed Central

Panchapakesan, U., Chen, X.M., and Pollock, C.A. (2005). Drug insight: thiazolidinediones and diabetic nephropathy-relevance to renoprotection. Nat. Clin. Pract. Nephrol. 1, 33–43.10.1038/ncpneph0029Search in Google Scholar PubMed

Panigrahy, D., Shen, L.Q., Kieran, M.W., and Kaipainen, A. (2003). Therapeutic potential of thiazolidinediones as anticancer agents. Expert. Opin. Investig. Drugs 12, 1925–1937.10.1517/13543784.12.12.1925Search in Google Scholar PubMed

Priebe, A., Tan, L., Wahl, H., Kueck, A., He, G., Kwok, R., Opipari, A., and Liu, J.R. (2011). Glucose deprivation activates AMPK and induces cell death through modulation of Akt in ovarian cancer cells. Gynecol. Oncol. 122, 389–395.10.1016/j.ygyno.2011.04.024Search in Google Scholar PubMed

Saito, S., Furuno, A., Sakurai, J., Sakamoto, A., Park, H.R., Shin-Ya, K., Tsuruo, T., and Tomida, A. (2009). Chemical genomics identifies the unfolded protein response as a target for selective cancer cell killing during glucose deprivation. Cancer Res. 69, 4225–4234.10.1158/0008-5472.CAN-08-2689Search in Google Scholar

Sano, R., Hou, Y.C., Hedvat, M., Correa, R.G., Shu, C.W., Krajewska, M., Diaz, P.W., Tamble, C.M., Quarato, G., Gottlieb, R.A., et al. (2012). Endoplasmic reticulum protein BI-1 regulates Ca2+-mediated bioenergetics to promote autophagy. Genes Dev. 26, 1041–1054.10.1101/gad.184325.111Search in Google Scholar

Shang, L., Chen, S., Du, F., Li, S., Zhao, L., and Wang, X. (2011). Nutrient starvation elicits an acute autophagic response mediated by Ulk1 dephosphorylation and its subsequent dissociation from AMPK. Proc. Natl. Acad. Sci. USA 108, 4788–4793.10.1073/pnas.1100844108Search in Google Scholar

Shiau, C.W., Yang, C.C., Kulp, S.K., Chen, K.F., Chen, C.S., and Huang, J.W. (2005). Thiazolidenediones mediate apoptosis in prostate cancer cells in part through inhibition of Bcl-xL/Bcl-2 functions independently of PPARγ. Cancer Res. 65, 1561–1569.10.1158/0008-5472.CAN-04-1677Search in Google Scholar

Sols, A. and Crane, R.K. (1954). Substrate specificity of brain hexokinase. J. Biol. Chem. 210, 581–595.10.1016/S0021-9258(18)65384-0Search in Google Scholar

Szegezdi, E., Logue, S.E., Gorman, A.M., and Samali, A. (2006). Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. 7, 880–885.10.1038/sj.embor.7400779Search in Google Scholar PubMed PubMed Central

Tower, D.B. (1958). The effects of 2-deoxy-D-glucose on metabolism of slices of cerebral cortex incubated in vitro. J. Neurochem. 3, 185–205.10.1111/j.1471-4159.1958.tb12625.xSearch in Google Scholar PubMed

Wang, D., Chu, P.C., Yang, C.N., Yan, R., Chuang, Y.C., Kulp, S.K., and Chen, C.S. (2012a). Development of a novel class of glucose transporter inhibitors. J. Med. Chem. 55, 3827–3836.10.1021/jm300015mSearch in Google Scholar PubMed PubMed Central

Wang, X., Zhang, A., and Sun, H. (2012b). Power of metabolomics in diagnosis and biomarker discovery of hepatocellular carcinoma. Hepatology [epub ahead of print].Search in Google Scholar

Wei, S., Yang, H.C., Chuang, H.C., Yang, J., Kulp, S.K., Lu, P.J., Lai, M.D., and Chen, C.S. (2008). A novel mechanism by which thiazolidinediones facilitate the proteasomal degradation of cyclin D1 in cancer cells. J. Biol. Chem. 283, 26759–26770.10.1074/jbc.M802160200Search in Google Scholar PubMed PubMed Central

Wei, S., Yang, J., Lee, S.L., Kulp, S.K., and Chen, C.S. (2009). PPARγ-independent antitumor effects of thiazolidinediones. Cancer Lett. 276, 119–124.10.1016/j.canlet.2008.08.008Search in Google Scholar PubMed PubMed Central

Wei, S., Kulp, S.K., and Chen, C.S. (2010). Energy restriction as an antitumor target of thiazolidinediones. J. Biol. Chem. 285, 9780–9791.10.1074/jbc.M109.065466Search in Google Scholar PubMed PubMed Central

Wei, S., Chu, P.C., Chuang, H.C., Hung, W.C., Kulp, S.K., and Chen, C.S. (2012a). Targeting the oncogenic e3 ligase skp2 in prostate and breast cancer cells with a novel energy restriction-mimetic agent. PloS One 7, e47298–e47309.10.1371/journal.pone.0047298Search in Google Scholar PubMed PubMed Central

Wei, S., Liu, L., Zhang, J., Bowers, J., Gowda, G.A., Seeger, H., Fehm, T., Neubauer, H.J., Vogel, U., Clare, S.E., and Raftery, D. (2012b). Metabolomics approach for predicting response to neoadjuvant chemotherapy for breast cancer. Mol. Oncol. [epub ahead of print].10.1016/j.molonc.2012.10.003Search in Google Scholar PubMed PubMed Central

Weng, J.R., Chen, C.Y., Pinzone, J.J., Ringel, M.D., and Chen, C.S. (2006). Beyond peroxisome proliferator-activated receptor γ signaling: the multi-facets of the antitumor effect of thiazolidinediones. Endocr. Relat. Cancer 13, 401–413.10.1677/erc.1.01182Search in Google Scholar PubMed

Whitlock, N.C. and Baek, S.J. (2012). The anticancer effects of resveratrol: modulation of transcription factors. Nutr. Cancer 64, 493–502.10.1080/01635581.2012.667862Search in Google Scholar PubMed PubMed Central

Received: 2013-2-20
Accepted: 2013-3-20
Published Online: 2013-3-24
Published in Print: 2013-7-1

©2013 by Walter de Gruyter Berlin Boston

Downloaded on 28.3.2024 from https://www.degruyter.com/document/doi/10.1515/hsz-2013-0139/html
Scroll to top button