9-ING-41, a small molecule inhibitor of GSK-3beta, potentiates the effects of anticancer therapeutics in bladder cancer

Hiroo Kuroki, Tsutomu Anraku, Akira Kazama, Vladimir Bilim, Masayuki Tasaki, Daniel Schmitt, Andrew P Mazar, Francis J Giles, Andrey Ugolkov, Yoshihiko Tomita, Hiroo Kuroki, Tsutomu Anraku, Akira Kazama, Vladimir Bilim, Masayuki Tasaki, Daniel Schmitt, Andrew P Mazar, Francis J Giles, Andrey Ugolkov, Yoshihiko Tomita

Abstract

Glycogen synthase kinase-3 beta (GSK-3β), a serine/threonine kinase, has been identified as a potential therapeutic target in human bladder cancer. In the present study, we investigated the antitumor effect of a small molecule GSK-3β inhibitor, 9-ING-41, currently in clinical studies in patients with advanced cancer, in bladder cancer cell lines. We found that treatment with 9-ING-41 leads to cell cycle arrest, autophagy and apoptosis in bladder cancer cells. The autophagy inhibitor chloroquine potentiated the antitumor effects of 9-ING-41 when tested in combination studies. Our findings also demonstrate that 9-ING-41 enhanced the growth inhibitory effects of gemcitabine or cisplatin when used in combination in bladder cancer cells. Finally, we found that 9-ING-41 sensitized bladder cancer cells to the cytotoxic effects of human immune effector cells. Our results provide a rationale for the inclusion of patients with advanced bladder cancer in clinical studies of 9-ING-41.

Conflict of interest statement

9-ING-41 has been licensed to Actuate Therapeutics, Inc. Andrew Mazar and Andrey Ugolkov hold an equity interest in Actuate Therapeutics, Inc. Daniel Schmitt and Francis Giles are employees of, and hold an equity interest in, Actuate Therapeutics, Inc. Hiroo Kuroki, Tsutomu Anraku, Akira Kazama, Vladimir Bilim, Masayuki Tasaki and Yoshihiko Tomita declare no competing interests.

Figures

Figure 1
Figure 1
Treatment with 9-ING-41 decreases proliferation and survival of bladder cancer cells. (A) Relative cell viability of bladder cancer cells treated with the indicated doses of 9-ING-41 for 96 hours was measured by MTS assay. *P < 0.05 by one-way ANOVA with Tukey post-hoc test. (B) BrdU colometric assay was done using bladder cancer cells treated with 9-ING-41 at indicated concentrations for 48 hours. Columns, mean; bars, SD. *P < 0.05 by one-way ANOVA with Tukey post-hoc test. (C) Flow cytometry was performed using T24, HT1376 and RT4 bladder cancer cells treated with 10 μM of 9-ING-41 for 72 hours. Cell cycle distribution and fraction of sub-G1, G1, S and G2 population are shown.
Figure 2
Figure 2
Antitumor effects of 9-ING-41 therapy in bladder cancer cells. (A) Bladder cancer cells were treated with indicated concentration of 9-ING-41 for 96 hrs. Post-treatment, whole cell lysate was prepared from the cells, separated by SDS-PAGE (20 μg/well), transferred to PVDF membrane, and immunoblotted as indicated. (B) T24 and HT1376 bladder cancer cells were treated with 9-ING-41 for 72 hours and mRNA expression of Bcl-2 and XIAP was analyzed by RT-PCR. Columns, mean; bars, SD. *P < 0.05 by one-way ANOVA with Tukey post-hoc test. (C) Activity of caspase-3 was detected in 9-ING-41-treated bladder cancer cells using colorimetric CaspACE Assay System. Columns, mean; bars, SD. *P < 0.05 by one-way ANOVA with Tukey post-hoc test.
Figure 3
Figure 3
Inhibition of autophagy potentiates the antitumor effects of 9-ING-41 in bladder cancer cells. (A) Brightfield images of T24 cancer cells treated with Vehicle (Control) and 9-ING-41 5 μM for 24 hours. (B) T24 bladder cancer cells were treated with 9-ING-41 10 μM for 48 hrs. Post-treatment, whole cell lysate was prepared from the cells, separated by SDS-PAGE (20 μg/well), transferred to PVDF membrane, and immunoblotted as indicated. (C) Relative cell growth was measured by MTS assay in bladder cancer cell lines T24 and HT1376 treated with 9-ING-41 in combination with autophagy inhibitor chloroquine for 72 hours as indicated. Columns, mean; bars, SD. *P < 0.05 by one-way ANOVA with Tukey post-hoc test.
Figure 4
Figure 4
Pharmacological inhibition of GSK-3β with 9-ING-41 potentiates the effect of anticancer therapeutics in bladder cancer cells. (A,B) Relative cell growth was measured by MTS assay in bladder cancer cell lines T24 and HT1376 treated with 9-ING-41 in combination with gemcitabine (A) and cisplatin (B) for 3 hours as indicated. After the treatment, drugs were replaced with fresh culture medium and relative cell growth was measured by MTS assay after 72 hours. Columns, mean; bars, SD. *P < 0.05 by one-way ANOVA with Tukey post-hoc test.
Figure 5
Figure 5
Treatment with 9-ING-41 sensitizes bladder cancer cells to cytotoxic effects of human immune cells. T24 (A) and HT1376 (B) bladder cancer cells were treated with 9-ING-41 for 48 hrs as indicated, harvested and mixed with human immune cells. LDH activity of supernatant was measured for evaluation of cytotoxicity. Columns, mean; bars, SD. Statistical analysis was performed using unpaired t-test. *P < 0.05.

References

    1. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68:394–424. doi: 10.3322/caac.21492.
    1. Sternberg CN, et al. M-VAC (methotrexate, vinblastine, doxorubicin and cisplatin) for advanced transitional cell carcinoma of the urothelium. The Journal of urology. 1988;139:461–469. doi: 10.1016/S0022-5347(17)42494-3.
    1. Seront E, Machiels JP. Molecular biology and targeted therapies for urothelial carcinoma. Cancer Treat Rev. 2015;41:341–353. doi: 10.1016/j.ctrv.2015.03.004.
    1. Woodgett JR, Cohen P. Multisite phosphorylation of glycogen synthase. Molecular basis for the substrate specificity of glycogen synthase kinase-3 and casein kinase-II (glycogen synthase kinase-5) Biochimica et biophysica acta. 1984;788:339–347. doi: 10.1016/0167-4838(84)90047-5.
    1. Gao C, Holscher C, Liu Y, Li L. GSK3: a key target for the development of novel treatments for type 2 diabetes mellitus and Alzheimer disease. Reviews in the neurosciences. 2011;23:1–11. doi: 10.1515/rns.2011.061.
    1. Hoeflich KP, et al. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature. 2000;406:86–90. doi: 10.1038/35017574.
    1. Ougolkov AV, Bone ND, Fernandez-Zapico ME, Kay NE, Billadeau DD. Inhibition of glycogen synthase kinase-3 activity leads to epigenetic silencing of nuclear factor kappaB target genes and induction of apoptosis in chronic lymphocytic leukemia B cells. Blood. 2007;110:735–742. doi: 10.1182/blood-2006-12-060947.
    1. Ougolkov AV, Fernandez-Zapico ME, Savoy DN, Urrutia RA, Billadeau DD. Glycogen synthase kinase-3beta participates in nuclear factor kappaB-mediated gene transcription and cell survival in pancreatic cancer cells. Cancer Res. 2005;65:2076–2081. doi: 10.1158/0008-5472.Can-04-3642.
    1. Aggarwal BB. Nuclear factor-kappaB: the enemy within. Cancer Cell. 2004;6:203–208. doi: 10.1016/j.ccr.2004.09.003.
    1. Taube JM, et al. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti-PD-1 Therapy. Clinical Cancer Research. 2014;20:5064–5074. doi: 10.1158/1078-0432.Ccr-13-3271.
    1. Naito S, et al. Glycogen Synthase Kinase-3: A Prognostic Marker and a Potential Therapeutic Target in Human Bladder Cancer. Clinical Cancer Research. 2010;16:5124–5132. doi: 10.1158/1078-0432.Ccr-10-0275.
    1. Hilliard TS, et al. Glycogen synthase kinase 3beta inhibitors induce apoptosis in ovarian cancer cells and inhibit in-vivo tumor growth. Anti-cancer drugs. 2011;22:978–985. doi: 10.1097/CAD.0b013e32834ac8fc.
    1. Gaisina IN, et al. From a natural product lead to the identification of potent and selective benzofuran-3-yl-(indol-3-yl)maleimides as glycogen synthase kinase 3beta inhibitors that suppress proliferation and survival of pancreatic cancer cells. Journal of medicinal chemistry. 2009;52:1853–1863. doi: 10.1021/jm801317h.
    1. Ugolkov AV, et al. 9-ING-41, a small-molecule glycogen synthase kinase-3 inhibitor, is active in neuroblastoma. Anti-cancer drugs. 2018;29:717–724. doi: 10.1097/cad.0000000000000652.
    1. Karmali R, et al. GSK-3beta inhibitor, 9-ING-41, reduces cell viability and halts proliferation of B-cell lymphoma cell lines as a single agent and in combination with novel agents. Oncotarget. 2017;8:114924–114934. doi: 10.18632/oncotarget.22414.
    1. Ugolkov A, et al. Combination Treatment with the GSK-3 Inhibitor 9-ING-41 and CCNU Cures Orthotopic Chemoresistant Glioblastoma in Patient-Derived Xenograft Models. Translational Oncology. 2017;10:669–678. doi: 10.1016/j.tranon.2017.06.003.
    1. Ding L, et al. Glycogen synthase kinase-3beta ablation limits pancreatitis-induced acinar-to-ductal metaplasia. J Pathol. 2017;243:65–77. doi: 10.1002/path.4928.
    1. Ugolkov A, et al. GSK-3 inhibition overcomes chemoresistance in human breast cancer. Cancer Lett. 2016;380:384–392. doi: 10.1016/j.canlet.2016.07.006.
    1. Galluzzi L, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34:856–880. doi: 10.15252/embj.201490784.
    1. Guo XL, et al. Targeting autophagy potentiates chemotherapy-induced apoptosis and proliferation inhibition in hepatocarcinoma cells. Cancer Lett. 2012;320:171–179. doi: 10.1016/j.canlet.2012.03.002.
    1. Amaravadi RK, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117:326–336. doi: 10.1172/JCI28833.
    1. Shimizu S, et al. Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. Int J Cancer. 2012;131:548–557. doi: 10.1002/ijc.26374.
    1. Walz A, et al. Molecular Pathways: Revisiting Glycogen Synthase Kinase-3β as a Target for the Treatment of Cancer. Clinical Cancer Research. 2017;23:1891–1897. doi: 10.1158/1078-0432.Ccr-15-2240.
    1. Wu Xiaosheng, Stenson Mary, Abeykoon Jithma, Nowakowski Kevin, Zhang Lianwen, Lawson Joshua, Wellik Linda, Li Ying, Krull Jordan, Wenzl Kerstin, Novak Anne J., Ansell Stephen M., Bishop Gail A., Billadeau Daniel D., Peng Kah Whye, Giles Francis, Schmitt Daniel M., Witzig Thomas E. Targeting glycogen synthase kinase 3 for therapeutic benefit in lymphoma. Blood. 2019;134(4):363–373. doi: 10.1182/blood.2018874560.
    1. Nakayama, Y. & Yamaguchi, N. In International Review of Cell and Molecular Biology Vol. 305 (ed. Kwang W. Jeon) 303–337 (Academic Press, 2013).
    1. Grassilli E, et al. Inhibition of GSK3B bypass drug resistance of p53-null colon carcinomas by enabling necroptosis in response to chemotherapy. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19:3820–3831. doi: 10.1158/1078-0432.CCR-12-3289.
    1. Tan J, et al. Pharmacologic Modulation of Glycogen Synthase Kinase-3β Promotes p53-Dependent Apoptosis through a Direct Bax-Mediated Mitochondrial Pathway in Colorectal Cancer Cells. Cancer Research. 2005;65:9012–9020. doi: 10.1158/0008-5472.Can-05-1226.
    1. Azoulay-Alfaguter I, Elya R, Avrahami L, Katz A, Eldar-Finkelman H. Combined regulation of mTORC1 and lysosomal acidification by GSK-3 suppresses autophagy and contributes to cancer cell growth. Oncogene. 2014;34:4613. doi: 10.1038/onc.2014.390.
    1. Marchand B, Arsenault D, Raymond-Fleury A, Boisvert F-M, Boucher M-J. Glycogen Synthase Kinase-3 (GSK3) Inhibition Induces Prosurvival Autophagic Signals in Human Pancreatic Cancer Cells. Journal of Biological Chemistry. 2015;290:5592–5605. doi: 10.1074/jbc.M114.616714.
    1. Sun A, et al. GSK-3beta controls autophagy by modulating LKB1-AMPK pathway in prostate cancer cells. Prostate. 2016;76:172–183. doi: 10.1002/pros.23106.
    1. Ghatalia P, Zibelman M, Geynisman DM, Plimack E. Approved checkpoint inhibitors in bladder cancer: which drug should be used when? Ther Adv Med Oncol. 2018;10:1758835918788310. doi: 10.1177/1758835918788310.
    1. Han S, et al. Lithium enhances the antitumour effect of temozolomide against TP53 wild-type glioblastoma cells via NFAT1/FasL signalling. Br J Cancer. 2017;116:1302–1311. doi: 10.1038/bjc.2017.89.
    1. Zhou Y, et al. Growth control of multiple myeloma cells through inhibition of glycogen synthase kinase-3. Leukemia & Lymphoma. 2008;49:1945–1953. doi: 10.1080/10428190802304966.
    1. Kaufmann L, et al. LiCl induces TNF-α and FasL production, thereby stimulating apoptosis in cancer cells. Cell Communication and Signaling. 2011;9:15. doi: 10.1186/1478-811X-9-15.

Source: PubMed

3
Iratkozz fel