Clinically relevant GSK‑3β inhibitor 9‑ING‑41 is active as a single agent and in combination with other antitumor therapies in human renal cancer

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

Abstract

Glycogen synthase kinase‑3 (GSK‑3), a serine/threonine kinase, is involved in a broad range of pathological processes including cancer. GSK‑3 has two isoforms, GSK‑3α and GSK‑3β, and GSK‑3β has been recognized as a therapeutic target for the development of new anticancer drugs. The present study aimed to investigate the antitumor effects of 9‑ING‑41, which is a maleimide‑based ATP‑competitive small molecule GSK‑3β inhibitor active in patients with advanced cancer. In renal cancer cell lines, treatment with 9‑ING‑41 alone induced cell cycle arrest and apoptosis, and autophagy inhibitors increased the antitumor effects of 9‑ING‑41 when used in combination. Treatment with 9‑ING‑41 potentiated the antitumor effects of targeted therapeutics and increased the cytotoxic effects of cytokine‑activated immune cells on renal cancer cell lines. These results provided a compelling rationale for the inclusion of patients with renal cancer in studies of 9‑ING‑41, both as a single agent and in combination with current standard therapies.

Figures

Figure 1
Figure 1
Treatment with 9-ING-41 inhibits the proliferation and survival of RCC cells. (A) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (B) Relative cell proliferation was measured by MTS assay in RCC cells treated with the indicated doses of 9-ING-41 for 24, 48, 72 and 96 h. Differences were analyzed by one-way ANOVA. (C) BrdU colorimetric assay was performed in RCC cells treated with diluent (DMSO) or 9-ING-41 at indicated concentrations for 48 h. *P<0.05, **P<0.01 and ***P<0.001. RCC, renal cell carcinoma; GS, glycogen synthase; p, phosphorylated; GSK-3β, glycogen synthase kinase-3β; GI50, concentration that inhibits cell proliferation by 50%; BrdU, 5-bromo-2-deoxyuridine; OD, optical density.
Figure 1
Figure 1
Treatment with 9-ING-41 inhibits the proliferation and survival of RCC cells. (A) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (B) Relative cell proliferation was measured by MTS assay in RCC cells treated with the indicated doses of 9-ING-41 for 24, 48, 72 and 96 h. Differences were analyzed by one-way ANOVA. (C) BrdU colorimetric assay was performed in RCC cells treated with diluent (DMSO) or 9-ING-41 at indicated concentrations for 48 h. *P<0.05, **P<0.01 and ***P<0.001. RCC, renal cell carcinoma; GS, glycogen synthase; p, phosphorylated; GSK-3β, glycogen synthase kinase-3β; GI50, concentration that inhibits cell proliferation by 50%; BrdU, 5-bromo-2-deoxyuridine; OD, optical density.
Figure 1
Figure 1
Treatment with 9-ING-41 inhibits the proliferation and survival of RCC cells. (A) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (B) Relative cell proliferation was measured by MTS assay in RCC cells treated with the indicated doses of 9-ING-41 for 24, 48, 72 and 96 h. Differences were analyzed by one-way ANOVA. (C) BrdU colorimetric assay was performed in RCC cells treated with diluent (DMSO) or 9-ING-41 at indicated concentrations for 48 h. *P<0.05, **P<0.01 and ***P<0.001. RCC, renal cell carcinoma; GS, glycogen synthase; p, phosphorylated; GSK-3β, glycogen synthase kinase-3β; GI50, concentration that inhibits cell proliferation by 50%; BrdU, 5-bromo-2-deoxyuridine; OD, optical density.
Figure 2
Figure 2
9-ING-41 induces cell cycle arrest and apoptosis in RCC cells. (A) Flow cytometry was performed in ACHN and KRCY renal cancer cells treated with 5 µM 9-ING-41 for 48 and 96 h. (B) Cell cycle distribution and rate of sub-G1, G1, S and G2 populations are presented following treatment of RCC cells with 9-ING-41 as indicated. RCC, renal cell carcinoma; FL2-A, total cell fluorescence area.
Figure 2
Figure 2
9-ING-41 induces cell cycle arrest and apoptosis in RCC cells. (A) Flow cytometry was performed in ACHN and KRCY renal cancer cells treated with 5 µM 9-ING-41 for 48 and 96 h. (B) Cell cycle distribution and rate of sub-G1, G1, S and G2 populations are presented following treatment of RCC cells with 9-ING-41 as indicated. RCC, renal cell carcinoma; FL2-A, total cell fluorescence area.
Figure 3
Figure 3
Treatment with 9-ING-41 suppresses the expression of cyclin-dependent kinases and anti-apoptotic proteins in RCC cells. (A and B) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (C) Reverse transcription-quantitative PCR was performed in RCC cells following treatment with the indicated doses of 9-ING-41 for 72 h. Gene expression of Bcl-2 and E2F-1 was significantly decreased in cells treated with 9-ING-41 compared with the control cells. *P<0.05. RCC, renal cell carcinoma; E2F-1, E2F transcription factor 1; CDK1, cell cycle controller 2; XIAP, X-linked inhibitor of apoptosis; PARP, poly (ADP-ribose) polymerase.
Figure 3
Figure 3
Treatment with 9-ING-41 suppresses the expression of cyclin-dependent kinases and anti-apoptotic proteins in RCC cells. (A and B) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (C) Reverse transcription-quantitative PCR was performed in RCC cells following treatment with the indicated doses of 9-ING-41 for 72 h. Gene expression of Bcl-2 and E2F-1 was significantly decreased in cells treated with 9-ING-41 compared with the control cells. *P<0.05. RCC, renal cell carcinoma; E2F-1, E2F transcription factor 1; CDK1, cell cycle controller 2; XIAP, X-linked inhibitor of apoptosis; PARP, poly (ADP-ribose) polymerase.
Figure 3
Figure 3
Treatment with 9-ING-41 suppresses the expression of cyclin-dependent kinases and anti-apoptotic proteins in RCC cells. (A and B) RCC cells were treated with the indicated concentrations of 9-ING-41 for 96 h, and protein expression was analyzed by western blotting. (C) Reverse transcription-quantitative PCR was performed in RCC cells following treatment with the indicated doses of 9-ING-41 for 72 h. Gene expression of Bcl-2 and E2F-1 was significantly decreased in cells treated with 9-ING-41 compared with the control cells. *P<0.05. RCC, renal cell carcinoma; E2F-1, E2F transcription factor 1; CDK1, cell cycle controller 2; XIAP, X-linked inhibitor of apoptosis; PARP, poly (ADP-ribose) polymerase.
Figure 4
Figure 4
Pharmacological inhibition of GSK-3 with 9-ING-41 potentiates the effect of anticancer therapeutics in RCC cells. Relative cell proliferation was measured by MTS assay in RCC cell lines (A and B) ACHN and (C and D) Caki-1 treated with 9-ING-41 in combination with (A and C) targeted therapeutics (sorafenib, sunitinib, pazopanib and cabozantinib) or (B and D) autophagy inhibitors (chloroquine and bafilomycin) for 72 h as indicated. RCC, renal cell carcinoma.
Figure 4
Figure 4
Pharmacological inhibition of GSK-3 with 9-ING-41 potentiates the effect of anticancer therapeutics in RCC cells. Relative cell proliferation was measured by MTS assay in RCC cell lines (A and B) ACHN and (C and D) Caki-1 treated with 9-ING-41 in combination with (A and C) targeted therapeutics (sorafenib, sunitinib, pazopanib and cabozantinib) or (B and D) autophagy inhibitors (chloroquine and bafilomycin) for 72 h as indicated. RCC, renal cell carcinoma.
Figure 5
Figure 5
Treatment with 9-ING-41 sensitizes renal cancer cells to the cytotoxic effects of human immune cells. ACHN and Caki-1 renal cancer cells were treated with 9-ING-41 as indicated, harvested and mixed with activated human immune cells. Lactate dehydrogenase activity of the supernatants was measured and cytotoxicity was calculated. The effector:target ratio indicates the proportion of immune to cancer cells. *P<0.05.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30. doi: 10.3322/caac.21387.
    1. Rodriguez-Vida A, Hutson TE, Bellmunt J, Strijbos MH. New treatment options for metastatic renal cell carcinoma. ESMO Open. 2017;2:e000185. doi: 10.1136/esmoopen-2017-000185.
    1. Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, Nathan P, Staehler M, de Souza P, Merchan JR, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med. 2013;369:722–731. doi: 10.1056/NEJMoa1303989.
    1. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–124. doi: 10.1056/NEJMoa065044.
    1. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: Results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–1068. doi: 10.1200/JCO.2009.23.9764.
    1. Eldar-Finkelman H. Glycogen synthase kinase 3: An emerging therapeutic target. Trends Mol Med. 2002;8:126–132. doi: 10.1016/S1471-4914(01)02266-3.
    1. Schlender KK, Beebe SJ, Willey JC, Lutz SA, Reimann EM. Isolation and characterization of cyclic AMP-independent glycogen synthase kinase from rat skeletal muscle. Biochim Biophys Acta. 1980;615:324–340. doi: 10.1016/0005-2744(80)90500-8.
    1. Nagini S, Sophia J, Mishra R. Glycogen synthase kinases: Moonlighting proteins with theranostic potential in cancer. Semin Cancer Biol. 2019;56:25–36. doi: 10.1016/j.semcancer.2017.12.010.
    1. de Groot RP, Auwerx J, Bourouis M, Sassone-Corsi P. Negative regulation of Jun/AP-1: Conserved function of glycogen synthase kinase 3 and the drosophila kinase shaggy. Oncogene. 1993;8:841–847.
    1. Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000;14:2501–2514. doi: 10.1101/gad.836800.
    1. Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 1998;12:3499–3511. doi: 10.1101/gad.12.22.3499.
    1. Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S, Polakis P. Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science. 1996;272:1023–1026. doi: 10.1126/science.272.5264.1023.
    1. Walz A, Ugolkov A, Chandra S, Kozikowski A, Carneiro BA, O'Halloran TV, Giles FJ, Billadeau DD, Mazar AP. Molecular pathways: Revisiting glycogen synthase kinase-3β as a target for the treatment of cancer. Clin Cancer Res. 2017;23:1891–1897. doi: 10.1158/1078-0432.CCR-15-2240.
    1. Bilim V, Ougolkov A, Yuuki K, Naito S, Kawazoe H, Muto A, Oya M, Billadeau D, Motoyama T, Tomita Y. Glycogen synthase kinase-3: A new therapeutic target in renal cell carcinoma. Br J Cancer. 2009;101:2005–2014. doi: 10.1038/sj.bjc.6605437.
    1. Hilliard TS, Gaisina IN, Muehlbauer AG, Gaisin AM, Gallier F, Burdette JE. Glycogen synthase kinase 3β inhibitors induce apoptosis in ovarian cancer cells and inhibit in-vivo tumor growth. Anticancer Drugs. 2011;22:978–985.
    1. Gaisina IN, Gallier F, Ougolkov AV, Kim KH, Kurome T, Guo S, Holzle D, Luchini DN, Blond SY, Billadeau DD, Kozikowski AP. 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. J Med Chem. 2009;52:1853–1863. doi: 10.1021/jm801317h.
    1. Ugolkov A, Qiang W, Bondarenko G, Procissi D, Gaisina I, James CD, Chandler J, Kozikowski A, Gunosewoyo H, O'Halloran T, et al. Combination treatment with the GSK-3 inhibitor 9-ING-41 and CCNU cures orthotopic chemoresistant glioblastoma in patient-derived xenograft models. Transl Oncol. 2017;10:669–678. doi: 10.1016/j.tranon.2017.06.003.
    1. Ugolkov AV, Bondarenko GI, Dubrovskyi O, Berbegall AP, Navarro S, Noguera R, O'Halloran TV, Hendrix MJ, Giles FJ, Mazar AP. 9-ING-41, a small-molecule glycogen synthase kinase-3 inhibitor, is active in neuroblastoma. Anticancer Drugs. 2018;29:717–724.
    1. Ugolkov A, Gaisina I, Zhang JS, Billadeau DD, White K, Kozikowski A, Jain S, Cristofanilli M, Giles F, O'Halloran T, 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. Pal K, Cao Y, Gaisina IN, Bhattacharya S, Dutta SK, Wang E, Gunosewoyo H, Kozikowski AP, Billadeau DD, Mukhopadhyay D. Inhibition of GSK-3 induces differentiation and impaired glucose metabolism in renal cancer. Mol Cancer Ther. 2014;13:285–296. doi: 10.1158/1535-7163.MCT-13-0681.
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–1108. doi: 10.1038/nprot.2008.73.
    1. Cormier KW, Woodgett JR. Recent advances in understanding the cellular roles of GSK-3. F1000Res. 2017;6 doi: 10.12688/f1000research.10557.1.
    1. Morais C, Gobe G, Johnson DW, Healy H. The emerging role of nuclear factor kappa B in renal cell carcinoma. Int J Biochem Cell Biol. 2011;43:1537–1549. doi: 10.1016/j.biocel.2011.08.003.
    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. Kawazoe H, Bilim VN, Ugolkov AV, Yuuki K, Naito S, Nagaoka A, Kato T, Tomita Y. GSK-3 inhibition in vitro and in vivo enhances antitumor effect of sorafenib in renal cell carcinoma (RCC) Biochem Biophys Res Commun. 2012;423:490–495. doi: 10.1016/j.bbrc.2012.05.147.
    1. Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, Zhang DM, Chen ZS. Autophagy and multidrug resistance in cancer. Chin J Cancer. 2017;36:52. doi: 10.1186/s40880-017-0219-2.
    1. Abdel-Mohsen MA, Ahmed OA, El-Kerm YM. BRCA1 gene mutations and influence of chemotherapy on autophagy and apoptotic mechanisms in Egyptian breast cancer patients. Asian Pac J Cancer Prev. 2016;17:1285–1292. doi: 10.7314/APJCP.2016.17.3.1285.
    1. Park JM, Huang S, Wu TT, Foster NR, Sinicrope FA. Prognostic impact of Beclin 1, p62/sequestosome 1 and LC3 protein expression in colon carcinomas from patients receiving 5-fluorouracil as adjuvant chemotherapy. Cancer Biol Ther. 2013;14:100–107. doi: 10.4161/cbt.22954.
    1. Gao S, Yang XJ, Zhang WG, Ji YW, Pan Q. Mechanism of thalidomide to enhance cytotoxicity of temozolomide in U251-MG glioma cells in vitro. Chin Med J (Engl) 2009;122:1260–1266.
    1. Sun A, Li C, Chen R, Huang Y, Chen Q, Cui X, Liu H, Thrasher JB, Li B. GSK-3β controls autophagy by modulating LKB1-AMPK pathway in prostate cancer cells. Prostate. 2016;76:172–183. doi: 10.1002/pros.23106.
    1. Marchand B, Arsenault D, Raymond-Fleury A, Boisvert FM, Boucher MJ. Glycogen synthase kinase-3 (GSK3) inhibition induces prosurvival autophagic signals in human pancreatic cancer cells. J Biol Chem. 2015;290:5592–5605. doi: 10.1074/jbc.M114.616714.
    1. Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3:17009. doi: 10.1038/nrdp.2017.9.
    1. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277–1290. doi: 10.1056/NEJMoa1712126.
    1. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–1813. doi: 10.1056/NEJMoa1510665.

Source: PubMed

3
Abonneren