Cancer Res Treat.  2019 Apr;51(2):451-463. 10.4143/crt.2017.341.

Pan-Pim Kinase Inhibitor AZD1208 Suppresses Tumor Growth and Synergistically Interacts with Akt Inhibition in Gastric Cancer Cells

Affiliations
  • 1Cancer Research Institute, Seoul National University, Seoul, Korea. moisa@snu.ac.kr
  • 2Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.
  • 3Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
Pim kinases are highly conserved serine/threonine kinases, and different expression patterns of each isoform (Pim-1, Pim-2, and Pim-3) have been observed in various types of human cancers, including gastric cancer. AZD1208 is a potent and selective inhibitor that affects all three isoforms of Pim. We investigated the effects of AZD1208 as a single agent and in combination with an Akt inhibitor in gastric cancer cells.
MATERIALS AND METHODS
The antitumor activity of AZD1208 with/without an Akt inhibitor was evaluated in a large panel of gastric cancer cell lines through growth inhibition assays. The underlying mechanism was also examined by western blotting, immunofluorescence assay, and cell cycle analysis.
RESULTS
AZD1208 treatment decreased gastric cancer cell proliferation rates and induced autophagy only in long-term culture systems. Light chain 3B (LC3B), a marker of autophagy, was increased in sensitive cells in a dose-dependent manner with AZD1208 treatment, which suggested that the growth inhibition effect of AZD1208 was achieved through autophagy, not apoptosis. Moreover, we found that cells damaged by Pim inhibition were repaired by activation of the DNA damage repair pathway, which promoted cell survival and led the cells to become resistant to AZD1208. We also confirmed that the combination of an Akt inhibitor with AZD1208 produced a highly synergistic effect in gastric cancer cell lines.
CONCLUSION
Treatment with AZD1208 alone induced considerable cell death through autophagy in gastric cancer cells. Moreover, the combination of AZD1208 with an Akt inhibitor showed synergistic antitumor effects through regulation of the DNA damage repair pathway.

Keyword

AZD1208; Pim kinase inhibitor; Autophagy; Akt; Stomach neoplasms

MeSH Terms

Apoptosis
Autophagy
Blotting, Western
Cell Cycle
Cell Death
Cell Line
Cell Proliferation
Cell Survival
DNA Damage
Fluorescent Antibody Technique
Humans
Phosphotransferases*
Protein Isoforms
Stomach Neoplasms*
Phosphotransferases
Protein Isoforms

Figure

  • Fig. 1. Effects of AZD1208 treatment on the inhibition of human gastric cancer cell growth. (A) Chemical structure of AZD1208. (B) The growth inhibition effects of AZD1208 were evaluated with a colony formation assay. The percentage of surviving cells was calculated by counting the number of colonies and are shown as a graph with standard error bars (n=3).

  • Fig. 2. Basal levels of Pim kinase protein expression in gastric cancer cell lines. Basal levels of Pim kinase proteins in human gastric cancer cell lines were confirmed by western blot analysis. BT-549 sample and α-tubulin were used as loading controls.

  • Fig. 3. Effects of AZD1208 treatment on the substrates of Pim kinase. Cells were treated with increasing doses of AZD1208 for 120 hours. Western blot analysis was performed with the indicated antibodies. α-Tubulin was used as a loading control.

  • Fig. 4. Induction of cell death by AZD1208 through stimulation of autophagy. (A) Cells were treated with dimethyl sulfoxide (DMSO; control) or 1 μM AZD1208 for 36 or 120 hours. The expression levels of light chain 3B (LC3B) and Beclin-1 were measured by western blot analysis. α-Tubulin was used as a loading control. (B) SNU-601 and SNU-638 cells transfected with GFP-LC3B were treated with 1 μM AZD1208 for 5 days. Confocal microscopy was used to observe the signals corresponding to LC3B expression (green fluorescence). DNA was counterstained with DAPI (blue). The merged images represent overlapping signals of the two channels. (C) SNU-638 cells were pre-treated with the autophagy inhibitor 3-methyladenine (3-MA; 10 μM) or caspase-3 inhibitor z-VAD-fmk (10 μM) for 24 hours. Next, the cells were treated with 1 μM AZD1208 every 3 days for 14 days. The percentages of surviving cells were calculated by counting the number of colonies and are presented in a bar graph with standard error bars (n=3). a)p=0.008.

  • Fig. 5. Association of the DNA damage repair pathway with AZD1208 resistance. Cells were treated with dimethyl sulfoxide (control) and 1 or 5 μM AZD1208 for 120 hours. The expression levels of ATM and Chk2 were measured by western blot analysis. α-Tubulin was used as a loading control.

  • Fig. 6. Enhanced antitumor effects of the combination of AZD1208 and an Akt inhibitor in gastric cancer cells. (A) Cells were seeded and cultured with increasing concentrations of AZD5363 and 1 μM AZD1208 every 3 days. The cells were cultured for 14 days until colonies formed and were then stained. The percentages of surviving cells were calculated by counting the number of colonies and are presented in a bar graph with standard error bars (n=3). a)p < 0.005. (B) Cells were treated for 120 hours with 1 μM AZD1208 and/or 100 nM AZD5363. The expression levels of the indicated proteins were determined by western blot analysis. α-Tubulin was used as a loading control. (C) Cells were treated with AZD1208 and AZD5363 alone or in combination for 5 days, and immunofluorescence analysis was subsequently performed. Confocal microscopy was used to observe the signals corresponding to p-Chk2 (red) and γ-H2AX (green). DAPI (blue) was used as a nuclear counterstain. CI, combination index. Scale bars=5 μm.


Reference

References

1. Narlik-Grassow M, Blanco-Aparicio C, Carnero A. The PIM family of serine/threonine kinases in cancer. Med Res Rev. 2014; 34:136–59.
Article
2. Brault L, Gasser C, Bracher F, Huber K, Knapp S, Schwaller J. PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica. 2010; 95:1004–15.
Article
3. Nawijn MC, Alendar A, Berns A. For better or for worse: the role of Pim oncogenes in tumorigenesis. Nat Rev Cancer. 2011; 11:23–34.
Article
4. Warnecke-Eberz U, Bollschweiler E, Drebber U, Metzger R, Baldus SE, Holscher AH, et al. Prognostic impact of protein overexpression of the proto-oncogene PIM-1 in gastric cancer. Anticancer Res. 2009; 29:4451–5.
5. Zheng HC, Tsuneyama K, Takahashi H, Miwa S, Sugiyama T, Popivanova BK, et al. Aberrant Pim-3 expression is involved in gastric adenoma-adenocarcinoma sequence and cancer progression. J Cancer Res Clin Oncol. 2008; 134:481–8.
Article
6. Khanna P, Chua PJ, Bay BH, Baeg GH. The JAK/STAT signaling cascade in gastric carcinoma (Review). Int J Oncol. 2015; 47:1617–26.
Article
7. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002; 2:489–501.
8. LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat. 2008; 11:32–50.
Article
9. Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011; 19:58–71.
Article
10. Muranen T, Selfors LM, Worster DT, Iwanicki MP, Song L, Morales FC, et al. Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. Cancer Cell. 2012; 21:227–39.
Article
11. Altomare DA, Khaled AR. Homeostasis and the importance for a balance between AKT/mTOR activity and intracellular signaling. Curr Med Chem. 2012; 19:3748–62.
12. Amaravadi R, Thompson CB. The survival kinases Akt and Pim as potential pharmacological targets. J Clin Invest. 2005; 115:2618–24.
Article
13. Hammerman PS, Fox CJ, Birnbaum MJ, Thompson CB. Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival. Blood. 2005; 105:4477–83.
Article
14. Cen B, Mahajan S, Wang W, Kraft AS. Elevation of receptor tyrosine kinases by small molecule AKT inhibitors in prostate cancer is mediated by Pim-1. Cancer Res. 2013; 73:3402–11.
Article
15. Meja K, Stengel C, Sellar R, Huszar D, Davies BR, Gale RE, et al. PIM and AKT kinase inhibitors show synergistic cytotoxicity in acute myeloid leukaemia that is associated with convergence on mTOR and MCL1 pathways. Br J Haematol. 2014; 167:69–79.
Article
16. Keeton EK, McEachern K, Dillman KS, Palakurthi S, Cao Y, Grondine MR, et al. AZD1208, a potent and selective pan-Pim kinase inhibitor, demonstrates efficacy in preclinical models of acute myeloid leukemia. Blood. 2014; 123:905–13.
Article
17. Dakin LA, Block MH, Chen H, Code E, Dowling JE, Feng X, et al. Discovery of novel benzylidene-1,3-thiazolidine-2,4-diones as potent and selective inhibitors of the PIM-1, PIM-2, and PIM-3 protein kinases. Bioorg Med Chem Lett. 2012; 22:4599–604.
Article
18. Davies BR, Greenwood H, Dudley P, Crafter C, Yu DH, Zhang J, et al. Preclinical pharmacology of AZD5363, an inhibitor of AKT: pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic background. Mol Cancer Ther. 2012; 11:873–87.
Article
19. Ku JL, Park JG. Biology of SNU cell lines. Cancer Res Treat. 2005; 37:1–19.
Article
20. Kang S, Min A, Im SA, Song SH, Kim SG, Kim HA, et al. TGFbeta suppresses COX-2 expression by tristetraprolin-mediated RNA destabilization in A549 human lung cancer cells. Cancer Res Treat. 2015; 47:101–9.
21. Wang Z, Zhang Y, Gu JJ, Davitt C, Reeves R, Magnuson NS. Pim-2 phosphorylation of p21(Cip1/WAF1) enhances its stability and inhibits cell proliferation in HCT116 cells. Int J Biochem Cell Biol. 2010; 42:1030–8.
Article
22. Bartek J, Lukas J. DNA damage checkpoints: from initiation to recovery or adaptation. Curr Opin Cell Biol. 2007; 19:238–45.
Article
23. Zirkin S, Davidovich A, Don J. The PIM-2 kinase is an essential component of the ultraviolet damage response that acts upstream to E2F-1 and ATM. J Biol Chem. 2013; 288:21770–83.
Article
24. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA. DNA repair pathways as targets for cancer therapy. Nat Rev Cancer. 2008; 8:193–204.
Article
25. Ang KL, Shi DL, Keong WW, Epstein RJ. Upregulated Akt signaling adjacent to gastric cancers: implications for screening and chemoprevention. Cancer Lett. 2005; 225:53–9.
Article
26. Xu N, Lao Y, Zhang Y, Gillespie DA. Akt: a double-edged sword in cell proliferation and genome stability. J Oncol. 2012; 2012:951724.
Article
27. Blanco-Aparicio C, Collazo AM, Oyarzabal J, Leal JF, Albaran MI, Lima FR, et al. Pim 1 kinase inhibitor ETP-45299 suppresses cellular proliferation and synergizes with PI3K inhibition. Cancer Lett. 2011; 300:145–53.
Article
28. Cervantes-Gomez F, Chen LS, Orlowski RZ, Gandhi V. Biological effects of the Pim kinase inhibitor, SGI-1776, in multiple myeloma. Clin Lymphoma Myeloma Leuk. 2013; 13 Suppl 2:S317–29.
Article
29. Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, et al. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell. 2006; 126:121–34.
Article
30. Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discov. 2014; 13:140–56.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr