Cancer Res Treat.  2023 Jul;55(3):851-864. 10.4143/crt.2022.1527.

Combination of the LARS1 Inhibitor, BC-LI-0186 with a MEK1/2 Inhibitor Enhances the Anti-Tumor Effect in Non–Small Cell Lung Cancer

Affiliations
  • 1Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
  • 2Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Korea
  • 3Department of Biotechnology, Yonsei University, Seoul, Korea

Abstract

Purpose
The mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth and proliferation by growth factor coordination and amino acid availability. Leucyl-tRNA synthetase 1 (LARS1) senses the intracellular leucine concentration and mediates amino acid-induced activation of mTORC1. Thus, LARS1 inhibition could be useful in cancer treatment. However, the fact that mTORC1 can be stimulated by various growth factors and amino acids suggests that LARS1 inhibition alone has limitations in inhibiting cell growth and proliferation. We investigated the combined effects of BC-LI-0186, a LARS1 inhibitor, and trametinib, an MEK inhibitor, on non–small cell lung cancer (NSCLC).
Materials and Methods
Protein expression and phosphorylation were observed by immunoblotting, and genes differentially expressed between BC-LI-0186–sensitive and –resistant cells were identified by RNA sequencing. The combined effect of the two drugs was inferred from the combination index values and a xenograft model.
Results
LARS1 expression was positively correlated with mTORC1 in NSCLC cell lines. BC-LI-0186 treatment of A549 and H460 cells maintained in media supplemented with fetal bovine serum revealed paradoxical phosphorylation of S6 and activation of mitogen- activated protein kinase (MAPK) signaling. Compared with BC-LI-0186–sensitive cells, –resistant cells showed enrichment of the MAPK gene set. The combination of trametinib and BC-LI-0186 inhibited the phosphorylation of S6, MEK, and extracellular signal-regulated kinase and their synergistic effects were confirmed in a mouse xenograft model.
Conclusion
The combination of BC-LI-0186 and trametinib inhibited the non-canonical mTORC1-activating function of LARS1. Our study demonstrated a new therapeutic approach for NSCLC without targetable driver mutations.

Keyword

BC-LI-0186; LARS1; Non-small cell lung cancer; mTORC1; Trametinib

Figure

  • Fig. 1 Expression of leucyl-tRNA synthetase 1 (LARS1) correlates positively with the mammalian target of rapamycin complex 1 (mTORC1) signal in non–small cell lung cancer (NSCLC). (A) Immunoblotting of LARS1 and molecules involved in mTOR signaling in various NSCLC cells. (B) The correlation plot between LARS1 and pS6 (r=0.8156, p=0.0040), pAkt (r=0.5879, p=0.0739), pERK (r=0.4092, p=0.2403), and pMEK (r=0.7478, p=0.0129). There was a significant positive correlation between LARS1 and pS6. p-values were obtained from Pearson’s correlation analysis and r denotes Pearson’s correlation coefficient. (C) Immunoblotting and heatmap of LARS1 and molecules associated with mTOR signaling according to leucine concentration in the A549 cell line and H460 cell line. (D) Effects of BC-LI-0186 (10 μM) on the A549 and H460 cell lines over time in fetal bovine serum (FBS) (−) media. (E) Effects of the BC-LI-0186 concentration on the A549 and H460 cell lines in FBS (+) media. It was observed that BC-LI-0186 concentrations less than 10 μM had a paradoxical effect on both A549 and H460 cell lines. R*, each cell line was treated with rapamycin (10 μM) for 10 minutes and subjected immunoblotting; S**, serum shock (used for positive control).

  • Fig. 2 Paradoxical response of BC-LI-0186 to lung cancer cell line in the presence of growth factors. (A) Immunoblotting and line graph of pMEK, pERK, and pS6 expression according to the concentration of BC-LI-0186 in the A549 and H460 cell lines. (B) H460 and A549 cell lines were treated with BC-LI-0186 (10 μM) or cisplatin (50 μM) for 48 hours, and cell death was measured by flow cytometry unsing annexin V and propidium iodide (PI) staining. (C) Volcano plot. The log2 fold-change indicates the mean expression level for each gene and each dot represents one gene. Genes with significantly increased expression in the A549 cell line relative to the H460 cell line was indicated. (D) Results of Kyoto Encyclopaedia of Genes and Genomes pathway enrichment analysis.

  • Fig. 3 The combination of BC-LI-0186 and trametinib shows a synergistic effect in human lung cancer cell lines. (A) Immunoblotting of pS6, pMEK, and pERK according to trametinib concentration in the A549 and H460 cell lines. (B) Colocalization images of LC3A/B and LAMP2, and histogram for the ratio of colocalization obtained from slides with 10 or more cells according to the use of BC-LI-0186 and trametinib in A549 and H460 cells. (C) Expression of p62, cleaved poly(ADP-ribose) polymerase (PARP), and cleaved caspase 3 according to the use of BC-LI-0186 and trametinib in A549 and H460 cells. (D) Immunoblotting results pAKT, pS6, pMEK, and pERK according to the use of BC-LI-0186 and trametinib. (E) Fa-dose plot and combination index plot using for treatment of A549, H460, H356, H1703, and H1650 cells.

  • Fig. 4 Synergistic anti-tumor effect of BC-LI-0186 and trametinib combination treatment. (A) Diagram of the treatment schedule in BALB/c nude mice lung cancer model. (B) Photographs of typical BALB/c nude mice bearing tumors. (C) Change rate in tumor volume of each group according to differences before and after treatment. There was a significant tumor volume change in control group comparing with combination group (p=0.004). (D) Representative photograph of haematoxylin and eosin and immunohistochemistry staining for activated caspase-3, pMEK, and pS6 on lung sections obtained from mice after 2 weeks’ treatment, and quantification of caspase-3, pMEK, and pS6 in the tumor of each treatment group. *p < 0.05. p-values were obtained by one-way analysis of variance followed by Tukey’s post-hoc multiple comparison tests. (E) Body weight change rate among mouse groups during the course of treatment.

  • Fig. 5 Schematic diagram of effective cancer suppression process through combination therapy of BC-LI-0186 and trametinib (created in BioRendor.com). mTOR, mammalian target of rapamycin; PI3K, phosphoinositide 3-kinase; Rag, Ras-related GTP-binding protein.


Reference

References

1. Bade BC, Dela Cruz CS. Lung cancer 2020: epidemiology, etiology, and prevention. Clin Chest Med. 2020; 41:1–24.
2. Kim EY, Lee JG, Lee JM, Kim A, Yoo HC, Kim K, et al. Therapeutic effects of the novel Leucyl-tRNA synthetase inhibitor BC-LI-0186 in non-small cell lung cancer. Ther Adv Med Oncol. 2019; 11:1758835919846798.
Article
3. Mamdani H, Matosevic S, Khalid AB, Durm G, Jalal SI. Immunotherapy in lung cancer: current landscape and future directions. Front Immunol. 2022; 13:823618.
Article
4. Alamgeer M, Ganju V, Watkins DN. Novel therapeutic targets in non-small cell lung cancer. Curr Opin Pharmacol. 2013; 13:394–401.
Article
5. Savas P, Hughes B, Solomon B. Targeted therapy in lung cancer: IPASS and beyond, keeping abreast of the explosion of targeted therapies for lung cancer. J Thorac Dis. 2013; 5 Suppl 5:S579–92.
6. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144:646–74.
Article
7. Xie J, Wang X, Proud CG. mTOR inhibitors in cancer therapy. F1000Res. 2016; 5:2078.
Article
8. Tian T, Li X, Zhang J. mTOR signaling in cancer and mTOR inhibitors in solid tumor targeting therapy. Int J Mol Sci. 2019; 20:755.
Article
9. Kim JH, Lee C, Lee M, Wang H, Kim K, Park SJ, et al. Control of leucine-dependent mTORC1 pathway through chemical intervention of leucyl-tRNA synthetase and RagD interaction. Nat Commun. 2017; 8:732.
Article
10. Han JM, Jeong SJ, Park MC, Kim G, Kwon NH, Kim HK, et al. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell. 2012; 149:410–24.
Article
11. Odogwu L, Mathieu L, Blumenthal G, Larkins E, Goldberg KB, Griffin N, et al. FDA approval summary: dabrafenib and trametinib for the treatment of metastatic non-small cell lung cancers harboring BRAF V600E mutations. Oncologist. 2018; 23:740–5.
12. Roman M, Baraibar I, Lopez I, Nadal E, Rolfo C, Vicent S, et al. KRAS oncogene in non-small cell lung cancer: clinical perspectives on the treatment of an old target. Mol Cancer. 2018; 17:33.
Article
13. Euhus DM, Hudd C, LaRegina MC, Johnson FE. Tumor measurement in the nude mouse. J Surg Oncol. 1986; 31:229–34.
14. Tomayko MM, Reynolds CP. Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol. 1989; 24:148–54.
Article
15. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010; 70:440–6.
Article
16. Sanaei MJ, Razi S, Pourbagheri-Sigaroodi A, Bashash D. The PI3K/Akt/mTOR pathway in lung cancer; oncogenic alterations, therapeutic opportunities, challenges, and a glance at the application of nanoparticles. Transl Oncol. 2022; 18:101364.
Article
17. Cheng H, Shcherba M, Pendurti G, Liang Y, Piperdi B, Perez-Soler R. Targeting the PI3K/AKT/mTOR pathway: potential for lung cancer treatment. Lung Cancer Manag. 2014; 3:67–75.
Article
18. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 2007; 1773:1263–84.
19. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008; 14:1351–6.
Article
20. Sos ML, Fischer S, Ullrich R, Peifer M, Heuckmann JM, Koker M, et al. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer. Proc Natl Acad Sci U S A. 2009; 106:18351–6.
Article
21. Grilley-Olson JE, Bedard PL, Fasolo A, Cornfeld M, Cartee L, Razak AR, et al. A phase Ib dose-escalation study of the MEK inhibitor trametinib in combination with the PI3K/mTOR inhibitor GSK2126458 in patients with advanced solid tumors. Invest New Drugs. 2016; 34:740–9.
22. Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J, et al. Dual PI3K/mTOR inhibitors induce rapid overactivation of the MEK/ERK pathway in human pancreatic cancer cells through suppression of mTORC2. Mol Cancer Ther. 2015; 14:1014–23.
Article
23. Lawal B, Lo WC, Mokgautsi N, Sumitra MR, Khedkar H, Wu AT, et al. Erratum: A preclinical report of a cobimetinib-inspired novel anticancer small-molecule scaffold of isoflavones, NSC777213, for targeting PI3K/AKT/mTOR/MEK in multiple cancers. Am J Cancer Res. 2021; 11:5761.
24. Blumenschein GR Jr, Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)dagger. Ann Oncol. 2015; 26:894–901.
25. Ekman S, Wynes MW, Hirsch FR. The mTOR pathway in lung cancer and implications for therapy and biomarker analysis. J Thorac Oncol. 2012; 7:947–53.
26. Paver E, O’Toole S, Cheng XM, Mahar A, Cooper WA. Updates in the molecular pathology of non-small cell lung cancer. Semin Diagn Pathol. 2021; 38:54–61.
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