DOI QR코드

DOI QR Code

Synergistic antitumor activity of sorafenib and MG149 in hepatocellular carcinoma cells

  • Moon, Byul (Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST)) ;
  • Park, Mijin (Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST)) ;
  • Cho, Seung-Hyun (Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST)) ;
  • Kim, Kang Mo (Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Seo, Haeng Ran (Advanced Biomedical Research Laboratory, Institut Pasteur Korea) ;
  • Kim, Jeong-Hoon (Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST)) ;
  • Kim, Jung-Ae (Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST))
  • 투고 : 2022.02.23
  • 심사 : 2022.06.09
  • 발행 : 2022.10.31

초록

Advanced hepatocellular carcinoma (HCC) is among the most challenging cancers to overcome, and there is a need for better therapeutic strategies. Among the different cancer drugs that have been used in clinics, sorafenib is considered the standard first-line drug for advanced HCC. Here, to identify a chemical compound displaying a synergistic effect with sorafenib in HCC, we screened a focused chemical library and found that MG149, a histone acetyltransferase inhibitor targeting the MYST family, exhibited the most synergistic anticancer effect with sorafenib on HCC cells. The combination of sorafenib and MG149 exerted a synergistic anti-proliferation effect on HCC cells by inducing apoptotic cell death. We revealed that cotreatment with sorafenib and MG149 aggravated endoplasmic reticulum (ER) stress to promote the death of HCC cells rather than adaptive cell survival. In addition, combined treatment with sorafenib and MG149 significantly increased the intracellular levels of unfolded proteins and reactive oxygen species, which upregulated ER stress. Collectively, these results suggest that MG149 has the potential to improve the efficacy of sorafenib in advanced HCC via the upregulation of cytotoxic ER stress.

키워드

과제정보

This work was supported by a grant (NRF-2019R1A2C10861 5114 to J.-A.K.) from the National Research Foundation, Ministry of Science and ICT, and by the Korea Research Institute of Bioscience and Biotechnology (KRIBB) Research Initiative Program (171134054 to J.-A.K. and 1711170633 to J.-H.K and J.-A.K.).

참고문헌

  1. Llovet JM, Ricci S, Mazzaferro V et al (2008) Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359, 378-390 https://doi.org/10.1056/NEJMoa0708857
  2. Cheng AL, Kang YK, Chen Z et al (2009) Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol 10, 25-34 https://doi.org/10.1016/S1470-2045(08)70285-7
  3. Rimassa L and Worns MA (2020) Navigating the new landscape of second-line treatment in advanced hepatocellular carcinoma. Liver International 40, 1800-1811 https://doi.org/10.1111/liv.14533
  4. Xia S, Pan Y, Liang Y, Xu J and Cai X (2020) The microenvironmental and metabolic aspects of sorafenib resistance in hepatocellular carcinoma. EBioMedicine 51, 102610 https://doi.org/10.1016/j.ebiom.2019.102610
  5. Huang A, Yang XR, Chung WY, Dennison AR and Zhou J (2020) Targeted therapy for hepatocellular carcinoma. Signal Transduct Target Ther 5, 146 https://doi.org/10.1038/s41392-020-00264-x
  6. Rahmani M, Davis EM, Crabtree TR et al (2007) The kinase inhibitor sorafenib induces cell death through a process involving induction of endoplasmic reticulum stress. Mol Cell Biol 27, 5499-5513 https://doi.org/10.1128/MCB.01080-06
  7. Zhou T, Lv X, Guo X et al (2015) RACK1 modulates apoptosis induced by sorafenib in HCC cells by interfering with the IRE1/XBP1 axis. Oncol Rep 33, 3006-3014 https://doi.org/10.3892/or.2015.3920
  8. Won JK, Yu SJ, Hwang CY et al (2017) Protein disulfide isomerase inhibition synergistically enhances the efficacy of sorafenib for hepatocellular carcinoma. Hepatology 66, 855-868 https://doi.org/10.1002/hep.29237
  9. Zhou B, Lu Q, Liu J, Fan L et al (2019) Melatonin increases the sensitivity of hepatocellular carcinoma to sorafenib through the PERK-ATF4-Beclin1 pathway. Int J Biol Sci 15, 1905-1920 https://doi.org/10.7150/ijbs.32550
  10. Hetz C and Papa FR (2018) The unfolded protein response and cell fate control. Mol Cell 69, 169-181 https://doi.org/10.1016/j.molcel.2017.06.017
  11. Shimizu S, Takehara T, Hikita H et al (2012) Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. Int J Cancer 131, 548-557 https://doi.org/10.1002/ijc.26374
  12. Ma MKF, Lau EYT, Leung DHW et al (2017) Stearoyl-CoA desaturase regulates sorafenib resistance via modulation of ER stress-induced differentiation. J Hepatol 67, 979-990 https://doi.org/10.1016/j.jhep.2017.06.015
  13. Tanaka K, Yu HA, Yang S et al (2021) Targeting Aurora B kinase prevents and overcomes resistance to EGFR inhibitors in lung cancer by enhancing BIM- and PUMA-mediated apoptosis. Cancer Cell 39, 1245-1261 e1246 https://doi.org/10.1016/j.ccell.2021.07.006
  14. Harris IS, Endress JE, Coloff JL et al (2019) Deubiquitinases maintain protein homeostasis and survival of cancer cells upon glutathione depletion. Cell Metab 29, 1166-1181 e1166 https://doi.org/10.1016/j.cmet.2019.01.020
  15. Broux M, Prieto C, Demeyer S et al (2019) Suz12 inactivation cooperates with JAK3 mutant signaling in the development of T-cell acute lymphoblastic leukemia. Blood 134, 1323-1336
  16. Niles AL, Moravec RA and Riss TL (2009) In vitro viability and cytotoxicity testing and same-well multi-parametric combinations for high throughput screening. Curr Chem Genomics 3, 33-41 https://doi.org/10.2174/1875397300903010033
  17. Sun Y, Jiang X, Chen S and Price BD (2006) Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation. FEBS Lett 580, 4353-4356 https://doi.org/10.1016/j.febslet.2006.06.092
  18. Ghizzoni M, Wu J, Gao T et al (2012) 6-alkylsalicylates are selective Tip60 inhibitors and target the acetyl-CoA binding site. Eur J Med Chem 47, 337-344 https://doi.org/10.1016/j.ejmech.2011.11.001
  19. Balasubramanyam K, Swaminathan V, Ranganathan A and Kundu TK (2003) Small molecule modulators of histone acetyltransferase p300. J Biol Chem 278, 19134-19140 https://doi.org/10.1074/jbc.M301580200
  20. Kusakabe Y, Chiba T, Oshima M et al (2021) EZH1/2 inhibition augments the anti-tumor effects of sorafenib in hepatocellular carcinoma. Sci Rep 11, 21396 https://doi.org/10.1038/s41598-021-00889-0
  21. Green AS, Maciel TT, Hospital MA et al (2015) Pim kinases modulate resistance to FLT3 tyrosine kinase inhibitors in FLT3-ITD acute myeloid leukemia. Sci Adv 1, e1500221 https://doi.org/10.1126/sciadv.1500221
  22. Yu H, Kim DJ, Choi HY et al (2021) Prospective pharmacological methodology for establishing and evaluating anti-cancer drug resistant cell lines. BMC Cancer 21, 1049 https://doi.org/10.1186/s12885-021-08784-7
  23. Chou TC (2010) Drug combination studies and their synergy quantification using the Chou-Talalay Method. Cancer Res 70, 440-446 https://doi.org/10.1158/0008-5472.CAN-09-1947
  24. Oslowski CM and Urano F (2011) Measuring ER stress and the unfolded protein response using mammalian tissue culture system. Methods Enzymol 490, 71-92 https://doi.org/10.1016/B978-0-12-385114-7.00004-0
  25. Lee CH, Han JH, Kim S et al (2020) Metformin ameliorates bile duct ligation-induced acute hepatic injury via regulation of ER stress. BMB Rep 53, 311-316 https://doi.org/10.5483/BMBRep.2020.53.6.169
  26. Oyadomari S and Mori M (2004) Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 11, 381-389 https://doi.org/10.1038/sj.cdd.4401373
  27. Novoa I, Zeng H, Harding HP and Ron D (2001) Feedback inhibition of the unfolded protein response by GADD34- mediated dephosphorylation of eIF2alpha. J Cell Biol 153, 1011-1022 https://doi.org/10.1083/jcb.153.5.1011
  28. Ogata M, Hino S, Saito A et al (2006) Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 26, 9220-9231 https://doi.org/10.1128/MCB.01453-06
  29. Hetz C, Chevet E and Oakes SA (2015) Proteostasis control by the unfolded protein response. Nat Cell Biol 17, 829-838 https://doi.org/10.1038/ncb3184
  30. Lee S, Jeong Y, Roe JS, Huh H, Paik SH and Song J (2021) Mitochondrial dysfunction induced by callyspongiolide promotes autophagy-dependent cell death. BMB Rep 54, 227-232 https://doi.org/10.5483/BMBRep.2021.54.4.037
  31. Kaizuka T, Morishita H, Hama Y et al (2016) An autophagic flux probe that releases an internal control. Mol Cell 64, 835-849 https://doi.org/10.1016/j.molcel.2016.09.037
  32. Shen D, Coleman J, Chan E et al (2011) Novel cell- and tissue-based assays for detecting misfolded and aggregated protein accumulation within aggresomes and inclusion bodies. Cell Biochem Biophys 60, 173-185 https://doi.org/10.1007/s12013-010-9138-4
  33. Wang C, Yu J, Huo L, Wang L, Feng W and Wang CC (2012) Human protein-disulfide isomerase is a redoxregulated Chaperone activated by oxidation of domain a'. J Biol Chem 287, 1139-1149 https://doi.org/10.1074/jbc.M111.303149
  34. Coriat R, Nicco C, Chereau C et al (2012) Sorafenib-induced hepatocellular carcinoma cell death depends on reactive oxygen species production in vitro and in vivo. Mol Cancer Ther 11, 2284-2293 https://doi.org/10.1158/1535-7163.MCT-12-0093
  35. Song Y, Kim JS, Kim SH et al (2018) Patient-derived multicellular tumor spheroids towards optimized treatment for patients with hepatocellular carcinoma. J Exp Clin Cancer Res 37, 109 https://doi.org/10.1186/s13046-018-0752-0
  36. Corazzari M, Gagliardi M, Fimia GM and Piacentini M (2017) Endoplasmic reticulum stress, unfolded protein response, and cancer cell fate. Front Oncol 7, 78 https://doi.org/10.3389/fonc.2017.00078
  37. Zou Y, Cong YS and Zhou J (2020) Implications of telomerase reverse transcriptase in tumor metastasis. BMB Rep 53, 458-465 https://doi.org/10.5483/BMBRep.2020.53.9.108
  38. Lin A, Giuliano CJ, Palladino A et al (2019) Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci Transl Med 11, eaaw8412 https://doi.org/10.1126/scitranslmed.aaw8412