FOXO3 mutation predicting gefitinib-induced hepatotoxicity in NSCLC patients through regulation of autophagy

Shaoxing Guan, Xi Chen, Youhao Chen, Guohui Wan, Qibiao Su, Heng Liang, Yunpeng Yang, Wenfeng Fang, Yan Huang, Hongyun Zhao, Wei Zhuang, Shu Liu, Fei Wang, Wei Feng, Xiaoxu Zhang, Min Huang, Xueding Wang, Li Zhang, Shaoxing Guan, Xi Chen, Youhao Chen, Guohui Wan, Qibiao Su, Heng Liang, Yunpeng Yang, Wenfeng Fang, Yan Huang, Hongyun Zhao, Wei Zhuang, Shu Liu, Fei Wang, Wei Feng, Xiaoxu Zhang, Min Huang, Xueding Wang, Li Zhang

Abstract

Hepatotoxicity is a common side effect for patients treated with gefitinib, but the related pathogenesis is unclear and lacks effective predictor and management strategies. A multi-omics approach integrating pharmacometabolomics, pharmacokinetics and pharmacogenomics was employed in non-small cell lung cancer patients to identify the effective predictor for gefitinib-induced hepatotoxicity and explore optional therapy substitution. Here, we found that patients with rs4946935 AA, located in Forkhead Box O3 (FOXO3) which is a well-known autophagic regulator, had a higher risk of hepatotoxicity than those with the GA or GG variant (OR = 18.020, 95%CI = 2.473 to 459.1784, P = 0.018) in a gefitinib-concentration dependent pattern. Furthermore, functional experiments identified that rs4946935_A impaired the expression of FOXO3 by inhibiting the promotor activity, increasing the threshold of autophagy initiation and inhibiting the autophagic activity which contributed to gefitinib-induced liver injury. In contrast, erlotinib-induced liver injury was independent on the variant and expression levels of FOXO3. This study reveals that FOXO3 mutation, leading to autophagic imbalance, plays important role in gefitinib-induced hepatotoxicity, especially for patients with high concentration of gefitinib. In conclusion, FOXO3 mutation is an effective predictor and erlotinib might be an appropriately and well-tolerated treatment option for patients carrying rs4946935 AA.

Keywords: Autophagy; FOXO3; Gefitinib; Hepatotoxicity; Pharmacogenomics; Pharmacokinetics; Pharmacometabolomic.

© 2022 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting by Elsevier B.V.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
The concentration of gefitinib/metabolites was not associated with gefitinib-induced hepatotoxicity in the general subjects (n = 180). (A) The grades of hepatotoxicity induced by gefitinib in NSCLC patients; (B–F) Neither the concentration of gefitinib nor the metabolites was associated with hepatotoxicity in the general subjects (n = 180). ns: no significant.
Figure 2
Figure 2
G>A rs4946935 was associated with gefitinib-induced hepatotoxicity. (A) Among 194 SNPs, only G>A rs4711998 in IL17, C>T rs4795896 in CCL11, G>A rs4946935 in FOXO3 and G>A rs12722604 in IL2RA were associated with gefitinib-induced hepatotoxicity; (B) G>A rs4711998, located in IL17A, was associated with gefitinib-induced hepatotoxicity; (C) C>T rs4795896, located in CCL11, was associated with gefitinib-induced hepatotoxicity; (D) G>A rs4946935, located in FOXO3, was correlated with gefitinib-induced hepatotoxicity; (E) G>A rs1272260, located in IL2RA, was correlated with gefitinib-induced hepatotoxicity; (F) G>A rs4946935, located in FOXO3, was significantly associated with gefitinib-induced hepatotoxicity by multivariate logistic regression; (G) Plasma AST/ALT level was significantly correlated with the concentration of gefitinib in FOXO3 AA carriers. ∗P < 0.05; ∗∗P < 0.01.
Figure 3
Figure 3
rs4946935_A impaired the expression of FOXO3. (A) All tag SNPs of FOXO3 in HCB; (B) G>A rs4946935 was located in intron 3 of FOXO3 on Chromosome 6; (C) FOXO3 rs4946935 was correlated to expression levels of FOXO3 in human spleen and brain caudate according to GTEx database; (D) FOXO3 rs4946935 was correlated to expression levels of FOXO3 in NSCLC patients; (E) Luciferase activity of rs4946935_ A and _G of FOXO3 reporter vectors in LO2 cells; (F–H) Overexpression of FOXO3 significantly increased the IC50 of gefitinib while knockout of FOXO3 decreased in LO2 cells. Data represent mean ± SD of three or more independent experiments; ∗P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001; ns: no significance.
Figure 4
Figure 4
gefitinib-induced hepatotoxicity was FOXO3-dependent by inhibiting autophagy. (A) The expression of FOXO3 was correlated with autophagy-related genes in liver tissue according to GTEx dataset; (B) FOXO3 directly regulates the expression of ATG3, ATG4A, ATG5, ATG7, ATG10, ATG12, ATG14, ATG16L1, and MAP1LC3B after treatment with gefitinib in LO2 cells; (C) Overexpression of FOXO3 decreased the threshold of autophagy initiation as indicated by LC3-II/I expression level; (D) Electron micrographs of FOXO3-knock out and -overexpression LO2 cells under incubation of gefitinib; (E) Inhibition of autophagy significantly suppressed proliferation of FOXO3 overexpression hepatocytes under incubation of gefitinib; (F) Inhibition of autophagy significantly increased the cytotoxicity of gefitinib in FOXO3 overexpression hepatocytes. Data represent mean ± SD of three or more independent experiments; ∗P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001; ns: no significance.

References

    1. Maemondo M., Inoue A., Kobayashi K., Sugawara S., Oizumi S., Isobe H., et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380–2388.
    1. Mitsudomi T., Morita S., Yatabe Y., Negoro S., Okamoto I., Tsurutani J., et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–128.
    1. Takeda M., Okamoto I., Fukuoka M., Nakagawa K. Successful treatment with erlotinib after gefitinib-related severe hepatotoxicity. J Clin Oncol. 2010;28:e273–e274.
    1. Ho C., Davis J., Anderson F., Bebb G., Murray N. Side effects related to cancer treatment: CASE 1. Hepatitis following treatment with gefitinib. J Clin Oncol. 2005;23:8531–8533.
    1. Shi Q., Yang X., Ren L., Mattes W.B. Recent advances in understanding the hepatotoxicity associated with protein kinase inhibitors. Expert Opin Drug Metab Toxicol. 2020;16:217–226.
    1. Lee K.W., Chan S.L. Hepatotoxicity of targeted therapy for cancer. Expert Opin Drug Metab Toxicol. 2016;12:789–802.
    1. Holt M.P., Ju C. Mechanisms of drug-induced liver injury. AAPS J. 2006;8:E48–E54.
    1. Li X., Kamenecka T.M., Cameron M.D. Bioactivation of the epidermal growth factor receptor inhibitor gefitinib: implications for pulmonary and hepatic toxicities. Chem Res Toxicol. 2009;22:1736–1742.
    1. Zhang J., Ren L., Yang X., White M., Greenhaw J., Harris T., et al. Cytotoxicity of 34 FDA approved small-molecule kinase inhibitors in primary rat and human hepatocytes. Toxicol Lett. 2018;291:138–148.
    1. McKillop D., McCormick A.D., Miles G., Phillips P.J., Pickup K.J., Bushby N., et al. In vitro metabolism of gefitinib in human liver microsomes. Xenobiotica. 2004;34:983–1000.
    1. Liu X., Lu Y., Guan X., Dong B., Chavan H., Wang J., et al. Metabolomics reveals the formation of aldehydes and iminium in gefitinib metabolism. Biochem Pharmacol. 2015;97:111–121.
    1. Kawamura T., Imamura C.K., Kenmotsu H., Taira T., Omori S., Nakashima K., et al. Evaluation of gefitinib systemic exposure in EGFR-mutated non-small cell lung cancer patients with gefitinib-induced severe hepatotoxicity. Cancer Chemother Pharmacol. 2020;85:605–614.
    1. Xin S., Zhao Y., Wang X., Huang Y., Zhang J., Guo Y., et al. The dissociation of gefitinib trough concentration and clinical outcome in NSCLC patients with EGFR sensitive mutations. Sci Rep. 2015;5:12675.
    1. Kobayashi H., Sato K., Niioka T., Takeda M., Okuda Y., Asano M., et al. Effects of polymorphisms in CYP2D6 and ABC transporters and side effects induced by gefitinib on the pharmacokinetics of the gefitinib metabolite, O-desmethyl gefitinib. Med Oncol. 2016;33:57.
    1. Takimoto T., Kijima T., Otani Y., Nonen S., Namba Y., Mori M., et al. Polymorphisms of CYP2D6 gene and gefitinib-induced hepatotoxicity. Clin Lung Cancer. 2013;14:502–507.
    1. Sugiyama E., Umemura S., Nomura S., Kirita K., Matsumoto S., Yoh K., et al. Impact of single nucleotide polymorphisms on severe hepatotoxicity induced by EGFR tyrosine kinase inhibitors in patients with non-small cell lung cancer harboring EGFR mutations. Lung Cancer. 2015;90:307–313.
    1. Wang J., Wu Y., Dong M., He X., Wang Z., Li J., et al. Observation of hepatotoxicity during long-term gefitinib administration in patients with non-small-cell lung cancer. Anti Cancer Drugs. 2016;27:245–250.
    1. U.S. Department of Health and Human Services . Ent Vocab Serv; 2009 May 28. Common Terminology Criteria for Adverse Events (CTCAE). Version 4.0. Available from:
    1. Hu X., Yang T., Li C., Zhang L., Li M., Huang W., et al. Human fetal hepatocyte line, L-02, exhibits good liver function in vitro and in an acute liver failure model. Transplant Proc. 2013;45:695–700.
    1. Luo P., Yan H., Du J., Chen X., Shao J., Zhang Y., et al. PLK1 (polo like kinase 1)-dependent autophagy facilitates gefitinib-induced hepatotoxicity by degrading COX6A1 (cytochrome c oxidase subunit 6A1) Autophagy. 2021;17:3221–3237.
    1. Ahmed M.M., Wang T., Luo Y., Ye S., Wu Q., Guo Z., et al. Aldo-keto reductase-7A protects liver cells and tissues from acetaminophen-induced oxidative stress and hepatotoxicity. Hepatology. 2011;54:1322–1332.
    1. McKillop D., Hutchison M., Partridge E.A., Bushby N., Cooper C.M., Clarkson-Jones J.A., et al. Metabolic disposition of gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor, in rat, dog and man. Xenobiotica. 2004;34:917–934.
    1. Guan S., Chen X., Wang F., Xin S., Feng W., Zhu X., et al. Development and validation of a sensitive LC–MS/MS method for determination of gefitinib and its major metabolites in human plasma and its application in non-small cell lung cancer patients. J Pharm Biomed Anal. 2019;172:364–371.
    1. Xin S., Zhao Y., Wang C., Huang Y., Zhuang W., Ma Y., et al. Polymorphisms of NF-κB pathway genes influence adverse drug reactions of gefitinib in NSCLC patients. Pharmacogenomics J. 2020;20:285–293.
    1. Kijima T., Shimizu T., Nonen S., Furukawa M., Otani Y., Minami T., et al. Safe and successful treatment with erlotinib after gefitinib-induced hepatotoxicity: difference in metabolism as a possible mechanism. J Clin Oncol. 2011;29:e588–e590.
    1. Kobayashi H., Sato K., Niioka T., Miura H., Ito H., Miura M. Relationship among gefitinib exposure, polymorphisms of its metabolizing enzymes and transporters, and side effects in Japanese patients with non-small-cell lung cancer. Clin Lung Cancer. 2015;16:274–281.
    1. Ma Y., Xin S., Huang M., Yang Y., Zhu C., Zhao H., et al. Determinants of gefitinib toxicity in advanced non-small cell lung cancer (NSCLC): a pharmacogenomic study of metabolic enzymes and transporters. Pharmacogenomics J. 2017;17:325–330.
    1. Seki N., Uematsu K., Shibakuki R., Eguchi K. Promising new treatment schedule for gefitinib responders after severe hepatotoxicity with daily administration. J Clin Oncol. 2006;24:3213–3214.
    1. Wandrer F., Frangez Z., Liebig S., John K., Vondran F., Wedemeyer H., et al. Autophagy alleviates amiodarone-induced hepatotoxicity. Arch Toxicol. 2020;94:3527–3539.
    1. Wang L., Wei W., Xiao Q., Yang H., Ci X. Farrerol ameliorates APAP-induced hepatotoxicity via activation of Nrf2 and autophagy. Int J Biol Sci. 2019;15:788–799.
    1. Zheng X.Y., Yang S.M., Zhang R., Wang S.M., Li G.B., Zhou S.W. Emodin-induced autophagy against cell apoptosis through the PI3K/AKT/mTOR pathway in human hepatocytes. Drug Des Devel Ther. 2019;13:3171–3180.
    1. Ni H.M., Bockus A., Boggess N., Jaeschke H., Ding W.X. Activation of autophagy protects against acetaminophen-induced hepatotoxicity. Hepatology. 2012;55:222–232.
    1. Ni H.M., McGill M.R., Chao X., Du K., Williams J.A., Xie Y., et al. Removal of acetaminophen protein adducts by autophagy protects against acetaminophen-induced liver injury in mice. J Hepatol. 2016;65:354–362.
    1. Chen Z.H., Wu Y.F., Wang P.L., Wu Y.P., Li Z.Y., Zhao Y., et al. Autophagy is essential for ultrafine particle-induced inflammation and mucus hyperproduction in airway epithelium. Autophagy. 2016;12:297–311.
    1. Shan S., Shen Z., Song F. Autophagy and acetaminophen-induced hepatotoxicity. Arch Toxicol. 2018;92:2153–2161.
    1. Mammucari C., Milan G., Romanello V., Masiero E., Rudolf R., Del Piccolo P., et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471.
    1. Pi H., Li M., Zou L., Yang M., Deng P., Fan T., et al. AKT inhibition-mediated dephosphorylation of TFE3 promotes overactive autophagy independent of MTORC1 in cadmium-exposed bone mesenchymal stem cells. Autophagy. 2019;15:565–582.
    1. Liu Y.M., Lv J., Zeng Q.L., Shen S., Xing J.Y., Zhang Y.Y., et al. AMPK activation ameliorates d-GalN/LPS-induced acute liver failure by upregulating Foxo3A to induce autophagy. Exp Cell Res. 2017;358:335–342.
    1. Manley S., Ding W. Role of farnesoid X receptor and bile acids in alcoholic liver disease. Acta Pharm Sin B. 2015;5:158–167.
    1. Zhong C., Pu L.Y., Fang M.M., Gu Z., Rao J.H., Wang X.H. Retinoic acid receptor α promotes autophagy to alleviate liver ischemia and reperfusion injury. World J Gastroenterol. 2015;21:12381–12391.
    1. Ni H.M., Du K., You M., Ding W.X. Critical role of FoxO3a in alcohol-induced autophagy and hepatotoxicity. Am J Pathol. 2013;183:1815–1825.
    1. Tsapras P., Nezis I.P. Caspase involvement in autophagy. Cell Death Differ. 2017;24:1369–1379.
    1. Fitzwalter B.E., Thorburn A. FOXO3 links autophagy to apoptosis. Autophagy. 2018;14:1467–1468.
    1. Sakata Y., Kawamura K., Shingu N., Hiroshige S., Yasuda Y., Eguchi Y., et al. The effects of switching EGFR-TKI treatments for non-small cell lung cancer because of adverse events. Asia Pac J Clin Oncol. 2020;16:e113–e117.
    1. Li X., Kamenecka T.M., Cameron M.D. Cytochrome P450-mediated bioactivation of the epidermal growth factor receptor inhibitor erlotinib to a reactive electrophile. Drug Metab Dispos. 2010;38:1238–1245.
    1. Duckett D.R., Cameron M.D. Metabolism considerations for kinase inhibitors in cancer treatment. Expert Opin Drug Metab Toxicol. 2010;6:1175–1193.
    1. Tan X., Thapa N., Sun Y., Anderson R.A. A kinase-independent role for EGF receptor in autophagy initiation. Cell. 2015;160:145–160.
    1. Chen X., Yang S., Pan Y., Li X., Ma S. Mitochondrial pathway-mediated apoptosis is associated with erlotinib-induced cytotoxicity in hepatic cells. Oncol Lett. 2018;15:783–788.

Source: PubMed

3
Prenumerera