The effect of rifampin on the pharmacokinetics of famitinib in healthy subjects

Ting Li, Xin Li, Xin Jiang, Chenjing Wang, Feifei Sun, Yanping Liu, Pingping Lin, Ping Shi, Yao Fu, Xiaomeng Gao, Yanyan Zhang, Yu Cao, Ting Li, Xin Li, Xin Jiang, Chenjing Wang, Feifei Sun, Yanping Liu, Pingping Lin, Ping Shi, Yao Fu, Xiaomeng Gao, Yanyan Zhang, Yu Cao

Abstract

Background: Famitinib is an oral, small-molecule, multi-targeted tyrosine kinase inhibitor under clinical investigation for the treatment of solid tumors. As famitinib is metabolized mainly by cytochrome P450 3A4 (CYP3A4), the study was conducted to investigate the effect of potent CYP3A4 inducer rifampin on the pharmacokinetics of famitinb.

Methods: This single-center, single-arm and fixed-sequence drug-drug interaction study enrolled 21healthy Chinese male subjects. Subjects received a single oral dose of famitinib 25 mg on days 1 and 16 and repeated administration of oral rifampin 600 mg once daily on days 10-23. Blood samples were collected and plasma concentrations of famitinib were measured by validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. Pharmacokinetic parameters were calculated using noncompartmental analysis and safety was assessed.

Results: In the presence of rifampin, the famitinib geometric mean maximum plasma concentration (Cmax) and area under the plasma concentration-time curve from time zero to infinity (AUC0-∞) decreased by 48% and 69%, respectively, and the mean elimination half-life was shortened from 33.9 to 18.2 h. The geometric mean ratio (GMR) of famitinib Cmax and AUC0-∞ and their 90% CI were 0.52 (0.50, 0.54) and 0.31 (0.29, 0.33). Single dose of famitinib 25 mg was well tolerated and eight subjects (38.1%) reported treatment emergent adverse events, which were all grade 1-2 in severity.

Conclusion: Co-administration of rifampin considerably reduces plasma concentration of famitinb due to CYP3A4 induction. Concomitant administration of famitinib and strong CYP3A4 inducers should be avoided, whereas when simultaneous use with inducers of CYP3A4, dose adjustment of famitinb is recommended.

Clinical trial registration number: NCT04494659 (July 31, 2020).

Keywords: CYP3A4; Drug-drug interaction; Famitinib; Pharmacokinetics; Rifampin.

Conflict of interest statement

Yanyan Zhang is an employee of Jiangsu Hengrui Pharmaceuticals Co. Ltd. The other authors report no conflicts of interest.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
The mean (± SD) plasma concentration–time curves of famitinib after a single oral administration of 25 mg famitinib in the presence and absence of rifampin (A linear and B semi-logarithmic scale)

References

    1. Huang L, Jiang S, Shi Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020) J Hematol Oncol. 2020;13(1):143. doi: 10.1186/s13045-020-00977-0.
    1. Ge S, et al. Famitinib exerted powerful antitumor activity in human gastric cancer cells and xenografts. Oncol Lett. 2016;12(3):1763–1768. doi: 10.3892/ol.2016.4909.
    1. Zhang M, et al. Third-generation EGFR inhibitor HS-10296 in combination with famitinib, a multi-targeted tyrosine kinase inhibitor, exerts synergistic antitumor effects through enhanced inhibition of downstream signaling in EGFR-mutant non-small cell lung cancer cells. Thorac Cancer. 2021;12(8):1210–1218. doi: 10.1111/1759-7714.13902.
    1. Qu YY, et al. Camrelizumab plus famitinib in patients with advanced or metastatic renal cell carcinoma: data from an Open-label, Multicenter Phase II Basket Study. Clin Cancer Res. 2021;27(21):5838–5846. doi: 10.1158/1078-0432.CCR-21-1698.
    1. Chen Q, et al. Famitinib in combination with concurrent chemoradiotherapy in patients with locoregionally advanced nasopharyngeal carcinoma: a phase 1, open-label, dose-escalation Study. Cancer Commun (Lond) 2018;38(1):66. doi: 10.1186/s40880-018-0330-z.
    1. Xu RH, et al. Famitinib versus placebo in the treatment of refractory metastatic colorectal cancer: a multicenter, randomized, double-blinded, placebo-controlled, phase II clinical trial. Chin J Cancer. 2017;36(1):97. doi: 10.1186/s40880-017-0263-y.
    1. Cao J, et al. Hypothyroidism as a potential biomarker of efficacy of famitinib, a novel VEGFR-2 inhibitor in metastatic breast cancer. Cancer Chemother Pharmacol. 2014;74(2):389–398. doi: 10.1007/s00280-014-2505-x.
    1. Zhou A, et al. Phase I study of the safety, pharmacokinetics and antitumor activity of famitinib. Cancer Chemother Pharmacol. 2013;72(5):1043–1053. doi: 10.1007/s00280-013-2282-y.
    1. Xie C, et al. Metabolism and bioactivation of famitinib, a novel inhibitor of receptor tyrosine kinase, in cancer patients. Br J Pharmacol. 2013;168(7):1687–1706. doi: 10.1111/bph.12047.
    1. Niemi M, et al. Pharmacokinetic interactions with rifampicin—clinical relevance. Clin Pharmacokinet. 2003;42(9):819–850. doi: 10.2165/00003088-200342090-00003.
    1. Teng R, Mitchell P, Butler K. Effect of rifampicin on the pharmacokinetics and pharmacodynamics of ticagrelor in healthy subjects. Eur J Clin Pharmacol. 2013;69(4):877–883. doi: 10.1007/s00228-012-1436-x.
    1. Huang SM, et al. Drug interaction studies: Study design, data analysis, and implications for dosing and labeling. Clin Pharmacol Ther. 2007;81(2):298–304. doi: 10.1038/sj.clpt.6100054.
    1. Gorski JC, et al. The effect of age, sex, and rifampin administration on intestinal and hepatic cytochrome P450 3A activity. Clin Pharmacol Ther. 2003;74(3):275–287. doi: 10.1016/S0009-9236(03)00187-5.
    1. Cotreau MM, von Moltke LL, Greenblatt DJ. The influence of age and sex on the clearance of cytochrome P450 3A substrates. Clin Pharmacokinet. 2005;44(1):33–60. doi: 10.2165/00003088-200544010-00002.
    1. Liu S, et al. LC-MS/MS method for simultaneous determination of famitinib and its major metabolites in human plasma. Bioanalysis. 2018;10(11):791–801. doi: 10.4155/bio-2017-0258.
    1. de Bruijn P, et al. Bioanalytical method for the quantification of sunitinib and its n-desethyl metabolite SU12662 in human plasma by ultra performance liquid chromatography/tandem triple-quadrupole mass spectrometry. J Pharm Biomed Anal. 2010;51(4):934–941. doi: 10.1016/j.jpba.2009.10.020.
    1. Lin JJ, et al. Mechanisms of resistance to selective RET tyrosine kinase inhibitors in RET fusion-positive non-small-cell lung cancer. Ann Oncol. 2020;31(12):1725–1733. doi: 10.1016/j.annonc.2020.09.015.
    1. Reitman ML, et al. Rifampin's acute inhibitory and chronic inductive drug interactions: experimental and model-based approaches to drug-drug interaction trial design. Clin Pharmacol Ther. 2011;89(2):234–242. doi: 10.1038/clpt.2010.271.
    1. Goodman VL, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007;13(5):1367–1373. doi: 10.1158/1078-0432.CCR-06-2328.
    1. Bello C, et al. Effect ofriFampin on the pharmacokinetics of SU11248 in healthy volunteers. J Clin Oncol. 2005;23:3078. doi: 10.1200/jco.2005.23.16_suppl.3078.
    1. Bolton AE, et al. Effect of rifampicin on the pharmacokinetics of imatinib mesylate (Gleevec, STI571) in healthy subjects. Cancer Chemother Pharmacol. 2004;53(2):102–106. doi: 10.1007/s00280-003-0722-9.
    1. Swaisland HC, et al. Pharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol. Clin Pharmacokinet. 2005;44(10):1067–1081. doi: 10.2165/00003088-200544100-00005.
    1. Hamilton M, et al. The effect of rifampicin, a prototypical CYP3A4 inducer, on erlotinib pharmacokinetics in healthy subjects. Cancer Chemother Pharmacol. 2014;73(3):613–621. doi: 10.1007/s00280-014-2390-3.
    1. Wind S, et al. Pharmacokinetic drug interactions of afatinib with rifampicin and ritonavir. Clin Drug Investig. 2014;34(3):173–182. doi: 10.1007/s40261-013-0161-2.
    1. Adams VR, Leggas M. Sunitinib malate for the treatment of metastatic renal cell carcinoma and gastrointestinal stromal tumors. Clin Ther. 2007;29(7):1338–1353. doi: 10.1016/j.clinthera.2007.07.022.

Source: PubMed

3
Předplatit