Pharmacokinetics and safety of revefenacin in subjects with impaired renal or hepatic function

Marie T Borin, Arthur Lo, Chris N Barnes, Srikanth Pendyala, David L Bourdet, Marie T Borin, Arthur Lo, Chris N Barnes, Srikanth Pendyala, David L Bourdet

Abstract

Purpose: Revefenacin, a long-acting muscarinic antagonist for nebulization, has been shown to improve lung function in patients with chronic obstructive pulmonary disease. Here we report pharmacokinetic (PK) and safety results from two multicenter, open-label, single-dose trials evaluating revefenacin in subjects with severe renal impairment (NCT02578082) and moderate hepatic impairment (NCT02581592).

Subjects and methods: The renal impairment trial enrolled subjects with normal renal function and severe renal impairment (estimated glomerular filtration rate <30 mL/min/1.73 m2). The hepatic impairment trial enrolled subjects with normal hepatic function and moderate hepatic impairment (Child-Pugh class B). Subjects received a single 175-µg dose of revefenacin through nebulization. PK plasma samples and urine collections were obtained at multiple time points for 5 days following treatment; all subjects were monitored for adverse events.

Results: In the renal impairment study, the maximum observed plasma revefenacin concentration (Cmax) was up to 2.3-fold higher and area under the concentration-time curve from time 0 to infinity (AUCinf) was up to 2.4-fold higher in subjects with severe renal impairment compared with those with normal renal function. For THRX-195518, the major metabolite of revefenacin, the corresponding changes in Cmax and AUCinf were 1.8- and 2.7-fold higher, respectively. In the hepatic impairment study, revefenacin Cmax and AUCinf were 1.03- and 1.18-fold higher, respectively, in subjects with moderate hepatic impairment compared with those with normal hepatic function. The corresponding changes in THRX-195518 Cmax and AUCinf were 1.5- and 2.8-fold higher, respectively.

Conclusion: Systemic exposure to revefenacin increased modestly in subjects with severe renal impairment but was similar between subjects with moderate hepatic impairment and normal hepatic function. The increase in plasma exposure to THRX-195518 in subjects with severe renal or moderate hepatic impairment is unlikely to be of clinical consequence given its low antimuscarinic potency, low systemic levels after inhaled revefenacin administration, and favorable safety profile.

Keywords: LAMA; chronic obstructive pulmonary disease; kidney disease; liver disease; long-acting muscarinic antagonist; revefenacin.

Conflict of interest statement

MTB is a consultant for Theravance Biopharma US, Inc. AL and DLB are employees of Theravance Biopharma US, Inc. CNB and SP were employees of Theravance Biopharma US, Inc. at the time this study was conducted. The authors report no other conflicts of interest in this work.

© 2019 Borin et al.

Figures

Figure 1
Figure 1
Mean ± SD plasma concentration–time profiles for revefenacin (A) and THRX-195518 (B) in subjects with and without severe renal impairment (n=8 per group). Abbreviations: h, hours; SD, standard deviation.
Figure 2
Figure 2
Comparison of Cmax (A) and AUCinfa (B) for revefenacin and THRX-195518 between cohorts with normal and severely impaired renal function. Notes:aAUCinf values predicted by the population PK model. Box represents the 25th–75th percentile of the data. Whiskers indicate the 1.5x interquartile range. The solid line in the box represents the median and each dot represents an individual subject. Abbreviations: AUCinf, area under the concentration–time curve from time 0 to infinity; Cmax, maximum observed plasma concentration.
Figure 3
Figure 3
Scatter plots of revefenacin Cmax (A) and AUCinfa (B) versus eGFR. Cmax (C) and AUCinfa (D) versus CLcr in the renal impairment study. Notes: Open circles represent individual subject data. aAUCinf values predicted by the population PK model. The line indicates linear least-squares fit; the shaded area indicates 95% CI. Abbreviations: AUCinf, area under the concentration–time curve from time 0 to infinity; CI, confidence interval; CLcr, creatinine clearance; Cmax, maximum observed plasma concentration; eGFR, estimated glomerular filtration rate; PK, pharmacokinetic.
Figure 4
Figure 4
Scatter plots of THRX-195518 Cmax (A) and AUCinfa (B) versus eGFR. Cmax (C) and AUCinfa (D) versus CLcr in the renal impairment study. Notes: Open circles represent individual subject data. aAUCinf values predicted by the population PK model. The line indicates linear least-squares fit; the shaded area indicates 95% CI. Abbreviations: AUCinf, area under the concentration–time curve from time 0 to infinity; CI, confidence interval; CLcr, creatinine clearance; Cmax, maximum observed plasma concentration; eGFR, estimated glomerular filtration rate; PK, pharmacokinetic.
Figure 5
Figure 5
Mean ± SD plasma concentration-time profiles for revefenacin (A) and THRX-195518 (B) in subjects with and without moderate hepatic impairment (n=8 per group). Abbreviations: h, hours; SD, standard deviation.
Figure 6
Figure 6
Comparison of Cmax (A) and AUCinfa (B) for revefenacin and THRX-195518 between cohorts with normal and moderately impaired hepatic function. Notes: The box represents the 25th–75th percentile of the data. Whiskers indicate the 1.5x interquartile range. The solid line in the box represents the median, while each dot represents an individual subject. aAUCinf values predicted by the population PK model. Abbreviations: AUCinf, area under the concentration–time curve from time 0 to infinity; Cmax, maximum observed plasma concentration; PK, pharmacokinetic.

References

    1. Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Pulmonary Disease; (2018 report) Available from: . Accessed May17, 2018.
    1. Mejza F, Gnatiuc L, Buist AS, et al. Prevalence and burden of chronic bronchitis symptoms: results from the BOLD study. Eur Respir J. 2017;50(5):1700621. doi:10.1183/13993003.00711-2017
    1. Naud J, Nolin TD, Leblond FA, Pichette V. Current understanding of drug disposition in kidney disease. J Clin Pharmacol. 2012;52(1 Suppl):10s–22s. doi:10.1177/0091270011413588
    1. Bonini M, Usmani OS. The importance of inhaler devices in the treatment of COPD. COPD Res Pract. 2015;1(1):9. doi:10.1186/s40749-015-0011-0
    1. Dhand R, Dolovich M, Chipps B, Myers TR, Restrepo R, Farrar JR. The role of nebulized therapy in the management of COPD: evidence and recommendations. COPD. 2012;9(1):58–72. doi:10.3109/15412555.2011.630047
    1. Tashkin DP. A review of nebulized drug delivery in COPD. Int J Chron Obstruct Pulmon Dis. 2016;11:2585–2596. doi:10.2147/COPD.S114034
    1. Pudi KK, Barnes CN, Moran EJ, Haumann B, Kerwin E. A 28-day, randomized, double-blind, placebo-controlled, parallel group study of nebulized revefenacin in patients with chronic obstructive pulmonary disease. Respir Res. 2017;18(1):182. doi:10.1186/s12931-017-0647-1
    1. Quinn D, Barnes CN, Yates W, et al. Pharmacodynamics, pharmacokinetics and safety of revefenacin (TD-4208), a long-acting muscarinic antagonist, in patients with chronic obstructive pulmonary disease (COPD): results of two randomized, double-blind, phase 2 studies. Pulm Pharmacol Ther. 2018;48:71–79. doi:10.1016/j.pupt.2017.10.003
    1. Ferguson GT, Feldman G, Pudi KK, et al. Improvements in lung function with nebulized revefenacin in the treatment of patients with moderate to very severe COPD: results from two replicate phase III clinical trials. Chronic Obstr Pulm Dis. 2019;6:154–165. doi:10.15326/jcopdf.6.2.2018.0152
    1. Donohue JF, Kerwin E, Sethi S, et al. Revefenacin, a once-daily, lung-selective, long-acting muscarinic antagonist for nebulized therapy: safety and tolerability results of a 52-week phase 3 trial in moderate to very severe chronic obstructive pulmonary disease. Respir Med. 2019;153:38–43.
    1. YUPELRI (revefenacin) Inhalation Solution, for Oral Inhalation [prescribing Information]. Morgantown, WV: Mylan Speciality L.P.; 2018. Available from: . Accessed November 16, 2018.
    1. Bourdet D, Yeola S, Colson P-J, Pendyala S, Barnes C, Borin M. Absorption, distribution, metabolism, and excretion of revefenacin, a long-acting muscarinic antagonist, in healthy male subjects. Presented at: Annual American Thoracic Society International Conference; May 18–23; 2018; San Diego, CA.
    1. Dreisbach AW. The influence of chronic renal failure on drug metabolism and transport. Clin Pharmacol Ther. 2009;86(5):553–556. doi:10.1038/clpt.2009.163
    1. Zhang Y, Zhang L, Abraham S, et al. Assessment of the impact of renal impairment on systemic exposure of new molecular entities: evaluation of recent new drug applications. Clin Pharmacol Ther. 2009;85(3):305–311. doi:10.1038/clpt.2008.208
    1. Lin S, Smith BS. Drug dosing considerations for the critically ill patient with liver disease. Crit Care Nurs Clin North Am. 2010;22(3):335–340. doi:10.1016/j.ccell.2010.04.006
    1. Palatini P, De Martin S. Pharmacokinetic drug interactions in liver disease: an update. World J Gastroenterol. 2016;22(3):1260–1278. doi:10.3748/wjg.v22.i37.8314
    1. Verbeeck RK, Horsmans Y. Effect of hepatic insufficiency on pharmacokinetics and drug dosing. Pharm World Sci. 1998;20(5):183–192. doi:10.1023/A:1008656930082
    1. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). Integrated addendum to ICH E6(R1): Guideline for Good Clinical Practice. E6 (R2); June 11, 2015. Available from: . Accessed July20, 2017.
    1. World Medical Association. World medical association declaration of helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191–2194. doi:10.1001/jama.2013.281053
    1. Lo A, Borin MT, Bourdet DL. Sequential modeling of the population pharmacokinetics of revefenacin and its major metabolite in COPD patients. Presented at: American Conference on Pharmacometrics; October 14–19, 2018; San Diego, CA.
    1. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH Harmonised Tripartite Guideline: Guideline for Good Clinical Practice E6(R1); June 10, 1996. Available from: . Accessed October5, 2018.
    1. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Guidance for Industry: Pharmacokinetics in Patients with Impaired Renal Function — Study Design, Data Analysis, and Impact on Dosing and Labeling; March, 2010. Available from: . Accessed December5, 2018.
    1. Bjermer L, Kuna P, Jorup C, Bengtsson T, Rosenborg J. Clinical pharmacokinetics of AZD3199, an inhaled ultra-long-acting beta2-adrenoreceptor agonist (uLABA). Drug Des Devel Ther. 2015;9:753–762. doi:10.2147/DDDT.S66049
    1. Borghardt JM, Weber B, Staab A, Kunz C, Kloft C. Model-based evaluation of pulmonary pharmacokinetics in asthmatic and COPD patients after oral olodaterol inhalation. Br J Clin Pharmacol. 2016;82(3):739–753. doi:10.1111/bcp.12975
    1. Brutsche MH, Brutsche IC, Munawar M, et al. Comparison of pharmacokinetics and systemic effects of inhaled fluticasone propionate in patients with asthma and healthy volunteers: a randomised crossover study. Lancet. 2000;356(9229):556–561. doi:10.1016/S0140-6736(00)02581-2
    1. Dalby C, Polanowski T, Larsson T, Borgström L, Edsbäcker S, Harrison TW. The bioavailability and airway clearance of the steroid component of budesonide/formoterol and salmeterol/fluticasone after inhaled administration in patients with COPD and healthy subjects: a randomized controlled trial. Respir Res. 2009;10:104. doi:10.1186/1465-9921-10-31
    1. Ferguson G, Pudi KK, Feldmann G, et al. Efficacy and safety of revefenacin (REV), a long-acting muscarinic antagonist for nebulization: results of replicate randomized, double-blind, placebo-controlled, parallel-group phase 3 trials in participants with moderate to very severe COPD. Eur Respir J. 2017;50(Suppl 61):PA1812. doi:10.1183/13993003.00711-2017
    1. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Guidance for Industry: Pharmacokinetics in Patients with Impaired Hepatic Function — Study Design, Data Analysis, and Impact on Dosing and Labeling; May, 2003. . Accessed July26, 2019.
    1. European Medicines Agency. Guideline on the evaluation of the pharmacokinetics of medicinal products in patients with decreased renal function; 2014; . Accessed July26, 2019.
    1. European Medicines Agency. Guideline on the Evaluation of the Pharmacokinetics of Medicinal Products in Patients with Impaired Hepatic Function; 2005; . Accessed July26, 2019.

Source: PubMed

3
Iratkozz fel