Population Pharmacokinetic and Exposure-Response Analysis of Finerenone: Insights Based on Phase IIb Data and Simulations to Support Dose Selection for Pivotal Trials in Type 2 Diabetes with Chronic Kidney Disease

Nelleke Snelder, Roland Heinig, Henk-Jan Drenth, Amer Joseph, Peter Kolkhof, Jörg Lippert, Dirk Garmann, Bart Ploeger, Thomas Eissing, Nelleke Snelder, Roland Heinig, Henk-Jan Drenth, Amer Joseph, Peter Kolkhof, Jörg Lippert, Dirk Garmann, Bart Ploeger, Thomas Eissing

Abstract

Background: Finerenone (BAY 94-8862) is a potent non-steroidal, selective mineralocorticoid receptor antagonist being developed for the treatment of patients with type 2 diabetes and chronic kidney disease.

Methods: We present the population pharmacokinetics and pharmacodynamics (PD) analysis for efficacy and safety markers based on data from two clinical phase IIb studies: ARTS-DN (NCT01874431) and ARTS-DN Japan (NCT01968668).

Results: The pharmacokinetics of finerenone were adequately characterized, with estimated glomerular filtration rate (eGFR) and body weight as influencing covariates. The area under the plasma concentration-time curve in Japanese patients did not differ from that in the global population, and the investigated pharmacokinetics were dose- and time-linear. In addition, the pharmacokinetic model provided robust individual exposure estimates to study exposure-response. The concentration-effect relationship over time for the efficacy marker urinary albumin:creatinine ratio (UACR) was well-characterized by a maximum effect model indicating saturation at high exposures. For the safety markers, a log-linear model and a power model were identified for serum potassium concentration and eGFR, respectively, indicating attenuation of effect gains at high exposures. There was no apparent ethnic effect on the investigated pharmacokinetic-pharmacodynamic relationships. The model-predicted times to reach the full (99%) steady-state drug effect on UACR, serum potassium, and eGFR were 138, 20, and 85 days, respectively, while the pharmacokinetic half-life was 2-3 h and steady state was achieved after 2 days, indicating timescale separation.

Conclusion: Our dose-exposure-response modeling and simulation indicates effects were largely saturated at finerenone 20 mg and doses of both 10 and 20 mg once daily appear safe and efficacious at reducing albuminuria.

Conflict of interest statement

NS and HJD were paid consultants for Bayer AG during the conduct of the analysis. RH, AJ, PK, JL, DG, BP, and TE are employed by Bayer AG. RH, AJ, JL, DG, BP, and TE are Bayer AG shareholders.

Figures

Fig. 1
Fig. 1
Box plot of dose-normalized a AUCss at steady state and bCmax,ss (day 30) of once-daily administration for the ARTS-DN and ARTS-DN Japan populations. The median dose-normalized AUCss and Cmax,ss values were 26.6 and 26.7 µg·h/L/mg and 6.8 and 7.3 µg/L/mg, for the ARTS-DN and ARTS-DN Japan studies, respectively. AUCss area under the plasma concentration–time curve at steady state, Cmax SS maximum concentration at steady state
Fig. 2
Fig. 2
Predicted and observed a UACR, b absolute serum potassium concentration, and c relative change from baseline in eGFR-EPI versus finerenone AUCss. The model was fitted to individual data. For plotting purposes, the data were binned in 20 categories based on equal numbers of records. Blue dashed lines: reference/threshold lines; dark gray dashed lines: simulated AUCss for a typical subject for doses of 10, 20, and 30 mg; black solid lines: 5th and 95th percentiles of the observations; black dots: medians of the observations; red solid line and dashed lines: median predictions and 5th and 95th percentiles of the predictions; gray areas: 90% confidence intervals of the median and 5th and 95th percentiles. AUCss area under the plasma concentration–time curve at steady state, eGFR-EPI estimated glomerular filtration rate according to the Chronic Kidney Disease Epidemiology Collaboration equation, UACR urinary albumin:creatinine ratio
Fig. 3
Fig. 3
Simulated dose–response curves for a UACR, b serum potassium concentration, and c eGFR-EPI for a phase III scenario (Table 3) following once-daily administration of placebo (0 mg) and different doses of finerenone (5.0, 7.5, 10, 15, 20, and 30 mg). Presented are the median predicted percentages of subjects (solid lines) with 90% confidence intervals (shaded areas), and the percentages of subjects reaching the thresholds following finerenone 10, 20, and 30 mg once daily. a Percentage of subjects who reach specified UACR targets at day 180. b Percentage of subjects who have one or more serum potassium concentration value > 5.5 mmol/L at any visit during treatment. c Percentage of subjects who have specified decreases from baseline in eGFR at day 180. eGFR estimated glomerular filtration rate, eGFR-EPI estimated glomerular filtration rate according to the Chronic Kidney Disease Epidemiology Collaboration equation, UACR urinary albumin:creatinine ratio

References

    1. Navaneethan SD, Nigwekar SU, Sehgal AR, Strippoli GF. Aldosterone antagonists for preventing the progression of chronic kidney disease: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2009;4(3):542–551. doi: 10.2215/CJN.04750908.
    1. Barfacker L, Kuhl A, Hillisch A, Grosser R, Figueroa-Perez S, Heckroth H, et al. Discovery of BAY 94-8862: a nonsteroidal antagonist of the mineralocorticoid receptor for the treatment of cardiorenal diseases. ChemMedChem. 2012;7(8):1385–1403. doi: 10.1002/cmdc.201200081.
    1. Kolkhof P, Delbeck M, Kretschmer A, Steinke W, Hartmann E, Barfacker L, et al. Finerenone, a novel selective nonsteroidal mineralocorticoid receptor antagonist protects from rat cardiorenal injury. J Cardiovasc Pharmacol. 2014;64(1):69–78. doi: 10.1097/FJC.0000000000000091.
    1. Pitt B, Kober L, Ponikowski P, Gheorghiade M, Filippatos G, Krum H, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J. 2013;34(31):2453–2463. doi: 10.1093/eurheartj/eht187.
    1. Bakris GL, Agarwal R, Chan JC, Cooper ME, Gansevoort RT, Haller H, et al. Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA. 2015;314(9):884–894. doi: 10.1001/jama.2015.10081.
    1. Katayama S, Yamada D, Nakayama M, Yamada T, Myoishi M, Kato M, et al. A randomized controlled study of finerenone versus placebo in Japanese patients with type 2 diabetes mellitus and diabetic nephropathy. J Diabetes Complic. 2017;31(4):758–765. doi: 10.1016/j.jdiacomp.2016.11.021.
    1. Lentini S, Heinig R, Kimmeskamp-Kirschbaum N, Wensing G. Pharmacokinetics, safety and tolerability of the novel, selective mineralocorticoid receptor antagonist finerenone - results from first-in-man and relative bioavailability studies. Fundam Clin Pharmacol. 2016;30(2):172–184. doi: 10.1111/fcp.12170.
    1. Heinig R, Kimmeskamp-Kirschbaum N, Halabi A, Lentini S. Pharmacokinetics of the novel nonsteroidal mineralocorticoid receptor antagonist finerenone (BAY 94-8862) in individuals with renal impairment. Clin Pharmacol Drug Dev. 2016;5(6):488–501. doi: 10.1002/cpdd.263.
    1. Heinig R, Lambelet M, Nagelschmitz J, Alatrach A, Halabi A. Pharmacokinetics of the novel nonsteroidal mineralocorticoid receptor antagonist finerenone (BAY 94-8862) in individuals with mild or moderate hepatic impairment. Eur J Drug Metab Pharmacokinet. 2019 doi: 10.1007/s13318-019-00547-x.
    1. Gerisch M, Heinig R, Engelen A, Lang D, Kolkhof P, Radtke M, et al. Biotransformation of finerenone, a novel nonsteroidal mineralocorticoid receptor antagonist, in dogs, rats, and humans, in vivo and in vitro. Drug Metab Dispos. 2018;46(11):1546–1555. doi: 10.1124/dmd.118.083337.
    1. Heinig R, Gerisch M, Engelen A, Nagelschmitz J, Loewen S. Pharmacokinetics of the Novel, selective, non-steroidal mineralocorticoid receptor antagonist finerenone in healthy volunteers: results from an absolute bioavailability study and drug-drug interaction studies in vitro and in vivo. Eur J Drug Metab Pharmacokinet. 2018;43(6):715–727. doi: 10.1007/s13318-018-0483-9.
    1. Heerspink HJ, Kropelin TF, Hoekman J, de Zeeuw D, Reducing Albuminuria as Surrogate Endpoint (REASSURE) Consortium Drug-induced reduction in albuminuria is associated with subsequent renoprotection: a meta-analysis. J Am Soc Nephrol. 2015;26(8):2055–2064. doi: 10.1681/ASN.2014070688.
    1. Carrero JJ, Grams ME, Sang Y, Arnlov J, Gasparini A, Matsushita K, et al. Albuminuria changes are associated with subsequent risk of end-stage renal disease and mortality. Kidney Int. 2017;91(1):244–251. doi: 10.1016/j.kint.2016.09.037.
    1. Coresh J, Heerspink HJL, Sang Y, Matsushita K, Arnlov J, Astor BC, et al. Change in albuminuria and subsequent risk of end-stage kidney disease: an individual participant-level consortium meta-analysis of observational studies. Lancet Diabetes Endocrinol. 2019;7(2):115–127. doi: 10.1016/S2213-8587(18)30313-9.
    1. Heerspink HJL, Greene T, Tighiouart H, Gansevoort RT, Coresh J, Simon AL, et al. Change in albuminuria as a surrogate endpoint for progression of kidney disease: a meta-analysis of treatment effects in randomised clinical trials. Lancet Diabetes Endocrinol. 2019;7(2):128–139. doi: 10.1016/S2213-8587(18)30314-0.
    1. Jamsen KM, McLeay SC, Barras MA, Green B. Reporting a population pharmacokinetic-pharmacodynamic study: a journal’s perspective. Clin Pharmacokinet. 2014;53(2):111–122. doi: 10.1007/s40262-013-0114-1.
    1. Ruilope LM, Agarwal R, Chan JC, Cooper ME, Gansevoort RT, Haller H, et al. Rationale, design, and baseline characteristics of ARTS-DN: a randomized study to assess the safety and efficacy of finerenone in patients with type 2 diabetes mellitus and a clinical diagnosis of diabetic nephropathy. Am J Nephrol. 2014;40(6):572–581. doi: 10.1159/000371497.
    1. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the Modification of Diet in Renal Disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247–254. doi: 10.7326/0003-4819-145-4-200608150-00004.
    1. Matsuo S, Imai E, Horio M, Yasuda Y, Tomita K, Nitta K, et al. Revised equations for estimated GFR from serum creatinine in Japan. Am J Kidney Dis. 2009;53(6):982–992. doi: 10.1053/j.ajkd.2008.12.034.
    1. Chudleigh RA, Ollerton RL, Dunseath G, Peter R, Harvey JN, Luzio S, et al. Performance of the revised ‘175’ Modification of Diet in Renal Disease equation in patients with type 2 diabetes. Diabetologia. 2008;51(9):1714–1718. doi: 10.1007/s00125-008-1086-9.
    1. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF, 3rd, Feldman HI, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–612. doi: 10.7326/0003-4819-150-9-200905050-00006.
    1. Horio M, Imai E, Yasuda Y, Watanabe T, Matsuo S. Modification of the CKD Epidemiology Collaboration (CKD-EPI) equation for Japanese: accuracy and use for population estimates. Am J Kidney Dis. 2010;56(1):32–38. doi: 10.1053/j.ajkd.2010.02.344.
    1. Grune J, Beyhoff N, Smeir E, Chudek R, Blumrich A, Ban Z, et al. Selective mineralocorticoid receptor cofactor modulation as molecular basis for finerenone’s antifibrotic activity. Hypertension. 2018;71(4):599–608. doi: 10.1161/HYPERTENSIONAHA.117.10360.
    1. de Zeeuw D, Remuzzi G, Parving HH, Keane WF, Zhang Z, Shahinfar S, et al. Proteinuria, a target for renoprotection in patients with type 2 diabetic nephropathy: lessons from RENAAL. Kidney Int. 2004;65(6):2309–2320. doi: 10.1111/j.1523-1755.2004.00653.x.
    1. de Zeeuw D, Remuzzi G, Parving HH, Keane WF, Zhang Z, Shahinfar S, et al. Albuminuria, a therapeutic target for cardiovascular protection in type 2 diabetic patients with nephropathy. Circulation. 2004;110(8):921–927. doi: 10.1161/01.CIR.0000139860.33974.28.
    1. Weir MR. Acute fall in glomerular filtration rate with renin-angiotensin system inhibition: a biomeasure of therapeutic success? Kidney Int. 2011;80(3):235–237. doi: 10.1038/ki.2011.132.
    1. Holtkamp FA, de Zeeuw D, Thomas MC, Cooper ME, de Graeff PA, Hillege HJ, et al. An acute fall in estimated glomerular filtration rate during treatment with losartan predicts a slower decrease in long-term renal function. Kidney Int. 2011;80(3):282–287. doi: 10.1038/ki.2011.79.

Source: PubMed

Подписаться