Evaluation of the Pharmacokinetics and Exposure-Response Relationship of Dapagliflozin in Patients without Diabetes and with Chronic Kidney Disease

Annemarie B van der Aart-van der Beek, Jeroen V Koomen, Claire C J Dekkers, Sean J Barbour, David W Boulton, Ron T Gansevoort, Peter J Greasley, Abdul Halim Abdul Gafor, Gozewijn D Laverman, Qiang Li, Soo Kun Lim, Jasper Stevens, Marc G Vervloet, Sunita Singh, Daniel C Cattran, Heather N Reich, David Z I Cherney, Hiddo J L Heerspink, Annemarie B van der Aart-van der Beek, Jeroen V Koomen, Claire C J Dekkers, Sean J Barbour, David W Boulton, Ron T Gansevoort, Peter J Greasley, Abdul Halim Abdul Gafor, Gozewijn D Laverman, Qiang Li, Soo Kun Lim, Jasper Stevens, Marc G Vervloet, Sunita Singh, Daniel C Cattran, Heather N Reich, David Z I Cherney, Hiddo J L Heerspink

Abstract

Background and objective: Dapagliflozin, a sodium-glucose co-transporter inhibitor, was originally developed as an oral glucose-lowering drug for the treatment of type 2 diabetes mellitus. Emerging data suggest that cardiovascular and kidney benefits extend to patients without diabetes. Limited pharmacological data are, however, available in patients without diabetes. We aimed to characterise the pharmacokinetic profile of dapagliflozin in patients with chronic kidney disease without type 2 diabetes.

Methods: Plasma samples were collected in a randomised, placebo-controlled, double-blind, cross-over trial (DIAMOND, NCT03190694, n = 53) that assessed the effects of 10 mg of dapagliflozin in patients with a glomerular filtration rate ≥ 25 mL/min/1.73 m2 and proteinuria > 500 mg/day. Mixed-effects models were used to develop a pharmacokinetic model and to evaluate the association between plasma exposure and response.

Results: Plasma concentrations (n = 430 observations) from 48 patients (mean age 50.8 years, mean glomerular filtration rate 57.9 mL/min/1.73 m2, median proteinuria 1115 mg/24 h) were best described using a two-compartment model with first-order elimination. Apparent clearance and volume of distribution were 11.7 (95% confidence interval 10.7-12.7) L/h and 44.9 (95% confidence interval 39.0-50.9) L, respectively. Median dapagliflozin plasma exposure was 740.9 ng h/mL (2.5th-97.5th percentiles: 434.0-1615.3). Plasma exposure increased with decreasing kidney function. Every 100-ng h/mL increment in dapagliflozin plasma exposure was associated with a decrease in the urinary albumin:creatinine ratio (β = - 2.8%, p = 0.01), glomerular filtration rate (β = - 0.5 mL/min/1.73 m2, p < 0.01) and systolic blood pressure (β = - 0.4 mmHg, p = 0.03).

Conclusions: The dapagliflozin plasma concentration-time profile in patients with non-diabetic kidney disease appears similar to the profile of patients with diabetic kidney disease described in the literature. Furthermore, the plasma exposure was associated with changes in risk markers for kidney disease.

Conflict of interest statement

Annemarie B. van der Aart-van der Beek, Abdul Halim Abdul Gafor, Claire C.J. Dekkers, Daniel C. Cattran, Jasper Stevens, Jeroen V. Koomen, Qiang Li, Sean J. Barbour and Sunita Singh have no conflicts of interest that are directly relevant to the content of this article. Hiddo J.L. Heerspink is a consultant to Abbvie, AstraZeneca, Boehringer Ingelheim, Bayer, Chinook, CSL Behring, Gilead, Janssen, Merck, Mundipharma, Mitsubishi Tanabe, Novo Nordisk and Retrophin. He received research support from AstraZeneca, Abbvie, Boehringer Ingelheim and Janssen. David W. Boulton is an employee and shareholder of AstraZeneca. David Z.I. Cherney has received honoraria from Boehringer Ingelheim-Lilly, Merck, AstraZeneca, Sanofi, Mitsubishi-Tanabe, Abbvie, Janssen, Bayer, Prometic, BMS and Novo-Nordisk and has received operational funding for clinical trials from Boehringer Ingelheim-Lilly, Merck, Janssen, Sanofi, AstraZeneca and Novo-Nordisk. Ron T. Gansevoort has consulting agreements with AstraZeneca, Bayer, Sanofi-Genzyme and Mundi Pharma; all fees are paid to his institution. Peter J. Greasley is employed by and owns shares in AstraZeneca. Gozewijn D. Laverman has received research grants and consulting fees from Sanofi and AstraZeneca, and research grants from Novo Nordisk. Marc G. Vervloet has received consulting fees from Amgen, Vifor, Fresenius Medical Care Renal Pharma, Medice, Kyowa Kirin and Astra Zeneca. Heather N. Reich has received consulting fees from Omeros and was involved in clinical trials supported by Omeros and Calliditas. Soo Kun Lim has received consulting fees or speaking honoraria from AstraZeneca, Boehringer Ingelheim, Novo Nordisk, Fresenius Kabi, Baxter and Sanofi.

Figures

Fig. 1
Fig. 1
Visual predictive check of the dapagliflozin population pharmacokinetic model. The points represent the observations of dapagliflozin plasma concentrations. Solid and dashed lines represent the observed 10th, 50th and 90th percentiles for all observations and the shaded area represents the 95% confidence interval for the 10th, 50th and 90th percentiles of the model predictions
Fig. 2
Fig. 2
Influence of kidney function and body weight on dapagliflozin plasma exposure. The points represent the estimated plasma exposure (area under the plasma–concentration time curve [AUC0inf]) per individual normalised by the estimated plasma exposure (AUC0inf) of a 70-kg individual (left plot) or normalised by a measured glomerular filtration rate (GFR) of 50 mL/min/1.73 m2 (right plot). The line and shaded area represent the population mean and 95% confidence interval
Fig. 3
Fig. 3
Relationship between exposure and response at week 6. Observations (black circle), reflected as change from baseline, are plotted vs the area under the plasma concentration–time curve (AUC0–inf). The dotted gray lines represent the individual difference between placebo and active treatment vs AUC0–inf. The dotted blue line represents the population difference between placebo and active treatment vs AUC0–inf. GFR glomerular filtration rate

References

    1. List JF, Woo V, Morales E, Tang W, Fiedorek FT. Sodium-glucose cotransport inhibition with dapagliflozin in type 2 diabetes. Diabetes Care. 2009;32:650–657. doi: 10.2337/dc08-1863.
    1. Zhang L, Feng Y, List J, Kasichayanula S, Pfister M. Dapagliflozin treatment in patients with different stages of type 2 diabetes mellitus: effects on glycaemic control and body weight. Diabetes Obes Metab. 2010;12:510–516. doi: 10.1111/j.1463-1326.2010.01216.x.
    1. Heerspink HJ, Perkins BA, Fitchett DH, Husain M, Cherney DZ. Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications. Circulation. 2016;134:752–772. doi: 10.1161/CIRCULATIONAHA.116.021887.
    1. Inzucchi SE, Zinman B, Fitchett D, et al. How does empagliflozin reduce cardiovascular mortality? Insights from a mediation analysis of the EMPA-REG OUTCOME trial. Diabetes Care. 2018;41:356–363. doi: 10.2337/dc17-1096.
    1. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393:31–39. doi: 10.1016/S0140-6736(18)32590-X.
    1. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380:2295–2306. doi: 10.1056/NEJMoa1811744.
    1. Heerspink HJ, Desai M, Jardine M, Balis D, Meininger G, Perkovic V. Canagliflozin slows progression of renal function decline independently of glycemic effects. J Am Soc Nephrol. 2017;28:368–375. doi: 10.1681/ASN.2016030278.
    1. Kasichayanula S, Liu X, Lacreta F, Griffen SC, Boulton DW. Clinical pharmacokinetics and pharmacodynamics of dapagliflozin, a selective inhibitor of sodium-glucose co-transporter type 2. Clin Pharmacokinet. 2014;53:17–27. doi: 10.1007/s40262-013-0104-3.
    1. Devineni D, Curtin CR, Marbury TC, et al. Effect of hepatic or renal impairment on the pharmacokinetics of canagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin Ther. 2015;37(610–28):e4.
    1. Ferrannini E, Veltkamp SA, Smulders RA, Kadokura T. Renal glucose handling: impact of chronic kidney disease and sodium-glucose cotransporter 2 inhibition in patients with type 2 diabetes. Diabetes Care. 2013;36:1260–1265. doi: 10.2337/dc12-1503.
    1. Kasichayanula S, Liu X, Pe Benito M, et al. The influence of kidney function on dapagliflozin exposure, metabolism and pharmacodynamics in healthy subjects and in patients with type 2 diabetes mellitus. Br J Clin Pharmacol. 2013;76:432–444. doi: 10.1111/bcp.12056.
    1. Macha S, Mattheus M, Halabi A, Pinnetti S, Woerle HJ, Broedl UC. Pharmacokinetics, pharmacodynamics and safety of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in subjects with renal impairment. Diabetes Obes Metab. 2014;16:215–222. doi: 10.1111/dom.12182.
    1. Scheen AJ. Pharmacokinetic/pharmacodynamic properties and clinical use of SGLT2 inhibitors in non-Asian and Asian patients with type 2 diabetes and chronic kidney disease. Clin Pharmacokinet. 2020;59:981–994. doi: 10.1007/s40262-020-00885-z.
    1. Petrie MC, Verma S, Docherty KF, et al. Effect of dapagliflozin on worsening heart failure and cardiovascular death in patients with heart failure with and without diabetes. JAMA. 2020;323:1353–1368. doi: 10.1001/jama.2020.1906.
    1. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383:1436–1446. doi: 10.1056/NEJMoa2024816.
    1. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;83:1413–1424. doi: 10.1056/NEJMoa2022190.
    1. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303.
    1. Heerspink HJL, Stefansson BV, Chertow GM, et al. Rationale and protocol of the Dapagliflozin And Prevention of Adverse outcomes in Chronic Kidney Disease (DAPA-CKD) randomized controlled trial. Nephrol Dial Transplant. 2020;35:274–282. doi: 10.1093/ndt/gfz290.
    1. Cherney DZI, Dekkers CCJ, Barbour SJ, et al. Effects of the SGLT2 inhibitor dapagliflozin on proteinuria in non-diabetic patients with chronic kidney disease (DIAMOND): a randomised, double-blind, crossover trial. Lancet Diabetes Endocrinol. 2020;8:582–593. doi: 10.1016/S2213-8587(20)30162-5.
    1. van der Aart-van der Beek AB, Wessels AMA, Heerspink HJL, Touw DJ. Simple, fast and robust LC–MS/MS method for the simultaneous quantification of canagliflozin, dapagliflozin and empagliflozin in human plasma and urine. J Chromatogr B Anal Technol Biomed Life Sci. 2020;1152:122257. doi: 10.1016/j.jchromb.2020.122257.
    1. Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development-part 2: introduction to pharmacokinetic modeling methods. CPT Pharmacometr Syst Pharmacol. 2013;2:e38. doi: 10.1038/psp.2013.14.
    1. van der Walt JS, Hong Y, Zhang L, Pfister M, Boulton DW, Karlsson MO. A nonlinear mixed effects pharmacokinetic model for dapagliflozin and dapagliflozin 3-O-glucuronide in renal or hepatic impairment. CPT Pharmacometr Syst Pharmacol. 2013;2:e42. doi: 10.1038/psp.2013.20.
    1. Porrini E, Ruggenenti P, Luis-Lima S, et al. Estimated GFR: time for a critical appraisal. Nat Rev Nephrol. 2019;15:177–190. doi: 10.1038/s41581-018-0080-9.
    1. Koomen JV, Stevens J, Heerspink HJL. Exposure–response relationships of dapagliflozin on cardiorenal risk markers and adverse events: a pooled analysis of 13 phase II/III trials. Br J Clin Pharmacol. 2020 doi: 10.1111/bcp.14318.
    1. Kroonen MYAM, Koomen JV, Petrykiv SI, Laverman GD, Heerspink HJL, Stevens J. Exposure–response relationships for the sodium-glucose co-transporter-2 inhibitor dapagliflozin with regard to renal risk markers. Diabetes Obes Metab. 2020;22:916–921. doi: 10.1111/dom.13976.
    1. Ghezzi C, Yu AS, Hirayama BA, et al. Dapagliflozin binds specifically to sodium-glucose cotransporter 2 in the proximal renal tubule. J Am Soc Nephrol. 2017;28:802–810. doi: 10.1681/ASN.2016050510.
    1. Committee for Medicinal Products for Human Use (CHMP). European public assessment report Forxiga dapagliflozin (Procedure No.: EMEA/H/C/002322, EMA/689976/2012). European medicines Agency (EmA), Amsterdam.

Source: PubMed

3
Předplatit