Bridging adults and paediatrics with secondary hyperparathyroidism receiving haemodialysis: a pharmacokinetic-pharmacodynamic analysis of cinacalcet

Ping Chen, Winnie Sohn, Adimoolam Narayanan, Per Olsson Gisleskog, Murad Melhem, Ping Chen, Winnie Sohn, Adimoolam Narayanan, Per Olsson Gisleskog, Murad Melhem

Abstract

Aims: The aims of this study were to develop a pharmacokinetic (PK) and PK-pharmacodynamic (PK/PD) model of cinacalcet in adults and paediatrics with secondary hyperparathyroidism (SHPT) on dialysis, to test covariates of interest, and to perform simulations to inform dosing in paediatrics with SHPT.

Methods: Cinacalcet PK, intact parathyroid hormone (iPTH) and corrected calcium (cCa) time courses following multiple daily oral doses (1-300 mg) were modelled using a nonlinear mixed effects modelling approach using data from eight clinical studies. Model-based trial simulations, using adult or paediatric titration schemas, predicted efficacy (iPTH change from baseline and proportion achieving iPTH decrease ≥30%) and safety (cCa change from baseline and proportion achieving cCa ≤8.4 mg/dL) endpoints at 24 weeks.

Results: Cinacalcet PK parameters were described by a two-compartment linear model with delayed first-order absorption-elimination (apparent clearance = 287.74 L h-1 ). Simulations suggested that paediatric starting doses (1, 2.5, 5, 10 and 15 mg) would provide PK exposures less than or similar to a 30 mg adult dose. The titrated dose simulations suggested that the mean (prediction interval) proportion of paediatric and adult subjects achieving ≥30% reduction in iPTH from baseline at Week 24 was 49% (36%, 62%), and 70.1% (62.5%, 77%), respectively. Additionally, the mean (confidence interval) proportion of paediatric and adult subjects achieving cCa ≤8.4 mg dL-1 at Week 24 was 8% (2%, 18%) and 23.6% (17.5%, 30.5%), respectively.

Conclusions: Model-based simulations showed that the paediatric cinacalcet starting dose (0.2 mg kg-1 ), titrated to effect, would provide the desired PD efficacy (PTH suppression <30%) while minimizing safety concerns (hypocalcaemia).

Keywords: PK/PD; chronic kidney disease; dialysis; modelling and simulation; paediatrics.

Conflict of interest statement

P.C., W.S. and A.N. are employees of Amgen Inc. M.M., currently with Vertex Pharmaceuticals, was an employee of Amgen at the time the work was completed. P.O.G. was a consultant on this analysis.

© 2019 Amgen Inc. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

Figure 1
Figure 1
PK/PD model schematic. CaS receptor, calcium‐sensing receptor; cCa, corrected calcium; CL/F, apparent systemic clearance; iPTH, intact parathyroid hormone; Kin cCa, the first order input rate for Ca; Kout cCa, the first order elimination rate for Ca; Q/F, apparent distribution clearance; Tlag, lag time
Figure 2
Figure 2
Prediction‐corrected visual predictive check for the final PK/PD model (combined dataset with additional 5 subjects in Study 20110100) for iPTH A, and corrected calcium B, separated by age group. Note that the bulk of the data fell within prediction‐corrected prediction intervals. cCa, corrected calcium; CI, confidence interval; iPTH, intact parathyroid hormone; Obs, observed; PK/PD, pharmacokinetics/pharmacodynamics; Sim, simulated
Figure 3
Figure 3
Simulated serum iPTH and cCa following Study 20070208 titrated dosing schema with weekly cCa monitoring in paediatric subjects (I), or following study titrated dosing schema in adults (II): A, PTH change from baseline vs time; B, fraction of subjects with iPTH decrease ≥30% vs time; C, corrected Ca change from baseline vs time; D, fraction of subjects with Ca ≤8.4 mg dL−1. Black line and shaded area indicates the mean and 90% prediction interval, respectively; cCa, corrected calcium; iPTH, intact parathyroid hormone
Figure 4
Figure 4
The predicted proportion of subjects to be on each dose level, over the course of the study. Simulations were performed following the design and six step‐wise titrated dosing schema from paediatric Study 20070208 A, or following dosing implemented in study 20000172 in adult subjects B. Subject to maintenance of serum iPTH and serum calcium values, subjects were eligible for a dose increase once every 4 weeks. For paediatric subjects, next dose level was based on subject post‐dialysis weight (see Supporting Information S4.1). cCa, corrected calcium, iPTH, intact parathyroid hormone

References

    1. Ketteler M, Block GA, Evenepoel P, et al. Executive summary of the 2017 KDIGO Chronic Kidney Disease‐Mineral and Bone Disorder (CKD‐MBD) Guideline Update: What's changed and why it matters. Kidney Int. 2017;92(1):26‐36.
    1. Goodman WG. The consequences of uncontrolled secondary hyperparathyroidism and its treatment in chronic kidney disease. Semin Dial. 2004;17(3):209‐216.
    1. Silver J, Naveh‐Many T. Regulation of parathyroid hormone synthesis and secretion. Semin Nephrol. 1994;14(2):175‐194.
    1. Cunningham J, Locatelli F, Rodriguez M. Secondary hyperparathyroidism: Pathogenesis, disease progression, and therapeutic options. Clin J Am Soc Nephrol. 2011;6(4):913‐921.
    1. Goodman WG, Goldin J, Kuizon BD, et al. Coronary‐artery calcification in young adults with end‐stage renal disease who are undergoing dialysis. N Engl J Med. 2000;342(20):1478‐1483.
    1. Goodman WG, Quarles LD. Development and progression of secondary hyperparathyroidism in chronic kidney disease: Lessons from molecular genetics. Kidney Int. 2008;74(3):276‐288.
    1. Moe SM. Disorders involving calcium, phosphorus, and magnesium. Prim Care. 2008;35(2):215‐237.
    1. Schmitt CP, Mehls O. Mineral and bone disorders in children with chronic kidney disease. Nat Rev Nephrol. 2011;7(11):624‐634.
    1. Denburg MR, Kumar J, Jemielita T, et al. Fracture burden and risk factors in childhood CKD: Results from the CKiD cohort study. J Am Soc Nephrol. 2016;27(2):543‐550.
    1. Groothoff JW, Offringa M, Van Eck‐Smit BL, et al. Severe bone disease and low bone mineral density after juvenile renal failure. Kidney Int. 2003;63(1):266‐275.
    1. Kuizon BD, Salusky IB. Growth retardation in children with chronic renal failure. J Bone Miner Res. 1999;14(10):1680‐1690.
    1. Mathias R, Salusky I, Harman W, et al. Renal bone disease in pediatric and young adult patients on hemodialysis in a children's hospital. J Am Soc Nephrol. 1993;3(12):1938‐1946.
    1. Sanchez CP. Secondary hyperparathyroidism in children with chronic renal failure: Pathogenesis and treatment. Paediatr Drugs. 2003;5(11):763‐776.
    1. Wesseling‐Perry K, Pereira RC, Tseng CH, et al. Early skeletal and biochemical alterations in pediatric chronic kidney disease. Clin J Am Soc Nephrol. 2012;7(1):146‐152.
    1. Wesseling‐Perry K, Salusky IB. Chronic kidney disease: Mineral and bone disorder in children. Semin Nephrol. 2013;33(2):169‐179.
    1. Brown EM, MacLeod RJ. Extracellular calcium sensing and extracellular calcium signaling. Physiol Rev. 2001;81(1):239‐297.
    1. Nemeth EF, Scarpa A. Cytosolic Ca2+ and the regulation of secretion in parathyroid cells. FEBS Lett. 1986;203(1):15‐19.
    1. Nemeth EF, Steffey ME, Fox J. The parathyroid calcium receptor: a novel therapeutic target for treating hyperparathyroidism. Pediatr Nephrol. 1996;10(3):275‐279.
    1. Nemeth EF, Bennett SA. Tricking the parathyroid gland with novel calcimimetic agents. Nephrol Dial Transplant. 1998;13(8):1923‐1925.
    1. Nemeth EF, Heaton WH, Miller M, et al. Pharmacodynamics of the type II calcimimetic compound cinacalcet HCl. J Pharmacol Exp Ther. 2004;308(2):627‐635.
    1. Nemeth EF, Steffey ME, Hammerland LG, et al. Calcimimetics with potent and selective activity on the parathyroid calcium receptor. Proc Natl Acad Sci USA. 1998;95(7):4040‐4045.
    1. Levi R, Ben‐Dov IZ, Lavi‐Moshayoff V, et al. Increased parathyroid hormone gene expression in secondary hyperparathyroidism of experimental uremia is reversed by calcimimetics: Correlation with posttranslational modification of the trans acting factor AUF1. J Am Soc Nephrol. 2006;17(1):107‐112.
    1. Block GA, Martin KJ, de Francisco AL, et al. Cinacalcet for secondary hyperparathyroidism in patients receiving hemodialysis. N Engl J Med. 2004;350(15):1516‐1525.
    1. Padhi D, Harris R. Clinical pharmacokinetic and pharmacodynamic profile of cinacalcet hydrochloride. Clin Pharmacokinet. 2009;48(5):303‐311.
    1. Padhi D, Harris RZ, Salfi M, Sullivan JT. No effect of renal function or dialysis on pharmacokinetics of cinacalcet (Sensipar/Mimpara). Clin Pharmacokinet. 2005;44(5):509‐516.
    1. Amgen Inc. Sensipar® (cinacalcet) prescribing information. 2011.
    1. Amgen Inc. Sensipar® (cinacalcet) Summary of Product Characteristics. 2018.
    1. Jain A, Bhayana S, Vlasschaert M, House A. A formula to predict corrected calcium in haemodialysis patients. Nephrol Dial Transplant. 2008;23(9):2884‐2888.
    1. Mouksassi MS, Marier JF, Cyran J, Vinks AA. Clinical trial simulations in pediatric patients using realistic covariates: Application to teduglutide, a glucagon‐like peptide‐2 analog in neonates and infants with short‐bowel syndrome. Clin Pharmacol Ther. 2009;86(6):667‐671.
    1. National Center for Health Statistics . National Health and Nutrition Examination Survey 2009–2010. .
    1. Amgen Inc. Clinical Study Report: A phase 3 study to assess the efficacy and safety of an oral calcimimetic agent (AMG 073) in secondary hyperparathyroidism of end stage renal disease treated with hemodialysis. 2003.
    1. Amgen Inc. Clinical Study Report: A randomized, double‐blind, placebo‐controlled study to assess the efficacy and safety of cinacalcet HCl in pediatric subjects with chronic kidney disease and secondary hyperparathyroidism receiving dialysis. 2016.
    1. Joborn H, Hjemdahl P, Wide L, Akerström G, Ljunghall S. Reduction of serum parathyroid hormone levels during sympathetic stimulation in man. J Endocrinol Invest. 1987;10(2):153‐156.
    1. Harding SD, Sharman JL, Faccenda E, et al. The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res. 2018;46(D1):D1091‐D1106.
    1. Alexander SPH, Christopoulos A, Davenport AP, et al. THE CONCISE GUIDE TO PHARMACOLOGY 2017/18: G protein‐coupled receptors. Br J Pharmacol. 2017;174(Suppl 1):S17‐S129.
    1. Chattopadhyay N. Biochemistry, physiology and pathophysiology of the extracellular calcium‐sensing receptor. Int J Biochem Cell Biol. 2000;32(8):789‐804.
    1. Ehlert FJ. Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. Mol Pharmacol. 1988;33(2):187‐194.
    1. Black JW, Leff P. Operational models of pharmacological agonism. Proc R Soc Lond B Biol Sci. 1983;220(1219):141‐162.
    1. Peterson MC, Riggs MM. A physiologically based mathematical model of integrated calcium homeostasis and bone remodeling. Bone. 2010;46(1):49‐63.
    1. Walter S, Baruch A, Dong J, et al. Pharmacology of AMG 416 (velcalcetide), a novel peptide agonist of the calcium‐sensing receptor, for the treatment of secondary hyperparathyroidism in hemodialysis patients. J Pharmacol Exp Ther. 2013;346(2):229‐240.
    1. Standing JF. Understanding and applying pharmacometric modelling and simulation in clinical practice and research. Br J Clin Pharmacol. 2017;83(2):247‐254.
    1. Chen P, Olsson Gisleskog P, Perez‐Ruixo JJ, et al. Population pharmacokinetics and pharmacodynamics of the calcimimetic etelcalcetide in chronic kidney disease and secondary hyperparathyroidism receiving hemodialysis. CPT Pharmacometrics Syst Pharmacol. 2016;5(9):484‐494.
    1. Wang B, Ludden T, Gonzalez M, Rein P, Harris R. A population pharmacokinetic (PK) and pharmacodynamic (PD) analysis of cinacalcet HCl in renal‐dialysis patients with secondary hyperparathyroidism (HPT). Clin Pharmacol Ther. 2004;75:62.
    1. Mahmood I. Dosing in children: A critical review of the pharmacokinetic allometric scaling and modelling approaches in paediatric drug development and clinical settings. Clin Pharmacokinet. 2014;53(4):327‐346.
    1. Raposo JF, Sobrinho LG, Ferreira HG. A minimal mathematical model of calcium homeostasis. J Clin Endocrinol Metab. 2002;87(9):4330‐4340.
    1. Riggs MM, Peterson MC, Gastonguay MR. Multiscale physiology‐based modeling of mineral bone disorder in patients with impaired kidney function. J Clin Pharmacol. 2012;52(S1):45s‐53s.
    1. Abraham AK, Mager DE, Gao X, Li M, Healy DR, Maurer TS. Mechanism‐based pharmacokinetic/pharmacodynamic model of parathyroid hormone‐calcium homeostasis in rats and humans. J Pharmacol Exp Ther. 2009;330(1):169‐178.
    1. Lalonde RL, Gaudreault J, Karhu DA, Marriott TB. Mixed‐effects modeling of the pharmacodynamic response to the calcimimetic agent R‐568. Clin Pharmacol Ther. 1999;65(1):40‐49.
    1. Abraham AK, Maurer TS, Kalgutkar AS, et al. Pharmacodynamic model of parathyroid hormone modulation by a negative allosteric modulator of the calcium‐sensing receptor. AAPS J. 2011;13(2):265‐273.
    1. Chen P, Narayanan A, Wu B, et al. Population pharmacokinetic and pharmacodynamic modeling of etelcalcetide in patients with chronic kidney disease and secondary hyperparathyroidism receiving hemodialysis. Clin Pharmacokinet. 2018;57(1):71‐85.
    1. Jong G. Pediatric development: Physiology, enzymes, drug metabolism, pharmacokinetics and pharmacodynamics In: Bar‐Shalom D, Rose K, eds. Pediatric Formulations: A Roadmap. AAPS Advances in the Pharmaceutical Sciences Series 11 New York: Springer; 2014.

Source: PubMed

3
Sottoscrivi