Population PK and PD Analysis of Domagrozumab in Pediatric Patients with Duchenne Muscular Dystrophy

Jessica Wojciechowski, Vivek S Purohit, Lutz O Harnisch, Pinky Dua, Beesan Tan, Timothy Nicholas, Jessica Wojciechowski, Vivek S Purohit, Lutz O Harnisch, Pinky Dua, Beesan Tan, Timothy Nicholas

Abstract

Myostatin, a negative regulator of skeletal muscle growth, is a therapeutic target in muscle-wasting diseases. Domagrozumab, a humanized recombinant monoclonal antibody, binds myostatin and inhibits activity. Domagrozumab was investigated in a phase II trial (NCT02310763) as a potential treatment for boys with Duchenne muscular dystrophy (DMD). Pharmacokinetic/pharmacodynamic (PK/PD) modeling is vital in clinical trial design, particularly for determining dosing regimens in pediatric populations. This analysis sought to establish the PK/PD relationship between free domagrozumab and total myostatin concentrations in pediatric patients with DMD using a prior semimechanistic model developed from a phase I study in healthy adult volunteers (NCT01616277) and following inclusion of phase II data. The refined model was developed using a multiple-step approach comprising structural, random effects, and covariate model development; assessment of model adequacy (goodness-of-fit); and predictive performance. Differences in PKs/PDs between healthy adult volunteers and pediatric patients with DMD were quantitatively accounted for and evaluated by predicting myostatin coverage (the percentage of myostatin bound by domagrozumab). The final model parameter estimates and semimechanistic target-mediated drug disposition structure sufficiently described both domagrozumab and myostatin concentrations in pediatric patients with DMD, and most population parameters were comparable with the prior model (in healthy adult volunteers). Predicted myostatin coverage for phase II patients with DMD was consistently > 90%. Baseline serum myostatin was ~ 65% lower than in healthy adult volunteers. This study provides insights into the regulation of myostatin in healthy adults and pediatric patients with DMD. Clinicaltrials.gov identifiers: NCT01616277 and NCT02310763.

Conflict of interest statement

B.T., P.D., and L.O.H. were employees of and hold stock in Pfizer; V.S.P., T.N., and J.W. are employees of and hold stock in Pfizer.

© 2022 Pfizer Inc. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Distribution density of empirical Bayes estimates (EBE) for baseline and internalization/degradation rate constants parameters. (a) Distribution density for EBEs on baseline. Red and blue lines are the distribution densities of individual random effects for baseline for healthy adult volunteers and pediatric patients with DMD, respectively. (b) Distribution density EBE on internalization/degradation rate constants. Red and blue lines are the distribution densities of individual random effects on internalization/degradation rate constants (kdeg  kint) for healthy adult volunteers and pediatric patients with DMD, respectively. DMD, Duchenne muscular dystrophy.
Figure 2
Figure 2
Evaluation of empirical Bayes estimates (EBEs) on domagrozumab clearance vs (a) age (years) and (b) total body weight (kg) for the final model and model with allometric scaling. Circles are the EBEs for clearance vs. age (a) and body weight (b) for the final model (left) and a model with allometric scaling (right). Blue circles represent pediatric patients with DMD from the phase II study and red circles represent the healthy adult volunteers from the phase I study. Black solid lines and shaded areas are the linear regression and 95% confidence interval, respectively, for the relationship of EBEs vs. covariate values. DMD, Duchenne muscular dystrophy.
Figure 3
Figure 3
Final TMDD model visual predictive check for free domagrozumab concentrations in pediatric patients with DMD. Visual predictive checks are presented for time after first dose (a) and time after dose (b). Observed data are represented by blue circles and the black lines (median (solid), 5th and 95th percentiles (dashed)). The simulated free domagrozumab concentrations for phase II patients with DMD are represented by the red line and red shaded ribbon (median and 95% prediction intervals (PIs) of the median, respectively), and the blue lines and blue shaded ribbons (median and 95% PIs of the 5th and 95th percentiles, respectively). Yellow indicators are the times where observed and simulated data are binned for summaries. DMD, Duchenne muscular dystrophy; TMDD, target‐mediated drug disposition.
Figure 4
Figure 4
Final TMDD model visual predictive check for total myostatin in pediatric patients with DMD. Visual predictive checks are presented for time after first dose (a) and time after dose (b). Observed data are represented by blue circles and the black lines (median (solid), 5th and 95th percentiles (dashed)). The simulated total myostatin concentrations for phase II patients with DMD are represented by the red line and red shaded ribbon (median and 95% prediction intervals (PIs) of the median, respectively), and the blue lines and blue shaded ribbons (median and 95% PIs of the 5th and 95th percentiles, respectively). Yellow indicators are the times where observed and simulated data are binned for summaries. DMD, Duchenne muscular dystrophy; TMDD, target‐mediated drug disposition.
Figure 5
Figure 5
Simulated myostatin coverage. (a) The predicted myostatin coverage following 4 doses of each of 5 mg/kg, 20 mg/kg, and 40 mg/kg every 28 days were simulated using the prior model for pediatric patients with DMD (green), the final model for healthy adult volunteers (blue), and the final model for pediatric patients with DMD (red). Lines represent the median, and ribbons depict the 95% prediction intervals (PIs) for a population of 500 subjects. (b) The predicted myostatin coverage following the administration of 0, 1, 2, 3, 5, 7, 10, 20, 30, 40, 60, and 80 mg/kg of domagrozumab every 4 weeks for 48 weeks using the final model for 500 pediatric patients with DMD. Lines represent the median, and ribbons depict the 95% prediction intervals. DMD, Duchenne muscular dystrophy.

References

    1. Gao, Q.Q. & McNally, E.M. The dystrophin complex: structure, function, and implications for therapy. Compr. Physiol. 5, 1223–1239 (2015).
    1. Mendell, J.R. & Lloyd‐Puryear, M. Report of MDA muscle disease symposium on newborn screening for Duchenne muscular dystrophy. Muscle Nerve 48, 21–26 (2013).
    1. Werneck, L.C. , Lorenzoni, P.J. , Ducci, R.D. , Fustes, O.H. , Kay, C.S.K. & Scola, R.H. Duchenne muscular dystrophy: an historical treatment review. Arq. Neuropsiquiatr. 77, 579–589 (2019).
    1. Koeks, Z. et al. Clinical outcomes in Duchenne muscular dystrophy: a study of 5345 patients from the TREAT‐NMD DMD global database. J. Neuromuscul. Dis. 4, 293–306 (2017).
    1. McNally, E.M. Powerful genes‐myostatin regulation of human muscle mass. N. Engl. J. Med. 350, 2642–2644 (2004).
    1. Mosher, D.S. et al. A mutation in the myostatin gene increases muscle mass and enhances racing performance in heterozygote dogs. PLoS Genet. 3, e79 (2007).
    1. Schuelke, M. et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N. Engl. J. Med. 350, 2682–2688 (2004).
    1. Bogdanovich, S. et al. Functional improvement of dystrophic muscle by myostatin blockade. Nature 420, 418–421 (2002).
    1. Wagner, K.R. et al. Randomized phase 2 trial and open‐label extension of domagrozumab in Duchenne muscular dystrophy. Neuromuscul. Disord. 30, 492–502 (2020).
    1. Huang, S.M. , Abernethy, D.R. , Wang, Y. , Zhao, P. & Zineh, I. The utility of modeling and simulation in drug development and regulatory review. J. Pharm. Sci. 102, 2912–2923 (2013).
    1. Bhattacharya, I. , Manukyan, Z. , Chan, P. , Heatherington, A. & Harnisch, L. Application of quantitative pharmacology approaches in bridging pharmacokinetics and pharmacodynamics of domagrozumab from adult healthy subjects to pediatric patients with Duchenne muscular disease. J. Clin. Pharmacol. 58, 314–326 (2018).
    1. Tiwari, A. , Bhattacharya, I. , Chan, P.L.S. & Harnisch, L. Comparing model performance in characterizing the PK/PD of the anti‐myostatin antibody domagrozumab. Clin. Transl. Sci. 13, 125–136 (2020).
    1. Bhattacharya, I. et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of domagrozumab (PF‐06252616), an antimyostatin monoclonal antibody, in healthy subjects. Clin. Pharmacol. Drug Dev. 7, 484–497 (2018).
    1. Mould, D.R. & Upton, R.N. Basic concepts in population modeling, simulation, and model‐based drug development‐part 2: introduction to pharmacokinetic modeling methods. CPT Pharmacometrics Syst. Pharmacol. 2, e38 (2013).
    1. Mariot, V. et al. Downregulation of myostatin pathway in neuromuscular diseases may explain challenges of anti‐myostatin therapeutic approaches. Nat. Commun. 8, 1859 (2017).
    1. Burch, P.M. et al. Reduced serum myostatin concentrations associated with genetic muscle disease progression. J. Neurol. 264, 541–553 (2017).
    1. Hanna, M.G. et al. Safety and efficacy of intravenous bimagrumab in inclusion body myositis (RESILIENT): a randomised, double‐blind, placebo‐controlled phase 2b trial. Lancet Neurol. 18, 834–844 (2019).
    1. Bi, Y. et al. Model‐informed drug development in pediatric dose selection. J. Clin. Pharmacol. 61(Suppl 1), S60–S69 (2021).
    1. Green, F.G. , Park, K. & Burckart, G.J. Methods used for pediatric dose selection in drug development programs submitted to the US FDA 2012–2020. J. Clin. Pharmacol. 61(Suppl 1), S28–S35 (2021).

Source: PubMed

Подписаться