Modelling and simulation as research tools in paediatric drug development

Francesco Bellanti, Oscar Della Pasqua, Francesco Bellanti, Oscar Della Pasqua

Abstract

Purpose: Although practical and ethical constraints impose special requirements for the evaluation of treatment safety and efficacy in children, the main issue remains the empirical basis for patient stratification and dose selection at the early stage of the development of new chemical and biological entities. The aim of this review is to highlight the advantages and limitations of modelling and simulation (M&S) in supporting decision making during paediatric drug development.

Methods: A literature search on Pubmed's database Medical Subject Headings (MeSH) has been performed to retrieve relevant publications on the use of model-based approaches in paediatric drug development and therapeutics.

Results: M&S enable the assessment of the impact of different regimens as well as of different populations on a drug's safety and efficacy profile. It has been widely used in the last two decades to support pre-clinical and early clinical drug development. In fact, M&S have been applied to drug development as decision tools, as study optimization tools and as data analysis tools. In particular, this approach can be used to support dose adjustment in specific subgroups of a population. M&S may therefore allow the individualisation of drug therapy in children, improving the risk-benefit ratio in this population.

Conclusions: The lack of consensus on how to assess the impact of developmental factors on pharmacokinetics, pharmacodynamics, efficacy and safety has so far prevented a broader use of M&S. This problem is compounded by the limited collaboration between stakeholders, which prevents data sharing in this field. In this article, we emphasise the need for a concerted effort to promote the effective use of this technology in paediatric drug development and avoid unnecessary exposure of children to clinical trials.

Figures

Fig. 1
Fig. 1
Simulations allow the assessment of a system’s performance under hypothetical and real-life scenarios (i.e. “what-if” scenarios), yielding information about the implications of different experimental designs and quantitative predictions about treatment outcome. In this example, a model of haematopoiesis is used to simulate the effects of darbepoetin alfa administered every 2 weeks in chemotherapy-induced anaemia based on weight-based fixed-dosing regimens. Adapted from Jumbe et al. [25]
Fig. 2
Fig. 2
Physiologically-based pharmacokinetic (PBPK) models provide an integrated view of drug disposition in vivo. In contrast to empirical compartmental models, a PBPK model is aimed at describing the in vivo behaviour of the drug before the acquisition of in vivo data. PBPK relies primarily on describing drug disposition in terms of organ distribution, blood flow and metabolic capacity. Actual experiments become confirmatory and can therefore be optimised in terms of dose range, sample size, frequency and sampling intervals. Diagram adapted from Theil et al [28]
Fig. 3
Fig. 3
Modelling and simulation can be used to support prediction and extrapolation of data in early clinical development. The graphs show the implications of pharmacokinetic differences for systemic exposure across different age groups in children. Based on pharmacokinetic parameter estimates systemic exposure can be simulated for a range of dosing regimens. Note the non-linearity in the dose range for the different age groups. Lines depict the fraction of patients categorised by body weight reaching target exposure criteria (a = AUC > 6.02 mg*h/L, b = plasma concentrations above IC80 for at least 3 h) following different doses of abacavir. circles 10 kg, crosses 20 kg, triangles 30 kg, squares 40 kg. From Cella et al [10]
Fig. 4
Fig. 4
The diagram depicts the major components of a clinical trial simulation (CTS). In model-based drug development, a CTS can be used to characterise the interactions between drug and disease, enabling among other things the assessment of disease-modifying effects, dose selection and covariate effects (e.g. age, body weight). In conjunction with a trial model, CTS allows the evaluation of such interactions, taking into account uncertainty and trial design factors, including the implications of different statistical methods for the analysis of the data
Fig. 5
Fig. 5
The concept of personalised medicines implies quantitative assessment of the risk–benefit ratio at the individual and patient population levels. M&S techniques are critical for such an evaluation. The use of a therapeutic utility index (TUI) illustrates how the safety–efficacy ratio of a treatment correlates with drug exposure. The graphs show a safety: probability of a major or minor bleeding event as a function of daily steady-state apixaban exposure (AUCss); b efficacy: probability of a venous thromboembolic event (VTE) as a function of daily apixaban AUCss and regimen. The shaded regions surrounding the regression lines represent the 90% bootstrap confidence intervals. The boxes at the bottom of each figure represent the distribution of apixaban exposures for the doses indicated. Exposure distributions are shown for the total daily dose (TDD) because the distributions of AUCss should be the same for b.i.d. and q.d. regimens for the same total daily dose. Subjects with moderate renal impairment are expected to have a 43% increase in apixaban exposure; however, apixaban’s therapeutic utility index suggests that dose adjustment is not needed in this population. From Leil et al. [53]

References

    1. Lalonde RL, Kowalski KG, Hutmacher MM, Ewy W, Nichols DJ, Milligan PA, Corrigan BW, Lockwood PA, Marshall SA, Benincosa LJ, Tensfeldt TG, Parivar K, Amantea M, Glue P, Koide H, Miller R. Model-based drug development. Clin Pharmacol Ther. 2007;82(1):21–32. doi: 10.1038/sj.clpt.6100235.
    1. . 2009
    1. Danhof M, de Lange EC, Della Pasqua OE, Ploeger BA, Voskuyl RA. Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modeling in translational drug research. Trends Pharmacol Sci. 2008;29(4):186–191. doi: 10.1016/j.tips.2008.01.007.
    1. Bakker BM, van Eunen K, Jeneson JA, van Riel NA, Bruggeman FJ, Teusink B. Systems biology from micro-organisms to human metabolic diseases: the role of detailed kinetic models. Biochem Soc Trans. 2010;38(5):1294–1301. doi: 10.1042/BST0381294.
    1. Musante CJ, Lewis AK, Hall K. Small- and large-scale biosimulation applied to drug discovery and development. Drug Discov Today. 2002;7(20 Suppl):S192–S196. doi: 10.1016/S1359-6446(02)02442-X.
    1. Michelson S. The impact of systems biology and biosimulation on drug discovery and development. Mol Biosyst. 2006;2:288–291. doi: 10.1039/b602194h.
    1. Manolis E, Pons G. Proposals for model-based paediatric medicinal development within the current European Union regulatory framework. Br J Clin Pharmacol. 2009;68:493–501. doi: 10.1111/j.1365-2125.2009.03484.x.
    1. Bruggeman FJ, Westerhoff HV. The nature of systems biology. Trends Microbiol. 2007;15:45–50. doi: 10.1016/j.tim.2006.11.003.
    1. Bangs A. Predictive biosimulation and virtual patients in pharmaceutical R and D. Stud Health Technol Inform. 2005;111:37–42.
    1. Cella M, de Vries Gorter F, Burger D, Danhof M, Della Pasqua O. A model-based approach to dose selection in early pediatric development. Clin Pharmacol Ther. 2010;87(3):294–302. doi: 10.1038/clpt.2009.234.
    1. Abernethy DR, Burckart GJ. Pediatric dose selection. Clin Pharmacol Ther. 2010;87(3):270–271. doi: 10.1038/clpt.2009.292.
    1. . 2009
    1. Rodriguez W, Selen A, Avant D, Chaurasia C, Crescenzi T, Gieser G, Di Giacinto J, Huang SM, Lee P, Mathis L, Murphy D, Murphy S, Roberts R, Sachs HC, Suarez S, Tandon V, Uppoor RS. Improving pediatric dosing through pediatric initiatives: what we have learned. Pediatrics. 2008;121(3):530–539. doi: 10.1542/peds.2007-1529.
    1. Ince I, de Wildt SN, Tibboel D, Danhof M, Knibbe CA. Tailor-made drug treatment for children: creation of an infrastructure for data-sharing and population PK-PD modeling. Drug Discov Today. 2009;14:316–320. doi: 10.1016/j.drudis.2008.11.004.
    1. Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: general principles. Eur J Pediatr. 2006;165:741–746. doi: 10.1007/s00431-006-0188-y.
    1. Bouzom F, Walther B. Pharmacokinetic predictions in children by using the physiologically based pharmacokinetic modelling. Fundam Clin Pharmacol. 2008;22:579–587. doi: 10.1111/j.1472-8206.2008.00648.x.
    1. de Wildt SN, Knibbe CA. Knowledge of developmental pharmacology and modeling approaches should be used to avoid useless trials in children. Eur J Clin Pharmacol. 2009;65:849–850. doi: 10.1007/s00228-009-0655-2.
    1. Cella M, Knibbe C, Danhof M, Della Pasqua O. What is the right dose for children? Br J Clin Pharmacol. 2010;70(4):597–603. doi: 10.1111/j.1365-2125.2009.03591.x.
    1. Slikker W, Jr, Young JF, Corley RA, Dorman DC, Conolly RB, Knudsen TB, Erstad BL, Luecke RH, Faustman EM, Timchalk C, Mattison DR. Improving predictive modeling in pediatric drug development: pharmacokinetics, pharmacodynamics, and mechanistic modeling. Ann N Y Acad Sci. 2005;1053:505–518. doi: 10.1196/annals.1344.044.
    1. ICH topic E11: clinical investigation of medicinal products in the paediatric population. 2001
    1. Johnson TN. Modelling approaches to dose estimation in children. Br J Clin Pharmacol. 2005;59:663–669. doi: 10.1111/j.1365-2125.2005.02429.x.
    1. Rooney KF, Snoeck E, Watson PH. Modelling and simulation in clinical drug development. Drug Discov Today. 2001;6:802–806. doi: 10.1016/S1359-6446(01)01855-4.
    1. Duan JZ. Applications of population pharmacokinetics in current drug labelling. J Clin Pharm Ther. 2007;32:57–79. doi: 10.1111/j.1365-2710.2007.00799.x.
    1. Laer S, Barrett JS, Meibohm B. The in silico child: using simulation to guide pediatric drug development and manage pediatric pharmacotherapy. J Clin Pharmacol. 2009;49:889–904. doi: 10.1177/0091270009337513.
    1. Jumbe N, Yao B, Rovetti R, Rossi G, Heatherington AC (2002) Clinical Trial Simulation of a 200-μg Fixed Dose of Darbepoetin Alfa in Chemotherapy-Induced Anemia. Oncology 16(Suppl 11): 37–44
    1. Laveille C, Vis P. Minutes from the round table held in Paris on July 6, 2007: ‘contribution of modelling in the paediatric drug development’. Fundam Clin Pharmacol. 2008;22:609–612. doi: 10.1111/j.1472-8206.2008.00653.x.
    1. Rajman I. PK/PD modelling and simulations: utility in drug development. Drug Discov Today. 2008;13:341–346. doi: 10.1016/j.drudis.2008.01.003.
    1. Theil FP, Guentert TW, Haddad S, Poulin P. Utility of physiologically based pharmacokinetic models to drug development and rational drug discovery candidate selection. Toxicol Lett. 2003;138:29–49. doi: 10.1016/S0378-4274(02)00374-0.
    1. van de Waterbeemd H, Gifford E. ADMET in silico modelling: towards prediction paradise. Nat Rev Drug Discov. 2003;2:192–204. doi: 10.1038/nrd1032.
    1. Jones HM, Gardner IB, Watson KJ. Modelling and PBPK simulation in drug discovery. AAPS J. 2009;11:155–166. doi: 10.1208/s12248-009-9088-1.
    1. Huntjens DR, Danhof M, Della Pasqua OE. Pharmacokinetic-pharmacodynamic correlations and biomarkers in the development of COX-2 inhibitors. Rheumatology (Oxford) 2005;44:846–859. doi: 10.1093/rheumatology/keh627.
    1. Brent RL. Utilization of juvenile animal studies to determine the human effects and risks of environmental toxicants during postnatal developmental stages. Birth Defects Res B Dev Reprod Toxicol. 2004;71:303–320. doi: 10.1002/bdrb.20020.
    1. . 2009
    1. Chatelut E. Population approaches in paediatrics. Fundam Clin Pharmacol. 2008;22:575–578. doi: 10.1111/j.1472-8206.2008.00647.x.
    1. Yim DS, Zhou H, Buckwalter M, Nestorov I, Peck CC, Lee H. Population pharmacokinetic analysis and simulation of the time-concentration profile of etanercept in pediatric patients with juvenile rheumatoid arthritis. J Clin Pharmacol. 2005;45(3):246–256. doi: 10.1177/0091270004271945.
    1. Knibbe CA, Melenhorst-de Jong G, Mestrom M, Rademaker CM, Reijnvaan AF, Zuideveld KP, Kuks PF, van Vught H, Danhof M. Pharmacokinetics and effects of propofol 6% for short-term sedation in paediatric patients following cardiac surgery. Br J Clin Pharmacol. 2002;54:415–422. doi: 10.1046/j.1365-2125.2002.01652.x.
    1. Ihmsen H, Viethen V, Forst J, Schwilden H, Schmitt HJ, Muenster T. Pharmacodynamic modelling of rocuronium in adolescents with Duchenne muscular dystrophy. Eur J Anaesthesiol. 2009;26:105–110. doi: 10.1097/EJA.0b013e32831aed11.
    1. Tod M. Evaluation of drugs in pediatrics using K-PD models: perspectives. Fundam Clin Pharmacol. 2008;22:589–594. doi: 10.1111/j.1472-8206.2008.00649.x.
    1. Aarons L, Karlsson MO, Mentre F, Rombout F, Steimer JL, van Peer A. Role of modelling and simulation in phase I drug development. Eur J Pharm Sci. 2001;13:115–122. doi: 10.1016/S0928-0987(01)00096-3.
    1. Colburn WA. Biomarkers in drug discovery and development: from target identification through drug marketing. J Clin Pharmacol. 2003;43:329–341. doi: 10.1177/0091270003252480.
    1. Krudys KM, Greenbaum CJ, Pihoker C, Vicini P. Use of oral glucose minimal model-derived index of insulin sensitivity in subjects with early type 1 diabetes mellitus. Metabolism. 2008;57(4):445–447. doi: 10.1016/j.metabol.2007.10.001.
    1. Breda E, Cavaghan MK, Toffolo G, Polonsky KS, Cobelli C. Oral glucose tolerance test minimal model indexes of beta-cell function and insulin sensitivity. Diabetes. 2001;50(1):150–158. doi: 10.2337/diabetes.50.1.150.
    1. Santen G, van Zwet E, Danhof M, Della Pasqua O. From trial and error to trial simulation. I. The importance of model-based drug development for antidepressant drugs. Clin Pharmacol Ther. 2009;86(3):248–254. doi: 10.1038/clpt.2009.105.
    1. Gobburu JVS, Lesko LJ (2009) Quantitative Disease, Drug and Trial Models. Annu Rev Pharmacol Toxicol 49: 291–301
    1. Girard P. Clinical trial simulation: a tool for understanding study failures and preventing them. Basic Clin Pharmacol Toxicol. 2005;96:228–234. doi: 10.1111/j.1742-7843.2005.pto960313.x.
    1. Onar A, Kocak M, Boyett JM. Continual reassessment method vs. traditional empirically based design: modifications motivated by phase I trials in pediatric oncology by the Pediatric Brain Tumor Consortium. J Biopharm Stat. 2009;19(3):437–455. doi: 10.1080/10543400902800486.
    1. Nguyen L. Integration of modelling and simulation into the development of intravenous busulfan in paediatrics: an industrial experience. Fundam Clin Pharmacol. 2008;22:599–604. doi: 10.1111/j.1472-8206.2008.00651.x.
    1. Laer S, Elshoff JP, Meibohm B, Weil J, Mir TS, Zhang W, Hulpke-Wette M. Development of a safe and effective pediatric dosing regimen for sotalol based on population pharmacokinetics and pharmacodynamics in children with supraventricular tachycardia. J Am Coll Cardiol. 2005;46:1322–1330. doi: 10.1016/j.jacc.2005.06.061.
    1. Maas HJ, Danhof M, Della Pasqua OE. Analysis of the relationship between age and treatment response in migraine. Cephalalgia. 2009;29(7):772–780. doi: 10.1111/j.1468-2982.2008.01823.x.
    1. Albers S, Meibohm B, Mir TS, Laer S. Population pharmacokinetics and dose simulation of carvedilol in paediatric patients with congestive heart failure. Br J Clin Pharmacol. 2008;65:511–522. doi: 10.1111/j.1365-2125.2007.03046.x.
    1. Standing JF, Howard RF, Johnson A, Savage I, Wong IC. Population pharmacokinetics of oral diclofenac for acute pain in children. Br J Clin Pharmacol. 2008;66:846–853. doi: 10.1111/j.1365-2125.2008.03289.x.
    1. Mussen F, Salek S, Walker S. A quantitative approach to benefit-risk assessment of medicines. I. The development of a new model using multi-criteria decision analysis. Pharmacoepidemiol Drug Saf. 2007;1(16 Suppl):S2–S15. doi: 10.1002/pds.1435.
    1. Leil TA, Feng Y, Zhang L, Paccaly A, Mohan P, Pfister M. Quantification of apixaban’s therapeutic utility in the prevention of venous thromboembolism: selection of phase III trial dose. Clin Pharmacol Ther. 2010;88(3):375–382. doi: 10.1038/clpt.2010.106.

Source: PubMed

3
Se inscrever