Population pharmacokinetic modelling of busulfan and the influence of body composition in paediatric Fanconi anaemia patients

Matthijs W van Hoogdalem, Chie Emoto, Tsuyoshi Fukuda, Tomoyuki Mizuno, Parinda A Mehta, Alexander A Vinks, Matthijs W van Hoogdalem, Chie Emoto, Tsuyoshi Fukuda, Tomoyuki Mizuno, Parinda A Mehta, Alexander A Vinks

Abstract

Aims: Fanconi anaemia (FA) is a rare disorder characterized by progressive bone marrow failure that requires haematopoietic cell transplantation (HCT). Busulfan is used in conditioning regimens prior to HCT. Doses used in non-FA patients cause life-threatening toxicities in FA patients and data on busulfan pharmacokinetics (PK) in this population are limited. This study characterized busulfan PK in paediatric FA patients using population PK modelling and evaluated the effect of body composition on steady-state concentrations (Css ).

Methods: A total of 200 busulfan plasma concentrations in 29 FA patients from a recent study (Clinicaltrials.gov; NCT01082133) were available for population PK modelling. The effect of different body size-scaled doses and body compositions on Css was investigated using population PK modelling.

Results: Fat free mass (FFM) was identified as the best size descriptor in a two-compartment busulfan PK model in FA patients. Conventional dosing, based on an amount of busulfan per kilogram of total body mass, resulted in higher Css in FA patients with higher body mass index (BMI). A newly proposed FFM-based dosing strategy would eliminate the observed trend of higher concentrations in high BMI patients, and achieve consistent Css across a wide BMI spectrum.

Conclusions: This is the first study to describe the population PK of busulfan in paediatric FA patients. The proposed model will facilitate PK model-informed precision dosing. FFM-based dosing is expected to improve the probability of achieving target Css , particularly in obese patients, while minimizing the risk of overdosing.

Keywords: Fanconi anaemia; busulfan; paediatrics; population pharmacokinetic modelling.

Conflict of interest statement

There are no competing interests to declare.

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

Figures

Figure 1
Figure 1
BMI distribution for male (A) and female (B) patients in the study population (black circles). Black lines are locally weighted scatterplot smoothing curves through international cut‐off points for BMI for overweight (lower curve) and obesity (upper curve) proposed by Cole et al.32 The yellow area indicates overweight, whereas patients in the red area are considered obese. One patient (male, 22.4 years of age, BMI = 23.9 kg m−2) is not included in this graph, since overweight and obesity in adulthood is determined differently, ie, BMI greater than 25 and 30 kg m−2, respectively
Figure 2
Figure 2
Age versus height (A, B), weight (C, D) and BMI (E, F) for male (left column) and female (right column) patients (black circles) plotted with the NHANES data (yellow and purple shading for men and women, respectively), executed by Centers for Disease Control and Prevention (CDC) over the years 2005‐2014. The dashed line is a locally weighted scatterplot smoothing curve through the NHANES data
Figure 3
Figure 3
Goodness‐of‐fit plots for the final PK model. Population‐predicted (A) and individual‐predicted (B) busulfan concentrations vs observed concentrations (solid line, line of identity; dashed line, linear regression; open circles, observed data). Conditional weighted residuals (CWRES) vs (C) population‐predicted busulfan concentration and (D) time after start of the infusion (solid line, line of identity; dashed line, second‐order polynomial; open circles, observed data)
Figure 4
Figure 4
Prediction‐corrected visual predictive check (pcVCP) for the final model of busulfan. Open circles represent observed concentration. The solid red line represents the median of the observed concentration, and the shaded red area represents a simulation‐based 95% CI for the median. The solid blue lines represent the 95th and 5th percentiles of the observed data, and the simulation‐based 95% CIs for the 95th and 5th percentiles are shown as the shaded blue areas
Figure 5
Figure 5
Plasma concentration‐time profiles of intravenous busulfan for underweight (A, B), normal weight (C, D), and obese (E, F) patients following the first dose. The left column represents male patients, female patients are represented by the right column. The dashed line is the population‐based prediction, black circles are observed data, and the solid line is the individually predicted fit
Figure 6
Figure 6
Individual steady‐state concentrations (Css) following the initial dose (A, 0.6 and 0.8 mg kg−1 TBM) and simulated doses (B, 0.6 mg kg−1 TBM; C, 0.6 mg kg−1 FFM; D, 0.8 mg kg−1 FFM) plotted against the body mass index (BMI) of the patient. Open circles represent non‐obese patients and obese patients are represented by black circles. The dotted line represents the proposed upper Css limit (target ≤ 0.35 mg L−1). The solid line represents the linear regression line
Figure 7
Figure 7
Individual steady‐state concentration (Css) following simulated FFM‐ and TBM‐based doses. Open circles represent non‐obese patients and obese patients are represented by black squares. Solid horizontal lines represent median with interquartile range (IQR) per dosing regimen. Solid vertical lines mark 0.6, 0.8, 1.0 and 1.2 mg kg−1 dosing scenarios. The dotted line represents the proposed upper Css limit (target ≤ 0.35 mg L−1)
Figure 8
Figure 8
Estimated clearance (A) and volume of distribution (B) related population pharmacokinetic parameters in FA patients compared to reported values in non‐FA paediatric patients.13, 14, 15, 16, 17 All values, which were often expressed corresponding to the median weight of the population, were allometrically scaled to a total body weight of 70 kg, using the theoretical power exponents of 0.75 and 1.0 for clearance and volume, respectively. Both one‐ and two‐compartment models have been reported. Since one‐compartment models inherently include only one volume of distribution (Vd), this value was compared to the sum of the estimated volumes for the central and peripheral compartment (Vss) in two‐compartment models. Black circles represent the estimated value, with whiskers showing the 95% confidence interval of the mean estimate, unless stated otherwise. †Whiskers show the 90% confidence interval

References

    1. Joenje H, Patel KJ. The emerging genetic and molecular basis of Fanconi anaemia. Nat Rev Genet. 2001;2(6):446‐457.
    1. Auerbach AD. Fanconi anemia and its diagnosis. Mutat Res. 2009;668(1‐2):4‐10.
    1. Gluckman E, Auerbach A, Horowitz M, et al. Bone marrow transplantation for Fanconi anemia. Blood. 1995;86(7):2856‐2862.
    1. Grochow LB, Jones RJ, Brundrett RB, et al. Pharmacokinetics of busulfan: correlation with veno‐occlusive disease in patients undergoing bone marrow transplantation. Cancer Chemother Pharmacol. 1989;25(1):55‐61.
    1. Andersson BS, Thall PF, Madden T, et al. Busulfan systemic exposure relative to regimen‐related toxicity and acute graft‐versus‐host disease: defining a therapeutic window for i.v. BuCy2 in chronic myelogenous leukemia. Biol Blood Marrow Transplant. 2002;8(9):477‐485.
    1. Slattery JT, Clift RA, Buckner CD, et al. Marrow transplantation for chronic myeloid leukemia: the influence of plasma busulfan levels on the outcome of transplantation. Blood. 1997;89(8):3055‐3060.
    1. Slattery JT, Sanders JE, Buckner CD, et al. Graft‐rejection and toxicity following bone marrow transplantation in relation to busulfan pharmacokinetics. Bone Marrow Transplant. 1995;16(1):31‐42.
    1. Gluckman E, Devergie A, Schaison G, et al. Bone marrow transplantation in Fanconi anaemia. Br J Haematol. 1980;45(4):557‐564.
    1. Walden H, Deans AJ. The Fanconi anemia DNA repair pathway: structural and functional insights into a complex disorder. Annu Rev Biophys. 2014;43:257‐278.
    1. Mehta PA, Emoto C, Fukuda T, et al. Busulfan pharmacokinetics and precision dosing: are patients with Fanconi anemia different? Biol Blood Marrow Transplant. 2019. ;25(12):2416‐2421.
    1. Nguyen L, Fuller D, Lennon S, Leger F, Puozzo C. I.V. busulfan in pediatrics: a novel dosing to improve safety/efficacy for hematopoietic progenitor cell transplantation recipients. Bone Marrow Transplant. 2004;33(10):979‐987.
    1. Trame MN, Bergstrand M, Karlsson MO, Boos J, Hempel G. Population pharmacokinetics of busulfan in children: increased evidence for body surface area and allometric body weight dosing of busulfan in children. Clin Cancer Res. 2011;17(21):6867‐6877.
    1. Booth BP, Rahman A, Dagher R, et al. Population pharmacokinetic‐based dosing of intravenous busulfan in pediatric patients. J Clin Pharmacol. 2007;47(1):101‐111.
    1. Paci A, Vassal G, Moshous D, et al. Pharmacokinetic behavior and appraisal of intravenous busulfan dosing in infants and older children: the results of a population pharmacokinetic study from a large pediatric cohort undergoing hematopoietic stem‐cell transplantation. Ther Drug Monit. 2012;34(2):198‐208.
    1. Long‐Boyle JR, Savic R, Yan S, et al. Population pharmacokinetics of busulfan in pediatric and young adult patients undergoing hematopoietic cell transplant: a model‐based dosing algorithm for personalized therapy and implementation into routine clinical use. Ther Drug Monit. 2015;37(2):236‐245.
    1. Savic RM, Cowan MJ, Dvorak CC, et al. Effect of weight and maturation on busulfan clearance in infants and small children undergoing hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2013;19(11):1608‐1614.
    1. McCune JS, Bemer MJ, Barrett JS, Baker KS, Gamis AS, Holford NHG. Busulfan in infant to adult hematopoietic cell transplant recipients: a population pharmacokinetic model for initial and Bayesian dose personalization. Am J Clin Cancer Res. 2014;20(3):754‐763.
    1. Bartelink IH, van Kesteren C, Boelens JJ, et al. Predictive performance of a busulfan pharmacokinetic model in children and young adults. Ther Drug Monit. 2012;34(5):574‐583.
    1. Neely M, Philippe M, Rushing T, et al. Accurately achieving target busulfan exposure in children and adolescents with very limited sampling and the BestDose software. Ther Drug Monit. 2016;38(3):332‐342.
    1. Mehta PA, Davies SM, Leemhuis T, et al. Radiation‐free, alternative‐donor HCT for Fanconi anemia patients: results from a prospective multi‐institutional study. Blood. 2017;129(16):2308‐2315.
    1. Slattery JT, Risler LJ. Therapeutic monitoring of busulfan in hematopoietic stem cell transplantation. Ther Drug Monit. 1998;20(5):543‐549.
    1. Holford NHG, Anderson BJ. Allometric size: the scientific theory and extension to normal fat mass. Eur J Pharm Sci. 2017;109S:S59‐S64.
    1. Green B, Duffull SB. What is the best size descriptor to use for pharmacokinetic studies in the obese? Br J Clin Pharmacol. 2004;58(2):119‐133.
    1. Janmahasatian S, Duffull SB, Ash S, Ward LC, Byrne NM, Green B. Quantification of lean bodyweight. Clin Pharmacokinet. 2005;44(10):1051‐1065.
    1. Al‐Sallami HS, Goulding A, Grant A, Taylor R, Holford N, Duffull SB. Prediction of fat‐free mass in children. Clin Pharmacokinet. 2015;54(11):1169‐1178.
    1. Kendrick JG, Carr RR, Ensom MHH. Pharmacokinetics and drug dosing in obese children. J Pediatr Pharmacol Ther. 2010;15(2):94‐109.
    1. Traub SL, Kichen L. Estimating ideal body mass in children. Am J Hosp Pharm. 1983;40(1):107‐110.
    1. Devine BJ. Gentamicin therapy. Drug Intell Clin Pharm. 1974;8:650‐655.
    1. Ette EI. Stability and performance of a population pharmacokinetic model. J Clin Pharmacol. 1997;37(6):486‐495.
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction‐corrected visual predictive checks for diagnosing nonlinear mixed‐effects models. AAPS j. 2011;13(2):143‐151.
    1. Palmer J, McCune JS, Perales MA, et al. Personalizing busulfan‐based conditioning: considerations from the American Society for Blood and Marrow Transplantation Practice Guidelines Committee. Biol Blood Marrow Transplant. 2016;22(11):1915‐1925.
    1. Cole TJ, Bellizzi MC, Flegal KM, Dietz WH. Establishing a standard definition for child overweight and obesity worldwide: international survey. BMJ. 2000;320(7244):1240‐1243.
    1. Centers for Disease Control and Prevention (CDC) . National Health and Nutrition Examination Survey 2005–2014. . Accessed May 4, 2017.
    1. Anderson BJ, Holford NH. Mechanism‐based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 2008;48:303‐332.
    1. Czerwinski M, Gibbs JP, Slattery JT. Busulfan conjugation by glutathione S‐transferases alpha, mu, and pi. Drug Metab Dispos. 1996;24(9):1015‐1019.
    1. Bussler S, Vogel M, Pietzner D, et al. New pediatric percentiles of liver enzyme serum levels (alanine aminotransferase, aspartate aminotransferase, γ‐glutamyltransferase): effects of age, sex, body mass index, and pubertal stage. Hepatology. 2018;68(4):1319‐1330.
    1. ten Brink MH, van Bavel T, Swen JJ, et al. Effect of genetic variants GSTA1 and CYP39A1 and age on busulfan clearance in pediatric patients undergoing hematopoietic stem cell transplantation. Pharmacogenomics. 2013;14(14):1683‐1690.
    1. Hassan M, Ljungman P, Bolme P, et al. Busulfan bioavailability. Blood. 1994;84(7):2144‐2150.
    1. Hassan M, Ehrsson H, Wallin I, Eksborg S. Pharmacokinetic and metabolic studies of busulfan in rat plasma and brain. Eur J Drug Metab Pharmacokinet. 1988;13(4):301‐305.

Source: PubMed

3
订阅