Development of a paediatric population-based model of the pharmacokinetics of rivaroxaban

Stefan Willmann, Corina Becker, Rolf Burghaus, Katrin Coboeken, Andrea Edginton, Jörg Lippert, Hans-Ulrich Siegmund, Kirstin Thelen, Wolfgang Mück, Stefan Willmann, Corina Becker, Rolf Burghaus, Katrin Coboeken, Andrea Edginton, Jörg Lippert, Hans-Ulrich Siegmund, Kirstin Thelen, Wolfgang Mück

Abstract

Background: Venous thromboembolism has been increasingly recognised as a clinical problem in the paediatric population. Guideline recommendations for antithrombotic therapy in paediatric patients are based mainly on extrapolation from adult clinical trial data, owing to the limited number of clinical trials in paediatric populations. The oral, direct Factor Xa inhibitor rivaroxaban has been approved in adult patients for several thromboembolic disorders, and its well-defined pharmacokinetic and pharmacodynamic characteristics and efficacy and safety profiles in adults warrant further investigation of this agent in the paediatric population.

Objective: The objective of this study was to develop and qualify a physiologically based pharmacokinetic (PBPK) model for rivaroxaban doses of 10 and 20 mg in adults and to scale this model to the paediatric population (0-18 years) to inform the dosing regimen for a clinical study of rivaroxaban in paediatric patients.

Methods: Experimental data sets from phase I studies supported the development and qualification of an adult PBPK model. This adult PBPK model was then scaled to the paediatric population by including anthropometric and physiological information, age-dependent clearance and age-dependent protein binding. The pharmacokinetic properties of rivaroxaban in virtual populations of children were simulated for two body weight-related dosing regimens equivalent to 10 and 20 mg once daily in adults. The quality of the model was judged by means of a visual predictive check. Subsequently, paediatric simulations of the area under the plasma concentration-time curve (AUC), maximum (peak) plasma drug concentration (C max) and concentration in plasma after 24 h (C 24h) were compared with the adult reference simulations.

Results: Simulations for AUC, C max and C 24h throughout the investigated age range largely overlapped with values obtained for the corresponding dose in the adult reference simulation for both body weight-related dosing regimens. However, pharmacokinetic values in infants and preschool children (body weight <40 kg) were lower than the 90 % confidence interval threshold of the adult reference model and, therefore, indicated that doses in these groups may need to be increased to achieve the same plasma levels as in adults. For children with body weight between 40 and 70 kg, simulated plasma pharmacokinetic parameters (C max, C 24h and AUC) overlapped with the values obtained in the corresponding adult reference simulation, indicating that body weight-related exposure was similar between these children and adults. In adolescents of >70 kg body weight, the simulated 90 % prediction interval values of AUC and C 24h were much higher than the 90 % confidence interval of the adult reference population, owing to the weight-based simulation approach, but for these patients rivaroxaban would be administered at adult fixed doses of 10 and 20 mg.

Conclusion: The paediatric PBPK model developed here allowed an exploratory analysis of the pharmacokinetics of rivaroxaban in children to inform the dosing regimen for a clinical study in paediatric patients.

Trial registration: ClinicalTrials.gov NCT01145859.

Figures

Fig. 1
Fig. 1
Generic workflow for the PBPK-based scaling of rivaroxaban pharmacokinetics from adults to children. ADME processes processes that involve absorption, distribution, metabolism and excretion of a drug, IV intravenous, PK pharmacokinetic, PBPK physiologically based pharmacokinetic
Fig. 2
Fig. 2
Individually observed (dots) and simulated (lines) plasma concentration–time profiles for rivaroxaban after a 30-min intravenous infusion of 1 mg rivaroxaban in healthy adults depicted as a linear (main graph) and semi-log plot (inset). Simulated data are represented as geometric means (black line), 90 % prediction interval (grey shaded area) and minimum and maximum values (dotted lines)
Fig. 3
Fig. 3
Individually observed (dots) and simulated (lines) plasma concentration–time profiles for rivaroxaban after oral administration of an immediate-release tablet to healthy adults depicted as a linear (main graph) and semi-log plot (inset). Simulated data are represented as geometric means (black line), 90 % prediction interval (grey shaded area) and minimum and maximum values (dotted lines). The graphs show concentration–time profiles of 10 mg (a, c) and 20 mg (b, d) rivaroxaban, under fasting (a, b) and fed (c, d) conditions
Fig. 4
Fig. 4
Gender-pooled paediatric simulations for maximum (peak) plasma drug concentration (Cmax) (a, b), area under the plasma concentration–time curve (AUC) (c, d) and concentration in plasma after 24 h (C24h) (e, f) versus body weight for two different doses of rivaroxaban: 0.143 mg/kg body weight (a, c, e) and 0.286 mg/kg body weight (b, d, f), simulated as oral suspension formulation compared with the adult reference population. Simulated data of the paediatric population are represented as geometric means (blue line) and 90 % prediction interval (grey shaded area). Simulated data of the adult reference population are represented as geometric means (thick red line) and 90 % confidence interval (red shaded area in the background of the graph). Expected body weight ranges for infants, preschool children, children and adolescents are indicated

References

    1. Raffini L, Huang YS, Witmer C, et al. Dramatic increase in venous thromboembolism in children’s hospitals in the United States from 2001 to 2007. Pediatrics. 2009;124:1001–1008. doi: 10.1542/peds.2009-0768.
    1. Goldenberg NA, Donadini MP, Kahn SR, et al. Post-thrombotic syndrome in children: a systematic review of frequency of occurrence, validity of outcome measures, and prognostic factors. Haematologica. 2010;95:1952–1959. doi: 10.3324/haematol.2010.026989.
    1. Kuhle S, Massicotte P, Chan A, et al. Systemic thromboembolism in children. Data from the 1-800-NO-CLOTS Consultation Service. Thromb Haemost. 2004;92:722–728.
    1. Rosendaal FR. Thrombosis in the young: epidemiology and risk factors. A focus on venous thrombosis. Thromb Haemost. 1997;78:1–6.
    1. van Ommen CH, Heijboer H, Büller HR, et al. Venous thromboembolism in childhood: a prospective two-year registry in The Netherlands. J Pediatr. 2001;139:676–681. doi: 10.1067/mpd.2001.118192.
    1. Andrew M, David M, Adams M, et al. Venous thromboembolic complications (VTE) in children: first analyses of the Canadian Registry of VTE. Blood. 1994;83:1251–1257.
    1. Streif W, Ageno W. Direct thrombin and Factor Xa inhibitors in children: a quest for new anticoagulants for children. Wien Med Wochenschr. 2011;161:73–79. doi: 10.1007/s10354-011-0879-5.
    1. Monagle P, Chan AK, Goldenberg NA, et al. Antithrombotic therapy in neonates and children: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141:e737S–e801S. doi: 10.1378/chest.11-2308.
    1. Conroy S, McIntyre J. The use of unlicensed and off-label medicines in the neonate. Semin Fetal Neonatal Med. 2005;10:115–122. doi: 10.1016/j.siny.2004.11.003.
    1. Choonara I. Unlicensed and off-label drug use in children: implications for safety. Expert Opin Drug Saf. 2004;3:81–83. doi: 10.1517/14740338.3.2.81.
    1. Conroy S, Choonara I, Impicciatore P, et al. Survey of unlicensed and off label drug use in paediatric wards in European countries. European Network for Drug Investigation in Children. BMJ. 2000;320:79–82. doi: 10.1136/bmj.320.7227.79.
    1. Cuzzolin L, Atzei A, Fanos V. Off-label and unlicensed prescribing for newborns and children in different settings: a review of the literature and a consideration about drug safety. Expert Opin Drug Saf. 2006;5:703–718. doi: 10.1517/14740338.5.5.703.
    1. ‘t Jong GW, Vulto AG, de Hoog M, et al. A survey of the use of off-label and unlicensed drugs in a Dutch children’s hospital. Pediatrics. 2001;108:1089–1093. doi: 10.1542/peds.108.5.1089.
    1. Roberts R, Rodriguez W, Murphy D, et al. Pediatric drug labeling: improving the safety and efficacy of pediatric therapies. JAMA. 2003;290:905–911. doi: 10.1001/jama.290.7.905.
    1. Kubitza D, Becka M, Wensing G, et al. Safety, pharmacodynamics, and pharmacokinetics of BAY 59-7939—an oral, direct Factor Xa inhibitor—after multiple dosing in healthy male subjects. Eur J Clin Pharmacol. 2005;61:873–880. doi: 10.1007/s00228-005-0043-5.
    1. Kubitza D, Becka M, Voith B, et al. Safety, pharmacodynamics, and pharmacokinetics of single doses of BAY 59-7939, an oral, direct Factor Xa inhibitor. Clin Pharmacol Ther. 2005;78:412–421. doi: 10.1016/j.clpt.2005.06.011.
    1. Kubitza D, Becka M, Zuehlsdorf M, et al. No interaction between the novel, oral direct Factor Xa inhibitor BAY 59-7939 and digoxin [abstract no. 11] J Clin Pharmacol. 2006;46:702.
    1. Kubitza D, Becka M, Zuehlsdorf M, et al. Effect of food, an antacid, and the H2 antagonist ranitidine on the absorption of BAY 59-7939 (rivaroxaban), an oral, direct Factor Xa inhibitor, in healthy subjects. J Clin Pharmacol. 2006;46:549–558. doi: 10.1177/0091270006286904.
    1. Kubitza D, Becka M, Mueck W, et al. Safety, tolerability, pharmacodynamics, and pharmacokinetics of rivaroxaban—an oral, direct Factor Xa inhibitor—are not affected by aspirin. J Clin Pharmacol. 2006;46:981–990. doi: 10.1177/0091270006292127.
    1. Kubitza D, Becka M, Mueck W, et al. Rivaroxaban (BAY 59-7939)—an oral, direct Factor Xa inhibitor—has no clinically relevant interaction with naproxen. Br J Clin Pharmacol. 2007;63:469–476. doi: 10.1111/j.1365-2125.2006.02776.x.
    1. Kubitza D, Mueck W, Becka M. No interaction between rivaroxaban—a novel, oral, direct Factor Xa inhibitor—and atorvastatin [abstract no. P062] Pathophysiol Haemost Thromb. 2008;36:A40.
    1. Kubitza D, Becka M, Mueck W, et al. Effect of co-administration of rivaroxaban and clopidogrel on bleeding time, pharmacodynamics and pharmacokinetics: a phase I study. Pharmaceuticals. 2012;5:279–296. doi: 10.3390/ph5030279.
    1. Perzborn E, Roehrig S, Straub A, et al. The discovery and development of rivaroxaban, an oral, direct Factor Xa inhibitor. Nat Rev Drug Discov. 2011;10:61–75. doi: 10.1038/nrd3185.
    1. Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med. 2008;358:2765–2775. doi: 10.1056/NEJMoa0800374.
    1. Kakkar AK, Brenner B, Dahl OE, et al. Extended duration rivaroxaban versus short-term enoxaparin for the prevention of venous thromboembolism after total hip arthroplasty: a double-blind, randomised controlled trial. Lancet. 2008;372:31–39. doi: 10.1016/S0140-6736(08)60880-6.
    1. Lassen MR, Ageno W, Borris LC, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty. N Engl J Med. 2008;358:2776–2786. doi: 10.1056/NEJMoa076016.
    1. Turpie AGG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised trial. Lancet. 2009;373:1673–1680. doi: 10.1016/S0140-6736(09)60734-0.
    1. The EINSTEIN Investigators Oral rivaroxaban for symptomatic venous thromboembolism. N Engl J Med. 2010;363:2499–2510. doi: 10.1056/NEJMoa1007903.
    1. The EINSTEIN-PE Investigators Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. N Engl J Med. 2012;366:1287–1297. doi: 10.1056/NEJMoa1113572.
    1. Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–891. doi: 10.1056/NEJMoa1009638.
    1. Mega JL, Braunwald E, Wiviott SD, et al. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012;366:9–19. doi: 10.1056/NEJMoa1112277.
    1. Stampfuss J, Kubitza D, Becka M, et al. The effect of food on the absorption and pharmacokinetics of rivaroxaban. Int J Clin Pharmacol Ther. 2013;51:549–61.
    1. Strougo A, Eissing T, Yassen A, et al. First dose in children: physiological insights into pharmacokinetic scaling approaches and their implications in paediatric drug development. J Pharmacokinet Pharmacodyn. 2012;39:195–203. doi: 10.1007/s10928-012-9241-9.
    1. Edginton AN, Schmitt W, Willmann S. Development and evaluation of a generic physiologically based pharmacokinetic model for children. Clin Pharmacokinet. 2006;45:1013–1034. doi: 10.2165/00003088-200645100-00005.
    1. Zhao P, Zhang L, Grillo JA, et al. Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review. Clin Pharmacol Ther. 2011;89:259–267. doi: 10.1038/clpt.2010.298.
    1. Leong R, Vieira ML, Zhao P, et al. Regulatory experience with physiologically based pharmacokinetic modeling for pediatric drug trials. Clin Pharmacol Ther. 2012;91:926–931. doi: 10.1038/clpt.2012.19.
    1. Weinz C, Schwarz T, Kubitza D, et al. Metabolism and excretion of rivaroxaban, an oral, direct Factor Xa inhibitor, in rats, dogs and humans. Drug Metab Dispos. 2009;37:1056–1064. doi: 10.1124/dmd.108.025569.
    1. Edginton AN. Knowledge-driven approaches for the guidance of first-in-children dosing. Paediatr Anaesth. 2011;21:206–213. doi: 10.1111/j.1460-9592.2010.03473.x.
    1. Thelen K, Coboeken K, Willmann S, et al. Evolution of a detailed physiological model to simulate the gastrointestinal transit and absorption process in humans, part 1: oral solutions. J Pharm Sci. 2011;100:5324–5345. doi: 10.1002/jps.22726.
    1. Thelen K, Coboeken K, Willmann S, et al. Evolution of a detailed physiological model to simulate the gastrointestinal transit and absorption process in humans, part II: extension to describe performance of solid dosage forms. J Pharm Sci. 2012;101:1267–1280. doi: 10.1002/jps.22825.
    1. Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94:1259–1276. doi: 10.1002/jps.20322.
    1. Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95:1238–1257. doi: 10.1002/jps.20502.
    1. Rodgers T, Rowland M. Mechanistic approaches to volume of distribution predictions: understanding the processes. Pharm Res. 2007;24:918–933. doi: 10.1007/s11095-006-9210-3.
    1. Kubitza D, Becka M, Mueck W, et al. Effects of renal impairment on the pharmacokinetics, pharmacodynamics and safety of rivaroxaban, an oral, direct Factor Xa inhibitor. Br J Clin Pharmacol. 2010;70:703–712. doi: 10.1111/j.1365-2125.2010.03753.x.
    1. Bayer Pharma AG. Xarelto® (rivaroxaban) summary of product characteristics; 2013. (Accessed 22 May 2013).
    1. Kubitza D, Roth A, Becka M, et al. Effect of hepatic impairment on the pharmacokinetics and pharmacodynamics of a single dose of rivaroxaban—an oral, direct Factor Xa inhibitor. Br J Clin Pharmacol. 2013;76:89–98. doi: 10.1111/bcp.12054.
    1. Gryback P, Hermansson G, Lyrenas E, et al. Nationwide standardisation and evaluation of scintigraphic gastric emptying: reference values and comparisons between subgroups in a multicentre trial. Eur J Nucl Med. 2000;27:647–655. doi: 10.1007/s002590050558.
    1. Thelen K, Coboeken K, Jia Y, et al. Dynamically simulating the effect of food on gastric emptying using a detailed physiological model for gastrointestinal transit and absorption [abstract no. II-36]. Population Approach Group Europe, Venice; 2012.
    1. Johnson KC. Dissolution and absorption modeling: model expansion to simulate the effects of precipitation, water absorption, longitudinally changing intestinal permeability, and controlled release on drug absorption. Drug Dev Ind Pharm. 2003;29:833–842. doi: 10.1081/DDC-120024179.
    1. Willmann S, Thelen K, Becker C, et al. Mechanism-based prediction of particle size-dependent dissolution and absorption: cilostazol pharmacokinetics in dogs. Eur J Pharm Biopharm. 2010;76:83–94. doi: 10.1016/j.ejpb.2010.06.003.
    1. Post TM, Freijer JI, Ploeger BA, et al. Extensions to the visual predictive check to facilitate model performance evaluation. J Pharmacokinet Pharmacodyn. 2008;35:185–202. doi: 10.1007/s10928-007-9081-1.
    1. Bergstrand M, Hooker AC, Wallin JE, et al. Prediction-corrected visual predictive checks for diagnosing nonlinear mixed-effects models. AAPS J. 2011;13:143–151. doi: 10.1208/s12248-011-9255-z.
    1. Edginton AN, Schmitt W, Voith B, et al. A mechanistic approach for the scaling of clearance in children. Clin Pharmacokinet. 2006;45:683–704. doi: 10.2165/00003088-200645070-00004.
    1. Pang KS, Rowland M. Hepatic clearance of drugs. I. Theoretical considerations of a “well-stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokinet Biopharm. 1977;5:625–653. doi: 10.1007/BF01059688.
    1. Edginton AN, Willmann S. Physiology-based simulations of a pathological condition: prediction of pharmacokinetics in patients with liver cirrhosis. Clin Pharmacokinet. 2008;47:743–752. doi: 10.2165/00003088-200847110-00005.
    1. McNamara PJ, Alcorn J. Protein binding predictions in infants. AAPS Pharm Sci. 2002;4:E4. doi: 10.1208/ps040104.
    1. Mahmood B, Daood MJ, Hart C, et al. Ontogeny of P-glycoprotein in mouse intestine, liver, and kidney. J Investig Med. 2001;49:250–257. doi: 10.2310/6650.2001.33969.
    1. Rosati A, Maniori S, Decorti G, et al. Physiological regulation of P-glycoprotein, MRP1, MRP2 and cytochrome P450 3A2 during rat ontogeny. Dev Growth Differ. 2003;45:377–387. doi: 10.1046/j.1440-169X.2003.00699.x.
    1. Pinto N, Halachmi N, Verjee Z, et al. Ontogeny of renal P-glycoprotein expression in mice: correlation with digoxin renal clearance. Pediatr Res. 2005;58:1284–1289. doi: 10.1203/01.pdr.0000188697.99079.27.
    1. Wagner JG. Inter- and intrasubject variation of digoxin renal clearance in normal adult males. Drug Intell Clin Pharm. 1988;22:562–567.
    1. MacFarland RT, Moeller VR, Pieniaszek HJ, Jr, et al. Assessment of the potential pharmacokinetic interaction between digoxin and ethmozine. J Clin Pharmacol. 1985;25:138–143. doi: 10.1002/j.1552-4604.1985.tb02814.x.
    1. Gorodischer R, Jusko WJ, Yaffe SJ. Renal clearance of digoxin in young infants. Res Commun Chem Pathol Pharmacol. 1977;16:363–374.
    1. Halkin H, Radomsky M, Millman P, et al. Steady state serum concentrations and renal clearance of digoxin in neonates, infants and children. Eur J Clin Pharmacol. 1978;13:113–117. doi: 10.1007/BF00609754.
    1. Collins-Nakai RL, Schiff D, Ng PK. Multiple-dose kinetics of digoxin in neonates. Pediatr Pharmacol (New York) 1985;5:117–122.
    1. Iisalo E, Dahl M. Serum levels and renal excretion of digoxin during maintenance therapy in children. Acta Paediatr Scand. 1974;63:699–704. doi: 10.1111/j.1651-2227.1974.tb16993.x.
    1. Willmann S, Lippert J, Schmitt W. From physicochemistry to absorption and distribution: predictive mechanistic modelling and computational tools. Expert Opin Drug Metab Toxicol. 2005;1:159–168. doi: 10.1517/17425255.1.1.159.
    1. Willmann S, Hohn K, Edginton A, et al. Development of a physiology-based whole-body population model for assessing the influence of individual variability on the pharmacokinetics of drugs. J Pharmacokinet Pharmacodyn. 2007;34:401–431. doi: 10.1007/s10928-007-9053-5.
    1. Damkier P, Brosen K. Quinidine as a probe for CYP3A4 activity: intrasubject variability and lack of correlation with probe-based assays for CYP1A2, CYP2C9, CYP2C19, and CYP2D6. Clin Pharmacol Ther. 2000;68:199–209. doi: 10.1067/mcp.2000.108532.
    1. Gaedigk A, Baker DW, Totah RA, et al. Variability of CYP2J2 expression in human fetal tissues. J Pharmacol Exp Ther. 2006;319:523–532. doi: 10.1124/jpet.106.109215.
    1. Rhodin MM, Anderson BJ, Peters AM, et al. Human renal function maturation: a quantitative description using weight and postmenstrual age. Pediatr Nephrol. 2009;24:67–76. doi: 10.1007/s00467-008-0997-5.
    1. Dorne JL, Walton K, Renwick AG. Human variability in CYP3A4 metabolism and CYP3A4-related uncertainty factors for risk assessment. Food Chem Toxicol. 2003;41:201–224. doi: 10.1016/S0278-6915(02)00209-0.
    1. Sy SK, Ciaccia A, Li W, et al. Modeling of human hepatic CYP3A4 enzyme kinetics, protein, and mRNA indicates deviation from log-normal distribution in CYP3A4 gene expression. Eur J Clin Pharmacol. 2002;58:357–365. doi: 10.1007/s00228-002-0487-9.
    1. Garzi A, Messina M, Frati F, et al. An extensively hydrolysed cow’s milk formula improves clinical symptoms of gastroesophageal reflux and reduces the gastric emptying time in infants. Allergol Immunopathol (Madr) 2002;30:36–41. doi: 10.1016/S0301-0546(02)79085-X.
    1. Shaaban SY, Nassar MF, Sawaby AS, et al. Ultrasonographic gastric emptying in protein energy malnutrition: effect of type of meal and nutritional recovery. Eur J Clin Nutr. 2004;58:972–978. doi: 10.1038/sj.ejcn.1601931.
    1. Vivatvakin B, Buachum V. Effect of carob bean on gastric emptying time in Thai infants. Asia Pac J Clin Nutr. 2003;12:193–197.
    1. Fallingborg J, Christensen LA, Ingeman-Nielsen M, et al. Measurement of gastrointestinal pH and regional transit times in normal children. J Pediatr Gastroenterol Nutr. 1990;11:211–214. doi: 10.1097/00005176-199008000-00010.
    1. Ofori-Kwakye K, Fell JT, Sharma HL, et al. Gamma scintigraphic evaluation of film-coated tablets intended for colonic or biphasic release. Int J Pharm. 2004;270:307–313. doi: 10.1016/j.ijpharm.2003.11.009.
    1. Hoekstra JH. Fructose breath hydrogen tests in infants with chronic non-specific diarrhoea. Eur J Pediatr. 1995;154:362–364. doi: 10.1007/BF02072103.
    1. Khin M, Bolin TD, Tin O, et al. Investigation of small-intestinal transit time in normal and malnourished children. J Gastroenterol. 1999;34:675–679. doi: 10.1007/s005350050318.
    1. Soares AC, Lederman HM, Fagundes-Neto U, et al. Breath hydrogen test after a bean meal demonstrates delayed oro-cecal transit time in children with chronic constipation. J Pediatr Gastroenterol Nutr. 2005;41:221–224. doi: 10.1097/01.mpg.0000167499.40074.d7.
    1. Van Den Driessche M, Van Malderen N, Geypens B, et al. Lactose-[13C]ureide breath test: a new, noninvasive technique to determine orocecal transit time in children. J Pediatr Gastroenterol Nutr. 2000;31:433–438. doi: 10.1097/00005176-200010000-00019.
    1. Vreugdenhil G, Sinaasappel M, Bouquet J. A comparative study of the mouth to caecum transit time in children and adults using a weight adapted lactulose dose. Acta Paediatr Scand. 1986;75:483–488. doi: 10.1111/j.1651-2227.1986.tb10234.x.
    1. Madsen JL. Effects of gender, age, and body mass index on gastrointestinal transit times. Dig Dis Sci. 1992;37:1548–1553. doi: 10.1007/BF01296501.
    1. Wagener S, Shankar KR, Turnock RR, et al. Colonic transit time—what is normal? J Pediatr Surg. 2004;39:166–169. doi: 10.1016/j.jpedsurg.2003.10.002.
    1. Helander HF. Quantitative morphological methods in intestinal research. Scand J Gastroenterol Suppl. 1985;112:1–5. doi: 10.3109/00365528509092206.
    1. Willmann S, Edginton AN, Kleine-Besten M, et al. Whole-body physiologically based pharmacokinetic population modelling of oral drug administration: inter-individual variability of cimetidine absorption. J Pharm Pharmacol. 2009;61:891–899. doi: 10.1211/jpp.61.07.0008.
    1. Wilding I, Hirst P, Connor A. Development of a new engineering-based capsule for human drug absorption studies. Pharm Sci Technol Today. 2000;3:385–392. doi: 10.1016/S1461-5347(00)00311-4.
    1. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER). General considerations for pediatric pharmacokinetic studies for drugs and biological products; 1998. (Accessed 19 Mar 2013).
    1. Mueck W, Borris LC, Dahl OE, et al. Population pharmacokinetics and pharmacodynamics of once- and twice-daily rivaroxaban for the prevention of venous thromboembolism in patients undergoing total hip replacement. Thromb Haemost. 2008;100:453–461.
    1. Mueck W, Becka M, Kubitza D, et al. Population model of the pharmacokinetics and pharmacodynamics of rivaroxaban—an oral, direct Factor Xa inhibitor—in healthy subjects. Int J Clin Pharmacol Ther. 2007;45:335–344. doi: 10.5414/CPP45335.
    1. Mueck W, Lensing AW, Agnelli G, et al. Rivaroxaban: population pharmacokinetic analyses in patients treated for acute deep-vein thrombosis and exposure simulations in patients with atrial fibrillation treated for stroke prevention. Clin Pharmacokinet. 2011;50:675–686. doi: 10.2165/11595320-000000000-00000.

Source: PubMed

3
구독하다