Human physiologically based pharmacokinetic model for propofol

David G Levitt, Thomas W Schnider, David G Levitt, Thomas W Schnider

Abstract

BACKGROUND: Propofol is widely used for both short-term anesthesia and long-term sedation. It has unusual pharmacokinetics because of its high lipid solubility. The standard approach to describing the pharmacokinetics is by a multi-compartmental model. This paper presents the first detailed human physiologically based pharmacokinetic (PBPK) model for propofol. METHODS: PKQuest, a freely distributed software routine http://www.pkquest.com, was used for all the calculations. The "standard human" PBPK parameters developed in previous applications is used. It is assumed that the blood and tissue binding is determined by simple partition into the tissue lipid, which is characterized by two previously determined set of parameters: 1) the value of the propofol oil/water partition coefficient; 2) the lipid fraction in the blood and tissues. The model was fit to the individual experimental data of Schnider et. al., Anesthesiology, 1998; 88:1170 in which an initial bolus dose was followed 60 minutes later by a one hour constant infusion. RESULTS: The PBPK model provides a good description of the experimental data over a large range of input dosage, subject age and fat fraction. Only one adjustable parameter (the liver clearance) is required to describe the constant infusion phase for each individual subject. In order to fit the bolus injection phase, for 10 or the 24 subjects it was necessary to assume that a fraction of the bolus dose was sequestered and then slowly released from the lungs (characterized by two additional parameters). The average weighted residual error (WRE) of the PBPK model fit to the both the bolus and infusion phases was 15%; similar to the WRE for just the constant infusion phase obtained by Schnider et. al. using a 6-parameter NONMEM compartmental model. CONCLUSION: A PBPK model using standard human parameters and a simple description of tissue binding provides a good description of human propofol kinetics. The major advantage of a PBPK model is that it can be used to predict the changes in kinetics produced by variations in physiological parameters. As one example, the model simulation of the changes in pharmacokinetics for morbidly obese subjects is discussed.

Figures

Figure 1
Figure 1
Schematic diagram of the PBPK model. The organ "portal" refers to all the organs drained by the portal vein. The connective tissue is divided between two organs: "tendon" with a relatively low blood flow and "other" with a higher blood flow.
Figure 5
Figure 5
Individual PBPK model fits to the experimental data for the young subjects. The values of the 3 parameters that provided the best fit for each individual subject are listed: Tclr = intrinsic liver clearance; frdose = fraction of the bolus dose sequestered in first passage through the lung; T = time constant for release of sequestered propofol. The value in parenthesis following Tclr is the fraction of the total liver blood flow that is cleared (eq. (10)).
Figure 6
Figure 6
Individual PBPK model fits to the experimental data for the middle aged subjects. The values of the 3 parameters that provided the best fit for each individual subject are listed (see fig. 5). For 6 of the 8 subjects there was no pulmonary sequestration of the bolus dose. In order to fit the data for subject 2–4, a small amount of extra-hepatic metabolism was required (kidney clearance = Tclrkid = 35).
Figure 7
Figure 7
Individual PBPK model fits to the experimental data for the old subjects. No pulmonary sequestration of the bolus dose was required in any subject in this age group. Therefore, there is only 1 adjustable parameter (intrinsic liver clearance, Tclr).
Figure 2
Figure 2
Error in fitting the kinetics following the bolus injection (0 to 60 minutes) using the PBPK parameters determined by fitting the constant infusion kinetics (>60 minutes) assuming no pulmonary sequestration. The solid lines are the PBPK model predictions and the squares are the experimental results of Schnider et. al. [20]. The top row shows the entire time course and the bottom shows the first 60 minutes. Absolute concentration on left and semi-log plot of concentration versus time on right.
Figure 3
Figure 3
Same experimental data as in figure 2 except that 60% of the bolus dose has been sequestered in the lung and released with a time constant of 80 minutes.
Figure 4
Figure 4
Effect of pulmonary sequestration of 0, 10, 25 or 50% of the continuously infused dose. Same experimental data as in figure 2 with 60% sequestration of the bolus dose. The sequestered propofol is released with a time constant of 80 minutes.
Figure 8
Figure 8
Age dependence of the fraction sequestered in lung. The dashed line is the linear regression for the data (eq. (13)).
Figure 9
Figure 9
PBPK model fits to the experimental data for the young subjects using the same age dependent parameters for all subjects: fractional liver clearance = 0.76; fraction sequestered described by eq. (13), and T = 80 minutes.
Figure 10
Figure 10
PBPK model fits to the experimental data for the middle-aged subjects using the same age dependent parameters for all subjects (see fig. 9).
Figure 11
Figure 11
PBPK model fits to the experimental data for the old subjects using the same age dependent parameters for all subjects (see fig. 9).
Figure 12
Figure 12
Comparison of NONMEM (Schnider et. al., Table 2 [20]) and PBPK model predictions for a standard male (21% body fat, age 53, weight 77 kg and height 177 cm). The top row shows the model predictions for a 1-hour constant infusion of 100 μg/kg/min followed by a 9 hour washout (left, absolute plot; right, semi-log plot). The middle row shows the model predictions when the washout period is extended to 49 hours. The bottom row compares the model predictions for a 5 day constant infusion of 100 μg/kg/min.
Figure 13
Figure 13
Same as fig. 12, except for an obese female with 47% body fat (age 53, weight 77 kg and height 150 cm).
Figure 14
Figure 14
Comparison of arterial blood concentration in normal weight (black) and morbidly obese subjects (red). The propofol was infused at a constant rate of 0.1 mg/kg/min for 180 minutes and the washout was followed for 8 hours (top panel) or 5 days (bottom panel).
Figure 15
Figure 15
Comparison of time for eye opening after a 180 minute constant infusion in normal weight (black) and morbidly obese subjects (red). Same conditions as fig. 14, but only the first 20 minutes of the washout is plotted. The dashed line indicates the arterial concentration associated with the first eye opening.
Figure 16
Figure 16
Comparison of arterial blood concentration in normal weight (black) and morbidly obese subjects (red). Ten-day constant infusion of 0.1 mg/kg/min.
Figure 17
Figure 17
Comparison of washout kinetics in normal weight (black) and morbidly obese subjects (red). The 10 day constant infusion rate has been adjusted so that the concentration at the end of 10 days is identical for the normal and obese subjects (normal: 0.1 mg/kg/min; obese: 0.058 mg/kg/min). Only the first 60 minutes of the washout is plotted.

References

    1. Weaver BM, Staddon GE, Mapleson WW. Tissue/blood and tissue/water partition coefficients for propofol in sheep. Br J Anaesth. 2001;86:693–703. doi: 10.1093/bja/86.5.693.
    1. Steward A, Allott PR, Cowles AL, Mapleson WW. Solubility coefficients for inhaled anaesthetics for water, oil and biological media. Br J Anaesth. 1973;45:282–293.
    1. Levitt DG. PKQuest: capillary permeability limitation and plasma protein binding - application to human inulin, dicloxacillin and ceftriaxone pharmacokinetics. BMC Clin Pharmacol. 2002;2:7. doi: 10.1186/1472-6904-2-7.
    1. Levitt DG. PKQuest: volatile solutes - application to enflurane, nitrous oxide, halothane, methoxyflurane and toluene pharmacokinetics. BMC Anesthesiol. 2002;2:5. doi: 10.1186/1471-2253-2-5.
    1. Levitt DG. PKQuest: measurement of intestinal absorption and first pass metabolism - application to human ethanol pharmacokinetics. BMC Clin Pharmacol. 2002;2:4. doi: 10.1186/1472-6904-2-4.
    1. Levitt DG. PKQuest: a general physiologically based pharmacokinetic model. Introduction and application to propranolol. BMC Clin Pharmacol. 2002;2:5. doi: 10.1186/1472-6904-2-5.
    1. Levitt DG. The use of a physiologically based pharmacokinetic model to evaluate deconvolution measurements of systemic absorption. BMC Clin Pharmacol. 2003;3:1. doi: 10.1186/1472-6904-3-1.
    1. Levitt DG. The pharmacokinetics of the interstitial space in humans. BMC Clin Pharmacol. 2003;3:3. doi: 10.1186/1472-6904-3-3.
    1. Levitt DG. Physiologically based pharmacokinetic modeling of arterial - antecubital vein concentration difference. BMC Clin Pharmacol. 2004;4:2. doi: 10.1186/1472-6904-4-2.
    1. Gargas ML, Burgess RJ, Voisard DE, Cason GH, Andersen ME. Partition coefficients of low-molecular-weight volatile chemicals in various liquids and tissues. Toxicol Appl Pharmacol. 1989;98:87–99. doi: 10.1016/0041-008X(89)90137-3.
    1. Poulin P, Theil FP. A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically-based pharmacokinetic models in drug discovery. J Pharm Sci. 2000;89:16–35. doi: 10.1002/(SICI)1520-6017(200001)89:1<16::AID-JPS3>;2-E.
    1. Yokogawa K, Ishizaki J, Ohkuma S, Miyamoto K. Influence of lipophilicity and lysosomal accumulation on tissue distribution kinetics of basic drugs: a physiologically based pharmacokinetic model. Methods Find Exp Clin Pharmacol. 2002;24:81–93.
    1. Ishizaki J, Yokogawa K, Nakashima E, Ichimura F. Prediction of changes in the clinical pharmacokinetics of basic drugs on the basis of octanol-water partition coefficients. J Pharm Pharmacol. 1997;49:762–767.
    1. de la Fuente L, Lukas JC, Vazquez JA, Jauregizar N, Calvo R, Suarez E. 'In vitro' binding of propofol to serum lipoproteins in thyroid dysfunction. Eur J Clin Pharmacol. 2002;58:615–619. doi: 10.1007/s00228-002-0520-z.
    1. de la Fuente L, Lukas JC, Jauregizar N, Vazquez JA, Calvo R, Suarez E. Prediction of unbound propofol concentrations in a diabetic population. Ther Drug Monit. 2002;24:689–695. doi: 10.1097/00007691-200212000-00002.
    1. Costela JL, Jimenez R, Calvo R, Suarez E, Carlos R. Serum protein binding of propofol in patients with renal failure or hepatic cirrhosis. Acta Anaesthesiol Scand. 1996;40:741–745.
    1. Altmayer P, Buch U, Buch HP. Propofol binding to human blood proteins. Arzneimittelforschung. 1995;45:1053–1056.
    1. Servin F, Desmonts JM, Haberer JP, Cockshott ID, Plummer GF, Farinotti R. Pharmacokinetics and protein binding of propofol in patients with cirrhosis. Anesthesiology. 1988;69:887–891.
    1. Mazoit JX, Samii K. Binding of propofol to blood components: implications for pharmacokinetics and for pharmacodynamics. Br J Clin Pharmacol. 1999;47:35–42. doi: 10.1046/j.1365-2125.1999.00860.x.
    1. Schnider TW, Minto CF, Gambus PL, Andresen C, Goodale DB, Shafer SL, Youngs EJ. The influence of method of administration and covariates on the pharmacokinetics of propofol in adult volunteers. Anesthesiology. 1998;88:1170–1182. doi: 10.1097/00000542-199805000-00006.
    1. Gallagher D, Visser M, Sepulveda D, Pierson RN, Harris T, Heymsfield SB. How useful is body mass index for comparison of body fatness across age, sex, and ethnic groups? Am J Epidemiol. 1996;143:228–239.
    1. Gallagher D, Heymsfield SB, Heo M, Jebb SA, Murgatroyd PR, Sakamoto Y. Healthy percentage body fat ranges: an approach for developing guidelines based on body mass index. Am J Clin Nutr. 2000;72:694–701.
    1. Claeys MA, Gepts E, Camu F. Haemodynamic changes during anaesthesia induced and maintained with propofol. Br J Anaesth. 1988;60:3–9.
    1. Price ML, Millar B, Grounds M, Cashman J. Changes in cardiac index and estimated systemic vascular resistance during induction of anaesthesia with thiopentone, methohexitone, propofol and etomidate. Br J Anaesth. 1992;69:172–176.
    1. Coates DP, Prys-Roberts C, Spelina KR, Monk CR, Norley I. Propofol ('Diprivan') by intravenous infusion with nitrous oxide: dose requirements and haemodynamic effects. Postgrad Med J. 1985;61 Suppl 3:76–79.
    1. Grounds RM, Twigley AJ, Carli F, Whitwam JG, Morgan M. The haemodynamic effects of intravenous induction. Comparison of the effects of thiopentone and propofol. Anaesthesia. 1985;40:735–740.
    1. Tonner PH, Poppers DM, Miller KW. The general anesthetic potency of propofol and its dependence on hydrostatic pressure. Anesthesiology. 1992;77:926–931.
    1. Hansch C, Leo A, Hoekman D. Exploring QSAR. Hydrophobic, Electronic and Steric Constants. Washington, DC, American Chemical Society; 1995.
    1. Fan SZ, Yu HY, Chen YL, Liu CC. Propofol concentration monitoring in plasma or whole blood by gas chromatography and high-performance liquid chromatography. Anesth Analg. 1995;81:175–178. doi: 10.1097/00000539-199507000-00036.
    1. Coetzee JF, Glen JB, Wium CA, Boshoff L. Pharmacokinetic model selection for target controlled infusions of propofol. Assessment of three parameter sets. Anesthesiology. 1995;82:1328–1345. doi: 10.1097/00000542-199506000-00003.
    1. Ledwozyw A, Michalak J, Stepien A, Kadziolka A. The relationship between plasma triglycerides, cholesterol, total lipids and lipid peroxidation products during human atherosclerosis. Clin Chim Acta. 1986;155:275–283. doi: 10.1016/0009-8981(86)90247-0.
    1. Oliver RE, Jones AF, Rowland M. A whole-body physiologically based pharmacokinetic model incorporating dispersion concepts: short and long time characteristics. J Pharmacokinet Biopharm. 2001;28:27–55.
    1. Roberts MS, Donaldson JD, Rowland M. Models of hepatic elimination: comparison of stochastic models to describe residence time distributions and to predict the influence of drug distribution, enzyme heterogeneity, and systemic recycling on hepatic elimination. J Pharmacokinet Biopharm. 1988;16:41–83. doi: 10.1007/BF01061862.
    1. Ludbrook GL, Upton RN, Grant C, Martinez A. Prolonged dysequilibrium between blood and brain concentrations of propofol during infusions in sheep. Acta Anaesthesiol Scand. 1999;43:206–211. doi: 10.1034/j.1399-6576.1999.430215.x.
    1. Zheng D, Upton RN, Martinez A. Skeletal muscle kinetics of propofol in anaesthetized sheep: effect of altered muscle blood flow. Xenobiotica. 2000;30:1079–1090. doi: 10.1080/00498250010006582.
    1. Gray PA, Park GR, Cockshott ID, Douglas EJ, Shuker B, Simons PJ. Propofol metabolism in man during the anhepatic and reperfusion phases of liver transplantation. Xenobiotica. 1992;22:105–114.
    1. He YL, Ueyama H, Tashiro C, Mashimo T, Yoshiya I. Pulmonary disposition of propofol in surgical patients. Anesthesiology. 2000;93:986–991. doi: 10.1097/00000542-200010000-00019.
    1. Dawidowicz AL, Fornal E, Mardarowicz M, Fijalkowska A. The role of human lungs in the biotransformation of propofol. Anesthesiology. 2000;93:992–997. doi: 10.1097/00000542-200010000-00020.
    1. Mapleson WW. Circulation-time models of the uptake of inhaled anaesthetics and data for quantifying them. Br J Anaesth. 1973;45:319–334.
    1. Servin F, Farinotti R, Haberer JP, Desmonts JM. Propofol infusion for maintenance of anesthesia in morbidly obese patients receiving nitrous oxide. A clinical and pharmacokinetic study. Anesthesiology. 1993;78:657–665.
    1. Rhode BM, Gimmon Z, Shizgal HM. The determination of body fat. J Parenter Enteral Nutr. 1987;11:7S.
    1. Virtanen KA, Lonnroth P, Parkkola R, Peltoniemi P, Asola M, Viljanen T, Tolvanen T, Knuuti J, Ronnemaa T, Huupponen R, Nuutila P. Glucose uptake and perfusion in subcutaneous and visceral adipose tissue during insulin stimulation in nonobese and obese humans. J Clin Endocrinol Metab. 2002;87:3902–3910. doi: 10.1210/jc.87.8.3902.
    1. Alexander JK, Dennis EW, Smith WG, Amad KH, Duncan WC, Austin RC. Blood volume, cardiac output and distribution of systemic blood flow in extreme obesity. Cardiovas Res Cent Bull. 1963;1:39–44.
    1. Zauner CW, Arborelius MJ, Swenson EW, Sundstrom. Lindell SE, Fried M. Arterial-venous differences across the lungs in plasma triglyceride concentration. Respiration. 1977;34:2–8.
    1. Zwetsch B, Bernath M, Decosterd L, Chassot PG, Ravussin P, Gardaz JP. First pass uptake of propofol in the human lung. Anesthesiology. 1996;85:A329.
    1. Gigon VJP, Enderlin F, Scheidegger S. Uber das Schicksal infundieter Fettemulsionen in der menschlichen Lunge. Schweiz med Wschr. 1966;96:71–75.
    1. Jonsson F, Johanson G. Physiologically Based Modeling of the Inhalation Kinetics of Styrene in Humans Using a Bayesian Population Approach. Toxicol Appl Pharmacol. 2002;179:35–49. doi: 10.1006/taap.2001.9331.
    1. Campbell GA, Morgan DJ, Kumar K, Crankshaw DP. Extended blood collection period required to define distribution and elimination kinetics of propofol. Br J Clin Pharmacol. 1988;26:187–190.
    1. Morgan DJ, Campbell GA, Crankshaw DP. Pharmacokinetics of propofol when given by intravenous infusion. Br J Clin Pharmacol. 1990;30:144–148.
    1. Albanese J, Martin C, Lacarelle B, Saux P, Durand A, Gouin F. Pharmacokinetics of long-term propofol infusion used for sedation in ICU patients. Anesthesiology. 1990;73:214–217.
    1. Andersen ME. Toxicokinetic modeling and its applications in chemical risk assessment. Toxicol Lett. 2003;138:9–27. doi: 10.1016/S0378-4274(02)00375-2.

Source: PubMed

3
Abonneren