On approximate solutions in pharmacokinetics

Similar documents
On drug transport after intravenous administration

The general concept of pharmacokinetics

Noncompartmental vs. Compartmental Approaches to Pharmacokinetic Data Analysis Paolo Vicini, Ph.D. Pfizer Global Research and Development David M.

Noncompartmental vs. Compartmental Approaches to Pharmacokinetic Data Analysis Paolo Vicini, Ph.D. Pfizer Global Research and Development David M.

Nonlinear pharmacokinetics

Marsh Model. Propofol

Quantitative Pooling of Michaelis-Menten Equations in Models with Parallel Metabolite Formation Paths

The Calculus Behind Generic Drug Equivalence

Multicompartment Pharmacokinetic Models. Objectives. Multicompartment Models. 26 July Chapter 30 1

Estimating terminal half life by non-compartmental methods with some data below the limit of quantification

APPLICATION OF LAPLACE TRANSFORMS TO A PHARMACOKINETIC OPEN TWO-COMPARTMENT BODY MODEL

PHARMACOKINETIC DERIVATION OF RATES AND ORDERS OF REACTIONS IN MULTI- COMPARTMENT MODEL USING MATLAB

Minto Model. Remifentanil

Fitting PK Models with SAS NLMIXED Procedure Halimu Haridona, PPD Inc., Beijing

PHA 4123 First Exam Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Chapter One. Introduction

MODELING THE ABSORPTION, CIRCULATION, AND METABOLISM OF TIRAPAZAMINE

Modelling a complex input process in a population pharmacokinetic analysis: example of mavoglurant oral absorption in healthy subjects

FOUNDATIONS OF PHARMACOKINETICS

On Assessing Bioequivalence and Interchangeability between Generics Based on Indirect Comparisons

Compartmental Modelling: Eigenvalues, Traps and Nonlinear Models

TK Solver Case Study: Pharmacokinetics

Using ODEs to Model Drug Concentrations within the Field of Pharmacokinetics

Love and differential equations: An introduction to modeling

The Simplex Method: An Example

IVIVC Industry Perspective with Illustrative Examples

NONLINEAR MODELS IN MULTIVARIATE POPULATION BIOEQUIVALENCE TESTING

ONE-COMPARTMENT KINETICS

How Critical is the Duration of the Sampling Scheme for the Determination of Half-Life, Characterization of Exposure and Assessment of Bioequivalence?

MA/ST 810 Mathematical-Statistical Modeling and Analysis of Complex Systems

Chapter 2 Direct Current Circuits

FUNDAMENTALS of SYSTEMS BIOLOGY From Synthetic Circuits to Whole-cell Models

PHAR 7632 Chapter 2 Background Mathematical Material

IDENTIFICATION OF PARAMETERS FOR HILL CURVE USED IN GENERAL ANESTHESIA

MIDW 125 Math Review and Equation Sheet

TMDD Model Translated from NONMEM (NM- TRAN) to Phoenix NLME (PML)

Feedback Control of Dynamic Bipedal Robot Locomotion

Functions. Copyright Cengage Learning. All rights reserved.

Estimation and Model Selection in Mixed Effects Models Part I. Adeline Samson 1

Solving the Van Der Pol nonlinear differential equation using first order approximation perturbation method

Distribution Theory. Comparison Between Two Quantiles: The Normal and Exponential Cases

Reaction Rate (Kinetics) Reaction Rate

PET Tracer Kinetic Modeling In Drug

- 1 - By H. S Steyn, Statistical Consultation Services, North-West University (Potchefstroom Campus)

ENZYME ACTIVITY AS A FUNCTION of ph

Evidence for the Existence of Non-monotonic Dose-response: Does it or Doesn t it?

Correction of the likelihood function as an alternative for imputing missing covariates. Wojciech Krzyzanski and An Vermeulen PAGE 2017 Budapest

Opportunities for Regulatory Relief via In Vitro Dissolution. James E. Polli June 10, 2015

SAMPLE SIZE RE-ESTIMATION FOR ADAPTIVE SEQUENTIAL DESIGN IN CLINICAL TRIALS

Beka 2 Cpt: Two Compartment Model - Loading Dose And Maintenance Infusion

As mentioned in the introduction of the manuscript, isoboles are commonly used to analyze

The Concept of Equilibrium

1 Lyapunov theory of stability

Radiological Control Technician Training Fundamental Academic Training Study Guide Phase I

13 TH NEW ENZYMOLOGY KINETICS WORKSHOP

BE 159: Signal Transduction and Mechanics in Morphogenesis

Singular perturbation theory

Hierarchical expectation propagation for Bayesian aggregation of average data

Mathematical Model of Catalytic Chemical Reactor. Sintayehu Agegnehu Matintu* School of Mathematical and Statistical Sciences, Hawassa University,

MULTISTAGE AND MIXTURE PARALLEL GATEKEEPING PROCEDURES IN CLINICAL TRIALS

Lecture Notes: Markov chains

SOLUTIONS MANUAL FOR. Thermodynamics in Material Science, Second Edition. Robert DeHoff

Lecture 2: Recurrence relations and population dynamics

A Dynamical Systems Analysis of the Indirect Response Model with Special Emphasis on Time to Peak Response 1

REVIEW REVIEW. Quantum Field Theory II

Quantum Field Theory II

ABC methods for phase-type distributions with applications in insurance risk problems

Review of scalar field theory. Srednicki 5, 9, 10

Early Stages of Drug Discovery in the Pharmaceutical Industry

In this section, thermoelasticity is considered. By definition, the constitutive relations for Gradθ. This general case

01 Harmonic Oscillations

Introduction to Pharmacokinetic Modelling Rationale

DRUG DRUG INTERACTION PREDICTION ASSESSMENT

State and Parameter Estimation Based on Filtered Transformation for a Class of Second-Order Systems

Hardy s Paradox. Chapter Introduction

Non-Instantaneous Impulses in Differential Equations

Fundamentals. Copyright Cengage Learning. All rights reserved.

Homework 1 (PHA 5127)

Correspondence should be addressed to M. G. Shambhu;

Partial Differential Equations with MATLAB

Principles of Drug Design

GI Simulation Methods

Geometric series and effective medicine dosage

Boltzmann Distribution Law (adapted from Nash)

Lecture 2 ENSO toy models

For the reaction: A B R f = R r. Chemical Equilibrium Chapter The Concept of Equilibrium. The Concept of Equilibrium

Carbon labeling for Metabolic Flux Analysis. Nicholas Wayne Henderson Computational and Applied Mathematics. Abstract

Exact Solution of a Constrained. Optimization Problem in Thermoelectric Cooling

Estimation of AUC from 0 to Infinity in Serial Sacrifice Designs

Reaxys Medicinal Chemistry Fact Sheet

CONTROL SYSTEMS, ROBOTICS AND AUTOMATION - Vol. VII - System Characteristics: Stability, Controllability, Observability - Jerzy Klamka

Engineering Pharmacology: Pharmacokinetic Models Using Recursive Finite Difference Equations

A nonlinear feedback model for tolerance and rebound

PODSYPANIN S PAPER ON THE LENGTH OF THE PERIOD OF A QUADRATIC IRRATIONAL. Copyright 2007

JUST THE MATHS UNIT NUMBER 1.3. ALGEBRA 3 (Indices and radicals (or surds)) A.J.Hobson

MEDCHEM 562 Kent Kunze Lecture 2. Physicochemical Properties of Drugs and Drug Disposition. Stereochemistry (Double the trouble. or double the fun?

! 4 4! o! +! h 4 o=0! ±= ± p i And back-substituting into the linear equations gave us the ratios of the amplitudes of oscillation:.»» = A p e i! +t»»

ON THE ARROW OF TIME. Y. Charles Li. Hong Yang

The Temperature of a System as a Function of the Multiplicity and its Rate of Change

Chapter 18. Remarks on partial differential equations

Transcription:

On separation of time scales in pharmacokinetics Piekarski S, Rewekant M. IPPT PAN, WUM Abstract A lot of criticism against the standard formulation of pharmacokinetics has been raised by several authors. It seems that the natural reaction for that criticism is to comment it from the point of view of the theory of conservation laws. Simple example of balance equations for the intravenous administration of drug has been given in 0 and the corresponding equations for extravasal administration are in the text. In principle, the equations of that kind allow one to describe in the self consistent manner different processes of administration, distribution, metabolism and elimination of drugs. Moreover, it is possible to model different pharmacokinetic parameters of the non-compatmental pharmacokinetics and therefore to comment Rosigno s criticism. However, for practical purposes one needs approximate methods, in particular, those based on separation of the time scales. In this text, such method is described and its effectiveness is discussed. Basic equations are in the next chapter. Final remarks are at the end of the text. On approximate solutions in pharmacokinetics In order to generalize system of equations describing intravenous administration [6] to the case of extravenous administration one has to add the additional term describing the rate of absorption of a drug in the plasma [5]. Moreover, in this text the processes of elimination of a drug from the central compartment shall be described by the arbitrary function [ ] instead of the linear expression - (this modification is not very important since most drugs are eliminated according to the linear formula).

Therefore, we start with the following equations: [ ] [ ] () () (3) [ ], (4) where s(t) denotes free drug concentration in the central compartment, 0,, 0 denotes protein concentration,, 0 denotes the concentration of the drug protein complex, k i k are the reaction rates ( on and off rates, k >0, k >0). is the quantity of a drug present in the peripheral compartment at the time instant, is the volume of the peripheral compartment, denoters the concentration of a free drug in the peryphecic compartment, (5) denotes the volume of the central compartment (usually 5 or 6 litres). The total quantity of a drug in the time instant therefore reads [ ] After comparing () and (3) one can see that the sum (6) (7) 0. (8) is a constant of motion which shall be denoted as : After insertion.

into () - (4) one arrives at e(t) c(t) [ c(t)] [ ] [ c(t)] and [ ] [ ] c(t) [ ] [ c(t)] [ c(t)] [ c(t)] (9) (0) () () (3) [ ] [ c(t)] [ c(t)] [ ]. (4) (5) (6) (7) It is worth to stress that the invariant is not a dynamical variable and equations (5) and (6) are dependent (they can be transformed one into another after multiplication with -) and therefore we are left with the following system of three equations: [ c(t)] [ ] [ c(t)] [ ] (8) (9)

[ ]. (0) In order to discuss the problem of time scales for the system (8) (0) one should take into account that according to the pharmacokinetic literature the processes of drug protein binding occur in the time scales of miliseconds. Let us start our discusion with writing equations () (4) in the following equivalent form: [ ] [ ] [ ]. () () (3) (4) The Guldberg - Waage law states that the condition of temporary chemical equilibrium between the concentrations of the complex and the concentrations of substrates and is. (5) Therefore, the state of chemical equilibrium is characterized by (6) while the relaxation of the system towards chemical equilibrium is characterized by value of the parameter. Now we want to determine the form of our equations in the limit, (7) without entering in rigorous derivations (some control on the validity of this approximation can be obtained by numerical experiments): in the limit (7) one

can assume that all quantities in cemtral compartment are in a temporary chemical equilibrium 0. (8) However, such an approximation has to take into account the exact identity (8) and therefore it is better to start with writing equations (8) (0) [ c(t)] [ c(t)] [ ] [ c(t)] [ ] [ ] in the form [ ] (30) (3) [ ] [ c(t)] (9) [ ]. (3) (33) (34) In order to get more insight in the nature of equations (3) (34) in the limit (7) let us write (35) and then [ c(t)] [ ] [ ] [ c(t)] (36) (37)

[ ]. (38) One can see that the perturbation introduces by small is singular and here we just quess the final form of the limit equations taking into account the constraint imposed by fixed value of the invariant for the condition of the temporal chemical equilibrium Those equations are [ c(t)] 0. [ ] [ ] (39),, [ ] (40) (4) and can bo solved for while c(t) can by determined from the condition of the temporal chemical equilibrium. Additionally, the initial condition has to be correspondingly changed and this change shall be discussed later by means of a simple example. The symbol,, in (40) denotes the fraction of the free form of a drug in central comparetment (which is represented here as a function of quantities,, ). In order to determine the explicit expression for,, let us consider first two arbitrary non negative quantities s(t) i c(t) and let and. (4) (43) After dividing simultaneously the nominator and denominator in (4) by s(t) one arives at.

In order to arrive at the explicit formula for the quotient (44) for Guldberg Waage formula for temporary chemical equilibrium one can apply the identities., (45) The identity (8) can be transformed to the form (46) and (46) can be inserted into (45) what results in and later Therefore [ ]. later and finally [, ] [ ] [. ] After inserting (5) into (44) one obtains (47) (48) (49) (50) (5) (5)

[ ] [ ( ( ] ). ) (53) Now all parts of the system (40) (4) are explicitly known and this system can be solved for and. Later can be determined from the identity (5). Let us illustrate this scheme on a simple example. The system (40),(4) describes extravenous administration of a drug (derscribed by the term ) and allows the nonlinear elimination elimination of a drug. It is well known that most drugs are eliminated according to the linear expression []: [ ]. (54) For the case of intravenous administration and fot the linear elimination our system reduces to [ ] [ ]. (55) (56) The initial conditions are of the form ( 0) > 0 ( 0) 0 (57) (58) Those equations can be easily solved but we assume further simplifications leading to (59)

[ ] (60) [ ]. (6) The solution of the system (60),(6) is (6) 4 One can see that (6),(63) for t0 give ( 0) (63) (63) ( 0) 0 (64) (65) After inserting (6) into (5) one can determine the corresponding concentration of the complex (66) ( is defined by (63)). For t 0, the concentration of the complex reads ( 0) [ ]

(67) Therefore, in our model the total concentration of a drug for t0 is ( 0) ( 0) [ ] (68) and the dose of a drug absorbed in a single intravascular administration for t0 is. [ ] (69) It should be stressed that in rigorous model (describing the states outside the chemical equilibrium) the dose absorbed during a single intravenous administration is given only in terms of the free form of a drug. The form of the expression (68) is a result of our approximate scheme of solution. One can see that (64) becomes the sum of exponential functions only in the limit that is, for (70) (7) The formula (59) can be equivalently written also in other forms and (7) which can be convenient for a more detailed discussion. Final remarks (73)

In standard pharmacokinetics one usually assumes that the solutions for the drug concentrations are the sums of the exponential functions [,8]. However, it is not so in our model and the consistency with literature takes place in the linear regime only. In seems important to make numerical simulations of our approximation scheme. We hope that this scheme requires further discussion and that it can be useful for finding approximate solutions of pharmacokinetic problems. Bibliography. T.W.Hermann, Farmakokinetyka, Teoria i Praktyka, Wydawnictwa Lekarskie PZWL, Warszawa 00.. A.Rescigno, Area under the curve and bioavailability, Pharmacological Research, Vol.4, No. 6, 000. 3. S.Piekarski, M.Rewekant, On inconsistencies in passive pharmacokinetics, arxiv:09.755 [pdf] 4. S.Piekarski, M.Rewekant, On the Way to More Convenient Description of Drug Plasma Protein Binding, arxiv:08.47 [pdf] 5. S.Piekarski, M.Rewekant, On applications of conservation laws in pharmacokinetics, arxiv:08.3847 [pdf] 6. S.Piekarski, M.Rewekant, On drug transport after intravenous administration,, arxiv:08.5746 [pdf] 7. A.Kallen, Computational Pharmacokinetics, Chapman&Hall/CRC, New York, 008. 8. P.Macheras, A.Iliadis, Modelling in Biopharmaceutics, Pharmacokinetics, and Pharmacodynamics, Homogeneous and Heterogeneous Approaches, Springer, 006. 9. T.Grabowski, Farmakokinetyka I Biofarmacja, www.biokinetica.pl Copyright Tomasz Grabowski, 000 00. 0.A.Rescigno, Clearance, turnover time, and volume of distribution. Pharmacol Res 997, 35: 89-93..A.Rescigno, Foundations of pharmacokinetics, Pharm.Res. Vol 4, 6, 000, 57-538.. L.Endrenyi, K.K. Midha, Individual bioequivalence Has its time come? Eur.J. of Pharma. Sci., 6, 998, 7-77. 3. S.M. Ebadi, Desk reference of clinical pharmacology, CRC Press Taylor &Francis Group, Boca Raton, 007. 4.A.Rescigno, The Rise and Fall of Compartmental Analysis, Pharmacol Res, Vol.44, No 4, 00. 5.A.Rescigno, G.Segre, On some topological properties of the systems of Compartments, Bulletin of Mathematical Biophysics, Volume 6, 964.

6.A.Rescigno, Pharmacokinetics, science or fiction? Pharmacol Res., Vol.33, No4/5, 996. 7.A.Rescigno, Compartmental Analysis and its Manifold Applications to Pharmacokinetics, The AAPS Journal, Vol, No, March 00. 8.A.Rescigno, Foundations of Pharmacokinetics, Pharmacol Res Vol.4, No 6, 000.