The general concept of pharmacokinetics

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

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

Nonlinear 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.

MIDW 125 Math Review and Equation Sheet

Homework 1 (PHA 5127)

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

Description of UseCase models in MDL

Introduction to PK/PD modelling - with focus on PK and stochastic differential equations

On drug transport after intravenous administration

Reaction Rate (Kinetics) Reaction Rate

Marsh Model. Propofol

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

Physiology. Biol 219 Lec 1 Fall The Science of Body Function. Themes of Physiology. Themes of Physiology

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

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

PHAR 7632 Chapter 2 Background Mathematical Material

IVIVC Industry Perspective with Illustrative Examples

Using ODEs to Model Drug Concentrations within the Field of Pharmacokinetics

3 Results. Part I. 3.1 Base/primary model

EUCAST. Linezolid Rationale for the EUCAST clinical breakpoints, version th November 2005

NONLINEAR MODELS IN MULTIVARIATE POPULATION BIOEQUIVALENCE TESTING

MODELING THE ABSORPTION, CIRCULATION, AND METABOLISM OF TIRAPAZAMINE

Nonparametric Bayes approach for a semimechanistic pharmacodynamic model

On approximate solutions in pharmacokinetics

Plot of Laser Operating Current as a Function of Time Laser Test Data

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

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

PET Tracer Kinetic Modeling In Drug

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

Population Pharmacokinetics in Support of Analgesics and Anaesthetics Studies in

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

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

Validation of the GastroPlus TM Software Tool and Applications

Love and differential equations: An introduction to modeling

IDENTIFICATION OF PARAMETERS FOR HILL CURVE USED IN GENERAL ANESTHESIA

ONE-COMPARTMENT KINETICS

PHAR 7633 Chapter 12 Physical-Chemical Factors Affecting Oral Absorption

Introduction to Pharmacokinetic Modelling Rationale

Predicting the Onset of Nonlinear Pharmacokinetics

The Calculus Behind Generic Drug Equivalence

Compartmental Modelling: Eigenvalues, Traps and Nonlinear Models

TK Solver Case Study: Pharmacokinetics

arxiv: v1 [q-bio.cb] 21 Dec 2015

Study No: Title : Rationale: Phase: Study Period: Study Design: Centres: Indication: Treatment: Objectives: Statistical Methods:

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

Author's personal copy. Mathematical and Computer Modelling

Performing Deconvolution Operations in SAS for Input Rate Computation

Determination of Design Space for Oral Pharmaceutical Drugs

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Nonlinear Mixed Effects Models

CTP-656 Tablet Confirmed Superiority Of Pharmacokinetic Profile Relative To Kalydeco in Phase I Clinical Studies

Tetracycline Rationale for the EUCAST clinical breakpoints, version th November 2009

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

Integration of SAS and NONMEM for Automation of Population Pharmacokinetic/Pharmacodynamic Modeling on UNIX systems

Gentamicin Rationale for the EUCAST clinical breakpoints, version th February, 2009

Testing for bioequivalence of highly variable drugs from TR-RT crossover designs with heterogeneous residual variances

Lecture 22: PHARMACY CALCULATIONS for Technicians Preparing Injectable Medications

Physical-Chemistry Factors Affecting Oral Absorption. Objectives. ph - Partition Theory. 27 September Chapter 23 1

Practice of SAS Logistic Regression on Binary Pharmacodynamic Data Problems and Solutions. Alan J Xiao, Cognigen Corporation, Buffalo NY

Nonlinear mixed-effects models using Stata

Minto Model. Remifentanil

Estimation of AUC from 0 to Infinity in Serial Sacrifice Designs

Benchmark Problem: A PK/PD Model and Safety Constraints for Anesthesia Delivery

Nitroxoline Rationale for the NAK clinical breakpoints, version th October 2013

Modeling biological systems - The Pharmaco-Kinetic-Dynamic paradigm

ADME studies with radiolabeled compounds. Biomedical Applications of Radioisotopes and Pharmacokinetics Unit

Most common dose (mg) 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1. Maximum dose schedule (mg) 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1 1g x 1

Basic Concepts in Population Modeling, Simulation, and Model-Based Drug Development Part 2: Introduction to Pharmacokinetic Modeling Methods

Challenges in modelling the pharmacokinetics of isoniazid in South African tuberculosis patients

Optimal Adaptive Designs for Dose Finding in Early Phase Clinical Trials

Handling parameter constraints in complex/mechanistic population pharmacokinetic models

In a linear pharmacokinetic system, plasma concentrations following bolus intravenous injection,

Geometric series and effective medicine dosage

Nonlinear Elimination of Drugs in One-Compartment Pharmacokinetic Models: Nonstandard Finite Difference Approach for Various Routes of Administration

The University of Jordan

CHAPTER 7 SUMMARY AND CONCLUSIONS. constitutes the objective of many a research project in the field of pharmaceutical

METHODS FOR POPULATION PHARMACOKINETICS AND PHARMACODYNAMICS

A primer on pharmacology pharmacodynamics

Alkalosis or alkalemia arterial blood ph rises above Acidosis or acidemia arterial ph drops below 7.35 (physiological acidosis)

Prediction of Oral Absorption: 6-Fluoroquinolones

Pharmacokinetics Calculations

An effective approach for obtaining optimal sampling windows for population pharmacokinetic experiments

MAT 17A - UHP - DISCUSSION #10 December 1, 2015

In my opinion this paper can become suitable for publication, provided below issues are adequately addressed.

PHARMACY CALCULATIONS

Journal home page:

Contribution of different oxazolidinones to the efficacy of novel regimens containing bedaquiline and pretomanid in murine models of TB

Supporting Information

Kinetic Modeling for Vesicle Drug Delivery to the Liver

SECTION 9: Kinetics. Chapter 12 in Chang Text

Mathematical Material. Calculus: Objectives. Calculus. 17 January Calculus

Impact of saturable distribution in compartmental PK models: dynamics and practical use

Principles of Drug Design

A nonlinear feedback model for tolerance and rebound

An Approach for Identifiability of Population Pharmacokinetic-Pharmacodynamic Models

Harmonic Regression in the Biological Setting. Michael Gaffney, Ph.D., Pfizer Inc

ANNOUNCEMENTS. HW5 is posted. It is due by 12:00 on 3/11.

Draft Guideline on Bioanalytical Method (Ligand Binding Assay) Validation in Pharmaceutical Development. (24 January, 2014, MHLW, Japan)

Transcription:

The general concept of pharmacokinetics Hartmut Derendorf, PhD University of Florida

Pharmacokinetics the time course of drug and metabolite concentrations in the body

Pharmacokinetics helps to optimize drug therapy: dose dosage regimen dosage form

What happens to a drug after its administration? ("Fate of drug") Liberation Absorption Distribution Metabolism Excretion

Goal to produce a desired drug concentration in the body using the optimum dosage regimen and dosage form Problem What is the optimum target concentration?

Pharmacokinetics in "normal" subjects Biological variability! Therapeutic drug level monitoring in individual patients

Pharmacokinetic Parameters Clearance Volume of distribution Half-life Bioavailability

Clearance! quantifies ELIMINATION! is the volume of body fluid cleared per time unit (L/h, ml/min)! is usually constant

Clearance Eliminating Organ CL = Q E Blood Flow Extraction Ratio

Clearance Clearance can be calculated from! Excretion rate / Concentration e.g. (mg/h) / (mg/l) = L/h! Dose / Area under the curve (AUC) e.g. mg / (mg h/l) = L/h

Clearance Total body clearance is the sum of the individual organ clearances CL = CL ren + CL hep + CL other

Volume of Distribution Vd = X / Cp - quantifies DISTRIBUTION - relates drug concentration (Cp) to amount of drug in the body (X) - gives information on the amount of drug distributed into the tissues

Apparent Volume of Distribution X X V C1 V C2 C1 = X / V V = X / C1 C1 > C2 V < Vd C2 = X / Vd Vd = X / C2

Volume of Distribution Ibuprofen Gentamicin (ECF) Antipyrine (TBW) Diazepam Digoxin Azithromycin 0.15 L/kg 0.25 L/kg 0.60 L/kg 1.1 L/kg 7.3 L/kg 31 L/kg

Half-Life t 1/ 2 = 0. 693 CL Vd Half-life is the time it takes for the concentration to fall to half of its previous value Half-life is a secondary pharmacokinetic parameter and depends on clearance and volume of distribution

Half-Life ln 2 t = = 1 / 2 k 0.693 k CL = k Vd k CL Vd elimination rate constant clearance volume of distribution

Zero order kinetics First order kinetics C = Co - ko t Co = 100 ng/ml ko = 10 ng/ml/h t [h] C [ng/ml] C = Co exp(-k t) t [h] C [ng/ml] 0 100 0 100 1 90 1 50 Co = 100 ng/ml 2 80 2 25 k = 0.693 1/h 3 70 3 12.5 4 60 4 6.25 5 50 5 3.125 6 40 6 1.563 100 80 100 80 C [ng/ml] 60 40 C [ng/ml] 60 40 20 20 0 0 2 4 6 t [h] 0 0 2 4 6 t [h]

Bioavailability f = AUC AUC po iv - quantifies ABSORPTION f is the fraction of the administered dose that reaches the systemic circulation

Bioavailability Rate and Extent of Absorption 70 Cmax 60 50 Cmax 40 30 20 10 0 0 2 4 6 8 10 12 tmax tmax Time (hours)

Models in Pharmacokinetics Compartment Models Physiological Models Statistical Models

Compartment Models Parameters: Rate constants, intercepts Linear and nonlinear regression Complete concentration-time-profiles Steady-state and non-steady-state

Physiological Models Q E = C i C i C o C i Eliminating Organ C o CL CL = = Q E Q Q + f u f u CL CL int int Parameters: Blood Flow, intrinsic clearance, protein binding Good prediction of changes in clearance Steady state

High-extraction drugs CL = Q << Q f Q + f u u f u CL CL CL int int int CL = Q Low-extraction drugs CL = Q >> Q f Q + f u u f u CL CL CL int int int CL = f u CL int

Statistical Models MRT AUMC = = AUC 0 Cp t dt 0 Cp dt Parameters: Mean residence time, mean absorption time Area calculation by trapezoidal rule Calculation of clearance and volume of distribution Additivity of parameters Steady state

Drug Input Intravenous Bolus First-Order Absorption Zero-Order Absorption

Intravenous bolus One compartment model D X k E D X E Dose Drug in the body Drug eliminated

Intravenous bolus Plasma concentration (single dose) C C = 0 e k t 10 2 C0 C (ng/ml) 10 1 -k/2.3 D 10 0 C = 0 Vd 10-1 0 1 2 3 4 5 6 7 8 Time (hours)

Intravenous bolus Multiple Dose

Intravenous bolus Plasma concentration (multiple dose, steady state) C = C 0 e kt ( 1 e k ) τ Peak Trough C C e 0 ( 1 e k τ ) Cmin = 0 ( 1 e k τ ) Cmax = k τ

The AUC within a dosing interval at steady state is equal to the total AUC of a single dose.

Intravenous bolus Average concentration (multiple dose, steady state) C = D CL τ

First-order absorption One compartment model D f A k a X k E Dose Drug at absorption site Drug in the body Drug eliminated

Oral administration Plasma concentration (single dose) C = F D k ( ) a k k Vd a ( e kt e k t) a

Oral administration

Oral administration Multiple Dose

Oral administration Average concentration (multiple dose, steady state) C = F CL D τ

Zero-order absorption One compartment model D f A R 0 X k E Dose Drug at absorption site Drug in the body Drug eliminated

Constant rate infusion Plasma concentration (during infusion) C R = 0 1 CL ( e kt )

Constant rate infusion

Constant rate infusion Plasma concentration (steady state) C = R0 CL

Two-compartment model D Xc k 10 E k 12 k 21 Xp Dose Xc Drug in the central compartment Xp Drug in the peripheral compartment Drug eliminated

Two-compartment model 10 3 C10 0 3 a C (ng/ml) 10 2 10 1 C (ng/ml) 10 2 b 10 1 β 10 0 10 0 α 10-1 0 2 4 6 8 10 Time (hours) 10-1 0 1 2 3 4 5 Time (hours)

Two-compartment model Plasma concentration (single i.v. bolus dose) α t β t C = a e + b e α-phase: β-phase: distribution phase elimination phase

Two-compartment model Volume of distribution Xc Xc Xc Xp Xp Xp initially steady state elimination phase V c = D C 0 Vd ss = 1 + k k 12 21 V c CL Vd = area β

Two-compartment model Amount of drug in tissue X T = k D 12 ( ) ( e α t e β t ) β α Free concentration in tissue C f T = ( ) k D f 21 1 V c ( β α) b ( e α t e β t )

Two-compartment model 10 3 10 2 C (ng/ml) 10 1 10 0 10-1 0 1 2 3 4 5 6 7 8 Time (hours)

Three-compartment model d Xp D k 31 k 13 Xc k 10 E k 12 k 21 D E Dose Drug eliminated Xp s Xc Drug in the central compartment Xps Drug in the shallow peripheral compartment Xpd Drug in the deep peripheral compartment

Significance of Pharmacokinetic Parameters for Dosing Maintenance Dose Loading Dose R 0 = C F D = τ LD = C desired C CL desired desired CL Vd Fluctuation Dosing Interval F = τ = C C max( desired ) min( desired ) 0 1/ 2.693 t ln( F)

Drug Delivery? Biopharmaceutics Pharmacokinetics? PK-PD-Modeling Pharmacodynamics

Conclusion Pharmacokinetic data is only therapeutically useful if there is a known relationship between drug concentration and drug effect or side effect.