On drug transport after intravenous administration

Similar documents
On approximate solutions in pharmacokinetics

The general concept of pharmacokinetics

Comparison of log P/D with bio-mimetic properties

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

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

Nonlinear pharmacokinetics

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

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.

CH MEDICINAL CHEMISTRY

In silico pharmacology for drug discovery

A primer on pharmacology pharmacodynamics

FRAUNHOFER IME SCREENINGPORT

CHEM 4170 Problem Set #1

Principles of Drug Design

Saba Al Fayoumi. Tamer Barakat. Dr. Mamoun Ahram + Dr. Diala Abu-Hassan

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

Introduction to medicinal chemisry. Mohammed Nooraldeen (PhD)

Marsh Model. Propofol

Introduction to Chemoinformatics and Drug Discovery

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

Targeted Covalent Inhibitors: A Risk-Benefit Perspective

Early Stages of Drug Discovery in the Pharmaceutical Industry

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

Introduction to medicinal chemisry

ENZYME KINETICS. Medical Biochemistry, Lecture 24

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

Retrieving hits through in silico screening and expert assessment M. N. Drwal a,b and R. Griffith a

Supporting Information

Topic 4: Equilibrium binding and chemical kinetics

On the status of the Michaelis-Menten equation and its implications for enzymology

PHYSIO-CHEMICAL PROPERTIES OF THE DRUG. It is the interaction of the drug with its environment

Research Article. Michaelis-Menten from an In Vivo Perspective: Open Versus Closed Systems. Johan Gabrielsson 1 and Lambertus A.

TRAINING REAXYS MEDICINAL CHEMISTRY

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

Lecture 11: Enzymes: Kinetics [PDF] Reading: Berg, Tymoczko & Stryer, Chapter 8, pp

ASSESSING THE DRUG ABILITY OF CHALCONES USING IN- SILICO TOOLS

MIDW 125 Math Review and Equation Sheet

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

Considering the Impact of Drug-like Properties on the Chance of Success

Optional Second Majors

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

Topology based deep learning for biomolecular data

Transfer Courses and their ISU Equivalent(s)

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

Kinetic Modeling for Vesicle Drug Delivery to the Liver

1.QSAR identifier 1.1.QSAR identifier (title): QSAR model for Human Serum Albumin Binding (logk(hsa))

Lecture 11: Protein Folding & Stability

Protein Folding & Stability. Lecture 11: Margaret A. Daugherty. Fall Protein Folding: What we know. Protein Folding

IVIVC Industry Perspective with Illustrative Examples

Quantum Mechanical Models of P450 Metabolism to Guide Optimization of Metabolic Stability

Receptor Based Drug Design (1)

Love and differential equations: An introduction to modeling

HPLC AND 13 C NMR FOR ANALYSIS OF TRANSAMIDATION IN A DYNAMIC COMBINATORIAL CHEMISTRY SYSTEM

Reaxys Medicinal Chemistry Fact Sheet

Minto Model. Remifentanil

Rapid LC/MS/MS Method. Development for Drug Discovery. Drug discovery is the process of generating compounds. A three-checkpoint paradigm

PHAR 7633 Chapter 12 Physical-Chemical Factors Affecting Oral Absorption

Fluorine in Peptide and Protein Engineering

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

COMBINATORIAL CHEMISTRY: CURRENT APPROACH

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

Problem solving steps

Using mathematical models & approaches to quantify BRAIN (dynamic) Positron Emission Tomography (PET) data

Quantitative structure activity relationship and drug design: A Review

Easier and Better Exploitation of PhysChem Properties in Medicinal Chemistry

Dongyue Cao,, Junmei Wang,, Rui Zhou, Youyong Li, Huidong Yu, and Tingjun Hou*,, INTRODUCTION

Protein Folding & Stability. Lecture 11: Margaret A. Daugherty. Fall How do we go from an unfolded polypeptide chain to a

Body Size is the most important quantitative determinant of drug dose

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

Hydrogen Bonding & Molecular Design Peter

Irreversible Inhibition Kinetics

Principles of Drug Design

Acid-Base Balance. Lecture # 5 Second class/ 2015

CHEM J-3 June Calculate the osmotic pressure of a 0.25 M aqueous solution of sucrose, C 12 H 22 O 11, at 37 C

Advanced Medicinal Chemistry SLIDES B

IDENTIFICATION OF PARAMETERS FOR HILL CURVE USED IN GENERAL ANESTHESIA

Using ODEs to Model Drug Concentrations within the Field of Pharmacokinetics

APPLICATION OF HIGH PERFORMANCE LIQUID CHROMATOGRAPHY FOR THE MEASUREMENTS OF LIPOPHILICITY AND BIO-MIMETIC BINDING PROPERTIES IN DRUG DISCOVERY

PET Tracer Kinetic Modeling In Drug

Introduction into Biochemistry. Dr. Mamoun Ahram Lecture 1

Predicting the Onset of Nonlinear Pharmacokinetics

DRUG DRUG INTERACTION PREDICTION ASSESSMENT

WALKUP LC/MS FOR PHARMACEUTICAL R&D

Relative Drug Likelihood: Going beyond Drug-Likeness

FRAGMENT SCREENING IN LEAD DISCOVERY BY WEAK AFFINITY CHROMATOGRAPHY (WAC )

Preparation And Characterization Of Simvastatin Nanosuspension By Homogenization Method

Lead- and drug-like compounds: the rule-of-five revolution

MODELING THE ABSORPTION, CIRCULATION, AND METABOLISM OF TIRAPAZAMINE

Elementary Binding Equations and Related Equations in Biochemistry

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

Bioengineering Laboratory I. Enzyme Assays. Part II: Determination of Kinetic Parameters Fall Semester

Course Plan for Pharmacy (Bachelor Program) No.: (2016/2017) Approved by Deans Council by decision (09/26/2016) dated (03/08/2016) )160) Credit Hours

Numerical Simulations of Stochastic Circulatory Models

Optimizing Solvent Blends for a Quinary System

arxiv: v1 [q-bio.cb] 13 Jan 2017

COMPUTER AIDED DRUG DESIGN (CADD) AND DEVELOPMENT METHODS

Comparative Analysis of Machine Learning Techniques for the Prediction of the DMPK Parameters Intrinsic Clearance and Plasma Protein Binding

Chemotherapy in heterogeneous cultures of cancer cells with interconversion

Electronegativity and Bond Polarity

Transcription:

On drug transport after intravenous administration S.Piekarski (1), M.Rewekant (2) Institute of Fundamental Technological Research Polish Academy of Sciences (1), Medical University of Warsaw, Poland Abstract A mathematical model of a drug transport after rapid injection is given. It takes into account three processes: - drug plasma protein binding in central compartment - transport processes between the central compartment and the peripheral compartment - elimination of a drug from the central compartment.. Introduction Drug protein binding is an reversible attachment resulting from electrostatic Londonvan der Waals's forces, hydrogen bond or their combinations. This reaction satisfy the law of mass action and lasts milliseconds (1). Unbound fraction of drug in vitro is calculated as a quotient of free drug serum concentration and total serum concentration of drug in the closed system (vials space) and given in percents. In design of molecular structures of new drugs, data from drug protein binding in vitro is not often used successfully (2). This approach was questioned by Smith and co-workers, authors of a free drug hypothesis." They indicated that in human organism (open system), it is free drug concentration, not unbound fraction of drug, that should be considered (3). Concise review of quantification methods used to assessing a degree of drug protein binding was described by Howard and co-workers (4).

Drug molecules weakly bonded with plasma protein may be displaced by strongly bonded molecules. This is an example of protein-drug pharmacokinetic interaction which may be simulated as Christensen and co-workers described (5). Interesting phenomenon of decreasing unbound fraction of drug when the total drug concentration increased in the limit range was recently described by Breshkovsky (6). He suggested the hypothesis of a strong binding site activation by weaker bonding site, present in protein molecules. Free drug concentration changes in time in a living organism because of dissociation of protein-drug complex, distribution and elimination processes and it has influence on the potency of pharmacological ect. How drug protein binding changes affect the pharmacokinetic and pharmacodynamic parameters has been the problem discussed in professional literature for many years. According to some earlier opinions, the drug protein binding in vivo was not clinically significant. There is an opinion that drug protein binding changes affect the clinical outcome due to changes of clearance of a free drug (7). According to Winter there is little clinical evidence of a sharp correlation between free drug concentration and clinical ects (8). Benet and Hoener believed that drug protein binding changes have little ect on bioavailability and clinical outcome of drugs (9). In 29 Buur and co-workers demonstrated physiologically based pharmacokinetic model that linked plasma protein binding interactions to drug disposition for SMZ and FLU in a swine. In a study in vivo they confirmed the presence of a drug interaction related to the problem of tissue residue (1). Recently, Schmidt and co-workers made a review of theoretical conceptions related to drug protein binding, describing a status of unbound fraction of drug in basic linear equations of the most important pharmacokinetic parameters such as: clearance, volume of distribution, steady-state concentration of drug and bioavailability and half-life (11). At present, it is necessary to create mathematical models describing not only the dynamic changes of drug protein binding in time but also ects in a range of pharmacokinetic and pharmacodynamic parameters simultaneously (12). There is a need to have a research tool for simulating drug interactions in all ect compartments. In the work presented below we proposed the mathematical modelling of a sequence of processes after intravenous drug administration.

Basic equations The standard equations of chemical kinetics, relating free drug concentration, protein concentration and the drug protein complex are s e c s k c e s k c e s k c e ( 1 ) ( 2 ) ( 3 ) where all quantities depend on time and k, k are the reaction rates ( k >, k >). The above equations describe the time evolution in an uniform system without external sources. Now, the elimination of a drug is modeled by an additional source term in the balance law 1. In the simplest possible version, such additional source term can by modeled by the term proportional to and the strength of a source can be measured by the proportionality coicient. Therefore, the resulting equations are now s e c e s kc αs ( 4 ) s k c e s k c e ( 5 ) ( 6 ) The equations ( 4) ( 6) still do not take into account the transport processes between the central compartment and the peripherial compartment. Let denote the volume of the central compartment (usually between 5 or 6 litres): then the number of molecules of a free fraction of a drug in a central compartment is. Let denote the nomer of molecules of a drug in a peripherial compartment.

In order to formulate the simplest possible dynamical model of the transport processes between both compartments let us assume that the elimination of a drugi is switched out. Then the conservation law for the transport of the free fraction of a drug between both compartments is [ s N ] =. ( 7 ) Every model of the transport processes between both compartments should identically satisfy the condition ( 7 ). The simplest such model depends On the two parameters and assumes that the volume of the peripherial compartment is and that molecules are distributed with a uniform density N = n ( 8 ) The explicit form of this model is s = β[ n s ] ( 9) n = β [ n s ] ( 1 ) where the real parameter models the rate of the transport processes between both compartments. Now one can see that the final system of evolution equations for the drug transport is s e e β s k c α s [ n s ] ( 11 ) s k c e ( 12 )

c s k c e n. ( 13 ) = β [ n s ] ( 14 ) In future we hope to interprete the solutions of this system in terms of the experimental results. References 1. S.M. Ebadi, Desk reference of clinical pharmacology. CRC Press Taylor & Francis Group, Boca Raton, 27. 2. N.A. Kratochwil, W. Huber, et al., Predicting plasma protein binding of drugs revisited, Curr Opin Drug Discov Devel 7(4) (24) 57-512. 3. D.A. Smith, L. Di, et al. The ect of plasma protein binding on in vivo icacy: misconceptions in drug discovery, Nat. Rev. Drug Discov. 9(12) (21) 929-939. 4. M.L. Howard, J.J. Hill, et al.. Plasma protein binding in drug discovery and development, Comb. Chem. High Throughput Screen 13(2) (21) 17-187. 5. H. Christensen, M. Baker, et al., Prediction of plasma protein binding displacement and its implications for quantitative assessment of metabolic drug-drug interactions from in vitro data, J. Pharm. Sci. 95(12) (26) 2778-2787. 6. L.M. Berezhkovskiy, On the possibility of self-induction of drug protein binding, J. Pharm. Sci., 99 (21) 44 445. 7. N.H.G Holford, Input from the deep south compartment: Personal viewpoint. Clin.Pharmacokinet. 29: (1995) 139. 8. M.E. Winter, Basic clinical pharmacokinetics, Applied Therapeutic, ancouver (1994). 9. L.Z. Benet, B. Hoener, Changes in plasma protein binding have little clinical relevance. Clin. Pharmacol. Ther. 71 (22) 115. 1. J.L.Buur, R.E. Baynes, G.W. Smith, J.E., A physiologically based pharmacokinetic model linking plasma protein binding interactions with drug disposition, Research in eterinary Science, 86 (2) (29) 293-31. 11. S. Schmidt, D. Gonzalez, H. Derendorf., Significance of protein binding in pharmacokinetics and pharmacodynamics, J.Pharm.Sci, 99 (21)117-1122. 12. M.A. Zeitlinger, H. Derendorf, et al., Protein Binding: Do We Ever Learn? Antimicrobial Agents and Chemotherapy, 55(7) 211, 367 374.