BASIC PHARMACOKINETICS

Similar documents
Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

BIOPHARMACEUTICS and CLINICAL PHARMACY

Applied Biopharmaceutics & Pharmacokinetics Sixth Edition

PHA5128 Dose Optimization II Case Study I Spring 2013

UNIVERSITY OF THE WEST INDIES, ST AUGUSTINE

Basic Concepts of TDM

General Principles of Pharmacology and Toxicology

PHARMACOKINETICS OF DRUG ABSORPTION

General Principles of Pharmacology and Toxicology

Pharmacokinetics Overview

TDM. Measurement techniques used to determine cyclosporine level include:

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

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors

PHAR 7633 Chapter 20 Non Compartmental Analysis

1. If the MTC is 100 ng/ml and the MEC is 0.12 ng/ml, which of the following dosing regimen(s) are in the therapeutic window?

PHA 5127 FINAL EXAM FALL On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Multiple IV Bolus Dose Administration

Define the terms biopharmaceutics and bioavailability.

Volume 1(3) May-June 2013 Page 351

Slide 1. Slide 2. Slide 3. Drug Action and Handling. Lesson 2.1. Lesson 2.1. Drug Action and Handling. Drug Action and Handling.

Biopharmaceutics Lecture-11 & 12. Pharmacokinetics of oral absorption

Basic Pharmacokinetic Principles Stephen P. Roush, Pharm.D. Clinical Coordinator, Department of Pharmacy

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

Determination of bioavailability

CLINICAL PHARMACOKINETICS INDEPENDENT LEARNING MODULE

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

C OBJECTIVES. Basic Pharmacokinetics LESSON. After completing Lesson 2, you should be able to:

Nonlinear Pharmacokinetics

WHY... 8/21/2013 LEARNING OUTCOMES PHARMACOKINETICS I. A Absorption. D Distribution DEFINITION ADME AND THERAPEUIC ACTION

Renal Function. 1. Glomerular filtration 2. Active tubular secretion 3. Passive tubular reabsorption 4. Excretion

Pharmaceutics I صيدالنيات 1. Unit 2 Route of Drug Administration

Guideline for Bioequivalence Studies of Generic Products

Basic Concepts in Pharmacokinetics. Leon Aarons Manchester Pharmacy School University of Manchester

Introduction to Pharmacokinetics (PK) Anson K. Abraham, Ph.D. Associate Principal Scientist, PPDM- QP2 Merck & Co. Inc., West Point, PA 5- June- 2017

Basic Biopharmaceutics, Pharmacokinetics, and Pharmacodynamics

Pharmacokinetics of Drugs. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

Drug Dosing in Renal Insufficiency. Coralie Therese D. Dimacali, MD College of Medicine University of the Philippines Manila

Osnove farmakokinetike. Aleš Mrhar. Prirejeno po. A First Course in Pharmacokinetics and Biopharmaceutics by David Bourne,

2- Minimum toxic concentration (MTC): The drug concentration needed to just produce a toxic effect.

Tamer Barakat. Abdul Aziz ALShamali. Abdul Aziz ALShamali

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

Introduction to. Pharmacokinetics. University of Hawai i Hilo Pre-Nursing Program NURS 203 General Pharmacology Danita Narciso Pharm D

PHARMACOKINETICS SMALL GROUP I:

Development of Canagliflozin: Mechanistic Absorption Modeling During Late-Stage Formulation and Process Optimization

Biomath M263 Clinical Pharmacology

Revised European Guideline on PK and Clinical Evaluation of Modified Release Dosage Forms

PHA Final Exam Fall 2006

Pharmacokinetics of drug infusions

FDB FOOD AND DRUGS BOARD G H A N A GUIDELINES FOR CONDUCTING BIOEQUIVALENCE STUDIES

Flecainide pharmacokinetics in healthy volunteers: the influence of urinary ph

It the process by which a drug reversibly leaves blood and enter interstitium (extracellular fluid) and/ or cells of tissues.

Tutorial. & In case studies 1 and 2, we explore intravenous iv. & Then, we move on to extravascular dosing in case

CEDIAMATE Metformin Tablets USP 500 mg

Nontraditional PharmD Program PRDO 7700 Pharmacokinetics Review Self-Assessment

PHA First Exam. Fall 2004

Dr. M.Mothilal Assistant professor

Understand the physiological determinants of extent and rate of absorption

Section 5.2: Pharmacokinetic properties

USING PBPK MODELING TO SIMULATE THE DISPOSITION OF CANAGLIFLOZIN

PHARMACOLOGY-1 PHL-313. Ali Alhoshani Office: 2B 84

Metformin Associated Lactic Acidosis. Jun-Ki Park 9/6/11

Lippincott Questions Pharmacology

1. Gastric Emptying Time Anatomically, a swallowed drug rapidly reaches the stomach. Eventually, the stomach empties its content in the small

Pharmacokinetics I. Dr. M.Mothilal Assistant professor

Principles of Toxicokinetics/Toxicodynanics

NONLINEAR PHARMACOKINETICS: INTRODUCTION

PHA Final Exam Fall 2001

PART 1. Pharmacology

Model Answer B.Pharm. VII sem, Examination 2013 Biopharmaceutics and Pharmacokinetics Paper code: AS-2532

PHA First Exam Fall 2003

COMPARTMENTAL ANALYSIS OF DRUG DISTRIBUTION Juan J.L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program September 23, 2010

Using Accelerator Mass Spectrometry to Explain the Pharmacokinetics of Vismodegib Cornelis E.C.A. Hop

Use of PBPK in simulating drug concentrations in pediatric populations: Case studies of Midazolam and Gabapentin

Pharmacokinetics Dr. Iman Lec. 3

Pharmacogenetics and Pharmacokinetics

PHAR 7632 Chapter 16

NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES S3A

Received: ; Revised; Accepted: A REVIEW ON BIOAVAILABILITY AND BIOEQUIVALENCE STUDY Shashi Kant*, Bharat Parashar

Rational Dose Prediction. Pharmacology. φαρμακον. What does this mean? pharmakon. Medicine Poison Magic Spell

1 Introduction: The Why and How of Drug Bioavailability Research

Mechanisms of Drug Action

Click to edit Master title style

ICU Volume 11 - Issue 3 - Autumn Series

Biopharmaceutics. Lec: 4

METHODS OF STUDYING BIOAVAILABILITY AND BIOEQUIVALENCE

Metformin: Mechanistic Absorption Modeling and IVIVC Development

Pharmacokinetics of strong opioids. Susan Addie Specialist palliative care pharmacist

Excretion of Drugs. Prof. Hanan Hagar Pharmacology Unit Medical College

Interchangeable Drug Products - Additional Criteria

One-Compartment Open Model: Intravenous Bolus Administration:

Assem Al Refaei. Sameer Emeish. Dr.Alia. Hodaifa Ababneh & Abdullah Shurafa

Pharmacokinetic Modeling & Simulation in Discovery and non-clinical Development

Introduction to. Pharmacokinetics. University of Hawai i Hilo Pre-Nursing Program NURS 203 General Pharmacology Danita Narciso Pharm D

Pharmacokinetics of Rifampicin in African Children Evaluation of the new WHO dosing guidelines

Biopharmaceutics. Tips Worth Tweeting. Contributor: Sandra Earle

Pharmacokinetics PCTH 325. Dr. Shabbits September 12, C t = C 0 e -kt. Learning Objectives

INTRODUCTION TO PHARMACOKINETICS

LD = (Vd x Cp)/F (Vd x Cp)/F MD = (Css x CL x T)/F DR = (Css x (Vm-DR))/Km Css = (F x D)/(CL x T) (Km x DR)/(Vm DR)

Renal Impairment From Dettli to Guideline: What can we learn?

The importance of clearance

Transcription:

BASIC PHARMACOKINETICS MOHSEN A. HEDAYA CRC Press Taylor & Francis Croup Boca Raton London New York CRC Press is an imprint of the Taylor & Francis Group, an informa business

Table of Contents Chapter 1 Introduction to Biopharmaceutics and Pharmacokinetics 1 1.1 Introduction 1 1.2 Application of Biopharmaceutic and Pharmacokinetic Principles in Biomedical Fields 2 1.2.1 Drug Formulation Design 2 1.2.2 Drug Dosage Form Design 2 1.2.3 Pharmacological Testing 3 1.2.4 Toxicological Testing 3 1.2.5 Evaluation of Organ Function 3 1.2.6 Dosing Regimen Design 3 1.3 Drug Concentration-Time Profile 3 1.4 Linear and Nonlinear Pharmacokinetics 3 1.4.1 Linear Pharmacokinetics 3 1.4.2 Nonlinear Pharmacokinetics 4 1.5 Pharmacokinetic Modeling 4 1.5.1 Compartmental Modeling 4 1.5.2 Physiological Modeling 4 1.5.3 Noncompartmental Approach 5 1.6 Pharmacokinetic Simulation 5 Questions 5 Chapter 2 Drug Pharmacokinetics Following Single Intravenous Administration 7 2.1 Introduction 7 2.2 Elimination Rate Constant 8 2.2.1 Rate of Drug Elimination 8 2.2.1.1 Rate Constant for Drug Elimination 8 2.2.2 Order of Drug Elimination 8 2.2.2.1 Zero-Order Elimination 8 2.2.2.2 First-Order Elimination 10 2.2.3 Determination of the First-Order Elimination Rate Constant k 13 2.2.4 Mathematical Expressions That Describe the Amount of the Drug in the Body When Elimination Process Follows First-Order Elimination 13 2.2.5 Clinical Importance of the Elimination Rate Constant 15 2.2.6 Summary 16

2.3 Volume of Distribution 16 2.3.1 Relationship between the Drug Amount in the Body and Drug Blood Concentration 16 2.3.2 Drug Protein Binding and Volume of Distribution 17 2.3.3 Determination of Volume of Distribution 19 2.3.4 Clinical Importance of Volume of Distribution 19 2.3.5 Summary 21 2.4 Half-Life 21 2.4.1 Half-Life during Zero-Order and First-Order Elimination 21 2.4.1.1 Zero-Order Elimination 21 2.4.1.2 First-Order Elimination 22 2.4.2 Graphical Determination of Half-Life 22 2.4.3 Clinical Importance of Half-Life 23 2.4.4 Summary 24 2.5 Total Body Clearance 24 2.5.1 Relationship between Total Body Clearance, Volume of Distribution, and the Elimination Rate Constant 24 2.5.2 Determination of Total Body Clearance 25 2.5.3 Total Body Clearance and Volume of Distribution Are Independent Pharmacokinetic Parameters 25 2.5.4 Clinical Importance of Total Body Clearance 26 2.5.5 Summary 26 2.6 Area Under the Curve 27 2.6.1 Factors Affecting Area Under the Curve after a Single IV Bolus Dose 27 2.6.2 Calculation of Area Under the Curve after a Single IV Bolus Dose 28 2.6.3 Clinical Importance of Area Under the Curve 29 2.7 Factors Affecting the Drug Blood Concentration-Time Profile after a Single IV Bolus Dose 30 2.7.1 Dose 30 2.7.2 Volume of Distribution 31 2.7.3 Total Body Clearance 31 Practice Problems 31 Chapter 3 Drug Absorption Following Oral Administration: Biopharmaceutical Considerations 37 3.1 Introduction 37 3.2 Physiological Factors Affecting Oral Drug Absorption 38 3.2.1 Nature of the GIT Membrane 38 3.2.1.1 Passive Diffusion 38 3.2.1.2 Carrier-Mediated Transport 39 3.2.1.3 Paracellular 39 3.2.1.4 Other Mechanisms 39 3.2.2 Gastrointestinal Physiology 39

3.2.2.1 Buccal Cavity 40 3.2.2.2 Esophagus 40 3.2.2.3 Stomach 40 3.2.2.4 Small Intestine 40 3.2.2.5 Large Intestine 41 3.2.2.6 Rectum 41 3.2.3 Effect of Food on Drug Absorption 41 3.2.4 Pathological Conditions Affecting Drug Absorption 41 3.3 Physical Factors Affecting Oral Drug Absorption 42 3.3.1 Drug Physicochemical Properties 42 3.3.1.1 Drug Lipid Solubility 42 3.3.1.2 ph Partition Theory 43 3.3.2 Dissolution of the Drug 44 3.3.2.1 Surface Area 45 3.3.2.2 Diffusion Coefficient 45 3.3.2.3 Thickness of the Unstirred Layer 45 3.3.2.4 Drug Solubility 45 3.4 Dosage Form Characteristics 46 3.4.1 Types of Oral Dosage Forms 47 3.4.1.1 Solutions 47 3.4.1.2 Suspensions 47 3.4.1.3 Capsules 47 3.4.1.4 Tablets 47 3.4.1.5 Coated Tablets 48 3.4.1.6 Sustained-Release Tablets 48 3.4.2 In Vitro Disintegration Test 48 3.4.3 In Vitro Dissolution Test 48 3.4.3.1 Rotating Basket 49 3.4.3.2 Paddle Method 49 3.4.3.3 Other Methods 49 3.4.3.4 Dissolution Requirements 49 3.4.3.5 Correlation of In Vitro Drug Dissolution with In Vivo Drug Absorption 49 Questions 50 Chapter 4 Drug Pharmacokinetics Following Single Oral Drug Administration: Rate of Drug Absorption 51 4.1 Introduction 51 4.2 Drug Absorption after Oral Administration 52 4.3 Plasma Concentration-Time Profile after a Single Oral Dose 54 4.4 Determination of Absorption Rate Constant 56 4.4.1 Method of Residuals 56 4.4.1.1 Lag Time 58 4.4.1.2 Flip Flop of k a and k 58 4.4.2 Wagner-Nelson Method 60

4.5 Clinical Importance of Absorption Rate Constant 62 4.6 Summary 63 Practice Problems 64 Chapter 5 Drug Pharmacokinetics Following Single Oral Drug Administration: Extent of Drug Absorption 67 5.1 Introduction 67 5.1.1 General Definitions 68 5.2 Purpose of Bioavailability and Bioequivalence Studies 69 5.3 Causes for Variation in Drug Bioavailability 69 5.3.1 Factors Related to Drug Formulation and Route of Administration 69 5.3.1.1 Route of Administration 69 5.3.1.2 Dosage Form 69 5.3.1.3 Excipient 70 5.3.2 Factors Related to the Drug 70 5.3.2.1 Drug Solubility 70 5.3.2.2 Drug Partition Coefficient 70 5.3.2.3 Stability and Drug Interaction 70 5.3.3 Factors Related to the Patient 71 5.3.3.1 Individual Variability 71 5.3.3.2 Site of Administration 71 5.3.3.3 Diseases 71 5.3.4 First-Pass Effect 71 5.4 Pharmacokinetic Basis of Drug Bioavailability and Bioequivalence 72 5.5 Determination of Drug Bioavailability 72 5.5.1 Expected Values for Drug Bioavailability 74 5.5.2 Clinical Importance of Bioavailability and Bioequivalence 74 5.6 Calculation of Area under the Curve (Linear Trapezoidal Rule) 75 5.7 Regulatory Requirements for Bioavailability and Bioequivalence 79 5.7.1 Design and Evaluation of Bioequivalence Studies 80 5.7.2 Criteria for Waiver of Bioavailability Requirements 81 5.8 Factors Affecting the Blood Concentration-Time Profile after a Single Oral Dose 81 5.8.1 Dose 82 5.8.2 Bioavailability 82 5.8.3 Total Body Clearance 82 5.8.4 Volume of Distribution 82 5.8.5 Absorption Rate Constant 82 Practice Problems 82

Chapter 6 Steady-State Principle and Drug Pharmacokinetics during Constant-Rate Intravenous Infusion 87 6.1 Introduction 87 6.2 Plasma Concentration during Continuous Constant-Rate IV Drug Administration 88 6.3 Time Required to Reach Steady State 89 6.4 Loading Dose 90 6.5 Determination of the Pharmacokinetic Parameters 92 6.5.1 Total Body Clearance 92 6.5.2 Elimination Rate Constant 92 6.5.3 Volume of Distribution 92 6.6 Effect of Changing the Pharmacokinetic Parameters on Steady-State Plasma Concentration during Constant-Rate IV Infusion 94 6.6.1 Infusion Rate 94 6.6.2 Volume of Distribution 94 6.6.3 Total Body Clearance 94 Practice Problems 95 Chapter 7 Steady State during Multiple Drug Administrations 99 7.1 Introduction 99 7.2 Drug Plasma Concentration-Time Profile during Multiple Drug Administrations 100 7.3 Average Plasma Concentration at Steady State 102 7.4 Time Required to Reach Steady State 104 7.5 Loading Dose 105 7.5.1 Intravenous Drug Administration 105 7.5.2 Extravascular Drug Administration 105 7.6 Drug Accumulation 105 7.7 Controlled-Release Formulations 106 7.8 Effect of Changing the Pharmacokinetic Parameters on Steady-State Plasma Concentration during Repeated Drug Administration 107 7.8.1 Dosing Rate 107 7.8.2 Total Body Clearance 107 7.8.3 Volume of Distribution 107 7.8.4 Absorption Rate Constant 107 7.9 Dosage Regimen Design 107 7.9.1 Factors to Be Considered 107 7.9.1.1 Therapeutic Range of the Drug 107 7.9.1.2 Required Onset of Effect 108 7.9.1.3 Drug Formulation 108 7.9.1.4 Patient Disease State 108

7.9.2 Estimation of Patient Pharmacokinetic Parameters 108 7.9.2.1 Lack of the Patient's Medical History 108 7.9.2.2 Information Available about the Patient's Medical History 108 7.9.2.3 Patient with History of Using the Drug 108 7.9.3 Selection of Dose and Dosing Interval 109 7.9.3.1 Controlled-Release Oral Formulation 109 7.9.3.2 Fast-Release Oral Formulations and IV Bolus Administration 109 7.9.4 Selection of Loading Dose 110 Practice Problems 111 Chapter 8 Renal Drug Elimination 115 8.1 Introduction 115 8.2 Mechanisms of Renal Excretion of Drugs 116 8.2.1 Glomerular Filtration 116 8.2.2 Tubular Secretion 116 8.2.3 Tubular Reabsorption 116 8.3 Determination of Renal Excretion Rate 117 8.3.1 Experimental Determination of Renal Excretion Rate 117 8.3.2 Renal Excretion Rate-Time Profile 118 8.4 Renal Clearance 119 8.4.1 Creatinine Clearance as a Measure of Kidney Function 120 8.5 Cumulative Amount of the Drug Excreted in Urine 121 8.5.1 Determination of Renal Clearance from the Cumulative Amount Excreted in Urine 122 8.6 Determination of Pharmacokinetic Parameters from Renal Excretion Rate Data 123 8.6.1 Elimination Rate Constant and Half-Life (k and t ]/2 ) 123 8.6.2 Renal Excretion Rate Constant k e 123 8.6.3 Volume of Distribution Vd 123 8.6.4 Renal Clearance CL R 123 8.6.5 Fraction of Dose Excreted Unchanged in Urine 124 8.6.6 Bioavailability 124 8.7 Effect of Changing the Pharmacokinetic Parameters on Urinary Excretion of Drugs 127 8.7.1 Dose 127 8.7.2 Total Body Clearance 127 8.7.3 Renal Clearance 127 Practice Problems 128 Chapter 9 Metabolite Pharmacokinetics 131 9.1 Introduction 131 9.2 Simple Model for Metabolite Kinetics 133

9.2.1 Elimination Rate Limitation 135 9.2.2 Formation Rate Limitation 136 9.2.3 Mathematical Description of Elimination Rateand Formation Rate-Limited Metabolites 137 9.2.4 Time to Achieve Maximum Metabolite Concentration 137 9.3 General Model for Metabolite Kinetics 138 9.4 Estimation of Metabolite Pharmacokinetic Parameters 140 9.4.1 Metabolite Elimination Rate Constant 140 9.4.1.1 Elimination Rate-Limited Metabolites 140 9.4.1.2 Formation Rate-Limited Metabolites 140 9.4.2 Fraction of the Parent Drug Converted to a Specific Metabolite (or Amount of Metabolite Formed) 141 9.4.3 Metabolite Clearance 142 9.4.4 Metabolite Volume of Distribution 142 9.4.5 Metabolite Formation Clearance 142 9.5 Effect of Changing the Pharmacokinetic Parameters on Drug and Metabolite Concentration-Time Profiles after a Single IV Drug Administration 145 9.5.1 Drug Dose 145 9.5.2 Drug Total Body Clearance CLp 146 9.5.3 Drug Volume of Distribution Vd 146 9.5.4 Fraction of Drug Dose Converted to Metabolite f m 146 9.5.5 Metabolite Total Body Clearance CL^, 147 9.5.6 Metabolite Volume of Distribution Vd (m, 147 9.6 Steady-State Metabolite Concentration during Repeated Administrations of Parent Drug 147 9.7 Effect of Changing the Pharmacokinetic Parameters on the Steady-State Drug and Metabolite Concentrations during Repeated Drug Administrations 150 9.7.1 Drug Dose 150 9.7.2 Drug Total Body Clearance CI^ 151 9.7.3 Drug Volume of Distribution Vd 151 9.7.4 Fraction of Drug Dose Converted to Metabolite f m 151 9.7.5 Metabolite Total Body Clearance CL^,, 151 9.7.6 Metabolite Volume of Distribution Vd, m, 151 9.8 Metabolite Kinetics after Extravascular Administration of the Parent Drug 151 9.9 Kinetics of Sequential Metabolism 152 Practice Problems 153 Chapter 10 Disease State and Drug Pharmacokinetics 159 10.1 Introduction 159 10.2 Patients with Kidney Dysfunction 159

10.2.1 Factors Affecting the Change in Drug Pharmacokinetics in Patients with Kidney Dysfunction 159 10.2.1.1 Fraction of Dose Excreted Unchanged in Urine 159 10.2.1.2 Degree of Kidney Dysfunction 160 10.2.2 Dosage Adjustment in Patients with Renal Dysfunction 160 10.2.2.1 Determination of Kidney Function 160 10.2.2.2 Determination of the Fraction of Dose Excreted Unchanged in Urine 161 10.2.2.3 Determination of Dosage Requirements in Patients with Reduced Kidney Function 161 10.3 Patients with Liver Diseases 163 10.3.1 Child-Pugh Score 164 Practice Problems 165 Chapter 11 Nonlinear Pharmacokinetics 169 11.1 Introduction 169 11.1.1 Causes of Nonlinear Pharmacokinetics 169 11.1.1.1 Saturable Drug Absorption 169 11.1.1.2 Saturable Protein Binding 169 11.1.1.3 Saturable Renal Elimination 170 11.1.1.4 Saturable Drug Metabolism 170 11.1.1.5 Others 170 11.1.2 Evidence of Nonlinear Pharmacokinetics 170 11.2 Michaelis-Menten Enzyme Kinetics 170 11.2.1 Pharmacokinetic Parameters 172 11.2.2 Plasma Concentration-Time Profile after a Single Intravenous Dose of a Drug Eliminated by a Metabolic Pathway That Follows Michaelis-Menten Kinetics 173 11.2.2.1 After a Single Drug Administration 173 11.2.2.2 After Multiple Drug Administrations 174 11.3 Determination of the Pharmacokinetic Parameters 175 11.3.1 Total Body Clearance 175 11.3.2 Half-Life 176 11.4 Effect of Changing the Pharmacokinetic Parameters on Plasma Concentration-Time Profile 176 11.4.1 Dose 176 11-4.2 V max 176 11.4.3 K m 176 11.5 Oral Administration of Drugs Eliminated by a Michaelis-Menten Process 177 11.6 Pharmacokinetic Parameter Determination and Dosage Recommendation 177 11.6.1 Mathematical Method 177 11.6.2 Direct Linear Plot 178 11.6.3 Linear Transformation Method 180

11.7 Multiple Elimination Pathways 180 Practice Problems 181 Chapter 12 Multicompartment Pharmacokinetic Models 185 12.1 Introduction 185 12.2 Two-Compartment Pharmacokinetic Model 186 12.3 Two-Compartment Pharmacokinetic Model Parameters 189 12.3.1 Definition of the Pharmacokinetic Parameters 189 12.3.2 Mathematical Equation That Describes the Plasma Concentration-Time Profile 190 12.4 Determination of Two-Compartment Pharmacokinetic Model Parameters 191 12.4.1 Method of Residuals 191 12.4.2 Determination of Model Parameters 192 12.4.2.1 Volume of Central Compartment V c 193 12.4.2.2 Area under the Curve (AUC) 193 12.4.2.3 Total Body Clearance CL[. 193 12.4.2.4 First-Order Elimination Rate Constant from Central Compartment k 3 193 12.4.2.5 First-Order Transfer Rate Constant from Peripheral Compartment to Central Compartment k 2 193 12.4.2.6 First-Order Transfer Rate Constant from Central Compartment to Peripheral Compartment k, 194 12.4.2.7 Volume of Distribution at Steady State Vd ss 194 12.4.2.8 Volume of Distribution in Elimination Phase Vd g 194 12.5 Effect of Changing the Pharmacokinetic Parameters on Drug Concentration-Time Profile after a Single IV Dose 196 12.5.1 Dose 196 12.5.2 Volume of Distribution 196 12.5.3 Hybrid Distribution Rate Constant a 197 12.5.4 Hybrid Elimination Rate Constant P 197 12.6 Oral Administration of Drugs That Follow the Two-Compartment Pharmacokinetic Model 197 12.7 Constant Rate IV Administration of Drugs That Follow the Two-Compartment Pharmacokinetic Model 198 12.8 Multiple Drug Administrations 199 12.9 Renal Excretion of Drugs That Follow the Two-Compartment Pharmacokinetic Model 199 12.10 Effect of Changing the Pharmacokinetic Parameters on Drug Distribution between Central and Peripheral Compartments 200 12.10.1 Dose 200 12.10.2 First-Order Transfer Rate Constant from Central to Peripheral Compartment k, 200

12.10.3 First-Order Transfer Rate Constant from Peripheral to Central Compartment k 2 200 12.10.4 First-Order Elimination Rate Constant from Central Compartment k 3 201 12.11 Three-Compartment Pharmacokinetic Model 201 Practice Problems 202 Chapter 13 Drug Pharmacokinetics Following Administration by Intermittent Intravenous Infusion 205 13.1 Introduction 205 13.2 Drug Concentration-Time Profile during Intermittent IV Infusion 206 13.2.1 After First Dose 206 13.2.2 After Repeated Administration before Reaching Steady State 208 13.2.3 At Steady State 209 13.3 Effect of Changing the Pharmacokinetic Parameters on Steady-State Plasma Concentration during Repeated Intermittent IV Infusion 210 13.3.1 Dose 210 13.3.2 Infusion Time 210 13.3.3 Total Body Clearance 210 13.3.4 Volume of Distribution 210 13.4 Application of Pharmacokinetic Principles for Intermittent IV Infusion to Therapeutic Use of Aminoglycoside 210 13.4.1 Pharmacokinetic Characteristics 211 13.4.1.1 Absorption 211 13.4.1.2 Distribution 211 13.4.1.3 Excretion 211 13.4.2 Guidelines for Aminoglycoside Plasma Concentration 211 13.4.3 Extended-Interval Aminoglycoside Dosing Regimen 212 13.5 Individualization of Aminoglycoside Therapy 212 13.5.1 Determination of Initial Dosing Regimen Based on Population Parameters 212 13.5.2 Determination of Patient-Specific Pharmacokinetic Parameters 213 13.5.2.1 If the Patient Is to Receive the First Aminoglycoside Dose 213 13.5.2.2 If the Patient Received Aminoglycosides before but the Steady State Was Not Achieved 215 13.5.2.3 If the Patient Received Aminoglycosides and Steady State Has Been Achieved 216 13.5.3 Determination of the Dosing Regimen Based on the Patient's Specific Parameters 217

13.5.3.1 Selection of Dosing Interval x 217 13.5.3.2 Selection of Dose 217 13.5.3.3 Selection of Loading Dose 217 Practice Problems 221 Chapter 14 Noncompartmental Approach to Pharmacokinetic Data Analysis 225 14.1 Introduction 225 14.2 Noncompartmental Approach in Data Analysis 226 14.3 Mean Residence Time 227 14.3.1 Calculation of AUC and AUMC 228 14.3.1.1 Area Under the Plasma Concentration-Time Curve...228 14.3.1.2 Area Under the First Moment-Time Curve 228 14.3.2 Mean Residence Time after Different Routes of Administration 231 14.3.2.1 Mean Residence Time after Extravascular Administration 231 14.3.2.2 The Mean Residence Time after Constant-Rate IV Infusion 233 14.4 Other Pharmacokinetic Parameters That Can Be Determined Using the Noncompartmental Approach 233 14.5 Determination of Mean Residence Time for Compartmental Models...234 Practice Problems 235 Chapter 15 Physiological Approach to Hepatic Clearance 237 15.1 Introduction 237 15.2 Organ Clearance 237 15.3 Hepatic Extraction Ratio 238 15.4 Intrinsic Clearance (CL int ) 239 15.5 Systemic Bioavailability 239 15.6 Effect of Change in Intrinsic Clearance and Hepatic Blood Flow on Hepatic Clearance, Systemic Availability, and Drug Concentration-Time Profile 240 15.6.1 Low Extraction Ratio Drugs 240 15.6.2 High Extraction Ratio Drugs 243 15.7 Protein Binding and Hepatic Extraction 250 Practice Problems 250 Chapter 16 Pharmacokinetic-Pharmacodynamic Relationship 253 16.1 Introduction 253 16.2 Pharmacodynamic Models 254 16.2.1 Fixed-Effect Model 255 16.2.2 Linear Model 255 16.2.3 Log-Linear Model 256

16.2.4 E max Model 257 16.2.5 Sigmoid E max Model 259 16.3 Link between Pharmacokinetic and Pharmacodynamic Models 260 16.4 Application of Pharmacodynamic Models 260 16.4.1 Duration of Drug Effect 260 16.4.2 Dosing Regimen 261 Practice Problems 261 Chapter 17 Therapeutic Drug Monitoring 263 17.1 Introduction 263 17.2 General Principles of Initiation and Management of Drug Therapy 263 17.3 Drug Blood Concentration versus Drug Dose 264 17.4 Therapeutic Range 265 17.5 Variability in Drug Pharmacokinetics and Response 267 17.5.1 Body Weight 267 17.5.2 Age 267 17.5.2.1 Pediatrics 267 17.5.2.2 Geriatrics 267 17.5.3 Drug-Drug Interactions 268 17.5.4 Genetic Factors 268 17.5.5 Pregnancy 268 17.5.6 Diseases 268 17.5.7 Other Factors 269 17.6 Advantages of Therapeutic Drug Monitoring 269 17.6.1 Facilitate Rapid Achievement of an Appropriate Dosing Regimen 269 17.6.2 Evaluate Existing Dosing Regimen 269 17.6.3 Prophylaxis against Toxicity 269 17.6.4 Distinguish between Pharmacokinetic and Pharmacodynamic Causes of Therapeutic Failure 269 17.6.5 Cost-Effectiveness 269 17.7 Candidate Drugs For Therapeutic Drug Monitoring 270 17.7.1 Drugs with Low Therapeutic Index 270 17.7.2 Drugs with Great Variability in Their Pharmacokinetic Properties 270 17.7.3 Drugs Used in Patients Who Are at High Risk of Toxicity 270 17.8 Methods for Measuring Drug Blood Concentrations 270 17.9 Establishing a Therapeutic Drug Monitoring Service 271 17.9.1 Major Requirements 271 17.9.2 Procedures 271 17.9.2.1 Determination of Initial Dosing Regimen 271

17.9.2.2 Determination of the Patient's Specific Pharmacokinetic Parameters 271 17.9.2.3 Calculation of Dosage Requirements Based on the Patient's Specific Pharmacokinetic Parameters of the Drug 272 Questions 272 Chapter 18 Solutions to Practice Problems 273 Index 285