450

Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

  • Upload
    others

  • View
    8

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW
Page 2: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

Page 3: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

BASIC PHARMACOKINETICSAND PHARMACODYNAMICS

Page 4: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

Page 5: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

BASIC PHARMACOKINETICSAND PHARMACODYNAMICS

An Integrated Textbook andComputer Simulations

SARA ROSENBAUM

A JOHN WILEY & SONS, INC., PUBLICATION

Page 6: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

Copyright C© 2011 by John Wiley & Sons, Inc. All rights reserved.

Published by John Wiley & Sons, Inc., Hoboken, New Jersey.Published simultaneously in Canada.

No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or byany means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permittedunder Section 107 or 108 of the 1976 United States Copyright Act, without either the prior writtenpermission of the Publisher, or authorization through payment of the appropriate per-copy fee to theCopyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978) 750-8400,fax (978) 750-4470, or on the web at www.copyright.com. Requests to the Publisher for permission shouldbe addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030,(201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permission.

Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts inpreparing this book, they make no representations or warranties with respect to the accuracy or completenessof the contents of this book and specifically disclaim any implied warranties of merchantability or fitness fora particular purpose. No warranty may be created or extended by sales representatives or written salesmaterials. The advice and strategies contained herein may not be suitable for your situation. You shouldconsult with a professional where appropriate. Neither the publisher nor author shall be liable for any loss ofprofit or any other commercial damages, including but not limited to special, incidental, consequential, orother damages.

For general information on our other products and services or for technical support, please contact ourCustomer Care Department within the United States at (800) 762-2974, outside the United States at(317) 572-3993 or fax (317) 572-4002.

Wiley also publishes its books in a variety of electronic formats. Some content that appears in print may notbe available in electronic formats. For more information about Wiley products, visit our web site atwww.wiley.com.

Library of Congress Cataloging-in-Publication Data:

Rosenbaum, Sara (Sara E.)Basic pharmacokinetics and pharmacodynamics : an integrated textbook and computer simulations /

by Sara Rosenbaum.p. ; cm.

Includes bibliographical references.ISBN 978-0-470-56906-1 (cloth)

1. Pharmacokinetics–Textbooks. 2. Pharmacokinetics–Computer simulation. 3. Drugs–Physiologicaleffect–Textbooks. 4. Drugs–Physiological effect–Computer simulation. I. Title.[DNLM: 1. Pharmacokinetics. 2. Computer Simulation. 3. Pharmacological Phenomena. QV 38RM301.5.R655 2011615′.7–dc22 2010043288

Printed in Singapore

eISBN: 9781118001059

10 9 8 7 6 5 4 3 2 1

Page 7: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

To S, M, and L

Page 8: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

Page 9: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

CONTENTS

Preface xvii

1 Introduction to Pharmacokinetics and Pharmacodynamics 1

1.1 Introduction: Drugs and Doses, 11.2 Introduction to Pharmacodynamics, 3

1.2.1 Drug Effects at the Site of Action, 31.2.2 Agonists, Antagonists, and Concentration–Response

Relationships, 61.3 Introduction to Pharmacokinetics, 9

1.3.1 Plasma Concentration of Drugs, 101.3.2 Processes in Pharmacokinetics, 11

1.4 Dose–Response Relationships, 131.5 Therapeutic Range, 14

1.5.1 Determination of the Therapeutic Range, 161.6 Summary, 18

2 Passage of Drugs Through Membranes 20

2.1 Introduction, 202.2 Structure and Properties of Membranes, 212.3 Passive Diffusion, 22

2.3.1 Transcellular Passive Diffusion, 242.3.2 Paracellular Passive Diffusion, 26

2.4 Carrier-Mediated Processes: Transport Proteins, 272.4.1 Uptake Transporters: SLC Superfamily, 282.4.2 Efflux Transporters: ABC Superfamily, 292.4.3 Characteristics of Transporter Systems, 31

vii

Page 10: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

viii CONTENTS

2.4.4 Simulation Exercise, 322.4.5 Clinical Examples of Transporter Involvement

in Drug Response, 33

3 Drug Administration, Absorption, and Bioavailability 36

3.1 Introduction: Local and Systemic Drug Administration, 373.2 Common Routes of Systemic Drug Administration, 37

3.2.1 Intravascular Direct Systemic Administration, 373.2.2 Extravascular Parenteral Routes, 383.2.3 Other Extravascular Routes, 38

3.3 Overview of Oral Absorption, 403.4 Extent of Drug Absorption, 41

3.4.1 Bioavailability Factor, 413.4.2 Individual Bioavailability Factors, 42

3.5 Determinants of the Bioavailability Factor, 433.5.1 Disintegration, 433.5.2 Dissolution, 433.5.3 Formulation Excipients, 433.5.4 Adverse Events Within the Gastrointestinal Lumen, 443.5.5 Transcellular Passive Diffusion, 463.5.6 Paracellular Passive Diffusion, 473.5.7 Uptake and Efflux Transporters, 473.5.8 Presytemic Intestinal Metabolism or Extraction, 503.5.9 Presystemic Hepatic Metabolism or Extraction, 52

3.6 Factors Controlling the Rate of Drug Absorption, 533.6.1 Dissolution-Controlled Absorption, 543.6.2 Membrane Penetration–Controlled Absorption, 553.6.3 Overall Rate of Drug Absorption, 55

3.7 Biopharmaceutics Classification System, 55Problems, 56References, 57

4 Drug Distribution 60

4.1 Introduction, 614.2 Extent of Drug Distribution, 61

4.2.1 Distribution Volumes, 624.2.2 Tissue Binding and Plasma Protein Binding: Concentrating

Effects, 644.2.3 Assessment of the Extent of Drug Distribution: Apparent

Volume of Distribution, 654.2.4 Plasma Protein Binding, 72

4.3 Rate of Drug Distribution, 794.3.1 Perfusion-Controlled Drug Distribution, 804.3.2 Diffusion-Controlled Drug Distribution, 82

4.4 Distribution of Drugs to the Central Nervous System, 83Problems, 86References, 87

Page 11: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

CONTENTS ix

5 Drug Elimination and Clearance 88

5.1 Introduction, 895.1.1 First-Order Elimination, 905.1.2 Determinants of the Elimination Rate Constant and the

Half-Life, 915.2 Clearance, 91

5.2.1 Definition and Determinants of Clearance, 915.2.2 Total Clearance, Renal Clearance, and Hepatic Clearance, 945.2.3 Relationships Among Clearance, Volume of Distribution,

Elimination Rate Constant, and Half-Life, 955.2.4 Primary and Secondary Parameters, 96

5.3 Renal Clearance, 975.3.1 Glomerular Filtration, 975.3.2 Tubular Secretion, 985.3.3 Tubular Reabsorption, 1005.3.4 Putting Meaning into the Value of Renal Clearance, 101

5.4 Hepatic Clearance, 1025.4.1 Phase I and Phase II Metabolism, 1035.4.2 The Cytochrome P450 Enzyme System, 1045.4.3 Glucuronidation, 1055.4.4 Drug–Drug Interactions, 1065.4.5 Hepatic Drug Transporters, 1075.4.6 Kinetics of Drug Metabolism, 1095.4.7 Hepatic Clearance, 111

5.5 Measurement of Clearances, 1155.5.1 Total Body Clearance, 1155.5.2 Renal Clearance, 1175.5.3 Fraction of the Drug Excreted Unchanged, 120

Problems, 121References, 124

6 Compartmental Models in Pharmacokinetics 126

6.1 Introduction, 1276.2 Expressions for Component Parts of the Dose–Plasma

Concentration Relationship, 1276.2.1 Effective Dose, 1276.2.2 Rate of Drug Absorption, 1286.2.3 Rate of Drug Elimination, 1296.2.4 Rate of Drug Distribution, 129

6.3 Putting Everything Together: Compartments and Models, 1306.3.1 One-Compartment Model, 1306.3.2 Two-Compartment Model, 1316.3.3 Three-Compartment Model, 131

6.4 Examples of Complete Compartment Models, 1336.4.1 Intravenous Bolus Injection in a One-Compartment Model

with First-Order Elimination, 133

Page 12: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

x CONTENTS

6.4.2 Intravenous Bolus Injection in a Two-Compartment Modelwith First-Order Elimination, 134

6.4.3 First-Order Absorption in a Two-Compartment Model withFirst-Order Elimination, 135

6.5 Use of Compartmental Models to Study MetabolitePharmacokinetics, 136

6.6 Selecting and Applying Models, 137Problems, 138Recommended Reading, 138

7 Pharmacokinetics of an Intravenous Bolus Injection in aOne-Compartment Model 139

7.1 Introduction, 1407.2 One-Compartment Model, 1407.3 Pharmacokinetic Equations, 142

7.3.1 Basic Equation, 1427.3.2 Half-Life, 1437.3.3 Time to Eliminate a Dose, 143

7.4 Simulation Exercise, 1447.5 Application of the Model, 145

7.5.1 Predicting Plasma Concentrations, 1457.5.2 Duration of Action, 1467.5.3 Value of a Dose to Give a Desired Initial Plasma

Concentration, 1477.5.4 Intravenous Loading Dose, 147

7.6 Determination of Pharmacokinetic Parameters Experimentally, 1487.6.1 Study Design for the Determination of Parameters, 1497.6.2 Pharmacokinetic Analysis, 149

7.7 Pharmacokinetic Analysis in Clinical Practice, 153Problems, 155Recommended Reading, 157

8 Pharmacokinetics of an Intravenous Bolus Injection in aTwo-Compartment Model 158

8.1 Introduction, 1598.2 Tissue and Compartmental Distribution of a Drug, 159

8.2.1 Drug Distribution to the Tissues, 1598.2.2 Compartmental Distribution of a Drug, 160

8.3 Basic Equation, 1628.3.1 Distribution: A, �, and the Distribution t1/2, 1638.3.2 Elimination:B, �, and the Beta t1/2, 163

8.4 Relationship Between Macro and Micro Rate Constants, 1648.5 Primary Pharmacokinetic Parameters, 165

8.5.1 Clearance, 1658.5.2 Distribution Clearance, 1668.5.3 Volume of Distribution, 167

8.6 Simulation Exercise, 170

Page 13: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

CONTENTS xi

8.7 Determination of the Pharmacokinetic Parameters of theTwo-Compartment Model, 1738.7.1 Determination of Intercepts and Macro Rate Constants, 1738.7.2 Determination of the Micro Rate Constants: k12, k21,

and k10, 1758.7.3 Determination of the Primary Pharmacokinetic

Parameters, 1758.8 Clinical Application of the Two-Compartment Model, 176

8.8.1 Measurement of the Elimination Half-Life in thePostdistribution Phase, 176

8.8.2 Determination of the Loading Dose, 1778.8.3 Evaluation of a Dose: Monitoring Plasma

Concentrations and Patient Response, 179Problems, 180Recommended Reading, 181

9 Pharmacokinetics of Extravascular Drug Administration 182

9.1 Introduction, 1839.2 Model for First-Order Absorption in a One-Compartment Model, 184

9.2.1 Model and Equations, 1849.2.2 Determination of the Model Parameters, 1869.2.3 Absorption Lag Time, 1929.2.4 Flip-Flop Model and Sustained-Release Preparations, 1929.2.5 Determinants of Tmax and Cmax, 194

9.3 Bioavailability, 1959.3.1 Bioavailability Parameters, 1959.3.2 Absolute Bioavailability, 1979.3.3 Relative Bioavailability, 1989.3.4 Bioequivalence, 1989.3.5 Example Bioavailability Analysis, 198

9.4 Simulation Exercise, 198Problems, 199Recommended Reading, 200

10 Introduction to Noncompartmental Analysis 201

10.1 Introduction, 20110.2 Mean Residence Time, 20210.3 Determination of Other Important Pharmacokinetic Parameters, 20510.4 Different Routes of Administration, 20710.5 Application of Noncompartmental Analysis to Clinical Studies, 208Problems, 210

11 Pharmacokinetics of Intravenous Infusion in a One-Compartment Model 212

11.1 Introduction, 21311.2 Model and Equations, 214

11.2.1 Basic Equation, 214

Page 14: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

xii CONTENTS

11.2.2 Application of the Basic Equation, 21611.2.3 Simulation Exercise: Part 1, 216

11.3 Steady-State Plasma Concentration, 21711.3.1 Equation for Steady-State Plasma Concentrations, 21711.3.2 Application of the Equation, 21711.3.3 Basic Formula Revisited, 21811.3.4 Factors Controlling Steady-State Plasma Concentration, 21811.3.5 Time to Steady State, 21911.3.6 Simulation Exercise: Part 2, 220

11.4 Loading Dose, 22111.4.1 Loading-Dose Equation, 22111.4.2 Simulation Exercise: Part 3, 223

11.5 Termination of Infusion, 22311.5.1 Equations for Termination Before and After

Steady State, 22311.5.2 Simulation Exercise: Part 4, 224

11.6 Individualization of Dosing Regimens, 22411.6.1 Initial Doses, 22411.6.2 Monitoring and Individualizing Therapy, 225

Problems, 227

12 Multiple Intravenous Bolus Injections in the One-Compartment Model 230

12.1 Introduction, 23112.2 Terms and Symbols Used in Multiple-Dosing Equations, 23212.3 Monoexponential Decay During a Dosing Interval, 234

12.3.1 Calculation of Dosing Interval to Give SpecificSteady-State Peaks and Troughs, 235

12.4 Basic Pharmacokinetic Equations for Multiple Doses, 23612.4.1 Principle of Superposition, 23612.4.2 Equations That Apply Before Steady State, 236

12.5 Steady State, 23812.5.1 Steady-State Equations, 23812.5.2 Average Plasma Concentration at Steady State, 24012.5.3 Fluctuation, 24212.5.4 Accumulation, 24312.5.5 Time to Reach Steady State, 24412.5.6 Loading Dose, 245

12.6 Basic Formula Revisited, 24512.7 Pharmacokinetic-Guided Dosing Regimen Design, 246

12.7.1 General Considerations for Selection of theDosing Interval, 246

12.7.2 Protocols for Pharmacokinetic-GuidedDosing Regimens, 247

12.8 Simulation Exercise, 251Problems, 253References, 253

Page 15: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

CONTENTS xiii

13 Multiple Intermittent Infusions 254

13.1 Introduction, 25413.2 Steady-State Equations for Multiple Intermittent

Infusions, 25613.3 Monoexponential Decay During a Dosing Interval:

Determination of Peaks, Troughs, and EliminationHalf-Life, 25913.3.1 Determination of Half-Life, 25913.3.2 Determination of Peaks and Troughs, 261

13.4 Determination of the Volume of Distribution, 26113.5 Individualization of Dosing Regimens, 26413.6 Simulation Exercise, 265

Problems, 265

14 Multiple Oral Doses 267

14.1 Introduction, 26714.2 Steady-State Equations, 268

14.2.1 Time to Peak Steady-State Plasma Concentration, 26914.2.2 Maximum Steady-State Plasma Concentration, 27014.2.3 Minimum Steady-State Plasma Concentration, 27114.2.4 Average Steady-State Plasma Concentration, 27114.2.5 Overall Effect of Absorption Parameters on a

Steady-State Dosing Interval, 27214.3 Equations Used Clinically to Individualize Oral Doses, 272

14.3.1 Protocol to Select an Appropriate Equation, 27314.4 Simulation Exercise, 274References, 265

15 Nonlinear Pharmacokinetics 277

15.1 Linear Pharmacokinetics, 27715.2 Nonlinear Processes in Absorption, Distribution, Metabolism,

and Elimination, 28015.3 Pharmacokinetics of Capacity-Limited Metabolism, 281

15.3.1 Kinetics of Enzymatic Processes, 28215.3.2 Plasma Concentration–Time Profile, 283

15.4 Phenytoin, 28415.4.1 Basic Equation for Steady State, 28515.4.2 Estimation of Doses and Plasma Concentrations, 28715.4.3 Influence of Km and Vmax and Factors That Affect

These Parameters, 28915.4.4 Time to Eliminate the Drug, 29015.4.5 Time to Reach Steady State, 29115.4.6 Individualization of Doses of Phenytoin, 292

Problems, 295References, 296

Page 16: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

xiv CONTENTS

16 Introduction to Pharmacodynamic Models and IntegratedPharmacokinetic–Pharmacodynamic Models 297

16.1 Introduction, 29816.2 Classic Pharmacodynamic Models Based on Traditional

Receptor Theory, 29916.2.1 Receptor Binding, 30016.2.2 Response–Concentration Models, 302

16.3 Empirical Pharmacodynamic Models Used Clinically, 30716.3.1 Sigmoidal Emax and Emax Models, 30816.3.2 Linear Adaptations of the Emax Model, 310

16.4 Integrated PK–PD Models: Emax Model Combined witha PK Model for Intravenous Bolus Injection in aOne-Compartment Model, 31216.4.1 Simulation Exercise, 314

16.5 Hysteresis and the Effect Compartment, 31516.5.1 Simulation Exercise, 318

Problems, 319References, 321

17 Mechanism-Based Integrated Pharmacokinetic–Pharmacodynamic Models 323

17.1 Introduction, 32417.2 Alternative Models for Drug–Receptor Interaction: Operational

Model of Agonism, 32517.2.1 Simulation Exercise, 329

17.3 Physiological Turnover Model and Its Characteristics, 32917.3.1 Points of Drug Action, 33017.3.2 System Recovery After Change in Baseline Value, 330

17.4 Indirect Effect Models, 33117.4.1 Characteristics of Indirect Effect Drug Responses, 33317.4.2 Characteristics of Indirect Effect Models Illustrated

Using Model I, 33417.4.3 Other Indirect Models, 340

17.5 Transduction and Transit Compartment Models, 34017.5.1 Simulation Exercise, 343

17.6 Tolerance Models, 34417.6.1 Counter-regulatory Force Model, 34517.6.2 Precursor Pool Model of Tolerance, 348

17.7 Irreversible Drug Effects, 35017.7.1 Application of the Turnover Model to Irreversible Drug

Action, 35017.7.2 Model for Hematological Toxicity of Anticancer Drugs, 352

17.8 Disease Progression Models, 35617.8.1 Generation of Drug Response, 35617.8.2 Drug Interaction with a Disease, 35617.8.3 Disease Progression Models, 356

Problems, 360References, 365

Page 17: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

CONTENTS xv

Appendix A Review of Exponents and Logarithms 368A.1 Exponents, 368A.2 Logarithms: log and ln, 369A.3 Performing Calculations in the Logarithmic Domain, 370

A.3.1 Multiplication, 370A.3.2 Division, 371A.3.3 Reciprocals, 371A.3.4 Exponents, 371

A.4 Calculations Using Exponential Expressions and Logarithms, 371A.5 Decay Function: e−kt , 373A.6 Growth Function: 1 − ekt , 374A.7 Decay Function in Pharmacokinetics, 374Problems, 375

Appendix B Rates of Processes 377B.1 Introduction, 377B.2 Order of a Rate Process, 378B.3 Zero-Order Processes, 378

B.3.1 Equation for Zero-Order Filling, 378B.3.2 Equation for Zero-Order Emptying, 379B.3.3 Time for Zero-Order Emptying to Go to 50%

Completion, 379B.4 First-Order Processes, 380

B.4.1 Equation for a First-Order Process, 380B.4.2 Time for 50% Completion: The Half-Life, 381

B.5 Comparison of Zero- and First-Order Processes, 382B.6 Detailed Example of First-Order Decay in Pharmacokinetics, 382

B.6.1 Equations and Semilogarithmic Plots, 382B.6.2 Half-Life, 383B.6.3 Fraction or Percent Completion of a First-Order Process

Using First-Order Elimination as an Example, 384B.7 Examples of the Application of First-Order Kinetics to

Pharmacokinetics, 385

Appendix C Creation of Excel Worksheets for Pharmacokinetic Analysis 387C.1 Measurement of AUC and Clearance, 387

C.1.1 Trapezoidal Rule, 388C.1.2 Excel Spreadsheet to Determine AUC0→∞ and

Clearance, 389C.2 Analysis of Data from an Intravenous Bolus Injection in a

One-Compartment Model, 393C.3 Analysis of Data from an Intravenous Bolus Injection in a

Two-Compartment Model, 394C.4 Analysis of Oral Data in a One-Compartment Model, 398C.5 Noncompartmental Analysis of Oral Data, 399

Appendix D Derivation of Equations for Multiple Intravenous Bolus Injections 403D.1 Assumptions, 403

Page 18: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

xvi CONTENTS

D.2 Basic Equation for Plasma Concentration After MultipleIntravenous Bolus Injections, 403

D.3 Steady-State Equations, 406

Appendix E Summary of the Properties of the Fictitious Drugs Usedin the Text 407

Appendix F Computer Simulation Models 409

Glossary of Abbreviations and Symbols 410

Index 415

Page 19: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

PREFACE

The behavior and characteristics of therapeutic drugs vary enormously. For example, dosesdiffer more than a thousandfold. Some drugs must be taken three times a day, othersonce daily, and some every month. The response to some therapies occurs immediately,whereas for others it may take days or even weeks for the response to be apparent. Somedrugs must be taken with food; others must be taken on an empty stomach. Concurrentmedications interact with some drugs but not with others. The study of pharmacokinetics(the dose–concentration relationship) and pharmacodynamics (the concentration–responserelationship), which have been referred to as the pillars of clinical pharmacology, unlocksthe mystery of this behavior and brings clarity to diverse patterns of drug action. The goalof this book is to provide straightforward, uncomplicated, but comprehensive coverage ofthe essentials of pharmacokinetics and pharmacodynamics. I hope the book will enable alarge and diverse group of students to develop an interest in this subject and gain a betterunderstanding of the properties and behaviors of drugs.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and ComputerSimulations is an introductory textbook suitable to accompany courses in pharmacokinet-ics, pharmacodynamics, and clinical pharmacology in pharmacy and medical schools. Itis also directed toward people in the pharmaceutical field who want to gain an under-standing of this area through self-study. The book is organized and written with severalobjectives in mind. First, as an introductory textbook, the intent is to present the ma-terial in as simple a way as possible, without compromising the accuracy and scope ofthe material. I think it is important that students not be overwhelmed during their initialexposure. Interested students can always find more advanced literature. Second, simula-tions are integrated into the text to allow students to visualize important concepts and topromote understanding. Pharmacokinetics and pharmacodynamics are subjects that mustbe approached with the goal of understanding, not memorizing, the material. The textprovides exercises to guide readers through simulations, but readers are also encouragedto experiment with simulations on their own. A third goal is to balance the qualitativeside of pharmacokinetics with the quantitative side, or equations. Although only a fraction

xvii

Page 20: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCfm JWBS062-Rosenbaum February 16, 2011 7:24 Printer Name: Yet to Come

xviii PREFACE

of students who study the subject will use pharmacokinetic equations in their daily work,the equations are an enormous help in understanding the concepts. Most of the pharma-cokinetic equations used in the book have been included for that purpose alone, and despitetheir sometimes hostile appearance, equations should be viewed as friends, not foes. Anyreader who is intimidated by pharmacokinetic equations and the expression e−kt is stronglyencouraged to read Appendix A and to understand the simple properties of the decayfunction. Demystifying this function and the growth function (1 − e−kt ) can transformpharmacokinetic equations into very simple expressions.

A final objective is to present some of the interesting recent developments in pharma-codynamics. Generally, clinical pharmacology courses at pharmacy and medical schoolsrequire that less time be spent discussing pharmacodynamics than discussing pharmacoki-netics. Yet in the last 10 to 20 years, the knowledge base in pharmacodynamics has grownconsiderably, and has enabled seemingly bizarre concentration–response relationships tobe understood. It is recognized that many readers may never use the pharmacodynamicmodels presented in this book to model data, but the models serve as excellent tools to gaina greater understanding of the mechanism of action of drugs and the time course of drugresponse. Combining a pharmacokinetic model with a pharmacodynamic model of drugresponse provides a complete model of the dose–response relationship and allows drugresponse to be estimated at any time after any dose. The models also provide an opportu-nity to apply and integrate several pharmaceutical disciplines, including pharmacokinetics,classical receptor theory, pharmacology, and therapeutics.

To provide some practical examples of the application of the concepts, as well as to tietogether the topics in the book, three fictitious drugs have been created and used as examplesthroughout. The characteristics and properties of the drugs are introduced in parallel withthe evolution of pharmacokinetic and pharmacodynamic principles. These drugs are usedas subject drugs in many of the end-of-chapter problems. Students are encouraged to solvegraphical problems using Excel worksheets. Detailed instructions are provided in AppendixC to guide students through the creation of these custom-made worksheets.

I would like to acknowledge and thank colleagues who provided feedback on some of thechapters, particularly Bob Rodgers, Diane Mould, Miroslav Dostalek, Abraham Kovoor,David Worthen, and Fatemeh Akhlaghi. I would also like to thank Ruohan Wang and JaredConnelly for their help with Flash programming, Linnae Anderson for her help with someof the diagrams, Brian L’Heureux and Christopher Barker for creating the background forthe fictitious drugs, and Ian Lester and Marian Gaviola for their help in setting up the Website for the computer simulation models.

Sara Rosenbaum

Page 21: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

1INTRODUCTION TOPHARMACOKINETICS ANDPHARMACODYNAMICS

1.1 Introduction: Drugs and Doses

1.2 Introduction to Pharmacodynamics1.2.1 Drug Effects at the Site of Action

1.2.1.1 Interaction of a Drug with Its Receptor1.2.1.2 Postreceptor Events

1.2.2 Agonists, Antagonists, and Concentration–Response Relationships

1.3 Introduction to Pharmacokinetics1.3.1 Plasma Concentration of Drugs1.3.2 Processes in Pharmacokinetics

1.4 Dose–Response Relationships

1.5 Therapeutic Range1.5.1 Determination of the Therapeutic Range

1.6 Summary

Objectives

The material in this chapter will enable the reader to:

1. Define pharmacodynamics and pharmacokinetics

2. Understand the processes that control the dose–response relationship

3. Appreciate in a general way how mathematical expressions in pharmacodynamicsand pharmacokinetics can be used for the rational determination of optimum dosingregimens

1.1 INTRODUCTION: DRUGS AND DOSES

Drugs may be defined as chemicals that alter physiological or biochemical processes in thebody in a manner that makes them useful in the treatment, prevention, or cure of diseases.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

1

Page 22: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

2 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

Based on this definition, any useful drug must affect body physiology or biochemistry,and by extension must, if used inappropriately, possess the ability to do harm. Drug actionbegins with administration of the drug (input) and concludes with the biological response(output, which can be a beneficial and/or an adverse effect). The inputs (dose, frequency ofadministration, route of administration) must be selected carefully to optimize the onset,intensity, and duration of therapeutic effects for a particular disease condition. At the sametime, the inputs selected must minimize any harmful effects of drugs.

The design of optimum dosing regimens requires a complete understanding of theprocesses and steps that translate the input into the output. It also requires an understandingof how the input–output relationship may be influenced by individual patient characteristicsthat may exist at the very beginning of therapy, as well as conditions that may arise duringthe course of drug therapy. These will include the age and weight of the patient, the presenceof other diseases, genetic factors, concurrent medications, and changes in the disease beingtreated over time.

The material presented in this book will address and explain why, as shown in Table 1.1,there is such tremendous variability in the value of drug doses and dosing frequencies amongtherapeutic drugs. Additionally, it will address why different routes of administration areused for different drugs and different indications (Table 1.1).

The steps between drug input and the emergence of the response can be broken downinto two phases: pharmacokinetic and pharmacodynamic. The pharmacokinetic phase en-compasses all the events between the administration of a dose and the achievement ofdrug concentrations throughout the body. The pharmacodynamic phase encompasses allthe events between the arrival of the drug at its site of action and the onset, magnitude, andduration of the biological response (Figure 1.1). The rational design of optimum dosingregimens must be based on a thorough understanding of these two phases, and ideallywill include the development of one or more mathematical expressions for the relationshipbetween dose and the time course of drug response.

Optimum drug administration is important not only for ensuring good patient outcomesin clinical practice but also in the design of clinical trials during drug development. Thecost of drug research and development is enormous, so it is critical that all drug candidatesselected for human trials be evaluated in the most efficient, cost-effective manner possible.

TABLE 1.1 Examples of Common Daily Doses and Dosing Intervals

Daily Dose Dose FrequencyDrug (mg) (h) Route

Calcium carbonate 3000 2 OralIbuprofen 1600 6 OralVancomycin (for MRSAa) 2000 12 IntravenousAmoxicillin 750 8 OralVancomycin (for pseudomembranous colitis) 1000 6 OralAtenolol 100 24 OralFluoxetine 20 24 OralRamipril 10 12 OralDigoxin 0.250 24 OralChloroquine 300 Weekly Oral

aMethicillin-resistant Staphylococcus aureus.

Page 23: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

INTRODUCTION TO PHARMACODYNAMICS 3

Drug in Systemic

Circulation

Drug at Site ofAction

DrugInteraction

With ReceptorRESPONSE

Pharmacokinetics Pharmacodynamics

DoseAdministered

FIGURE 1.1 The two phases of drug action. The pharmacokinetic phase is concerned with therelationship between the value of the dose administered and the value of the drug concentrationsachieved in the body; the pharmacodynamic phase is concerned with the relationship between drugconcentrations at the site of action and the onset, intensity, and duration of drug response.

The application of pharmacokinetic and pharmacodynamic principles to this process hasbeen shown to enhance the selection of optimum doses and the optimum design of phase IIclinical trials.

1.2 INTRODUCTION TO PHARMACODYNAMICS

Pharmaco- comes from the Greek word for “drug,” pharmackon, and dynamics means “ofor relating to variation of intensity.” Pharmacodynamics (PD) is the study of the magnitudeof drug response. In particular, it is the study of the onset, intensity, and duration of drugresponse and how these are related to the concentration of a drug at its site of action. Anoverview of some basic drug terminology and the drug response–concentration relationshipis provided below.

1.2.1 Drug Effects at the Site of Action

Note that although some references and textbooks distinguish the terms drug effect anddrug response, this distinction has not been adopted universally. In this book, effect andresponse are used interchangeably.

1.2.1.1 Interaction of a Drug with Its ReceptorDrug response is initiated by a chemical interaction between a drug and a special bindingsite on a macromolecule in a tissue. This macromolecule is known as a drug receptor. Thedrug–receptor interaction results in a conformational change in the receptor, which resultsin the generation of a stimulus, which ultimately leads to a biochemical or physiologicalresponse (Figure 1.2). Most receptors (over 95%) are proteins, but other types of receptorsexist. For example, the receptors of the alkylating agents used in cancer chemotherapyare on DNA. The drug–receptor interaction involves chemical bonding, which is usuallyreversible in nature and can be expressed using the law of mass action (Figure 1.2). Thus,at the site of action the drug binds to its receptor and equilibrium is established between thebound and the unbound drug. As the drug is eliminated from the body and removed fromits site of action, it dissociates from the receptor, which is left unchanged, and the responsedissipates.

Page 24: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

4 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

SIGNAL

Cell Membrane

Receptor

C + R RC

RESPONSE

Drug

kon

koff

FIGURE 1.2 Drug–receptor interaction. Here AG signifies a drug agonist, C is the free drugconcentration (not bound to the receptor), R the concentration of free receptors, RC the concentrationof the drug–receptor complex, and kon and koff are the rate constants for the forward and backwardprocesses, respectively.

In contrast, a few drugs form irreversible covalent bonds with their receptors. Forexample, aspirin inhibits platelet aggregation by inhibiting the formation of thromboxanein the platelets. It accomplishes this by binding covalently to and blocking the catalyticactivity of cyclooxygenase, the enzyme that produces thromboxane. The effect of a singledose of aspirin will persist long after the drug has been removed from its site of actionand will continue until new cyclooxygenase molecules are synthesized, which can thenresume the production of thromboxane. Other examples of drugs that bind irreversibly totheir receptors include the alkylating agents mentioned above, and proton pump inhibitorssuch as omeprazole, which block the secretion of gastric acid by binding irreversibly to theH+,K+-ATPase pumps of parietal cells.

Owing to the chemical nature of drug–receptor interaction, it is highly dependent on thechemical structure of both the drug and the receptor; small changes in the structure of thedrug can reduce or destroy activity. For example, the drug–receptor interaction can distin-guish between the R- and S-isomers of drugs that have chiral carbon atoms. Usually, oneisomer is much more active than the other. The S-isomer of warfarin, for example, is two tofive times more active than the R-isomer. The development and promotion of S-omeprazole(Nexium) is based on the premise that the S-isomer has the higher affinity for the bindingsite and thus offers therapeutic advantages over preparations containing racemic mixtures(equal quantities of each isomer) of omeprazole, such as Prilosec and its generic equivalents.

Receptors are assumed to exist for all active endogenous compounds (natural ligands),such as neurotransmitters and hormones. The interaction of the natural ligands with theirreceptors controls and/or regulates physiological and biochemical processes in the body.In most cases, drugs mimic or antagonize the actions of endogenous ligands by interactingwith their cognate receptors. For example, epinephrine is the natural ligand that interactswith the �2-adrenergic receptors in bronchial smooth muscle to bring about bronchialdilation. The drug albuterol also interacts with this receptor to produce bronchial dilation.

Page 25: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

INTRODUCTION TO PHARMACODYNAMICS 5

Acetylcholine transmits signals through a synapse by interacting with its nicotinic receptoron the postsynaptic neuronal membrane. The interaction results in the production of anaction potential, the response. The action of acetylcholine is mimicked by the drug nicotine.

It should be noted that there are a few drugs that do not act on receptors but that exert theiraction by bringing about physicochemical changes in the body. For example, conventionalantacids such as calcium carbonate act as buffers to reduce acidity in the stomach, andpolyethylene glycol is an osmotic laxative that acts by preventing the absorption of waterin the large intestine.

1.2.1.2 Postreceptor EventsDrugs almost always bring about some type of change in the intracellular environmentof cells, but the lipophilic cell membrane presents a physical barrier to most drugs andendogenous ligands. As a result, most receptors are located on the cell membrane itself. Theultimate action of a drug is dependent on the initial stimulus that results from the interactionof the drug with its membrane-bound receptor being relayed to the inside of the cell. Therelaying of the initial stimulus, known as coupling or signal transduction, often involves acascade of different steps during which the initial signal may be amplified or diminished.Some important transduction mechanisms are summarized below (see Figure 1.3).

1. Interaction of a drug with a receptor can lead directly to the opening or closing ofan ion channel that lies across a cell membrane. In this case the signal is relayedby changes in the ion concentration within the cell. For example, the interaction ofacetylcholine with its nicotinic receptor results in the opening of the ion channel. Thiscauses Na+ to move into the cell and results in the initiation of an action potential.

Receptor

Drug

1 2 3 4

Opens Ion Channels

Stimulatesa G-Protein

Activates ProteinKinase

Drug Penetratesthe Membrane

FIGURE 1.3 Diagrammatic representations of how a drug receptor interaction brings about intra-cellular events. The intracellular relay of the initial signal resulting from the interaction of a drugwith a membrane-bound receptor can be accomplished in one of three ways: (1) the direct openingof ion channels; (2) the activation of a G-protein that may lead to the activation of another enzyme orto a modulation of an ion channel; (3) the activation of protein kinase. Alternatively, (4), some drugsare able to penetrate membranes and directly activate intracellular receptors.

Page 26: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

6 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

2. Signal transduction for a large number of drugs involves the activation of aG-protein (guanine nucleotide-binding protein). The drug–receptor interaction onthe membrane triggers the activation of a G-protein on the cytoplasmic side of themembrane, which then initiates a series of events that culminate in the biologicalresponse. Activated G-protein can produce a variety of effects, including stimula-tion or inhibition of enzymes, and the opening or closing of ion channels. Theseevents usually result in changes in the concentration of an intracellular compoundknown as the second messenger. Examples of second messengers include cyclicadensine-3′,5′-monophosphate (cAMP), calcium, and phosphoinositides. The sec-ond messengers then relay the response further through a series of complex steps.For example, the interaction of catecholamines such as norepinephrine with certain�-receptor subtypes involves G-protein activation. This then stimulates adenylatecyclase to convert adenosine triphosphate (ATP) to cAMP, which acts as the secondmessenger. Subsequent events include the stimulation of specific protein kinases, acti-vation of calcium channels, and modification of cellular proteins. Other examples ofG-protein–coupled receptors (GPCRs) are the action of acetylcholine on its mus-carinic receptors and the action of serotonin on 5-HT receptors.

3. The interaction of a drug with its receptor can also result in the stimulation of areceptor-associated enzyme, tyrosine kinase. The activated tyrosine kinase phophory-lates key macromolecules, which are often a part of the receptor itself, to relay thesignal. Insulin and peptide growth factors, for example, use this form of signal trans-duction.

Some drugs are lipophilic enough to penetrate the cell membrane; others may be trans-ported across the cell membrane by uptake transporters. Drugs that are able to enter acell can interact directly with intracellular receptors. Examples include many steroids,such as glucocorticoid steroids, sex hormones, and thyroid hormones, and drugs such asthe HMG-CoA reductase inhibitors (commonly known as statins), which act on enzymeswithin the hepatocyte, and metformin, which is transported across the hepatocyte by anuptake transporter.

1.2.2 Agonists, Antagonists, and Concentration–Response Relationships

A drug that mimics the endogenous receptor ligand to activate the receptor is referred to asan agonist. The typical relationship between the drug effect and the agonist concentrationat the receptor site is shown in Figure 1.4a. Note that as the concentration of the drugincreases, the effect increases. At low concentrations, there is a linear relationship betweenconcentration and effect (i.e., the response is proportional to the concentration). At higherdrug concentrations, increases in concentration bring about much smaller changes in effect(the law of limited returns). Eventually, at very high concentrations, the effect achieves amaximum value and then remains constant and independent of concentration. In this areaof the curve, increases in concentration will not result in further increases in response.This relationship is observed because response is generated by a saturable, capacity-limitedprocess. For example, the response may be limited by the number of receptors that a tissuecontains. At low drug concentrations there are many free receptors, so as the drug con-centration increases, the drug can bind to the free receptors, and response can increase

Page 27: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

INTRODUCTION TO PHARMACODYNAMICS 7

Res

pons

e

Res

pons

e

Concentration Logarithm Concentration

(a)

MaximumResponse

MaximumResponse

(b)

FIGURE 1.4 Plots of response versus drug concentration: on a linear scale (a) and on a semiloga-rithmic scale (b).

proportionally. At higher concentrations more and more of the receptors are occupied, soincreases in the drug concentration produce much less increase in effect. Eventually, all ofthe receptors are occupied (or saturated) and a maximum effect is observed. To accommo-date a wide range of concentrations, the relationship between effect and concentration isusually plotted on a semilogarithmic scale, which transforms the plot to a sigmoidal shape(Figure 1.4b).

Many agonists are able to produce the system’s maximum response without fully oc-cupying all the receptors. In these systems the maximum response of the drug must bethe result of some other saturable, capacity-limited process that occurs after receptorbinding. These tissues or systems are said to have spare receptors. Experimentally, thepresence of spare receptors can be demonstrated by destroying some of the receptors. Ifan agonist is still able to produce a maximum response, the system must contain sparereceptors.

The efficiency with which a drug’s interaction with the receptor is converted into theinitial stimulus or biosignal is a function of the number of receptors at the site of actionand a drug’s intrinsic efficacy, which can be defined as the magnitude of the stimulusproduced per unit receptor occupied. The value of the stimulus that results from a specificconcentration of a drug is also a function of the drug’s affinity for its receptors. Affinity canbe defined as the extent or fraction to which a drug binds to receptors at any given drugconcentration. Drugs that have high affinity require less drug to produce a certain degreeof binding and to elicit a certain response compared to drugs with low affinity. Affinity isone of the factors that determine potency (see Chapter 16).

A drug that binds to a receptor but does not activate it is referred to as an antagonist. Thepresence of an antagonist at the receptor site blocks the action of the agonist (Figure 1.5),and higher concentrations of the agonist are needed to displace the antagonist and toproduce the effect that it elicited when the antagonist was absent. The antagonist shiftsthe concentration–response curve of an agonist to the right (Figure 1.6). At sufficientlyhigh concentrations of the antagonist, the agonist’s action may be blocked completely andthe effect of even high concentrations of the agonist reduced to zero. Some drugs bindto receptors but the binding is less efficient, and a full response cannot be achieved evenwhen the drug’s concentration is very high and all the receptors are occupied (Figure 1.7).These drugs are referred to as partial agonists. A partial agonist will block the effect of

Page 28: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

8 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

Cell Membrane

Receptor

FIGURE 1.5 Diagrammatic representation of the action of an antagonist. The antagonist (ATG)binds to the receptor but does not produce a signal. Its presence on the receptor blocks the action ofagonists (AG), including the natural ligand.

Res

pons

e to

an

Ago

nist

Logarithm of Agonist Concentration

Increasing Concentrationof Antagonist

No Antagonist

FIGURE 1.6 Plot of response versus logarithm concentration for an agonist in the absence andpresence of increasing concentrations of an antagonist.

Res

pons

e

Logarithm of Concentration

Full Agonist

Parital Agonist

FIGURE 1.7 Plot of response versus logarithm concentration for a full and a partial agonist.

Page 29: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

INTRODUCTION TO PHARMACOKINETICS 9

a full agonist. In the presence of high concentrations of a partial agonist, the action ofa full agonist can be reduced to the maximum response elicited by the partial agonist.Clinically, partial agonists are used to act as buffers to avoid full stimulation of a system.Examples of partial agonists include several �-blockers, including pindolol, and the opioidbuprenorphine. The latter is a partial agonist on the �-opioid receptors and is considered asafer alternative to morphine because it does not produce as much respiratory depression.

In summary, drug action is mediated primarily by the interaction of a drug withmembrane-bound receptors at its site of action. This produces conformational changesin the receptor which lead to the generation of an initial signal. The signal is then relayedto the intracellular environment by means of a variety of transduction processes. The re-sponse increases with increases in drug concentration until enough receptors are occupiedto generate the maximal response. The response to a specific concentration of drug isdependent on drug-specific properties (e.g., intrinsic efficacy, affinity) and tissue-specificproperties (e.g., number or density of receptors, amplification or diminution of the initialsignal during transduction). An important goal in a study of pharmacodynamics is to de-rive a mathematical expression for the magnitude of drug response as a function of drugconcentration:

E = fPD(C) (1.1)

where E is the drug effect or response, C the drug concentration, and f PD a pharmacodynamicfunction that links these two variables and contains the drug-specific parameters of intrinsicefficacy and affinity. In equation (1.1), E is the dependent variable because it is dependenton all the other components of the equation. The drug concentration at the site of action (C)is the independent variable because it is independent of all the other components of equation(1.1). This expression would allow the effect to be estimated at any drug concentration andwould allow the required concentrations for optimum response to be identified.

1.3 INTRODUCTION TO PHARMACOKINETICS

Pharmaco- comes from the Greek word for “drug,” pharmackon, and kinetics comes fromthe Greek word for “moving,” kinetikos. Pharmacokinetics (PK) is the study of drugmovement into, around, and out of the body. By extension it involves the study of drugabsorption, distribution, and elimination (metabolism and excretion) (ADME).

Pharmacokinetics involves the study of how drugs enter the body, distribute throughoutthe body, and leave the body. It is concerned with the driving forces for these processes andthe rate at which they occur. Pharmacokinetics is the study of the time course of drug con-centrations in body compartments. From a therapeutic perspective, the drug concentrationat the site of action is by far the most important: Concentrations should be sufficiently highto produce a response but not so high as to produce toxicity. Because it is not possible rou-tinely to measure this concentration clinically, the plasma concentration of the drug is themain focus in pharmacokinetics. It is often assumed that the plasma concentration reflectsthe drug concentration at the site of action. This is generally true, and the relationship isoften linear. Increases or decreases in the plasma concentration will be reflected by propor-tional increases or decreases, respectively, at the site of action. However, as discussed insubsequent chapters, this is not always the case and a more complex relationship betweenthese two concentrations may exist. It is important to note that although changes in the

Page 30: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

10 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

plasma concentration will usually result in proportional changes in the drug concentrationat the site of action, the reverse is not true. Because the amount of drug that is delivered tothe site of action is usually such a very small fraction of the total amount of drug in the body(in other tissues and the systemic circulation), local changes in the amount of drug at thesite of action are generally not reflected by noticeable changes in the plasma concentration.

1.3.1 Plasma Concentration of Drugs

As stated above, pharmacokinetics is concerned with the body’s exposure to a drug and howdrug concentrations change over time. For the most part, drug concentrations in the plasmaare the focus in pharmacokinetics. The rationale for this is twofold. First, blood is one ofthe few body fluids that can be obtained and analyzed repeatedly for drug concentrations atspecified times after the administration of a dose. The concentration of drug in whole bloodis not commonly used in pharmacokinetics because blood is a complex physical systemthat consists of red blood cells, white blood cells, and platelets suspended in plasma water.Blood with the cellular elements removed, either by centrifugation (plasma) or clotting(serum), is preferred. The collection of plasma requires the use of an anticoagulant suchas heparin, which can interfere with the assay of some drugs. In these cases (e.g., formeasuring digoxin concentration) serum rather than plasma is used as the reference fluid.In this book no distinction will be made between plasma and serum, and the term plasmaconcentration will be used almost universally.

The second rationale for focusing on plasma concentrations in pharmacokinetics isthat the circulatory system is the central fluid for the receipt and distribution of drugs(Figure 1.8). All drug input processes conclude when drug reaches the plasma, and alldisposition (distribution and elimination) processes begin once drug is present in the plasma.Thus, drugs in absorption sites such as the gastrointestinal tract or subcutaneous tissue areabsorbed into the circulatory system. Once in the blood, drugs undergo distribution tovarious tissues in the body, and elimination, primarily through the liver and/or kidneys.

Plasma or plasma water consists of small dissolved molecules (e.g., glucose, ions,nutrients, drugs) and suspended substances such as proteins which are too large to dissolve.Many drugs can bind or associate with the plasma proteins. The binding is reversible andmay be expressed according to the law of mass action:

[D] + [P]k1−→←−k2

[DP] (1.2)

where D is the free drug concentration, P the concentration of the protein not involvedin binding, DP the concentration of the drug–protein complex, and k1 and k2 are the rateconstants for the forward and backward reaction, respectively.

Thus, many drugs exist in the plasma in an equilibrium between two forms: one compo-nent dissolved in the plasma water (free drug) and one component associated or bound toplasma proteins (bound drug). The term plasma concentration (Cp) in pharmacokineticsrefers to the total drug concentration of the drug, that is, the bound plus the free. Totaldrug concentrations are reported routinely because they are much easier and less expen-sive to measure than are free drug concentrations. However, as presented in subsequentchapters, the free concentration is the clinically important component, as only free un-bound drug is able to pass biological membranes, interact with the receptor, and generatea pharmacological response.

Page 31: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

INTRODUCTION TO PHARMACOKINETICS 11

Drug in Tablet

Distribution

Absorption

G.I. Membrane

Liver

EliminationDrug

MetabolismDrug

Excretion

Drug inTissues

Drug inTissues

Disposition

Drug Removedfrom Body

Drug inSystemic

Circulation

FIGURE 1.8 Processes of drug absorption, distribution and elimination (metabolism and excre-tion) (ADME). Drug contained within the tablet must undergo absorption. It must penetrate thegastrointestinal membrane and pass through the liver before reaching the systemic circulation. Oncein the blood it has the opportunity to distribute to the tissues, including the site of action. As soon asdrug is present in the systemic circulation, it is subject to elimination. This occurs primarily in theliver and kidney, where drugs undergo metabolism and/or excretion, respectively. The fate of a drugin the systemic circulation (distribution and elimination) is referred to as drug disposition.

1.3.2 Processes in Pharmacokinetics

Pharmacokinetics involves the study of the processes that affect the plasma concentrationof a drug at any time after the administration of a dose. These processes are summarized inFigure 1.8. Most drugs are administered orally as tablets. A tablet is a compressed powdermass that consists of the active drug, which usually comprises only a small portion ofthe overall tablet, and other compounds required for either the manufacture of the tablet(i.e., diluents, lubricants, etc.) or to optimize the characteristics of the finished product (i.e.,color, taste, and hardness). Once a tablet is swallowed, it enters the stomach, where the drugcontained within the hard powder mass must be exposed and released. The tablet must firstdisintegrate into small particles to enable the drug to dissolve in the gastrointestinal fluid.These initial processes of disintegration and dissolution are part of biopharmaceutics, whichmay be defined as the study of how a drug’s chemical and physical properties influence boththe administration of the drug and the pharmacokinetic behavior of the dosage form in vivo.

Page 32: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

12 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

When the drug is dissolved in the gastrointestinal fluid it has the opportunity to pass acrossthe epithelial cell lining of the gastrointestinal membrane and get taken up into the bloodon the other side. Once in the circulatory system, the drug has to pass through the liver,which a major organ of drug elimination. The absorbed drug may undergo elimination bymetabolism during its first pass through the liver. After the liver the drug is taken to the heart,which pumps the drug throughout the entire circulatory system. At this point the drug hasbeen absorbed. The rate and extent of absorption of a drug are very important determinantsof the early plasma concentrations of a drug. Rapid rates of absorption will promote highearly plasma concentrations. Once the heart pumps the drug around the body, the drug isgiven the opportunity to distribute to all the tissues, including the biophase or site of action.A drug’s distribution pattern, particularly the rate and extent to which it distributes to thetissues, is also an important determinant of the early plasma concentrations. The greaterits distribution to the tissues, the lower will be the remaining plasma concentration. Theplasma concentration will also be influenced by drug elimination, which occurs as soonas the drug is in the plasma. The main pathways of elimination are hepatic metabolismand renal excretion. The process of drug elimination will continue to affect the plasmaconcentration until the drug has been removed from the body completely.

In summary, a drug’s pharmacokinetics are determined by the simultaneous processes ofabsorption, distribution, metabolism, and excretion (ADME) (Figure 1.8). The combinedprocesses of drug elimination and drug distribution, or the fate of a drug once it is present, isreferred to as drug disposition (Figure 1.8). The individual pharmacokinetic steps associatedwith the administration of a tablet are summarized in Table 1.2.

The goal of pharmacokinetics is to study each of the ADME processes with the aim of:

1. Identifying the drug and patient factors that determine the rate and extent of eachprocess. Topics to be considered include:� How does a drug’s lipophilicity influence absorption, distribution, and elimination?� What factors determine a drug’s distribution pattern?� Is the whole of a dose absorbed into the body?� Does a drug get to every tissue in the body?� To what extent do drugs undergo renal as opposed to hepatic elimination?� How are pharmacokinetic processes affected by patient characteristics, such as the

age of the patient, renal or hepatic impairment, ethnicity, and genetics?

TABLE 1.2 Pharmacokinetic Processes That Control the Dose–Plasma ConcentrationRelationship After the Consumption of a Tablet

Process Type of Process

1 Release of drug: tablet disintegration Biopharmaceutics2 Dissolution of tablet Biopharmaceutics3 Absorption of drug through gastrointestinal

membrane into the bloodAbsorption

4 Passage through the liver Absorption5 Entry to systemic circulation Absorption6 Distribution to the biophase Biophase distribution7 Distribution throughout the body Distribution8 Elimination (metabolism and excretion) Elimination

Page 33: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

DOSE–RESPONSE RELATIONSHIPS 13

2. Identifying a way to quantify or summarize each process in ADME using a singleparameter. Issues to be considered include:� How can the extent of absorption of a drug be quantified?� How can the extent to which a drug distributes the tissues be quantified?

3. Deriving a mathematical expression for the rate of each process in ADME andfor the overall relationship between a drug’s plasma concentration and time afterany dose:

Cp = fPK(dose, time) (1.3)

where, Cp is the plasma concentration, and f PK is a function that contains expressionsand parameters for ADME. In equation (1.3), Cp is the dependent variable, becauseit is dependent on all the other components of the equation, time is the independentvariable, and dose is a constant in a given situation. The dependent variable is relatedto the independent variable and the constant by means of an expression that containsparameters for each of the processes of ADME. The parameters are assumed to beconstants for a given drug under normal circumstances.

1.4 DOSE–RESPONSE RELATIONSHIPS

It will become apparent in subsequent chapters that for most drugs, the drug concentrationin the body at any time is proportional to the dose. As a result, plots of response at a certaintime, as a function of dose (Figure 1.9), resemble the plots of response versus concentration(Figure 1.4): A hyperbolic plot is often observed on the linear scale and a sigmoidal plot,on the semilogarithmic scale. Thus, dose–response curves are analogous, but not identical,to pharmacodynamic concentration–effect curves.

In contrast to the plots of response versus concentration, which are purely dependenton a drug’s pharmacodynamics, a dose–response curve is a function of both the drug’spharmacodynamic characteristics (intrinsic efficacy and affinity) and its pharmacokinetic

Res

pons

e

Logarithm of Dose

FIGURE 1.9 Graph of response versus logarithm of dose.

Page 34: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

14 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

characteristics (the fraction of the dose absorbed, the extent to which a drug distributesthroughout the body, etc.). Note that low doses produce no effect, and as the maximumresponse is approached, increasing the dose produces little change in the response (limitingreturns). Based on the characteristics shown in Figure 1.9, doses must be selected to avoidthe subtherapeutic areas of the plot and to avoid doses that approach or lie on the plateauand provide little or no additional benefit over lower doses. Most drugs also producetoxicity at higher concentrations. It is important that doses be selected that minimize thistoxicity. The toxicity may be an extension of the drug’s pharmacological action (e.g.,the major adverse effects of warfarin, digoxin, and anticholinergic drugs), in which caseit will be important to avoid areas on the dose–response curve close to the maximumeffect. Alternatively, the toxicity may arise because the drug may interact with multiplereceptors of different types, particularly at higher concentrations, to produce undesiredeffects. Examples of this type of toxicity include muscle toxicity associated with the statins,and drowsiness associated with first-generation antihistamines. The development of modelsand mathematical expressions of the pharmacokinetic and pharmacodynamic phases of drugresponse provides an opportunity for the rational selection of optimum dosing regimens.

The expression for a drug’s pharmacokinetics [equation (1.3)] can be combined withthe expression for a drug’s pharmacodynamics [equation (1.1)] to produce a completeexpression for the dose–response relationship:

E = fPD( fPK(dose, time)) (1.4)

Note that in this equation the plasma concentration of the drug (Cp) has been substitutedfor the drug concentration at the site of action (C) in the pharmacodynamic equation. Thisassumes that the concentration at the site is always proportional to the plasma concentration.The validity and limitations of this are discussed in subsequent chapters. Equation (1.4)would enable the full time course of drug response to be estimated after any dose. It couldalso be used to estimate the dose and dosing interval to produce optimum response. Ifthese relationships are identified early in the course of drug development, they can be usedto determine optimum doses for clinical trials. This in turn will increase the efficiency ofthe trials, reduce the time for drug development, and decrease the price of these highlycostly studies. The expressions can also be used to simulate response data for situations notyet studied clinically. For example, if a drug’s pharmacokinetics and pharmacodynamicsare known after a single dose, it is possible to use a combined PK–PD equation to simulatethe type of response that may be expected during multiple dosing therapy. Simulations canbe performed using different dosing regimens to try to obtain an estimate of what may bethe most effective dosing regimen.

1.5 THERAPEUTIC RANGE

In vivo pharmacodynamic studies aimed at the development of mathematical expressionsof drug response are relatively new. Historically, in vivo pharmacodynamic studies havebeen very difficult to perform, for several reasons:

1. Creating meaningful models and mathematical expressions for drug response re-quires that response data be collected on a continuous scale. It also requires that thedata possess a reasonable degree of precision. All-or-none responses and subjectivedata, based largely on a patient’s or a physician’s opinion, have limited value in this

Page 35: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

THERAPEUTIC RANGE 15

application. Responses to only a handful of drugs (e.g., anticoagulants, hypoglycemicagents) meet these criteria. In the last 10 to 20 years this problem has been over-come by the development and use of biomarkers (see Chapter 16) of drug response.Biomarkers are parallel changes in the levels or intensities of concrete measurable bi-ological molecules or other effects that have been found to be predictably associatedwith a drug’s biological response. Biomarkers may, for example, be cells, proteins,antibodies, body temperature, or features of an electroencephalograph.

2. The mathematical expressions derived from pharmacodynamic models are mainlynonlinear and could not be applied to clinical data until computer software becameavailable for nonlinear regression analysis.

3. Each drug or drug class has a unique mechanism of action and way of relaying orcoupling the initial drug effect. Signal transduction may take less than a second forsome drugs, several minutes for others, or up to several hours for others. As a result,summarizing the characteristics of the concentration–response relationship can becomplex.

4. In many cases a drug’s response lags behind the plasma concentration. This canconfound the concentration–response relationship and add an additional layer ofcomplexity to modeling response as a function of plasma concentration.

By contrast, pharmacokinetic studies are relatively simple to perform. Blood is easilysampled, drug assays for most drugs are fairly easily developed, and even before thewide availability of computers and software for pharmacokinetic analysis, the analysiscould be performed by linearizing the mathematical expressions and analyzing the datausing simple linear regression analysis. Furthermore, the pharmacokinetics of most drugscan be modeled using one of about three basic well-established models. As a result,pharmacokinetic studies and modeling have been a central part of the drug developmentprocess for decades. In order to use pharmacokinetic models for the design of dosingregimens, it is necessary to have target-optimal plasma concentrations or some idea ofthe concentration–response relationship. In the absence of mathematical expressions forthis relationship, a very simple approach for linking drug concentrations to response wasdeveloped and termed the therapeutic range, defined as the range of plasma concentrationsthat are associated with optimum response and minimal toxicity in most patients. Mostcommonly, the goal of therapy is to maintain drug concentrations within the therapeuticrange at all times. There are a small number of drugs for which this is not desirable,such as certain antibiotics and drugs such as nitroglycerin, where tolerance develops withcontinuous exposure to the drug.

The therapeutic range is illustrated in Figure 1.10, which shows:

� The minimum effective concentration (MEC), the lower boundary for effective drugconcentrations; plasma concentrations below the MEC have a high probability ofbeing subtherapeutic.

� The maximum tolerated concentration (MTC) is the upper boundary for optimumdrug concentrations; plasma concentrations above the MTC have a high probabilityof producing adverse effects or toxicity.

� The onset of action of a drug, which may be estimated as the time it takes for plasmaconcentrations to reach the MEC.

� The duration of action of a drug, which may be estimated as the time during whichplasma concentrations remain within the therapeutic range.

Page 36: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

16 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

MEC

MTC

Pla

sma

Con

cent

rati

on

Time

Onset ofAction

ActionCeases

Duration ofAction

FIGURE 1.10 Therapeutic range. The therapeutic range of a drug is the range of plasma con-centrations bounded by a lower minimum effective concentration (MEC) and an upper maximumtolerated concentration (MTC). The typical plasma concentration–time profile observed with the ad-ministration of a single oral dose is also shown. The therapeutic range allows the onset and durationof action of a drug to be estimated.

1.5.1 Determination of the Therapeutic Range

To apply the therapeutic range appropriately, and to understand both its value and itslimitations, it is necessary to appreciate how it is typically derived. It is usually determinedby studying the effects of a drug in a large population and noting the plasma concentrationsat which patients:

� Experience therapeutic effects� Experience side effects or toxicity

The cumulative plot of the percentage of all patients who experience a therapeuticresponse is then plotted as a function of plasma concentration (Figure 1.11). The cumulativeplot of the percentage of patients experiencing adverse effects at the various concentrationsis then added to the same graph (Figure 1.11). Similar sigmoidal shapes are obtained forboth curves, but the plot for toxicity is always displaced to the right. Higher concentrationsare needed for adverse compared to therapeutic effects (if this were not the case, the drugwould not be of therapeutic value). A frequent characteristic of these plots is that whereas100% of patients experience toxicity if concentrations are high enough, fewer than 100%of patients experience therapeutic effects even at high concentrations. Patients who do notrespond therapeutically even to high concentrations are referred to as nonresponders.

This plot is then used to estimate a drug’s therapeutic range. The MEC and MTCare usually chosen at concentrations where a high percentage of patients experience atherapeutic effect and a small percentage of patients experience toxicity, respectively. Thespecific concentrations selected for the MEC and the MTC will depend on the margin of

Page 37: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

THERAPEUTIC RANGE 17

0

20

40

60

80

100

% P

atie

nts

Res

pond

ing

70% Respond

MEC MTC

10% ExperienceToxicity

100% of patientsmay not respond

TherapeuticResponse

ToxicResponse

FIGURE 1.11 Identification of the therapeutic range. A drug’s therapeutic range is based onstudying the concentrations associated with response and toxicity in a large group of patients. TheMEC is selected at a concentration at which a large fraction of the population respond (70% is usedin the diagram). The MTC is selected at a concentration where a significant fraction of the populationexperience toxicity. In the diagram the MTC was selected at the concentration where 10% of thepopulation experience toxicity.

safety and the risk–benefit ratio acceptable for a given indication. For example, the MTCfor an over-the-counter analgesic or nonsteroidal anti-inflammatory drug will be chosenat a concentrations associated with much less toxicity than that of a drug used to treata life-threatening condition such as cancer. In Figure 1.11 the MEC was selected as theconcentration at which 70% of the population experienced a therapeutic benefit, and theMTC was selected as the concentration at which 10% of the population experienced someadverse effects.

The therapeutic range has been enormously useful clinically, particularly in helpingclinicians determine optimum doses of drugs that have both narrow therapeutic ranges andwide interpatient variability in dose requirements. Examples of these drugs are shown inTable 1.3. The optimum dose in one patient may produce concentrations below in MEC ina second and concentrations above the MTC in a third. As a result, doses are frequentlyindividualized by measuring plasma concentrations achieved by a typical dose and thenapplying pharmacokinetic principles to target concentrations in the therapeutic range.

TABLE 1.3 Therapeutic Ranges of Example Drugs (1)

Drug Therapeutic Range

Cyclosporine 100–400a �g/L, whole blood HPLCb analysisDigoxin 0.5–2c �g/LLithium 0.6–1.5 mEq/LPhenytoin 10–20 mg/LTacrolimus 5–20a �g/L, whole bloodTheophylline 5–15 mg/L

aDepending on the time after transplant, the type of transplant, and the preference of the center.bHigh-performance liquid chromatography.cDepending on the indication.

Page 38: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

18 INTRODUCTION TO PHARMACOKINETICS AND PHARMACODYNAMICS

It is, however, important to recognize that the therapeutic range has limitations, whichinclude:

1. It represents the range of concentrations that are optimum for most people. Certainpatients will, however, experience therapeutic effects at concentrations below theMEC, and others will experience toxicity below the MTC. Some patients neverrespond therapeutically to a drug even at concentrations well above the MTC.

2. It does not incorporate a graded concentration-related response (i.e., a response thatincreases with increases in concentration). It is an all-or-nothing response: Patientsare predicted to respond when the plasma concentration is within the establishedtherapeutic plasma concentration range, and not to respond when the plasma concen-tration is below the MEC.

3. It only applies to plasma concentrations that are in equilibrium with the drug con-centrations at the site of action. It can take a long time for some drugs to distribute totheir site of action. For example, it takes about 6 to 8 h for digoxin to fully distributeto its site of action (the myocardium of the heart). During this distribution period thetherapeutic range will not apply. For example, serum concentrations above the MTCin this period will not necessarily be associated with toxicity.

Therapeutic Index (TI) or Therapeutic Ratio. Like the therapeutic range, the therapeuticindex or therapeutic ratio is a way to express the safety margin offered by a drug. It is theratio of the dose of the drug that produces toxicity in 50% of patients to the dose of thedrug that produces therapeutic response in 50% of patients:

TI = TD50

ED50(1.5)

where TD50 is the dose that produces toxicity in half the patients and ED50 is the therapeuticdose in half the patients. If, for example, a drug has a therapeutic index of 100, this indicatesthat the toxic dose is about 100 times larger than the effective dose, and illustrates that thedrug has a wide safety margin. Conversely, a therapeutic index of 3 would indicate a smallmargin of safety. A drug with a small therapeutic ratio will have a narrow therapeutic range.

1.6 SUMMARY

In summary:

� Pharmacokinetics may be defined as a study of the relationship between drug concen-tration and time after the administration of a given dose. It involves the study of allthe processes that affect this relationship: that is, the drug ADME. Pharmacokineticsrepresents the first stage in the process of drug response.

� In pharmacokinetics, the plasma concentrations of a drug are usually studied. Agoal is to derive a mathematical expression for the relationship between the plasmaconcentration and the dose and time:

Cp = fPK(dose, time) (1.6)

Page 39: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc01 JWBS062-Rosenbaum February 17, 2011 8:12 Printer Name: Yet to Come

REFERENCE 19

where Cp is the plasma concentration of the drug, and f PK is a function that describesthe relationships among Cp, dose, and time and incorporates the drug’s pharmacoki-netic parameters.

� Pharmacodynamics may be defined as a study of the relationship between drug con-centration at the site of action and the onset, duration, and intensity of response tothe drug. The pharmacodynamic phase constitutes the second and final step in drugresponse.

� A goal is to derive a mathematical expression for the relationship between the responseand the drug concentration:

E = fPD(C) (1.7)

where E is the drug effect or response, C the concentration at the site of action, and f PD

a function that describes the relationship between the two and incorporates a drug’spharmacodynamic parameters.

� Integrating pharmacokinetics and pharmacodynamics covers the entire dose–responserelationship. Mathematical expressions for the pharmacokinetic and pharmacody-namic phases can be combined to provide a complete mathematical expression of thedose–response relationship:

E = fPD( fPK(dose, time)) (1.8)

� Equation (1.8) provides a complete expression for the time course of drug response. Itwill allow the drug response to be calculated at any time after any dose. It will allowoptimum dosing regimens to be determined and can be used to simulate drug responsedata in situations not studied clinically.

REFERENCE

1. Bauer, L. A. (2008) Applied Clinical Pharmacokinetics, 2nd ed., McGraw-Hill, New York.

Page 40: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

2PASSAGE OF DRUGSTHROUGH MEMBRANES

2.1 Introduction

2.2 Structure and Properties of Membranes

2.3 Passive Diffusion2.3.1 Transcellular Passive Diffusion2.3.2 Paracellular Passive Diffusion

2.4 Carrier-Mediated Processes: Transport Proteins2.4.1 Uptake Transporters: SLC Superfamily2.4.2 Efflux Transporters: ABC Superfamily2.4.3 Characteristics of Transporter Systems2.4.4 Simulation Exercise2.4.5 Clinical Examples of Transporter Involvement in Drug Response

Objectives

The material in this chapter will enable the reader to:

1. Distinguish diffusion and transporter-mediated passage

2. Distinguish transcellular and paracellular transport

3. Identify membrane and drug factors that control passive diffusion

4. Distinguish uptake and efflux transporters

5. Become familiar with members of the SLC and ABC superfamilies

6. Understand how transporters affect pharmacokinetics and pharmacodynamics

2.1 INTRODUCTION

A knowledge of how drugs penetrate membranes is fundamental to understanding theprocesses of drug absorption, distribution, metabolism, and excretion (ADME). The sys-temic circulation is the central transport medium for drugs. The process of drug absorption

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

20

Page 41: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

STRUCTURE AND PROPERTIES OF MEMBRANES 21

Plasma

Absorption

Tissue Distribution and

Redistribution

Elimination(Metabolism and Excretion)

Biological Membranes

Distributionto and from

Site of Action

FIGURE 2.1 Points of membrane penetration in pharmacokinetics. Drugs must pass one or moremembranes during absorption, distribution, metabolism, excretion, and delivery to the site of action.

results in drugs entering the systemic circulation from the place of drug administration (e.g.,gastrointestinal tract, intramuscular site, etc.). Drug distribution is the transport of drugsfrom the circulatory system to other parts of the body. Drug elimination (metabolism andexcretion) is a process by which drugs in the circulatory system are removed from the body(either by chemical modification, primarily in the liver, or by excretion of the parent drug,which occurs primarily in the kidney). All of these processes require that drugs penetratemembranes (Figure 2.1). Drugs taken orally must penetrate the gastrointestinal membrane.The first step in drug distribution involves the passage of drugs across the membranes of thecapillaries that bathe the various tissues. Drug metabolism requires that drugs penetrate thehepatocyte membrane. Finally, the ability of drugs to pass across the glomerular membraneand the membranes in the renal tubule controls a drug’s renal excretion. Most important,drugs must pass all the membranes that separate the site of action from the systemic circu-lation. Centrally acting drugs must penetrate the blood–brain barrier, drugs that act withina cell must penetrate cell membranes, and drugs used in the treatment of solid tumors mustpenetrate the tumor mass and remain within a cancer cell long enough to elicit a response.

2.2 STRUCTURE AND PROPERTIES OF MEMBRANES

To get into the body and get taken up by the tissues, drugs have to penetrate the epithelialmembranes that line the major organs and body cavities. Epithelial membranes consist ofa series of cells joined by water-filled junctions (Figure 2.2). The membranes of these cellsconsist of a bimolecular layer of lipoproteins. The two layers of lipids are oriented suchthat the polar ends of the molecule point out toward the aqueous medium or the outsideof the membrane, and the lipid component makes up the inner core or the matrix of themembrane. Various protein molecules are embedded at different points in the membranes

Page 42: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

22 PASSAGE OF DRUGS THROUGH MEMBRANES

Lumen:Apical Side

Capillary: Basolateral Side

Individual Cells

Water-Filled Junctions

Polar Heads of Lipid

TransmembraneProtein

Bimolecular lipoprotein membrane

LipophilicCore

FIGURE 2.2 Cell membrane. Individual epithelial cells are joined together by water-filled junc-tions. The membranes of the cells consist of bimolecular layers of lipoproteins. The polar portion ofthe lipid point toward the outer side of the membrane, and the lipid component makes up the innercore of the membrane. The apical side of the membrane points toward the outside, or lumen, and theremaining sides are known as the basolateral sides. (Diagram drawn by Linnea E. Anderson.)

(Figure 2.2). The portion of the epithelial membrane that faces the outside of the organor body (i.e., the side facing the gastrointestinal lumen) is called the apical side. Theremaining sides, including the side that faces the circulatory system, are the basolateralsides. Individual membranes may consist of a single layer or multiple layers of cells.During each process in ADME, a drug may have to pass across several membranes. Forexample, orally administered drugs must pass through the gastrointestinal wall into theinterstitial fluid and then through the capillary membrane to enter the blood. From therethey may have to pass several other membranes to access their site of action and to beremoved from the body. Drugs penetrate these membranes by either passive diffusion or bya transport-mediated process.

2.3 PASSIVE DIFFUSION

Passive diffusion is the most common way for drugs to pass through biological membranes.The concentration gradient across a membrane is the driving force for the process, whichtries to equalize the drug concentrations on either side of the membrane. As a result, thereis a net movement of drug from the side with a high concentration to the side with alow concentration. Any drug that is bound to tissue macromolecules or plasma proteins isessentially taken out of circulation and does not participate in the concentration gradient.The process of diffusion is governed by Fick’s law and may be expressed as:

dAb

dt= Pm · S Am · (Cu1 − Cu2) (2.1)

where dAb/dt is the amount of drug diffusing per unit time (mg/h), Pm the permeability ofthe drug through the membrane (cm/h), SAm the surface area of the membrane (cm2) Cu1

Page 43: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

PASSIVE DIFFUSION 23

the higher unbound drug concentration (mg/mL), and Cu2 the lower unbound concentration(mg/mL).

In most situations (e.g., during drug absorption and the initial phases of drug distribution),drug that diffuses across the membrane is diluted into a very large volume. Thus, Cu2 canbe considered to be considerably smaller that Cu1:

Cu1 � Cu2 and Cu1 − Cu2 ≈ Cu1

Thus, equation (2.1) may be written

dAb

dt= Pm · S Am · Cu1 (2.2)

From equation (2.2) it can be seen that under these circumstances, the rate of diffusionapproximates a first-order process:

dAb

dt= k · Cu1 where k = Pm · S Am (2.3)

The rate of diffusion is proportional to the driving force for the process: that is, theconcentration of drug. The constant of proportionality is the product of a drug’s permeability(the relative ease with which the drug passes the membrane) and the surface area of themembrane.

Surface Area According to equation (2.2), the rate of diffusion increases as a membrane’ssurface area increases. The importance of surface area is illustrated by considering thestructure of the membrane of the small intestine, a tissue whose function is to absorbessential nutrients from digested foods. To assist in this function, the membrane of thesmall intestine is folded into villi, or fingers, which are estimated to increase the surfacearea of the small intestine about 10-fold. Each of the villi is folded further into microvilli(Figure 2.3), which are estimated to increase the surface area an additional 20-fold. Thisextremely large surface area greatly enhances the absorptive properties, a feature that canalso be taken advantage of by drugs administered orally. The very favorable absorption ofthe small intestine results in its being the primary site for the absorption of drugs takenorally.

Permeability Permeability reflects the ability or speed with which a drug can pass througha membrane. It is dependent on both the characteristics of the membrane and the physic-ochemical properties of the drug: specifically, lipophilicity, charge, and size. The impactof these variables on diffusion depends on whether a drug passes through the membraneby the transcellular route or by the paracellular route. In transcellular transport, drugs dif-fuse through the matrix or core of the membrane. In paracellular transport, drugs diffusethrough the water-filled gaps between adjacent cells (Figure 2.4).

The nature of the core of the membrane is essentially constant from one type of mem-brane to another. As a result, the principles of transcellular diffusion are the same forall membranes, including the gastrointestinal membrane, the blood–brain barrier, and therenal tubular membrane. Paracellular diffusion is controlled by the nature of the junctionsbetween adjacent cells of the membrane. These vary from tissue to tissue. As a result, and

Page 44: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

24 PASSAGE OF DRUGS THROUGH MEMBRANES

Villi

Brush BorderCells

VenuleArteriole

Blood Vessels

Brush Border Cells

Microvilli

FIGURE 2.3 Villi and microvilli in the small intestine. The membrane of the small intestine isfolded into villi, which are then folded further into microvilli. This provides the membrane with avery large surface area, ideally suited for the absorption of nutrients and drugs. (Diagram drawn byLinnea E. Anderson.)

in contrast to transcellular diffusion, the ability of a drug to pass through a membrane bythe paracellular route will vary from one tissue to another.

2.3.1 Transcellular Passive Diffusion

The ease of transcellular diffusion is determined by a drug’s permeability across thelipophilic matrix of the membrane. As such, it depends on the lipophilicity, polarity, andsize of the drug molecule. A drug’s lipophilicity is probably the most important determi-nant of permeability. A drug’s lipophilicity, or fat-loving nature, is traditionally assessed by

Apical Side of Membrane

Basolateral Side of Membrane

TranscellularPenetration

ParacellularPenetration

Water Filled Junctions

FIGURE 2.4 Transcellular and paracellular diffusion. Passive diffusion is the most common waythat drugs penetrate membranes. They can pass though the matrix of the cell (transcellular passage)or through the water-filled junctions between adjacent cells (paracellular transport). (Diagram drawnby Linnea E. Anderson.)

Page 45: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

PASSIVE DIFFUSION 25

TABLE 2.1 Calculated Values of Log P, Log D7.4, and Log D6.0 of Several Drugs

Drug Log P Log D7.4 Log D6.0

Atenolol 0.10 −1.66 −2.74Famotidine −0.40 −1.02 −2.06Felodipine 4.83 4.83 4.82Furosemide 3.00 −0.12 0.26Ibuprofen 3.72 0.80 2.12Ondansetron 2.49 2.14 1.02Pindolol 1.97 0.18 −0.88Theophylline −0.17 −0.20 −0.18Valproic acid 2.72 0.16 1.51

Source: Ref. (1).

measuring its distribution between the immiscible phases of n-octanol and water. The ratioof the drug’s concentration in n-octanol and water is the drug’s partition coefficient (P):

P = Cn-octanol

Cwater(2.4)

Because of the very wide range of P values among therapeutic drugs, P values areexpressed most conveniently on a log scale. Table 2.1 provides the log P values of sometherapeutic drugs.

Drugs with large positive log P values (felodipine) preferentially partition into the lipidlayer. They are lipophilic, would have high permeability across the lipophilic core of themembrane, and would be expected to diffuse easily. As the log P value decreases amonga series of drugs, lipophilicity and permeability both decrease and transcellular membranepenetration would become increasingly difficult. Drugs with very low or negative log Pvalues (e.g., atenolol and famotidine) partition primarily into the aqueous phase, are morepolar in nature, and would be expected to have poor membrane permeability.

The disadvantage of the partition coefficient is that it measures the distribution of drugsbetween the two phases when the drug is completely in the nonionized state. Thus, it is ameasure of a drug’s inherent or intrinsic lipophilicity. But most drugs are either weak acidsor bases and, as a result, exist in biological fluids in equilibrium between their ionized andnonionized forms. For example, a drug that is a weak acid would ionize as follows in anaqueous medium:

DH −→←− D− + H+ (2.5)

The degree of ionization will influence a drug’s permeability because only the nonionizedform of the drug would be able to penetrate a lipophilic membrane. A more useful measureof membrane permeability is the partition coefficient measured at a specific, biologicallyrelevant pH. The partition coefficient of a drug between a lipid phase and an aqueous phaseat a specific pH is referred to as the distribution coefficient (D), or log D on the log scale.The log D7.4 and log D6.0 values of several drugs are shown in Table 2.1. It can be seenthat a drug’s log D value is generally less than its log P value, which reflects the fact thatwhen the drug is partially ionized, the ionized form cannot partition into the lipid phase,which makes the drug effectively less lipophilic than is the totally nonionized form of the

Page 46: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

26 PASSAGE OF DRUGS THROUGH MEMBRANES

drug. It can also be seen that the log D values of drugs that are weak acids (e.g., ibuprofen,furosemide, valproic acid) are higher at the more acidic pH of 6.0 than at pH 7.4. Thisreflects the fact that drugs are less ionized at more acidic pH’s. Conversely, the log D valuesof drugs that are weak bases (e.g., atenolol, ondansetron) are lower at a more acidic pH of6.0, owing to the greater degree of ionization.

The size or mass of the drug molecules also affects the permeability, as large moleculesexperience difficulty diffusing. Studies suggest, however, that mass does not appear tobe important if it is below about 400 daltons (Da), which includes many of the drugspresently used therapeutically. Transcellular diffusion has been studied most extensivelyin the context of the absorption of orally administered drugs. Pharmaceutical companieswant to maximize the possibility that newly developed drugs can be given orally, andmuch research has been conducted to try to identify critical physiochemical properties ofdrugs. Studies have emphasized the importance of a high log D value. These studies alsoindicate that mass and polarity are important and suggest that drugs with a molecular massgreater than 500 Da are likely to have poor membrane permeability, particularly if theyalso possess additional adverse physicochemical characteristics, such as polarity or poorlipophilicity (2).

In summary, transcellular permeability is highest for small lipophilic, nonpolar drugs.

2.3.2 Paracellular Passive Diffusion

Paracellular transport involves to the passage of drugs through the junctions between thecells of the membrane (Figure 2.4). It is dependent on the size of the junction and on the sizeof the drug molecule. The junctions between adjacent cells of the epithelium membranevary from one tissue to another. The junctions between the cells in the gastrointestinalmembrane and skin are very tight and serve to hold transcellular proteins in place andalso, presumably, to protect the body from the penetration of foreign substances acrossthese outside membranes. As a result, paracellular diffusion of drugs across the intestinalmembrane is a very minor route of absorption. Atenolol [molecular weight or molecularmass (MW) = 266 Da; log D6.5 =−2] and terbutaline (MW = 180 Da; log D6.5 =−1.3) werethought to be absorbed by this route. However, recent evidence has cast doubt on this (3) andsuggests that they may be too large for paracellular diffusion across the intestinal membrane.The aminoglycoside antibiotics, which are both too polar (log D7.4 ≈ −10) and too large(MW 450 to over 1000) for transcellular or paracellular diffusion, must be administeredparenterally for the treatment of systemic infections. The gaps between adjacent cells in thenasal membrane are looser in nature, and the cutoff for paracellular absorption is around1000 Da. However, even at this site, paracellular diffusion of polar drugs is much lessefficient than transcellular absorption of lipophilic drugs. A very high percentage of a doseof a lipophilic drug such as pentazocine and fentanyl can be absorbed by the transcellularroute through the nasal membrane, compared to only about 10% of a polar drug such asmorphine. The use of absorption enhancers to improve the intranasal delivery of polardrugs has met with some success. Chitosan, a compound derived from crustacean shells,has produced substantial increases in morphine penetration through the nasal membrane(4). It is believed that the effect of chitosan is due in part to a transient opening of thetight junctions and increased paracellular diffusion. Clinical trials of intranasal morphinepreparations containing chitosan suggest that it will offer an alternative to intravenousmorphine for the treatment of postoperative pain (5).

Page 47: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

CARRIER-MEDIATED PROCESSES: TRANSPORT PROTEINS 27

Capillary membranes in most tissues have fairly loose membrane junctions that allow freepassage of even large molecular mass (high molecular weight) peptides and polypeptidesinto and out of the circulatory system. Large molecular mass drug molecules such as theaminoglycosides are able to pass through these membranes by the paracellular route. Thehydrophilicity of the aminoglycosides will prevent them from diffusing passively throughthe membranes within the tissues, and as a result their distribution is limited primarily toextracellular fluid. The aminoglycosides are active against bacteria because bacterial cellmembranes contain an oxygen-dependent transport process that carries the drugs to theinside of the bacterial cell. The membrane in the renal glomerulus is another example ofa very loose membrane that allows the paracellular passage of even large drug molecules.Plasma water and solutes undergo paracellular filtration across this membrane into therenal tubule. The paracellular space in the glomerular membrane allows the passage ofmolecules with molecular weights up to about 5000 Da. For example, insulin is able to passthrough the glomerular membrane. In contrast, in certain sensitive or delicate tissues, suchas the central nervous system, the retina, the placenta, and the testes, the capillary epithelialmembrane is very tightly knit and limits paracellular diffusion, presumably to protect thesetissues from potentially toxic drugs and other xenobiotics.

2.4 CARRIER-MEDIATED PROCESSES: TRANSPORT PROTEINS

Transporters are proteins that reside in cell membranes and serve to facilitate the passageof chemicals into or out of a cell. It is likely that transporters have evolved to protectcells against potentially harmful xenobiotics as well as to assist in the absorption anddistribution of essential nutrients. Over the last 15 to 20 years, it has become apparent thatdrug transporters exert a profound influence on the body’s exposure to drugs and on theaccess of drugs to their site of action and/or toxicity. Transporters located in the intestinalmembrane, the hepatocyte, and the renal tubular membrane influence drug absorption,metabolism, and excretion, respectively, and as a result, the body’s overall exposure todrugs. In other locations, such as the central nervous system, the placenta, and the testis,transporters may have only subtle or negligible influence on the body’s overall exposure toa drug, but by virtue of their control of the access of a small fraction of the dose to theseareas are critical in controlling therapeutic and/or toxicological effects.

There are two broad classes of transporters: uptake transporters, which transport drugsinto the cell, and efflux transporters, which extrude or transport drugs out of the cell.In gastrointestinal, hepatocytic, and renal tubular membranes, transporters may resideon either the basolateral (blood) side or on the apical (luminal) side of the membrane.The four possible arrangements of drug transporters in an epithelial membrane such asthe gastrointestinal tract are shown in Figure 2.5. The location of a transporter in themembrane determines its impact on the body’s exposure to a substrate. For example, anefflux transporter on the apical side of a membrane would restrict the body’s exposure to itssubstrates. In contrast, an efflux transporter on the basolateral side would serve to preservea drug. If the drug were a substrate for both an efflux transporter on the basolateral side ofthe membrane and an uptake transporter on the luminal side, the two transporter systemscould work in concert to absorb and/or retain the drug in the body.

A very large number of transporters have been identified in human tissue, but the roleof many is not fully understood at this time. The discussion that follows is restricted to

Page 48: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

28 PASSAGE OF DRUGS THROUGH MEMBRANES

UApical Membrane

Basolateral Membrane

Lumen

Blood

E

UE

Out

Out

In

In

FIGURE 2.5 Theoretical placements of uptake and efflux transporters in an epithelial cell mem-brane. E represents an efflux transporter such as permeability glycoprotein (P-gp) or breast cancerresistance protein (BCRP); U represents an uptake transporter such as organic anion transportingpolypeptide (OATP) or organic cation transporter (OCT). Uptake transporters on the apical mem-brane and efflux transporters on the basolateral side promote the retention of drugs in the body.Uptake transporters on the basolateral side of the membrane and efflux transporters on the apical sidepromote the removal of drug from the body. (Diagram drawn by Linnea E. Anderson.)

those transporters whose involvement in drug response and/or pharmacokinetics has beendemonstrated in clinical studies. The role of specific transporters in drug absorption, drugexcretion in the kidney, drug elimination in the liver, and drug uptake into the centralnervous system is discussed in greater detail in later chapters.

2.4.1 Uptake Transporters: SLC Superfamily

The main uptake transporters belong to the solute carrier (SLC) superfamily and operateprimarily by bidirectional facilitated diffusion, although in some cases the transport maybe active and unidirectional. As a result, these proteins generally transport drug moleculesalong their concentration gradient but at a higher rate than that observed for passive dif-fusion. These transporters do not require adenosine triphosphate (ATP), and they transportsubstances into the cell. Of the very large number of SLC transporters that have beenidentified, four families appear to be involved in drug absorption and disposition (distri-bution and elimination) in humans: the organic anion transporting polypeptide (OATP,gene symbol SLCO), the organic anion transporter (OAT, gene symbol SLC22), the or-ganic cation transporter (OCT, gene symbol SLC22), and the peptide transporter (PEPT;gene SLC15A). Some examples of specific transporters, their tissue distribution, and someexample substrates are shown in Table 2.2.

Organic anions are transported by the OATP and the OAT families. The OAT1 and OAT3are important in the kidney, where they facilitate the uptake of drugs from the blood intothe renal tubular cells. The OATP family is important in drug absorption in the intestine andthe uptake of drugs into the hepatocyte, presumably to facilitate metabolism. Small organiccations such as metformin, cisplatin, procainamide, and cimetidine are transported by theOCT family (OCT1 and OCT2). These two carriers display different tissue distributions:OCT1 is expressed primarily in the liver, where it enhances hepatic uptake of drugs, and

Page 49: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

CARRIER-MEDIATED PROCESSES: TRANSPORT PROTEINS 29

TABLE 2.2 Primary Locations of Major Drug Uptake Transporters and Some ExampleSubstrates and Inhibitorsa

Transporter Gene Name Location Example Substrates Inhibitors

OATP1A2 SLCO1A2 Brain: ASIntestine: AS

Fexofenadine, digoxin,saquinavir

Fruit (apple, grapefruit,orange) juices

OATP1B1 SLCO1B1 Liver: BS Pravastatin, atorvastatin,olmesartan, valsartan

CyclosporineRifampin

OATP1B3 SLCO1B3 Liver: BS Pravastatin, atorvastatin,olmesartan, valsartan,fexofenadine

OAPT2B1 SLCO2B1 Liver: BSIntestine: AS

Pravastatin, atorvastatin,benzylpenicillin

OATP4C1 SLCO4C1 Kidney: BS DigoxinOCT1 SLC22A1 Liver: BS Metformin, cimetidine Ritonavir

CimetidineOCT2 SLC22A2 Kidney: BS Metformin, cimetidine,

cisplatinCimetidine

OAT1 SLC22A6 Kidney: BS Overlaps with OAT3:furosemide, adefovir,acylcovir,cephalosporins

ProbenecidCephalosporins

OAT3 SLC22A8 Kidney: BS Overlaps with OAT1:furosemide, adefovir,pravastatin, olmesartan,fexofenadine,H2-receptor blockers,benzylpenicillin

ProbenecidCimetidineNSAIDsCephalosporins

PEPT1 SLC15A1 Intestine: AS Cephalosporins,penicillins, valacyclovir

Cephalosporins

Source: The data in the table were obtained from several sources (6–11).aAS is the apical or luminal side of the membrane; BS is the basolateral side of the membrane NSAIDs isnonsteroidal anti-inflamatory drugs.

OCT2 is expressed primarily in the kidney, where it enhances the uptake of its substratesinto the renal tubular membrane from the blood. There appears to be some but not completeoverlap in the substrate specificity of these two transporters. The location of the main uptaketransporters in the intestinal, hepatocytic, and proximal renal tubular cells are shown inFigure 2.6.

2.4.2 Efflux Transporters: ABC Superfamily

Efflux transporters were first identified in the 1970s when a glycoprotein was found toprovide some cancer cells with resistance to certain anticancer drugs. The protein, known aspermeability glycoprotein (P-gp) or multidrug resistance protein 1 (MDR 1; gene ABCB1),was found to operate by reducing cellular exposure to a particular drug by actively extrudingit from the cell. Since that time, P-gp and several other efflux transporters have been foundnot only in cancer cells, but also in many normal tissues, where they are thought to functionto protect healthy cells from damage by drugs and other xenobiotics. The efflux transporters

Page 50: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

30 PASSAGE OF DRUGS THROUGH MEMBRANES

FIGURE 2.6 Location of some of the major uptake (U) and efflux (E) transporters involvedin drug absorption, metabolism, and excretion. Efflux transporters include permeability glyco-protein (P-gp), multidrug resistance–associated protein family (MRP), breast cancer resistanceprotein (BCRP), and bile salt export pump (BSEP). Uptake transporters include organic an-ion transporting polypeptide (OATP), organic anion transporter (OAT), organic cation transporter(OCT), and peptide transporter (PEPT). An interactive version of this diagram may be found athttp://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter2 and can be used to ob-serve how transporters at the various locations influence the plasma concentration–time profile.

belong to the ATP-binding cassette (ABC) superfamily. These ABC efflux transportersfacilitate the removal of drugs from cells by an active transport process. Clinically importantmembers of this superfamily include P-gp, multidrug resistance-associated protein (MRP;gene ABCC) family, breast cancer resistance protein (BCRP; gene ABCG2), and bile saltexport pump (BSEP; gene ABCB11; also known as sister P-glycoprotein). The locationof the efflux transporters and some examples of substrates and modifiers are shown inTables 2.3 and 2.4.

P-gp is found in many areas of the body, including the intestinal membrane, theblood–brain barrier, the placenta, the kidney, and the liver. In these locations it serveseither to prevent drugs from being absorbed into the body or to facilitate their removal fromthe body or individual cells. It transports such a wide variety of different molecules that ithas been termed the “promiscuous” transporter. This broad substrate specificity, combinedwith the large number of compounds known to modify its activity, make P-gp an importanttarget for drug–drug interactions. In view of its importance and the relatively large bodyof information on this transporter, a separate list of its substrates, inhibitors, and inducersis provided in Table 2.4. The very large overlap in the substrates, inducers, and inhibitorsof P-gp with those of cytochrome P450 3A4 (CYP3A4), a hepatic and intestinal enzyme,suggest that P-gp and CYP3A4 may have evolved to work in concert to reduce the body’s

Page 51: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

CARRIER-MEDIATED PROCESSES: TRANSPORT PROTEINS 31

TABLE 2.3 Primary Locations of Major Drug Efflux Transporters and SomeExample Substratesa

TransporterGeneName Location Example Substrates Inhibitors

P-gp MDR1ABCB1

Brain: ASLiver: ASKidney: ASIntestine: ASPlacentaTestis

See Table 2.4 See Table 2.4

MRP2 ABCC2 Liver: ASKidney: ASIntestine: AS

Pravastatin, olmesartan,indinavir, conjugates ofgluathione andglucuronidesulfasalazine

Cyclosporine

MRP3 ABCC3 Liver: BSIntestine: BS

Methorexate, glucoronides

MRP4 ABCC4 Liver: BSKidney: AS

Methotrexate, adefovir

BCRP ABCG2 Brain: ASLiver: ASIntestine: AS

Irinotecan, topotecan,daunorubicin, imatinib,mitoxantron, quinolones,pitavastatin, rosuvastatinsulfasalazine

Proton pumpInhibitorsProtease

inhibitorsElacridar

BSEP ABCB11 Liver: AS Vinblastine, tamoxifen CyclosporineTroglitazoneGlyburide

Source: The data in the table were obtained from several sources (6,7,12,13).aAS is the apical or luminal side of the membrane; BS is the basolateral side of the membrane.

exposure to harmful chemicals. MRP2 is found in the apical membrane in the kidney, liver,and intestine. In the liver it promotes the biliary excretion of anionic drugs and conjugatesof glucoronide, glutathione, and sulfate. Some of the MRP transporters, such as MRP4 inthe hepatocyte and intestine, are located on the basolateral side of the membrane, wherethey promote the absorption or retention of substrates in the systemic circulation. BCRPis found in many tissues, including the intestinal membrane, liver, brain, and placenta, andhas been studied primarily in relation to cancer treatment because several anticancer drugs,including irinotecan and topotecan, are substrates. However, many other drugs, includingcimetidine and rosuvastatin, are substrates of BCRP. Apart from its involvement in thebiliary secretion of bile salts, the role of BSEP is not fully understood. The location of themain efflux transporters in the intestine, hepatocytic, and proximal renal tubular cells isshown in Figure 2.6.

2.4.3 Characteristics of Transporter Systems

Transporter-mediated membrane permeation differs in several ways from diffusion-mediated transport. First, transporters are substrate-specific and only carry drugs that havespecific molecular features. Second, there is only a finite amount of a transporter at a given

Page 52: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

32 PASSAGE OF DRUGS THROUGH MEMBRANES

TABLE 2.4 Examples of Substrates, Inhibitors, and Inducers of P-gp

SubstratesAnticancer drugs Cardiovascular drugs H2-receptor antagonists

Paclitaxel Verapamil CimetidineDoxorubicin Diltiazem RanitidineVinblastine Digoxin ImmunosuppressantsEtoposide Talinolol CyclosporineIrinotecan Quinidine Tacrolimus

Antiemetics Amiodarone Protease inhibitorsOndansetron Losartan IndinavirAntihistamines Atorvastatin NelfinavirFexofenadine Saquinavir

RitonavirInhibitors

Cyclosporine Second generation Third generationVerapamil Valspodar ElacridarQuinidineItraconazoleKetoconazoleRitonavir

InducersRifampin St. John’s wort

Source: The data in the table were obtained from several sources (8,12,14).

location, and if the substrate concentration is high, it may saturate the transporter. For ex-ample, consider a drug that is a substrate for an intestinal efflux transporter, which limits itsabsorption. If the concentration of the drug in the intestine becomes so high that it saturatesthe transporter, the efflux process will no longer work efficiently and the drug will be ableto undergo a greater degree of absorption. Competition between two drugs for the sametransporter could also result in reduced transport of a drug. Finally, because transporters arebiological in nature, their expression is under genetic control and their activity may varysubstantially within a population. Their activity can be further modified by concomitantdrugs, nutritional products, foods, and environmental factors that either inhibit or inducetransport systems.

2.4.4 Simulation Exercise

An interactive model of the major uptake and efflux transporters involved in absorptionand elimination is provided at the following link:

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter2

Simulations can be performed to observe how the transporters at the intestine andrenal tubule influence the plasma concentration–time profile.

1. Go to the Single Oral Dose page and perform a simulation with all the transportersystems switched on. Run four more simulations in which each of the transportersis switched off in turn. Observe how the plasma concentration profile and half-lifeare affected in each case.

2. Repeat the exercise with multiple oral and intravenous doses.

Page 53: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

CARRIER-MEDIATED PROCESSES: TRANSPORT PROTEINS 33

3. Digoxin is a substrate for P-gp in the intestinal membrane and renal tubule. Usethe single oral dose model to observe how digoxin’s plasma concentration–timerelationship and half-life may be affected when:

(a) Intestinal P-gp is inhibited

(b) Renal P-gp is inhibited

Comment on any differences in the two profiles.

4. Repeat the exercise above with the multiple intravenous model.

5. How would it be possible to distinguish between effects on intestinal and renalP-gp?

6. Give four examples of modifiers of P-gp.

2.4.5 Clinical Examples of Transporter Involvement in Drug Response

In an effort to identify clinically important drug transporters and delineate their role indrug response, there has been an explosive growth in the number of research paperspublished in this area during the past 15 years. It is likely that our knowledge of thesesystems and the specific functions of individual transporters will grow at a rapid ratein the coming years. The transporters are frequently studied in vitro, using human cellsand tissues, and in animals, in which the activity of a transporter is altered either bychemical inhibitors or by breeding animals deficient in the transporter (e.g., knockoutmice). Clinical studies are then performed to try to confirm the findings of the in vitro andanimal studies.

The clinical role played by transporters is usually studied by observing the effects ofaltered activity, either in people who have genetically determined low activity or usingchemical modifiers (inhibitors and inducers) of specific transport systems. These studieshave revealed many interesting and important characteristics of transporters. Inhibitors ofintestinal P-gp have been found to increase the absorption of many drugs, which in somecases (e,g., digoxin) can lead to toxicity. Conversely, inducers of P-gp, such as rifampin,can reduce the absorption of P-gp substrates and in some cases (e.g., cyclosporine) can leadto dangerously low, subtherapeutic blood concentrations of a drug. The hepatic transportersOATP1B1 and OCT1 are important for the delivery of HMG-CoA reductase inhibitors(statins) and metformin, respectively, to their site of action in the liver. Patients withreduced OCT1 activity may have reduced response to metformin because of lower hepaticuptake (11). The hepatic uptake of the statins by OATP1B1 also facilities their eliminationin the liver, where they either undergo metabolism or are excreted into the bile. Reducedactivity of OATP1B1 can inhibit the elimination and produce large increases in systemicconcentrations of these drugs, and predispose individuals to an increased risk of muscletoxicity. Several of the hepatic efflux transporters facilitate secretion into the bile of drugs,their metabolites, and other metabolic waste products. Inhibition of the activity of thesetransporters can lead to a buildup of these compounds in the liver and predispose patients tocholestasis. Uptake and efflux transporters on the basolateral and apical sides, respectively,of the renal tubular membrane can act in concert to enhance the renal excretion of drugs.The uptake transporters carry drugs from the circulatory system into the tubular membrane,and the efflux transporters continue the elimination process by secreting the drugs intothe renal tubule. However, the high drug concentrations in renal tabular cells that resultfrom this uptake have been implicated as a possible cause of the renal toxicity of cisplatin,cidofovir, and cephaloridine.

Page 54: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

34 PASSAGE OF DRUGS THROUGH MEMBRANES

Transporters at the blood–brain barrier restrict the uptake of potentially toxic substancesinto this very sensitive organ. The opioid loperamide is devoid of central effects becauseP-gp prevents its accumulation in the brain, and as a result it is readily available over thecounter. However, when coadministered with quinidine, an inhibitor of P-gp, loperamideproduces respiratory depression, which demonstrates that significant amounts are able toaccess the central nervous system. P-gp may also play a role in excluding the nonsedating,second-generation antihistamines from the central nervous system (15), which raises thepossibility that inhibitors of P-gp could produce sedation from these drugs. By limitingaccess to the central nervous system, the efflux transporters also create a significant barrierfor the treatment of such conditions of the brain as cancer, neurological diseases, mooddisorders, and infections.

REFERENCES

1. Linnankoski, J., Makela, J. M., Ranta, V. P., Urtti, A., and Yliperttula, M. (2006) Computationalprediction of oral drug absorption based on absorption rate constants in humans, J Med Chem49, 3674–3681.

2. Lipinski, C. A., Lombardo, F., Dominy, B. W., and Feeney, P. J. (2001) Experimental and com-putational approaches to estimate solubility and permeability in drug discovery and developmentsettings, Adv Drug Deliv Rev 46, 3–26.

3. Lennernas, H. (2007) Intestinal permeability and its relevance for absorption and elimination,Xenobiotica 37, 1015–1051.

4. Illum, L., Watts, P., Fisher, A. N., Hinchcliffe, M., Norbury, H., Jabbal-Gill, I., Nankervis, R.,and Davis, S. S. (2002) Intranasal delivery of morphine, J Pharmacol Exp Ther 301, 391–400.

5. Stoker, D. G., Reber, K. R., Waltzman, L. S., Ernst, C., Hamilton, D., Gawarecki, D., Mer-melstein, F., McNicol, E., Wright, C., and Carr, D. B. (2008) Analgesic efficacy and safety ofmorphine–chitosan nasal solution in patients with moderate to severe pain following orthopedicsurgery, Pain Med 9, 3–12.

6. Ho, R. H., and Kim, R. B. (2005) Transporters and drug therapy: implications for drug dispositionand disease, Clin Pharmacol Ther 78, 260–277.

7. Kusuhara, H., and Sugiyama, Y. (2009) In vitro–in vivo extrapolation of transporter-mediatedclearance in the liver and kidney, Drug Metab Pharmacokinet 24, 37–52.

8. Oostendorp, R. L., Beijnen, J. H., and Schellens, J. H. (2009) The biological and clinical role ofdrug transporters at the intestinal barrier, Cancer Treat Rev 35, 137–147.

9. United States Food and Drug Administration. (2006) Drug Development and Drug Interactions:Table of Substrates, Inhibitors and Inducers, U.S. FDA, Washington, DC.

10. Yasui-Furukori, N., Uno, T., Sugawara, K., and Tateishi, T. (2005) Different effects of threetransporting inhibitors, verapamil, cimetidine, and probenecid, on fexofenadine pharmacokinet-ics, Clin Pharmacol Ther 77, 17–23.

11. Choi, M. K., and Song, I. S. (2008) Organic cation transporters and their pharmacokinetic andpharmacodynamic consequences, Drug Metab Pharmacokinet 23, 243–253.

12. Marchetti, S., Mazzanti, R., Beijnen, J. H., and Schellens, J. H. (2007) Concise review: clinicalrelevance of drug drug and herb drug interactions mediated by the ABC transporter ABCB1(MDR1, P-glycoprotein), Oncologist 12, 927–941.

13. Murakami, T., and Takano, M. (2008) Intestinal efflux transporters and drug absorption, ExpertOpin Drug Metab Toxicol 4, 923–939.

Page 55: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc02 JWBS062-Rosenbaum February 2, 2011 15:28 Printer Name: Yet to Come

REFERENCES 35

14. Eyal, S., Hsiao, P., and Unadkat, J. D. (2009) Drug interactions at the blood–brain barrier: factor fantasy? Pharmacol Ther 123, 80–104.

15. Polli, J. W., Baughman, T. M., Humphreys, J. E., Jordan, K. H., Mote, A. L., Salisbury, J. A.,Tippin, T. K., and Serabjit-Singh, C. J. (2003) P-glycoprotein influences the brain concentra-tions of cetirizine (Zyrtec), a second-generation non-sedating antihistamine, J Pharm Sci 92,2082–2089.

Page 56: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

3DRUG ADMINISTRATION, ABSORPTION,AND BIOAVAILABILITY

3.1 Introduction: Local and Systemic Drug Administration

3.2 Common Routes of Systemic Drug Administration3.2.1 Intravascular Direct Systemic Administration3.2.2 Extravascular Parenteral Routes3.2.3 Other Extravascular Routes

3.3 Overview of Oral Absorption

3.4 Extent of Drug Absorption3.4.1 Bioavailability Factor3.4.2 Individual Bioavailability Factors

3.5 Determinants of the Bioavailability Factor3.5.1 Disintegration3.5.2 Dissolution3.5.3 Formulation Excipients3.5.4 Adverse Events Within the Gastrointestinal Lumen3.5.5 Transcellular Passive Diffusion3.5.6 Paracellular Passive Diffusion3.5.7 Uptake and Efflux Transporters

3.5.7.1 Uptake Transporters3.5.7.2 Efflux Transporters

3.5.8 Presytemic Intestinal Metabolism or Extraction3.5.9 Presystemic Hepatic Metabolism or Extraction

3.6 Factors Controlling the Rate of Drug Absorption3.6.1 Dissolution-Controlled Absorption3.6.2 Membrane Penetration–Controlled Absorption3.6.3 Overall Rate of Drug Absorption

3.7 Biopharmaceutics Classification System

Problems

Objectives

The material in this chapter will enable the reader to:

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

36

Page 57: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

COMMON ROUTES OF SYSTEMIC DRUG ADMINISTRATION 37

1. Define local and systemic drug administration

2. Describe common routes of drug administration and their characteristics

3. Characterize the steps involved in oral absorption

4. Define the bioavailability factor (F)

5. Categorize the components of F: Fa, Fg, and Fh

6. Identify the drug, formulation, and biological determinants of bioavailability

7. Understand the determinants of the rate of drug absorption

8. Define the biopharmaceutical classification system

9. Understand how the BCS and BDDCS can be applied

3.1 INTRODUCTION: LOCAL AND SYSTEMIC DRUG ADMINISTRATION

Drugs can be administered using a variety of routes. The route must be selected so that thedrug is delivered to its site of action in a time frame appropriate for the clinical indication.For some indications, such as the treatment of life-threatening cardiac arrhythmias, it iscritical that a drug reaches its site of action within a few minutes of administration. In othersituations the patient will not be adversely affected if it takes 0.5 to 1 h or more for thedrug to elicit an effect. Drug administration may be broadly classified as local or systemic.Local administration refers to the direct application of a drug to its site of action, andit is much less common than systemic administration. Examples of local administrationinclude the use of creams, ointments, or gels to apply drugs topically: for example, theuse of lidocaine gel to provide anesthesia at the site of application. Other examples are thetreatment of ulcerative colitis with oral sulfasalazine, a drug that is not absorbed throughthe intestinal membrane and is delivered to the colon; and the treatment of asthma withinhaled �-agonists and corticosteroids. In contrast, the vast majority of drugs rely on thecirculatory system to deliver drugs to their site of action. This is referred to as systemicadministration, and in many cases the uptake or absorption of the drug into the systemiccirculation is a critical first step in response process. In pharmacokinetics, drug absorptionrefers specifically to the uptake of drug to the systemic circulation and not just its entryinto the body. For example, in the case of orally administered drugs, drug absorption is notsimply passage of the drug across the intestinal membrane—this is only an intermediarystep in the overall absorption of the drug into the systemic circulation.

3.2 COMMON ROUTES OF SYSTEMIC DRUG ADMINISTRATION

3.2.1 Intravascular Direct Systemic Administration

Intravenous and Intraarterial The administration of drugs directly into the systemic cir-culation using the intravenous route, or the less common intraarterial route, bypasses theabsorption process completely. These routes of administration are invasive and require askilled health professional and are generally not used unless there is a specific reason.Intravenous administration is often used when an immediate effect is desired and/or whenit is important that the dose be administered with a high degree of accuracy. It may alsobe used for drugs that are poorly or incompletely absorbed from other routes. Intraarterial

Page 58: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

38 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

administration is not common but may be used if a therapy is to be directed to a specificorgan. For example, it has been used to administer anticancer drugs to target liver tumors.Intravascular administration is the only route that guarantees that the entire dose adminis-tered will reach the systemic circulation. For all other routes of drug administration, poormembrane penetration and/or loss of drug at the absorption site may result in the incom-plete absorption of a dose. An extreme example of this is provided by alendronate, usedin the treatment of osteoporosis. Only about 0.7% of an oral dose of this drug reachesthe systemic circulation. As a result of the complete absorption, doses of drugs given in-travenously are often much lower than those used in other routes, where absorption maybe incomplete. For example, the intravenous dose of morphine is about one-third of theoral dose.

3.2.2 Extravascular Parenteral Routes

Intramuscular and Subcutaneous Administration Like intravenous and intraarterial, in-tramuscular and subcutaneous drug delivery are examples of parenteral routes. Parenteralis defined as “outside the gastrointestinal tract” and, strictly speaking, would include allnonoral routes. However, in the pharmaceutical field, the term is limited to injectable routes.The intramuscular and subcutaneous routes are frequently used to avoid the gastrointestinaltract, either because the drug would be destroyed in the gastrointestinal fluid or because adrug is too polar or too large to penetrate the gastrointestinal membrane. Drugs adminis-tered by the intramuscular and subcutaneous routes must still undergo absorption into thebloodstream, but generally, loose capillary membranes at their site of administration allowsparacellular penetration even of polar and/or large drug molecules. Drugs administered bythe intramuscular and subcutaneous routes often reach the bloodstream faster than do orallyadministered drugs. However, the physicochemical properties of a drug (e.g., particle size)can be manipulated to provide a more gradual absorption from these routes.

3.2.3 Other Extravascular Routes

The convenience of oral administration makes it the most common route of drug admin-istration. As such, the bulk of this chapter is devoted to discussing the principles of oraldrug absorption. However, many drugs cannot be administered orally, either because theyare destroyed in the gastrointestinal fluid, unable to penetrate the intestinal membrane, oreliminated in the liver during the absorption process. Most of the other extravascular routesof drug administration were developed initially to provide a nonparenteral alternative forthe administration drugs that cannot be given orally.

Buccal and Sublingual The buccal (between the gums and cheek) and sublingual (underthe tongue) routes of administration take advantage of relatively porous, well-perfusedmucous membranes in the mouth. The major advantage of these routes is that the capil-laries in this area do not drain into the hepatic artery, and thus they offer an alternativeoral route for drugs such as nitroglycerin that undergo extensive metabolism in the liverand are essentially removed from the body before they reach the systemic circulation(see Section 1.3.2). Drug absorption from these routes is fairly rapid, a feature that has

Page 59: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

COMMON ROUTES OF SYSTEMIC DRUG ADMINISTRATION 39

enables buccal and sublingual preparations of fentanyl to be used for the treatment ofbreakthrough pain.

Rectal The rectal route of administration has the advantage of bypassing the liver. It isalso a particularly useful route for patients who cannot take drugs orally, either because theyare experiencing nausea and vomiting or are unconscious. Prochlorperazine, for example,is available in suppository form for the treatment of nausea and vomiting.

Transdermal The transdermal route was initially developed as a way to bypass the liver andadminister drugs that experienced extensive hepatic first pass extraction, but it has becomepopular because of the convenience it offers. A single patch can provide continuous andconstant delivery of drug over an extended period, making it particularly attractive for drugsthat are eliminated rapidly and usually require frequent administration. Examples of drugsthat are available as transdermal patches include methylphenidate, estradiol/norethindrone,fentanyl, and nitroglycerin. This route is limited to small lipophilic drugs, which canpenetrate the poorly perfused, tightly knit stratum corneum layer of the skin. For thepractical reason of accommodating the drug in the patch, this route is also limited todrugs that are used in relatively small doses. Rotigotine, a new drug for the treatment ofParkinson’s disease, was recently launched as a transdermal patch because it experiencedextensive hepatic extraction from the oral route. Unfortunately, the product had to bewithdrawn due to crystallization of the drug in the patch.

Intranasal Drugs administered by the intranasal route take advantage of the large surfacearea and highly permeable nature of the nasal membrane. This area is also very wellperfused with blood, which enables drugs to be absorbed rapidly. The intranasal route hasbeen pursued aggressively as a means of administering protein and peptide drugs, whichgenerally have to be administered parenterally. Intranasal formulations of calcitonin anddesmopressin are currently on the market, and the intranasal delivery of insulin is beingpursued. The rapid absorption of traditional small drug molecules from this route hasresulted in its use when a rapid onset of action is required. For example, nasal formulationsare available for analgesics (morphine), antimigraine drugs (sumatriptan and zolmitriptan),and the intranasal delivery of naloxone has been used as an alternative to the intravenousroute in the treatment of heroin overdose. Because a portion of the dose administered bythe intranasal route gains direct access to the central nervous system, this route is beinginvestigated as a means of avoiding the blood–brain barrier in the delivery of drugs used inthe treatment of such diseases as Alzheimer’s disease and Parkinson’s disease (1). Intranasaldelivery is limited by the local irritation caused by the drugs and excipients, and constraintson the amount of drug that can be formulated in the small volume of an intranasal unit dose.

Pulmonary The pulmonary route is used primarily for the local administration of agentsto treat asthma and other respiratory conditions. The large surface area, good permeability,and extremely high perfusion of the alveolar membrane make it ideally suited for drugabsorption. As a result, it has been investigated as a route for the administration of ther-apeutic agents, particularly proteins and peptides, which generally must be administeredparenterally. An inhalation form of insulin (Exubera) was approved by the U.S. Food andDrug Administration but subsequently withdrawn due to poor sales, as it failed to gain

Page 60: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

40 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

acceptance among health professionals and patients. Concern has also been expressedabout the long-term health effects of using the delicate alveoli to transport proteins.

3.3 OVERVIEW OF ORAL ABSORPTION

Orally administered drugs are formulated into convenient, palatable dosage forms, suchas tablets (compressed powder mass), capsules, solutions, suspensions, and emulsions.The remainder of the chapter is devoted to discussing the absorption of drugs from theoral route. The discussion focuses on absorption from standard dosage forms rather thanthe more specialized topic of controlled-release products. When an individual dose isconsumed, the drug must go through a series of processes and steps before reaching thesystemic circulation. These steps control both the amount of the dose that is eventuallyabsorbed into the systemic circulation and the rate of drug absorption.

After a patient has swallowed a tablet, it enters the stomach and is wet by the gastricfluid. The tablet must break up or disintegrate in the fluid so that the drug contained withinits matrix is exposed or released. Thus, disintegration of the tablet is the first step in drugabsorption. Once the tablet breaks up into small particles, the drug must dissolve in thegastrointestinal fluid. Once in solution, the drug now has the opportunity to pass through thegastrointestinal membrane. Drug absorption through the membrane most commonly occursby passive diffusion, driven by the concentration gradient across the membrane. Uptaketransporters in the luminal side of the enterocyte (intestinal epithelial absorptive cell) mem-brane may facilitate absorption. Once in the enterocyte membrane, the drug may be subjectto the action of efflux transporters, which will reduce absorption by secreting the drug backinto the lumen. The enterocytes of the intestine also contain some drug-metabolizing en-zymes, which may metabolize and inactivate a drug before it passes through the basolateral(blood-side) membrane. When the drugs pass through the basolateral membrane, they aretaken up into the capillaries bathing the tissue. These capillaries ultimately drain into thehepatic artery, which takes drugs through the liver before they reach the systemic circu-lation. The liver is a major organ of drug elimination, and any drugs that are metabolizedextensively by the liver may be essentially lost before reaching the systemic circulation.

The key steps involved in drug absorption are summarized in Figure 3.1 and listed below.

1. Disintegration of tablet

2. Dissolution of drug

3. Diffusion of drug across the gastrointestinal membrane

4. Active uptake of drug into the gastrointestinal membrane

5. Active efflux of drug from the gastrointestinal membrane back into the lumen

6. Metabolism of drug in the gastrointestinal membrane

7. Metabolism of drug during the first passage through the liver (first pass)

Drugs that are already either in the disperse state (suspension formulations) or in solution(solution formulations, e.g., syrups) will be farther along the absorption chain than soliddosage forms such as tablets. As a result, drugs formulated as suspensions and solutions,in particular, often display more rapid and/or more complete absorption than do theirsolid dosage formulations. The absorption of itraconazole, for example, which has poordissolution properties, is better from a solution formulation than from a tablet.

Page 61: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

EXTENT OF DRUG ABSORPTION 41

D(aq)

1.

3.

4.

5.

6.

Tablet Consumed: Dose = D

Disintegration andDissolution

MembranePenetration (Fa)

Uptake (U) and Efflux (E)Transporters

Metabolism in theEnterocyte (Fg)

MetabolismDuring First PassThrough Liver (Fh)

LIVER

UE

D

D

D

Compliance

D

D

D

M

D DD

D

D

DD

D

DDD

DD

D D

D

DD

D

D

DD

MM

D

7. Drug in systemic circulation: Effective Dose = Fa•Fg•Fh•D

2.

FIGURE 3.1 Steps involved in the oral absorption of a dose of a drug formulated as an oraltablet. After consumption the tablet must disintegrate in the gastrointestinal fluid so that the drug candissolve. Once in solution, the drug has the opportunity to pass the intestinal membrane. The fractionof the dose penetrating the membrane is Fa. In the membrane the drug may be subject to uptake andefflux transporters. In the enterocyte cell the drug may be subject to metabolism. The fraction of thedrug in the enterocyte that escapes metabolism is Fg. When the drug passes into the capillary vesselsit is taken to the liver where it may undergo metabolism. Fh is the fraction of the drug entering theliver that escapes metabolism.

3.4 EXTENT OF DRUG ABSORPTION

3.4.1 Bioavailability Factor

The extent of drug absorption is assessed by means of its bioavailability factor, which isusually referred to simply as “bioavailability,” although strictly speaking, a drug’s bioavail-ability refers to both the rate and extent of drug absorption. The bioavailability factor, Fis the fraction of the dose that reaches the systemic circulation intact. As a factor it canachieve a value between zero (none of the dose reaches the systemic circulation) and 1 (the

Page 62: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

42 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

entire dose reaches the systemic circulation). Thus, if a drug’s F value is 0.80, 80% of thedose administered reaches the circulation as intact drug. Since the body is exposed only tothe dose that is absorbed, the effective dose of a drug may be defined as the product of thebioavailability and the dose administered:

effective dose = F · dose administered

Thus, if 100 mg of a drug is administered and if F = 0.8,

effective dose = 0.8 × 100 = 80 mg

As pharmacokinetic equations are introduced throughout the book, all formulas willinclude the bioavailability factor as a potential qualifier of the dose. The factor will even beincluded in a formula specifically derived for intravenous administration, where bioavail-ability is always 1. This is because an intravenous formula may be applied to other routesof administration that may not have complete bioavailability, and the presence of the factorin the formula serves as a reminder to account for bioavailability if necessary.

The determinants of the bioavailability factor can be classified into three broad areas:

1. Physicochemical characteristics of the drug

2. Biological factors such as whether or not a drug is a substrate for the membranetransporters, and the drug-metabolizing enzymes

3. The manufacturing process and formulation of a specific dosage form. As such thiscomponent of bioavailability may vary from one brand of dosage form to another

Under normal circumstances the bioavailability factor for a given brand is a constantand, very important, does not change with dose. A dose-dependent bioavailability factor isan example of nonlinear pharmacokinetics, discussed in Chapter 15.

3.4.2 Individual Bioavailability Factors

A portion of the drug dose may be lost at several points during the absorption process.Overall, the losses are usually classified into three categories, which results in three typesof bioavailability:

1. Fraction absorbed (Fa): the fraction of the dose that is absorbed intact across theapical membrane into the cells (enterocytes) of the intestinal membrane (2)

2. Intestinal bioavailability (Fg): the fraction of the drug in the enterocytes that escapesmetabolism

3. Hepatic bioavailability (Fh): the fraction of the dose that enters the liver and escapesmetabolism during the first pass

Overall bioavailability (F) may thus be expressed

F = Fa · Fg · Fh (3.1)

Page 63: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

DETERMINANTS OF THE BIOAVAILABILITY FACTOR 43

Example 3.1 Consider a drug that has the following characteristics: 20% of a dose of adrug is destroyed by acid in the stomach, the remaining drug is able to penetrate the apicalmembrane completely, 10% of the drug passing though the membrane is metabolized, and40% of the drug entering the liver is metabolized. Find the effective dose when 100 mg isadministered.

Solution

F = Fa · Fg · Fh

= 0.8 × 0.9 × 0.6 = 0.432

If 100 mg was administered, the effective dose would be F · D = 43.2 mg.

This categorization is useful because it separates the physicochemical factors that controlFa from the biological factors that control Fg and Fh. It is useful to distinguish Fg from Fh

because they are influenced by different factors. For example, Fh may be altered by changesin hepatic blood flow, which would have no effect on Fg.

3.5 DETERMINANTS OF THE BIOAVAILABILITY FACTOR

3.5.1 Disintegration

The disintegration of the tablet into small particles suitable for drug dissolution is anessential step in the absorption process. If disintegration does not occur, or if it is very slow,the absorption of the drug will be compromised. Most tablets contain special ingredientsor excipients called disintigrants (e.g., starch), which swell when wet by the gastric fluid.As a result, tablet disintegration is usually a straightforward, unproblematic step in theabsorption process.

3.5.2 Dissolution

The dissolution characteristics are determined primarily by a drug’s aqueous solubility,which in turn is determined primarily by its hydrophilicity or lipophilicity, with morehydrophilic drugs dissolving in the aqueous gastrointestinal fluid more readily than dolipophilic drugs. Other factors that determine dissolution include the crystalline form of thedrug and the particle size of the powdered drug. For example, the dissolution properties ofpoorly soluble drugs can be improved by using a process known as micronization to greatlyreduce the particle size. This method is used to increase the dissolution of progesterone.Most drugs are either weak acids or weak bases and are often formulated as salts, to improvetheir dissolution properties. As a result of improved dissolution, the rate and extent ofabsorption of drugs can be increased. The specific salt form can be selected to optimizethese qualities. Examples of salts include propranolol hydrochloride, phenytoin sodium,naproxen sodium, and aminophylline, which is the ethylenediamine salt of theophylline.

3.5.3 Formulation Excipients

Dosage forms contain many other ingredients in addition to active drug. These ingredients,or excipients, are pharmacologically inert and are added to assist in the manufacture ofthe dosage form or to impart certain properties to the finished product. For example, many

Page 64: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

44 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

powder-based dosage forms (tablets and capsules) contain lubricants to improve the flowproperties of the powers so that they can be poured and transferred more easily duringmanufacture. Most solid dosage forms also contain diluents to increase the size of theunit dosage form, to make it a more manageable size. The importance of assessing thepotential influence of inert excipients on bioavailability was demonstrated very poignantlyin 1968 when over 50 Australian patients developed serious phenytoin toxicity after themanufacturer simply changed the diluent in the formulation from calcium sulfate to themore hydrophilic lactose. Other excipients that may be added to improve the propertiesof the final dosage form include wetting agents, dissolution enhancers, and substances topromote membrane permeation.

3.5.4 Adverse Events Within the Gastrointestinal Lumen

The gastrointestinal lumen is an extremely harsh environment. It contains enzymes, foods invarious stages of digestion, and displays fairly wide changes in pH. This hostile environmentcan be beneficial in that it can destroy potentially harmful bacteria but can also be verydetrimental for drugs.

pH The pH along the gastrointestinal tract varies from around 1.5 to 7. The gastric pH canapproach about 1.5 in the presence of food as a result of gastric acid secretion stimulatedby gastrin and histamine. In the fasting state, the pH generally rests at around a pH of 2 to6. The high acidity of the stomach can be particularly problematic for drugs. The early ornatural penicillins are very susceptible to acid hydrolysis, particularly penicillin G, which asa consequence is best administered parenterally. Oral preparations of penicillin V should beadministered on an empty stomach (1 to 2 h before food) to minimize hydrolysis. The newersemisynthetic penicillins such as amoxicillin are much less susceptible to hydrolysis andprovide better and more consistent oral bioavailability. Drugs susceptible to acid hydrolysisin the stomach can be formulated into enteric-coated tablets. The coating protects the drugfrom the acid in the stomach, since it will dissolve only in the higher pH of the smallintestine, which generally lies in the region of 6 to 7. In another approach to protectingacid-labile drugs, antacids can be included in their formulation. This is used for the acid-labile reverse transcriptase inhibitor didanosine. The increase in the gastric pH broughtabout by treatment with proton pump inhibitors, H2 antagonists, and antacids can affect thedissolution and bioavailability of some drugs. The absorption of drugs such as delavirdineand the poorly soluble antifungal agents itroconazole and ketoconazole, which displaymuch better dissolution in acidic pH, has been found to be reduced by low gastric acidity.The absorption of other drugs, such as alendronate has been found to be increased (3). Theless acidic environment created by proton pump inhibitors may also reduce the absorptionof calcium, which can make patients who take these drugs for extended periods moresusceptible to bone fractures and osteoporosis (4).

Enzymatic Attack Digestive enzymes such as pepsin, trypsin, and chymotrypsin in thegastrointestinal fluid are the main enzymes that affect drug absorption. These enzymes pre-vent protein drugs such as insulin from being given orally. Drugs can also be affected by theenzymes of the microflora, which are found primarily in the large intestine. Since most drugsare absorbed before they reach the large intestine, these enzymes generally do not affect thefraction of the dose absorbed initially. They can be important for drugs that are conjugatedwith glucuronide and subsequently excreted in the bile and then into the intestine. The

Page 65: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

DETERMINANTS OF THE BIOAVAILABILITY FACTOR 45

Pla

sma

Con

cent

rati

on

Time

Secondary peakwhen drug is reabsorbed

(a) (b)

BiliaryExcretion

Liver

D-GD-G

D + GD

IntestinalLumnen

IntestinalFlora

FIGURE 3.2 Diagrammatic representation of enterohepatic recirculation (a). The glucuronideconjugate of the drug is hydrolyzed by the gut flora. The reabsorption of the drug results in a secondpeak on the plasma concentration–time curve (b).

gut flora can hydrolyze the conjugates and release the drug, which can then be reabsorbedback into the body. This process, known as enterohepatic recirculation (Figure 3.2), oftenresults in the appearance of a secondary peak in the plasma concentration–time profile of adrug (Figure 3.2). The process results in the reabsorption of drug that has been eliminatedfrom the body. Thus, in theory, enteroheaptic recirculation could result in a drug havinga bioavailability factor greater than 1. The concurrent administration of broad-spectrumantibiotics such as the quinolones can reduce the population of the gut flora, which in turncan lead to reduced enterohepatic recirculation and a reduction in the body’s exposure to adrug. The enterohepatic recirculation of ethinyl estradiol, a component in oral contracep-tives, can be reduced by concurrent antibiotics, which could compromise the effectivenessof the drug. Generally, this is thought to be a problem only in patients who are alreadypredisposed to low bioavailability of ethinyl estradiol, or when the antibiotic rifampin isused. In addition to reducing the gut flora, this compound induces liver enzymes and canincrease the metabolism of estradiol to further reduce the body’s exposure to estradiol.

Interaction with Other Components of the Gastrointestinal Fluid Drugs may interactadversely with other components of the gastric fluid, such as digested and undigestedfoods and other drugs. The classical example of this type of interaction is the complexationreaction between tetracycline and di- and trivalent ions (Ca2+, Mg2+, Al3+) found indairy products and antacids. The tetracycline complex is insoluble and the drug cannotbe absorbed. Similar complexes are formed with the quinolone antibiotics. Antacids canreduce the absorption of so many other drugs, including phenytoin, several �-blockers,isoniazid, and digoxin, that as a general rule, patients should be advised to stagger thedosing of antacids and other prescription drugs by a period of at least 2 h.

Page 66: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

46 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

Food Effects Food can affect drug absorption through effects on gastrointestinal physiol-ogy and through physical effects on drugs in the lumen. The magnitude and direction of afood effect depends on the specific characteristics of the meal, including the compositionof the meal (proteins, carbohydrates, or fats), calorie content, and the total volume of foodand fluid ingested. Physiological changes brought about by food include a delay in gastricemptying; changes in gastrointestinal pH, stimulation of bile flow, and increased liver bloodflow, all of which can affect drug absorption. Absorption can also be affected by the phys-ical presence of food in the gastrointestinal lumen. Food can impede drug diffusion to themembrane and reduce absorption; and constituents in food, such as ions, can chelate drugsand reduce absorption. Increased gastric residence time may increase dissolution in gastricfluid but can also increase the destruction of acid-labile drugs. As a general rule, the delayin gastric emptying brought about by food delays the absorption of most drugs because itslows their delivery to the small intestine, which is where most drugs are absorbed. Theeffect of food on the extent of absorption is more variable. A classification system in whichdrugs are categorized according to their permeability and dissolution characteristics hasbeen used to help predict whether food will increase, decrease, or not affect the absorptionof individual drugs (see Section 3.7).

3.5.5 Transcellular Passive Diffusion

Most drugs penetrate the gastrointestinal membrane by transcellular passive diffusion,driven by the concentration gradient across the membrane. As discussed in Chapter 2, thesmall intestine (duodenum, jejunum, and ileum) has an extremely large absorptive area,which results in it being the primary site for drug absorption. The small intestine is alsohighly perfused with blood. This allows absorbed drug to be rapidly carried away fromthe absorption site, and enables a high concentration gradient to be maintained acrossthe intestinal membrane. Because of the more extensive absorption of the drug from theintestine, the rate of stomach emptying plays an important role in controlling the speedwith which drugs are absorbed. Slow stomach emptying delays the delivery of drug to theprimary site of absorption and slows down absorption, whereas rapid stomach emptyingcan speed up drug absorption.

A drug’s transcellular permeability is determined primarily by its lipophilicity, whichas discussed in Chapter 2, can be quantified using the drug’s partition coefficient (P) ordistribution coefficient (D). Drugs with negative log P or log D values, indicative of highhydrophilicity and poor lipophilicity, cannot easily penetrate the lipid matrix of a membrane.Conversely, drugs with high log P or log D values generally have good permeability. Astudy of almost 500 therapeutic drugs found that 50% had log P � 2 and 80% had log P � 0(2). However, compounds with high log P or log D values are also most likely to have poordissolution in aqueous media (5). As a result, values of log D7.4 in the range of about 1 to 3 areconsidered optimum for balancing permeability and dissolution. The size of a drug moleculeis also important. Large molecules such as peptides and proteins are unable to penetratethe intestinal membrane. Drugs with a molecular mass greater than 500 Da are predictedto have poor membrane permeability, particularly if they also possess additional adversephysicochemical characteristics, such as polarity or low lipophilicity (6). Studies indicatethat for drugs with molecular masses below around 400 Da, size is not an important factorfor membrane penetration (7). Under these circumstances, a drug’s log P or log D value ata physiological pH, combined with a measure of the drug’s polarity (polar surface area ornumber of hydrogen bond donors), can be used to predict transcellular passive diffusion.

Page 67: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

DETERMINANTS OF THE BIOAVAILABILITY FACTOR 47

3.5.6 Paracellular Passive Diffusion

The junctions between the endothelial cells of the gastrointestinal membrane are very tight,which makes it extremely difficult for all but very small drugs to penetrate. Furthermore,the gaps between adjacent cells in the small intestine constitute only a very small fraction(0.01%) of the total absorptive area, which makes it a very unappealing and inefficient routeof drug penetration (2). Small polar drugs such as atenolol (MW 266 Da; log D7.4 ≈ −1.5)were believed to be absorbed by this route (5). However, recent work has cast doubt on thisand suggests that the enormous absorptive area available for transcellular absorption mayenable small hydrophilic drugs such as atenolol to undergo some transcellular absorption(2). The aminoglycoside antibiotics, which are both very polar (log D7.4 ≈ −10) andvery large (MW 450 to �1000 Da), are unable to penetrate the intestinal membrane byeither route. These drugs must be given parenterally and are usually administered as short,intermittent intravenous infusions for the treatment of systemic infections.

The use of excipients to increase the size of the tight junctions in the intestinal lumenis being investigated as a means of opening up the paracellular route to large and/or polardrugs, which normally cannot penetrate the membrane. However, this would also allowother compounds, normally excluded from the body, to be absorbed, and concern has beenexpressed about the possible health risks associated with this approach to improving drugabsorption.

3.5.7 Uptake and Efflux Transporters

Drug transporters within the enterocyte membrane can also play an important role in theabsorption process. Over 20 individual transporters have been identified on the apical andbasolateral sides of the cell membrane. The clinical role of many of these has yet to beestablished, and this discussion will generally be limited to uptake and efflux transporters,whose role in drug absorption has been demonstrated in clinical studies. All these wereintroduced in Chapter 2 (Section 2.4). The main transporters involved in drug absorptionfrom the gastrointestinal tract are OATPs, PEPT1, P-gp, BCRP, and MRP2 (Figure 3.3)and they are all located on the apical side of the membrane. Thus, the uptake transporters(OATPs, PEP1) enhance drug absorption, and the efflux transporters (P-gp, BCRP, MRP2)impede absorption. The efflux transporter MRP3, which is located on the basolateral sideof the membrane (Figure 3.3), would in theory serve to enhance the absorption of itssubstrates, but at this time, clinical evidence for its role in drug absorption is lacking.

3.5.7.1 Uptake TransportersOATP1A2 and OATP2B1 are present on the apical side of the intestinal membrane, andthere appears to be some overlap in their substrate specificity. Fexofenadine and saquinavirare transported by OATP1A2, and the concomitant administration of fruit (apple, grapefruit,or orange) juices, which inhibit OATP1A2, decreases the absorption of fexofenadine bybetween 30 and 50% (8,9). Atorvastatin and other HMG-CoA reductase inhibitors (statins)are substrates for OAP2B1, but its role, if any, in the absorption of these drugs is not knownat this time. The PEPT1 transporter plays an important role in the absorption of digestedproteins (dipeptides and tripeptides). However, it can also transport drugs that possess di-and tripeptide-like structures such as cephalosporins, penicllins, and angiotensin-convertingenzyme inhibitors such as captopril (10). Valacylovir, the l-valine ester prodrug of acyclovir,was developed specifically to take advantage of the PEPT1 carrier. The absorption of the

Page 68: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

48 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

Intestinal Lumen

Apical Side

Basolateral Side

IntestinalMembrane

Capillary

P-gpOATP1A2OATP2B1

E E UU E

E

PEPT1MRP2

MRP3

BCRP

FIGURE 3.3 Main transporters in the human intestine involved in drug absorption. Uptaketransporters (U) include organic anion transporting polypeptides (OATPs) and peptide trans-porters (PEPTs). Efflux transporters include permeability glycoprotein (P-gp), multidrug resistance–associated protein (MRP), and breast cancer resistance protein (BCRP). (Diagram drawn by LinneaAnderson.)

valacyclovir is about double that of acyclovir (11). Some of the substrates of PEPT1 (e.g.,the cephalosporins) also act as inhibitors of the transporter. Generally, information on therole of uptake transporters in drug absorption in humans is still rather limited.

3.5.7.2 Efflux TransportersThe efflux transporters on the apical side of the intestinal membrane serve to reduce theabsorption of drugs by pumping substances that have penetrated the cell membrane backinto the lumen (Figure 3.3). The greatest amount of clinical evidence is available on therole of P-gp, which is able to transport a wide variety of drugs with diverse chemicalstructures. Clinical studies have demonstrated that P-gp plays an important role in limitingthe oral absorption of many drugs, including cyclosporine, tacrolimus, paclitaxel, irinotecan,digoxin, and talinolol (10,12,13).

The levels of P-gp vary throughout the intestine and are present in greater amounts inthe distal portion (ileum) than in the proximal portion (duodenum and jejunum) of thesmall intestine. A drug’s exposure to the higher P-gp concentration in the distal portion ofthe small intestine will depend on how rapidly it is absorbed. Highly soluble, lipophilicP-gp substrates such as diltiazem and verapamil are absorbed rapidly in the upper smallintestine and will have less exposure to P-gp efflux. Additionally, highly soluble, lipophilicP-gp substrates may be absorbed so rapidly that they saturate P-gp during absorption andlimit its negative effects on bioavailability. The higher concentration of P-gp in the distalsmall intestine has been postulated to explain why the extent of absorption of talinolol islower from slow-release than from immediate-release preparations: The greater exposureof the drug to the distal small intestine in the slow-release preparation leads to greater P-gp

Page 69: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

DETERMINANTS OF THE BIOAVAILABILITY FACTOR 49

TABLE 3.1 Examples of Clinically Important Changes in Bioavailability Resulting fromAltered P-gp Activity

Drug Concomitant Drug Outcome Mechanism Proposed

Digoxin Quinidine Increased F Inhibition of P-gpSt. John’s wort Decreased F Induction of P-gp

Cyclosporine Rifampin Decreased F Induction of P-gpTacrolimus Rifampin Decreased F Induction of P-gpPaclitaxel Cyclosporine Increased F Inhibition of P-gp

Elacridar Increased F Inhibition of P-gpDocetaxel Cyclosporine Increased F Inhibition of P-gpTopotecan Elacridar Increased F Inhibition of P-gp, inhibition of BCRP

Source: Refs. (10,13).

efflux and less absorption (12). The greater bioavailability of cyclosporine from the Neoralformulation compared to Sandimmune is also believed to result, in part, from less P-gpefflux. Cyclosporine is absorbed faster from Neoral, which results in less drug reachingthe distal intestine, where the concentration of P-gp is greatest (12). Table 3.1 lists someexamples of modifiers of P-gp activity that have been found to cause clinically importantchanges in bioavailability.

It is difficult to quantify definitively the effect of P-gp on drug absorption because thereis extensive overlap of its substrates, inhibitors, and inducers with those of cytochromeP450 (CYP3A4), which can also reduce drug absorption. Thus, metabolism by intestinalCYP3A4 will also play a role in limiting the absorption of many of the drugs listed in Ta-ble 3.1. Indeed, the wide overlap in the substrate specificity of these two systems suggeststhat they may have evolved to work together to provide a concerted effort to limit the absorp-tion of foreign chemicals (xenobiotics) into the body. Talinolol, digoxin, and fexofenadinehave been used in clinical studies as probes for P-gp because these drugs do not undergosubstantial metabolism by CYP3A4. Intestinal P-gp reduces the bioavailability of digoxin,but digoxin is also a substrate for renal P-gp, which, like intestinal P-gp, reduces the body’sexposure to digoxin, in this case by promoting elimination. Reduced P-gp activity at eithersite would increase the body’s exposure to digoxin (increased absorption and/or decreasedelimination), and increased P-gp activity would have the opposite effect. Rifampin, an in-ducer of P-gp activity, was indeed found to decrease the systemic concentrations of digoxin(14). Three observations indicated that the effects on intestinal, as opposed to renal, P-gpappeared to dominate. First, digoxin’s renal clearance and half-life were not altered byrifampin. Second, rifampin had much less effect on the pharmacokinetics of intravenousdigoxin. Third, the increase in intestinal P-gp content mediated by rifampin correlated withthe increase in digoxin concentration after oral administration. In other studies, the relativecontribution of altered renal and/or intestinal P-gp on digoxin’s pharmacokinetics is notclear. From a clinical standpoint, modifiers of P-gp should be used cautiously with digoxin.Irrespective of the mechanism, inhibitors and inducers may increase and decrease, respec-tively, the body’s exposure to digoxin. Similarly, modifiers of P-gp and CYP3A4 should beused cautiously with the compounds listed Table 3.1. Irrespective of the mechanism (P-gpor CYP3A4), inhibitors will tend to increase absorption and inducers to reduce absorption.

Some of the changes in bioavailability brought about by modifiers of P-gp activityare small. For example, pretreatment with St. John’s wort or rifampin, both of whichinduce P-gp, reduced the bioavailability of talinolol by 25 and 35%, respectively (15,16).

Page 70: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

50 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

In other cases, modifiers can bring about large changes in bioavailability. For example,cyclosporine, an inhibitor of P-gp, increased the bioavailability of the anticancer drugspaclitaxel and docetaxel from a negligible value (0.04 and 0.08, respectively) to a substantialvalue (0.47 and 0.88, respectively) (10). Clinically, the coadministration of P-gp inhibitorswith anticancer drugs not only offers the advantage of improving oral bioavailability butcould also improve their effectiveness by preventing the efflux of drugs from cancer cellsand/or the central nervous system. The use of cyclosporine as an inhibitor P-gp is limitedby its immunosuppressant activity, and newer P-gp inhibitors have been developed that aredevoid of this activity. Second-generation inhibitors such as valspodar cause unacceptableside effects because they inhibit the elimination of some drugs. Third-generation P-gpinhibitors such as zosuquidar and elacridar are potent P-gp inhibitors but do not inhibithepatic enzymes. They are being tested clinically as a means of increasing the effectivenessand bioavailability of anticancer drugs (17). Elacridar, an inhibitor of both P-gp and BCRP,increased the bioavailability of topotecan from 0.4 to 0.97 (10).

Other efflux transporters that have been shown to have clinically important effects onabsorption include BCRP and MRP2. BCRP is one of the most abundant efflux transportersin the human intestinal lumen, particularly the jejunum. Animal studies have demonstratedthat BCRP reduces the absorption of a number of anticancer drugs, including topotecanand irinotecan. Clinical studies in people with a genetically determined low activity levelof BCRP were found to have markedly higher plasma concentrations of atorvastatin androsuvastatin (18). These patients had higher peak plasma concentrations but not alteredelimination half-lives, which suggests that BCRP affects the absorption but not the elimi-nation of these drugs. Sulfasalazine, a drug used in the treatment of ulcerative colitis andCrohn’s disease, is a substrate for both BCRP and MRP2. Despite good inherent membranepermeability characteristics (log P = 3.88), only a small fraction of the dose is absorbedafter oral administration. It is thought that BCRP and/or MRP2 (19,20) reduce its absorp-tion and enable a large portion of the dose to reach its site of action in the colon, wherebacteria cleave to its active metabolites, sulfapyridine and mesalamine. Indomethacin, aninhibitor of MRP2, was found to increase the absorption of sulfasalazine (21). Thus, theconcomitant consumption of indomethacin with sulfasalazine may reduce the effectivenessof sulfasalazine.

3.5.8 Presytemic Intestinal Metabolism or Extraction

The enterocytes of the small intestine contain several enzymes that can metabolize drugsthat get inside the cell. These include cytochrome P450 (CYP) isozymes, glucuronosyl-transferases, and alcohol dehydrogenase. For many drugs, intestinal extraction is the leastimportant component of overall bioavailability. However, for some drugs, the fraction ofa dose lost at this point in absorption is substantial and results in a large loss of dose.Extensive intestinal extraction makes the subject drugs susceptible to clinically importantdrug–drug interactions. CYP3A4 and CYP2C9 are the most abundant phase I enzymes inthe enterocytes, comprising 80% and 15% of total intestinal CYP, respectively. The highconcentration of CYP3A4 and the large number of drugs metabolized by this isozymecombine to make CYP3A4 metabolism the most important intestinal extraction process.In contrast to P-gp, the levels of CYP3A4 are higher in the upper part of the small intes-tine (duodenum) and decrease progressively to the distal ileum. There appears to be wideinterindividual variability in its expression, which will result in wide interindividual vari-ability in the bioavailability of drugs that undergo significant intestinal metabolism (22).

Page 71: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

DETERMINANTS OF THE BIOAVAILABILITY FACTOR 51

TABLE 3.2 Estimated Values of Overall (F), Intestinal (Fg), andHepatic (Fh) Bioavailability for Several CYP3A4 Substrates

CYP3A4 Substrate F Fg Fh

Tacrolimus 0.14 0.14 0.96Buspirone N.A.a 0.21 0.24Atorvastain 0.14 0.24 0.58Cyclosporine 0.22–0.36 0.33–0.48 0.75–0.88Felodipine 0.14 0.45 0.34Midazolam 0.25–0.41 0.40–0.79 0.49–0.74Simvastatin N.A.a 0.66 0.67Triazolam 0.55 0.75 0.75Nifedipine 0.41 0.78 0.53Quinidine 0.78 0.90 0.86Alprozolam 0.84 0.94 0.97

Source: Ref. (24).aN.A., not available.

Despite its low concentration relative to the liver (about 1%) (23), intestinal CYP3A4 canmetabolize a very substantial part of an oral dose of its substrates. Table 3.2 shows thevalues of Fg and Fh calculated for a number of CYP3A4 substrates (24). It can be seen thatfor some drugs, such as tacrolimus, buspirone, atorvastatin, and cyclosporine, intestinalextraction is extensive and is estimated to remove 50% or more of an oral dose.

Many of the drugs that are substrates for intestinal CYP3A4 are also substrates for P-gp,and as mentioned previously, the two processes may have evolved to work synergisticallywithin the enterocyte to prevent the absorption of drugs and other xenobiotics. The effluxof a drug by P-gp followed by its reabsoprtion creates a recycling effect that can prolongthe overall time that a drug spends in the enterocyte and provide CYP3A4 with moreopportunity to metabolize it. This may explain, in part, why despite its relatively lowconcentration compared to the liver, intestinal CYP3A4 is able to metabolize such a largefraction of the dose of some drugs. There are drugs, however, such as midazolam, felodipine,and nifedipine, that are CYP3A4 substrates but do not appear to be substrates for intestinalP-gp, whereas others, including talinolol, digoxin, and fexofenadine, are substrates for P-gpbut not CYP3A4 (17).

Clinical studies have demonstrated that inhibitors of CYP3A4, such as ketoconazole,itroconazole, the macrolide antibiotics, and cyclosporine, increase the bioavailability ofmany CYP3A4 substrates. There is wide interindividual variability in the magnitude ofthe effect of inhibitors, presumably because of interpatient variability in the expressionof intestinal CYP3A4. Drugs that undergo significant metabolism will be particularlysusceptible to clinically important interactions. For example, in theory, if the intestinalextraction of tacrolimus was completely inhibited, its bioavailability would increase fromaround 0.14 to almost 1, which translates into a sevenfold (1/0.14) increase in the effectivedose. Large increases (in some cases over 10-fold) in the bioavailability of buspirone,tacrolimus, sirolimus, lovastatin, and simvastatin have been observed in the presence ofinhibitors. Reduced P-gp activity as well as reduced hepatic CYP3A4 activity could alsocontribute to these effects.

Grapefruit juice is an irreversible inhibitor of CYP3A4, and evidence suggests that itaffects primarily intestinal rather than hepatic CYP3A4, although consumption of large

Page 72: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

52 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

quantities may also inhibit hepatic CYP3A4. There is no strong evidence that it inhibitsP-gp (9), although, along with other fruit juices, it inhibits OATP1A2. Thus, concomitantadministration of drugs with grapefruit juice may provide a good indication of the role ofintestinal CYP3A4 in drug absorption. Grapefruit juice increases the oral bioavailability ofmany drugs, including simvastatin, lovastatin, atorvastatin, felodipine, cyclosporine, andbuspirone. The effect appears to be concentration dependent, as double-strength juice hasa greater effect than regular-strength juice. Large increases of more than 10-fold have beenobserved in the plasma concentrations of simvastatin, lovastatin, and buspirone (25–27).

3.5.9 Presystemic Hepatic Metabolism or Extraction

After drugs pass through the basolateral side of the intestinal membrane, they are takenup into the mesenteric vessels surrounding the small intestine. These vessels drain into thehepatic portal system, which then takes them directly to the liver. The liver is a major organfor drug elimination and contains the full complement of the drug-metabolizing enzymes(see Chapter 5), which are present in a higher concentration than anywhere else in the body.As a result, a portion of an oral dose can be lost before it reaches systemic circulation duringthis first pass through the liver. The extent of presystemic hepatic extraction (the first-passeffect) depends on the ability of the liver enzymes to metabolize a specific drug. This isexpressed using the drug’s hepatic extraction ratio (E), which is defined as the fraction ofthe incoming drug that is metabolized during a single pass through the liver. As a fraction,E can achieve values between 0 and 1, and under normal circumstances it is a constantfor a particular drug. If the liver enzymes have a large capacity to metabolize a particulardrug, its extraction ratio will be large, and a large fraction of the incoming drug will belost during its first pass through the liver. Recall that a drug’s hepatic bioavailability (Fh)is defined as the fraction of the dose enetering the liver that escapes extraction. Thus, Fh

is equal to 1 − E and a high level of extraction will be associated with a small degree ofhepatic bioavailability. For example, when

E = 0.8, 80% of the drug entering the liver is eliminated by metabolismduring its first pass

Fh is the fraction of the drug entering the liver that escapes metabolism:

Fh = 1 − E = 1 − 0.8 = 0.2 20% of the drug entering the liver escapes metabolismand reaches the systemic circulation

Poor oral bioavailability due to extensive first-pass extraction may preclude the oral routeof administration for some drugs. Examples of drugs that experience extensive first-passextraction and cannot be given orally include lidocaine, nitroglycerin, and naloxone. Alter-native routes for these three drugs include the parenteral, buccal, intranasal, and transdermalroutes. Interestingly, an oral preparation containing the opiate antagonist naloxone is avail-able. It is present in combination with the opioid, pentazocine in Talwin tablets. The tabletswere developed specifically to prevent the misuse of pentazocine, particularly its combi-nation with the antihistamine tripelennamine (no longer available in the United States)to produce a heroin substitute. When Talwin tablets are taken orally, naloxone is inactivebecause of extensive first-pass extraction in the liver. However, if attempts are made toadminister the preparation intravenously or subcutaeously, naloxone will be completely

Page 73: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

FACTORS CONTROLLING THE RATE OF DRUG ABSORPTION 53

TABLE 3.3 Effect of a 10% Reduction in Hepatic Extraction on the Effective Oral Dose ofHigh- and Low-Extraction Drugsa

Control 10% Reduction in E

Drug 1 Drug 2 Drug 1 Drug 2

E 0.1 0.9 0.09 0.81Fh 0.9 0.1 0.91 0.19Effective dose from a 100-mg dose 90 mg 10 mg 91 mg 19 mgChange in effective dose — — 1.1% 90%

aE is the hepatic extraction ratio and Fh is the hepatic bioavailability (1 − E). It is assumed that the entire dosereaches the liver.

bioavailable and will block the action of pentazocine. Despite extensive first-passmetabolism, many other drugs, including propranolol, meperidine, and verapamil, are stilladministered orally. Oral doses of these drugs greatly exceed the value of intravenous doses.

Altered activity of the liver enzymes as a result of concomitant medication, hepatic dis-ease, and/or age can bring about clinically important changes in a drug’s hepatic bioavail-ability. These effects are particularly important for drugs that experience high hepaticextraction and have very low hepatic bioavailability. Table 3.3 shows how a 10% changein the fraction of the dose extracted by the liver could affect bioavailability. It can be seenthat although the effect on drugs with a low extraction ratio is minimal, a 10% reduction inthe extraction of a highly extracted drug can essentially double the bioavailable dose. Thefactors that influence hepatic metabolism and first-pass extraction are discussed in moredetail in Chapter 5.

3.6 FACTORS CONTROLLING THE RATE OF DRUG ABSORPTION

Drug absorption involves several steps, including tablet disintegration, drug dissolution,and membrane penetration. The overall rate of drug absorption is controlled by the sloweststep in the process, usually either dissolution or membrane penetration. As discussed previ-ously, owing to its large surface area and extremely large blood supply, the small intestine isthe primary site for drug absorption. When dissolution is rapid and absorption is controlledby membrane penetration, stomach emptying can exert an important influence on the rateof drug absorption. Stomach emptying time varies widely. Food is a particularly importantfactor. The consumption of food, particularly high-fat meals, slows stomach emptying toallow digestion to occur and generally delays drug absorption. Opiates and anticholiner-gic drugs such as propanthiline also delay stomach emptying. In contrast, in the fastingstate, when there is less need to hold drug in the stomach, emptying time is much shorter.Metoclopramide increases gastrointestinal motility and speeds up stomach emptying. Therelationship between stomach emptying and the rate of drug absorption is particularlystrong for drugs that have a rapid dissolution rate and permeability-controlled absorption.

The absorption of these drugs, such as acetaminophen, can be used to assess stomach-emptying time. Figure 3.4 shows how the absorption of acetaminophen was influencedby the coadministration of either metoclopramide or propantheline. Propantheline, whichslows stomach emptying, slowed the absorption of acetaminophen. The size and time ofthe peak plasma concentration were smaller and longer, respectively. In contrast, when

Page 74: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

54 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

Pla

sma

Con

cent

rati

on (

mg/

L)

Time (h)

0

5

10

15

50

with metoclopramideacetaminophen alonewith propantheline

FIGURE 3.4 Use of acetaminophen to assess the rate of drug absorption. Acetaminophen(1500 mg) was administered alone and concurrently with propantheline or metoclopramide in a22-year-old man. [Redrawn with permission from ref. (28).]

coadministered with metoclopramide, acetaminophen’s absorption was faster: The peakplasma concentration was larger and occurred earlier than in the absence of metoclo-pramide. The determinants of dissolution and permeability controlled absorption will nowbe presented.

3.6.1 Dissolution-Controlled Absorption

The rate of dissolution of a drug is described by the Noyes–Whitney equation. Accordingly,the rate may be expressed

dC

dt= D · S

V · h· (Cs − C) (3.2)

where C is the concentration of the drug in the gastrointestinal fluid, t the time, S the surfacearea of the solid undergoing dissolution, h the thickness of a diffusion layer surroundingthe solid, D the diffusion coefficient of the drug, V the volume of the gastrointestinal fluid,Cs the solubility of the drug in the gastrointestinal fluid, and C the concentration of drug inthe bulk of the fluid.

When absorption is dissolution controlled, any dissolved drug is rapidly absorbed andremoved from the intestinal fluid. As a result, C will be much, much less than Cs. Thus,the equation reduces to

dC

dt= D · S

V · h· Cs (3.3)

As dissolution proceeds, the surface area of the solid will decrease and the dissolution willdecrease proportionally. Under these circumstances, dissolution approximates a first-orderprocess.

Page 75: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

BIOPHARMACEUTICS CLASSIFICATION SYSTEM 55

3.6.2 Membrane Penetration–Controlled Absorption

Since passive diffusion is the most common method of membrane penetration, the rate ofpenetration-controlled absorption can be approximated by Fick’s law of diffusion [equation(2.2)]. Diffusion is driven by the concentration gradient across the intestinal membrane.However, as discussed in Chapter 2, because the drug absorbed is rapidly diluted into avery large volume, diffusion approximates a first-order process driven by the concentrationof drug in the intestinal fluid [equation (2.3)].

3.6.3 Overall Rate of Drug Absorption

As discussed above, both dissolution and membrane penetration approximate first-orderprocesses. As a result, the absorption of orally administered drugs is often assumed tofollow first-order kinetics. The rate of drug absorption is proportional to the amount of drugin the gastrointestinal tract:

d Ab

dt= AGI · ka (3.4)

where Ab is the amount of drug in the body, AGI the amount of drug in the gastrointestinaltract, ka the first-order rate constant for absorption, and t the time.

This simplistic approach to a complex process that involves several steps, each of whichcan be influenced by a variety of factors, may not hold on all occasions. The rate of absorp-tion of some drugs may require more complex approaches, which could include absorptionlag times, zero-order absorption, and multiple concurrent first- and/or zero-order processes.

3.7 BIOPHARMACEUTICS CLASSIFICATION SYSTEM

In view of the importance of dissolution and gastrointestinal permeability in controlling therate and extent of drug absorption, the Biopharmaceutics Classification System (BCS) hasbeen developed, in which drugs are placed into one of four groups, depending on whetherthey possess high or low solubility and permeability (29) (Table 3.4). Drugs are definedas highly soluble if the highest dose strength is soluble in 250 mL or less of an aqueousmedium over the pH range 1 to 7.5 at 37◦C. Drugs are defined as highly permeable if theextent of absorption (parent drug plus metabolites) is greater than or equal to 90%.

Drugs in class I (high solubility and high permeability) are absorbed rapidly and ex-tensively unless they are subject to presystemic extraction, and the rate of absorption iscontrolled by dissolution, or stomach emptying if dissolution is very fast. In the case ofclass II drugs (low solubility and high permeability), dissolution is the rate-controlling step

TABLE 3.4 Biopharmaceutics Classification System

Class Solubility Permeability Examples

I High High Acetaminophen, desimipramine, fluoxetineII Low High Digoxin, ibuprofen, naproxen, warfarin

III High Low Atenolol, cimetidine, nadolol, penicillinsIV Low Low Amphotericin B, chorothiazine, neomycin

Source: Examples in the table are drawn from the literature (30).

Page 76: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

56 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

TABLE 3.5 Use of the BCS and BDDCS Classification Systems toPredict the Effect of Food on Drug Absorption

Class Solubility PermeabilityEffect of Permeability on

Extent of Absorption

I High High ↔II Low High ↑

III High Low ↓IV Low Low ?

in absorption. In contrast, the rate absorption of class III drugs (high solubility and low per-meability) is controlled by membrane permeability. The poor solubility and low membranepermeability of class IV drugs makes oral administration problematic. The classificationsystem has been used by the U.S. Food and Drug Administration to simplify the assessmentof the bioavailability of immediate-release oral preparations In the case of class I drugs(highly permeable and highly soluble), if the product can exhibit rapid dissolution, it isassumed that the absorption process should be straightforward, and the requirements forhuman trials to demonstrate the equivalence of their bioavailability (bioequivalence) withother products may be waived.

Benet and his colleagues noted that most the drugs in classes I and II (highly permeable)are eliminated primarily by metabolism, and that drugs in classes III and IV (low perme-ability) are almost all eliminated by the renal or biliary excretion of unchanged drug. Theyproposed a modification of the BCS in which the route of elimination is substituted for per-meability (30). In this system, which is known as the Biopharmaceutics Drug DispositionClassification System (BDDCS), if a drug is ≥ 90% eliminated in the form of metabolitesin the urine or feces, it is classified as being highly permeable, because in order to bemetabolized, it must have been absorbed through the intestinal membrane. The BDDCSsystem has the advantage of making it easier to classify drugs because assessment of theextent of metabolism is much more straightforward than the assessment of permeability,and it is evaluated routinely during drug development. The BDDCS system has also beenused to make predictions about a drug’s distribution and elimination in vivo.

These classifications have also been used to predict the potential effect of food (high-fat meals) on the extent of drug absorption (30) (Table 3.5). Accordingly, the extent ofabsorption of class I drugs is predicted to be least affected by food because they dissolverapidly and their high permeability allows them to be absorbed quickly. However, since thepresence of food delays stomach emptying, the absorption of these drugs may be delayedby food. The extent of absorption of class II drugs (low solubility and high permeability)is predicted to increase in the presence of high-fat meals as a result of increased solubilitydue to increased bile flow. Drugs in class III dissolve easily but have poor membranepermeability, and the presence of food is only predicted to impede membrane permeabilityfurther and reduce absorption of these drugs. The effect of food on the absorption of classIV drugs is more difficult to predict. As mentioned previously, the delay in gastric emptyingbrought about by food generally delays the absorption of most drugs.

PROBLEMS

3.1 A drug has a log D6.0 value of 3.7 and is poorly soluble in aqueous media. Whenadministered orally, approximately 30% of a dose is lost due to incomplete dissolution.

Page 77: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

REFERENCES 57

TABLE P3.2

Lipoamide Nosolatol Disolvprazole

Acid or Base Base Acid BaseMolecular mass 396 Da 365 Da 221 DaHighest dose strength 150 mg 250 mg 50 mglog P 3.2 2.1 0.2log D6.0 3.0 1.8 −2.8Solubility pH 1 to 7.5 High: 1.0 g/1000 mL Low: 0.5 g/1000 mL High: 5 g/1000 mLFraction of oral dose recovered

as metabolites in humans99.0% 99.2% 3–5%

Main enzyme involved in itsmetabolism

CYP3A4 CYP3A4 None

Substrate for intestinal uptaketransporter

None known None known OATP1A2

Substrate for intestinal effluxtransporter

P-gp P-gp None known

Bioavailability factor (F) 0.21 0.7 0.5

It encounters no further problems during absorption, but it is a CYP3A4 substrate, andabout 25% of the drug passing through the membrane undergoes intestinal metabolism.During its initial pass through the liver, about 70% of the drug is lost due to metabolism.

(a) Calculate Fa, Fg, Fh, and F for this drug.

(b) Determine the effective dose when 50 mg is given orally.

(c) Determine the value of an intravenous dose that is equivalent to a 100-mg oraldose.

3.2 Three fictitious drugs are used as examples throughout this book: lipoamide, nosolatol,and disolvprazole. Details of these drugs are provided in Appendix E. Lipoamideis a novel antipyretic drug, nosolatol is a cardioselective �1-adrenergic antagonist,and disolvprazole is a proton pump inhibitor. Table P3.2 lists the physicochemicalcharacteristics of the three drugs.

(a) Use this information to discuss their potential for oral administration. Addressin detail how the information provides insight into how they may penetrate theintestinal membrane, their expected extent of absorption, and how they would beclassified according to the BCS or BDDCS system.

(b) Discuss how you would predict food to affect their absorption.

(c) Suggest possible explanations for the value of bioavailability reported for eachdrug.

REFERENCES

1. Costantino, H. R., Illum, L., Brandt, G., Johnson, P. H., and Quay, S. C. (2007) Intranasal delivery:physicochemical and therapeutic aspects, Int J Pharm 337, 1–24.

2. Lennernas, H. (2007) Intestinal permeability and its relevance for absorption and elimination,Xenobiotica 37, 1015–1051.

3. Lahner, E., Annibale, B., and Delle Fave, G. (2009) Systematic review: impaired drug absorptionrelated to the co-administration of antisecretory therapy, Aliment Pharmacol Ther 29, 1219–1229.

Page 78: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

58 DRUG ADMINISTRATION, ABSORPTION, AND BIOAVAILABILITY

4. Yang, Y. X. (2008) Proton pump inhibitor therapy and osteoporosis, Curr Drug Saf 3, 204–209.

5. Smith, D. (2007) Introduction to drug metabolism enzymes, in Drug Metabolizing Enzymes:Fundamentals, Study Methods, Recent Advances and Clinical Significance ( Daly, A., Ed.), TheBiomedical & Life Sciences Collection, Henry Stewart Talks Ltd., London.

6. Lipinski, C. A., Lombardo, F., Dominy, B. W., and Feeney, P. J. (2001) Experimental and com-putational approaches to estimate solubility and permeability in drug discovery and developmentsettings, Adv Drug Deliv Rev 46, 3–26.

7. Linnankoski, J., Makela, J. M., Ranta, V. P., Urtti, A., and Yliperttula, M. (2006) Computationalprediction of oral drug absorption based on absorption rate constants in humans, J Med Chem49, 3674–3681.

8. Glaeser, H., Bailey, D. G., Dresser, G. K., Gregor, J. C., Schwarz, U. I., McGrath, J. S., Jolicoeur,E., Lee, W., Leake, B. F., Tirona, R. G., and Kim, R. B. (2007) Intestinal drug transporterexpression and the impact of grapefruit juice in humans, Clin Pharmacol Ther 81, 362–370.

9. Farkas, D., and Greenblatt, D. J. (2008) Influence of fruit juices on drug disposition: discrepanciesbetween in vitro and clinical studies, Expert Opin Drug Metab Toxicol 4, 381–393.

10. Oostendorp, R. L., Beijnen, J. H., and Schellens, J. H. (2009) The biological and clinical role ofdrug transporters at the intestinal barrier, Cancer Treat Rev 35, 137–147.

11. Rautio, J., Kumpulainen, H., Heimbach, T., Oliyai, R., Oh, D., Jarvinen, T., and Savolainen, J.(2008) Prodrugs: design and clinical applications, Nat Rev Drug Discov 7, 255–270.

12. Murakami, T., and Takano, M. (2008) Intestinal efflux transporters and drug absorption, ExpertOpin Drug Metab Toxicol 4, 923–939.

13. Marchetti, S., Mazzanti, R., Beijnen, J. H., and Schellens, J. H. (2007) Concise review: clinicalrelevance of drug drug and herb drug interactions mediated by the ABC transporter ABCB1(MDR1, P-glycoprotein), Oncologist 12, 927–941.

14. Greiner, B., Eichelbaum, M., Fritz, P., Kreichgauer, H. P., von Richter, O., Zundler, J., andKroemer, H. K. (1999) The role of intestinal P-glycoprotein in the interaction of digoxin andrifampin, J Clin Invest 104, 147–153.

15. Schwarz, U. I., Hanso, H., Oertel, R., Miehlke, S., Kuhlisch, E., Glaeser, H., Hitzl, M., Dresser,G. K., Kim, R. B., and Kirch, W. (2007) Induction of intestinal P-glycoprotein by St John’s wortreduces the oral bioavailability of talinolol, Clin Pharmacol Ther 81, 669–678.

16. Westphal, K., Weinbrenner, A., Zschiesche, M., Franke, G., Knoke, M., Oertel, R., Fritz, P., vonRichter, O., Warzok, R., Hachenberg, T., Kauffmann, H. M., Schrenk, D., Terhaag, B., Kroemer,H. K., and Siegmund, W. (2000) Induction of P-glycoprotein by rifampin increases intestinalsecretion of talinolol in human beings: a new type of drug/drug interaction, Clin Pharmacol Ther68, 345–355.

17. Fischer, V., Einolf, H. J., and Cohen, D. (2005) Efflux transporters and their clinical relevance,Mini Rev Med Chem 5, 183–195.

18. Keskitalo, J. E., Zolk, O., Fromm, M. F., Kurkinen, K. J., Neuvonen, P. J., and Niemi, M. (2009)ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin,Clin Pharmacol Ther 86, 197–203.

19. Urquhart, B. L., Ware, J. A., Tirona, R. G., Ho, R. H., Leake, B. F., Schwarz, U. I., Zaher, H.,Palandra, J., Gregor, J. C., Dresser, G. K., and Kim, R. B. (2008) Breast cancer resistance protein(ABCG2) and drug disposition: intestinal expression, polymorphisms and sulfasalazine as an invivo probe, Pharmacogenet Genomics 18, 439–448.

20. Dahan, A., and Amidon, G. L. (2009) Small intestinal efflux mediated by MRP2 and BCRPshifts sulfasalazine intestinal permeability from high to low, enabling its colonic targeting, Am JPhysiol Gastrointest Liver Physiol 297, G371–G377.

21. Dahan, A., and Amidon, G. L. (2010) MRP2 mediated drug–drug interaction: indomethacin in-creases sulfasalazine absorption in the small intestine, potentially decreasing its colonic targeting,Int J Pharm 386, 216–220.

Page 79: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc03 JWBS062-Rosenbaum February 16, 2011 7:32 Printer Name: Yet to Come

REFERENCES 59

22. Thelen, K., and Dressman, J. B. (2009) Cytochrome P450–mediated metabolism in the humangut wall, J Pharm Pharmacol 61, 541–558.

23. Paine, M. F., Khalighi, M., Fisher, J. M., Shen, D. D., Kunze, K. L., Marsh, C. L., Perkins, J.D., and Thummel, K. E. (1997) Characterization of interintestinal and intraintestinal variationsin human CYP3A-dependent metabolism, J Pharmacol Exp Ther 283, 1552–1562.

24. Galetin, A., Hinton, L. K., Burt, H., Obach, R. S., and Houston, J. B. (2007) Maximal inhibition ofintestinal first-pass metabolism as a pragmatic indicator of intestinal contribution to the drug–druginteractions for CYP3A4 cleared drugs, Curr Drug Metab 8, 685–693.

25. Bressler, R. (2006) Grapefruit juice and drug interactions: exploring mechanisms of this interac-tion and potential toxicity for certain drugs, Geriatrics 61, 12–18.

26. Neuvonen, P. J., Backman, J. T., and Niemi, M. (2008) Pharmacokinetic comparison of thepotential over-the-counter statins simvastatin, lovastatin, fluvastatin and pravastatin, Clin Phar-macokinet 47, 463–474.

27. Ando, H., Tsuruoka, S., Yanagihara, H., Sugimoto, K., Miyata, M., Yamazoe, Y., Takamura,T., Kaneko, S., and Fujimura, A. (2005) Effects of grapefruit juice on the pharmacokinetics ofpitavastatin and atorvastatin, Br J Clin Pharmacol 60, 494–497.

28. Nimmo, J., Heading, R. C., Tothill, P., and Prescott, L. F. (1973) Pharmacological modificationof gastric emptying: effects of propantheline and metoclopromide on paracetamol absorption, BrMed J 1, 587–589.

29. Amidon, G. L., Lennernas, H., Shah, V. P., and Crison, J. R. (1995) A theoretical basis for abiopharmaceutic drug classification: the correlation of in vitro drug product dissolution and invivo bioavailability, Pharm Res 12, 413–420.

30. Wu, C. Y., and Benet, L. Z. (2005) Predicting drug disposition via application of BCS: trans-port/absorption/elimination interplay and development of a biopharmaceutics drug dispositionclassification system, Pharm Res 22, 11–23.

Page 80: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

4DRUG DISTRIBUTION

4.1 Introduction

4.2 Extent of Drug Distribution4.2.1 Distribution Volumes4.2.2 Tissue Binding and Plasma Protein Binding: Concentrating Effects4.2.3 Assessment of the Extent of Drug Distribution: Apparent Volume of Distribution

4.2.3.1 Fraction of Drug in the Plasma and Tissues4.2.3.2 Influence of Tissue and Plasma Protein Binding

4.2.4 Plasma Protein Binding4.2.4.1 Factors Controlling Binding4.2.4.2 Clinical Consequences of Changes in Plasma Protein Binding

4.3 Rate of Drug Distribution4.3.1 Perfusion-Controlled Drug Distribution4.3.2 Diffusion-Controlled Drug Distribution

4.4 Distribution of Drugs to the Central Nervous System

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand the factors that control a drug’s distribution from the plasma to thetissues

2. Know the main physiological volumes that a drug may access

3. Understand the influence of plasma protein and tissue binding on the distributionprofile

4. Understand how the apparent volume of distribution expresses the distribution of adrug between plasma and the rest of the body

5. Understand the factors that control plasma protein binding

6. Appreciate the clinical significance of altered protein binding

7. Understand the factors that control the rate of drug distribution

8. Appreciate some unique aspects of drug distribution to the central nervous system

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

60

Page 81: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 61

4.1 INTRODUCTION

As a result of either direct systemic administration or absorption from an extravascularroute, drug reaches the systemic circulation, where it very rapidly distributes throughoutthe entire volume of plasma water and is delivered to tissues around the body. Two aspectsof drug distribution need to be considered: how rapidly, and to what extent, the drugin the plasma gets taken up by the tissues. Clinically, it is rarely possible to measuretissue concentrations of a drug, and consequently, distribution patterns have to be inferredfrom measurements of the plasma concentrations of the drug. A lot of information on therate of drug distribution can be obtained by observing the pattern of the changes in theplasma concentrations in the early period following drug administration. Information aboutthe extent of drug distribution can be obtained by considering the value of the plasmaconcentration once distribution is complete. Thus, the plasma concentration constitutes a“window” for obtaining information on the distribution of the bulk of the drug in the bodyand how it changes over time.

4.2 EXTENT OF DRUG DISTRIBUTION

Two aspects of a drug’s distribution pattern are of greatest interest:

� The access of the drug to its site of action� The relative distribution of a drug between plasma and the rest of the body

A drug must reach its site of action to produce an effect. Generally, this involves onlya very small amount of the overall drug in the body, and access to the site of action isgenerally a problem only if the site is located in a specialized area or space. For example,drug access to certain poorly perfused areas such as inner ear fluid or solid tumor massesmay be problematic. Additionally, the specialized membrane (the blood–brain barrier) thatseparates the brain from the systemic circulation limits the access of many drugs to thecentral nervous system.

The second important aspect of the extent of drug distribution is the relative distributionof a drug between plasma and the rest of the body. This affects the plasma concentration ofthe drug and is important because:

1. As discussed above, the plasma concentration is the “window” through which we areable to “see” the drug in the body. It is important to know how a measured plasmaconcentration is related to the total amount of drug in the body.

2. Drug is delivered to the organs of elimination via the blood. If a drug distributesextensively from the plasma to the tissues, the drug in the plasma will constitute onlya small fraction of the drug in the body. Little drug will be delivered to the organs ofelimination, and this will hamper elimination. Conversely, if a drug is very limitedin its ability to distribute beyond the plasma, a greater fraction of the drug in thebody will be physically located in the plasma. The organs of elimination will be wellsupplied with drug, and this will enhance the elimination processes.

It is important to appreciate that as long as the drug reaches its site of action, withinreason there is no “good” or “bad” distribution pattern. It is simply important to understandthe distribution pattern for a given drug.

Page 82: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

62 DRUG DISTRIBUTION

Dru

g C

once

ntra

tion

Time After Dose

Plasma Concentration

Concentration in a Tissue

DistributionPhase

PostdistributionPhase

FIGURE 4.1 Drug concentrations in the plasma and a theoretical tissue during the early periodafter an intravenous dose of a drug. The plasma concentration falls steeply and the tissue concentrationrises during the distribution phase. During the postdistribution phase, distribution is complete and theplasma and tissue concentrations fall in parallel.

Drug distribution to the tissues is driven primarily by the passive diffusion of free,unbound drug along its concentration gradient. Consider the administration of a singleintravenous dose of a drug. In the early period after administration, the concentration ofdrug in the plasma is much higher than that in the tissues, and there is a net movement ofdrug from the plasma to the tissues (Figure 4.1); this period is known as the distributionphase. Eventually, a type of equilibrium is established between the tissues and plasma, atwhich point the ratio of the tissue to plasma concentration remains constant. At this timethe distribution phase is complete and the tissue and plasma concentrations rise and fall inparallel; this period is known as the postdistribution phase (Figure 4.1). It should be notedthat after a single dose, true equilibrium between the tissues and the plasma is not achievedin the postdistribution phase because the plasma concentration falls continuously as drugis eliminated from the body. This breaks the equilibrium between the two and results inthe redistribution of drug from the tissues to the plasma. Uptake and efflux transportersin certain tissues may also be involved in the distribution process and may enhance orlimit a drug’s distribution to specific tissues. Our discussion of the factors controlling drugdistribution is presented through a consideration of the major physiological volumes thatdrugs can potentially access.

4.2.1 Distribution Volumes

Three important physiological volumes—plasma water, extracellular fluid, and total bodywater—are shown in Figure 4.2. In the figure, these volumes are drawn to scale.

Plasma In the systemic circulation, drugs distribute throughout the volume of plasmawater (about 3 L) (Figure 4.2). Where a drug goes beyond this, including distribution to the

Page 83: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 63

Plasma

Extracellular Fluid

Total Body Water

Capillary∗

EpitheliumCell∗∗Membranes

40L3L 15L

FIGURE 4.2 Physiological volumes drugs may access. *Most capillary membranes are loose andpermit the paracellular passage of even large polar drugs. Notable exceptions include the brain, testes,and placenta. **Possible only for lipophilic drugs unless a specialized transport system is present for adrug. Efflux transporters may extrude drugs that penetrate the membrane. Note that the three volumesare drawn to scale.

cellular elements of the blood, depends on the physicochemical properties of the drug andthe permeability characteristics of individual membranes.

Distribution to the Extracellular Fluid The membranes of the capillary epithelial cellsare generally very loose in nature and permit the paracellular passage of even polar and/orlarge drug molecules, including the aminoglycosides (log D7.4 ≈ −10; MW 450 to 1000 Da)and protein molecules. Thus, most drugs are able to distribute throughout the volume ofextracellular fluid, a volume of about 15 L (Figure 4.2). However, the capillary membranesof certain tissues, notably delicate tissues such as the central nervous system, the placenta,and the testes, have much more tightly knit membranes, which may limit the access ofcertain drugs, particularly large and/or polar drugs.

Distribution to Intracellular Fluid Once in the extracellular fluid, drugs are exposed tothe individual cells of tissues. The ability of drugs to penetrate the membrane of thesecells is dependent on a drug’s physicochemical properties (Figure 4.2). Polar drugs andlarge molecular mass drugs will be unable to pass cell membranes by passive diffusion.For example, the extremely polar aminoglycosides cannot penetrate cell membranes and,as a result, distribute into a volume that is approximately equal to that of extracellular fluid.Polar drugs may enter cells if they are substrates for specialized uptake transporters. Theantidiabetic drug metformin is a small polar molecule (MW 129 Da; log D7.4 ≈ −3.4) thatwould be expected to have difficulty diffusing through cell membranes. However, it is ableto access its site of action in the hepatocyte because it is a substrate for the hepatic organiccation transporter (OCT1), which transports it across the hepatocyte membrane. On theother hand, efflux transporters will restrict the distribution of their substrates. For example,P-glycoprotein (P-gp) at the blood–brain barrier limits the access of a large number ofdrugs, including ritonavir, loperamide, and many anticancer drugs. Small lipophilic drugsthat can easily penetrate cell membranes can potentially distribute throughout the total bodywater, which is around 40 L.

In summary, drugs are able to pass through most of the capillary membranes in thebody and distribute into a volume approximately equal to that of the extracellular fluid

Page 84: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

64 DRUG DISTRIBUTION

(about 15 L). The ability of a drug to distribute beyond this depends primarily on itsphysicochemical characteristics. Small, lipophilic drug molecules should penetrate biolog-ical membranes with ease and distribute throughout the total body water (about 40 L). Adrug’s distribution to specific tissues may be enhanced by uptake transporters. Conversely,efflux transporters will restrict the tissue distribution of their substrates. Total body water,about 40 L, represents the maximum volume into which a drug can distribute.

4.2.2 Tissue Binding and Plasma Protein Binding: Concentrating Effects

Given that drug distribution is driven primarily by passive diffusion, it would be reasonableto assume that once distribution has occurred, the concentration of drug would be the samethroughout its distribution volume. This is rarely the case because of tissue and plasmaprotein binding. Drugs frequently bind in a reversible manner to sites on proteins andother macromolecules in the plasma and tissues (binding is discussed in more detail inSection 4.2.4). At this time it is important to appreciate that bound drug cannot participatein the concentration gradient that drives the distribution process. The bound drug can beconsidered to be secreted away or hidden in tissue or plasma. Binding has a very importantinfluence on a drug’s distribution pattern. Consider a drug that binds extensively (90%) to theplasma proteins but does not bind to tissue macromolecules. In the plasma, 90% of the drugis bound and only 10% is free and able to diffuse to the tissues. At equilibrium, the unboundconcentrations in the plasma and tissues will be the same, but the total concentration ofdrug in the plasma will be much higher than that in the tissues.

Plasma protein binding has the effect of limiting distribution and concentrating drugin the plasma (Figure 4.3). On the other hand, consider a drug that binds extensively tomacromolecules in the tissues but does not bind to the plasma proteins. Assume that overall90% of the drug in the tissues is bound and only 10% is free. As the distribution processoccurs, a large fraction of the drug in the tissues will bind and be removed from participationin the diffusion gradient. As a result, more and more drug will distribute to the tissues.When distribution is complete, the unbound concentrations in the plasma and tissues willbe the same, but the total (bound plus free) average tissue concentration will be much largerthan the plasma concentration (Figure 4.3). Tissue binding essentially draws drug fromthe plasma and concentrates it in the tissues. Drugs often bind to both the plasma proteinsand tissue macromolecules. In this case the final distribution pattern will be determined bywhich is the dominant process.

In summary, the binding of drugs to plasma proteins and tissue macromolecules exertsan important influence on the pattern of drug distribution. Plasma protein binding tends toconcentrate drug in the plasma and limit its distribution to the tissues. Tissue binding tendsto concentrate a large amount of drug in a tissue. Drug concentration will vary from tissueto tissue and be greatest in those tissues where binding is most extensive.

Overall, drug distribution is driven primarily by the passive diffusion of a drug alongthe concentration gradient created by the unbound drug in the plasma and tissues. Theoverall pattern of drug distribution is determined by the physiological volumes that a drugis able to access and by the concentrating effects of drug binding to plasma protein and/ortissues. Once distribution is complete, the unbound drug concentration should be the samethroughout the physiological volumes where the drug is found. However, uptake and effluxtransporters may also be involved and may, respectively, either promote or limit a drug’sdistribution beyond that predicted by passive diffusion alone.

Page 85: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 65

Dru

g C

once

ntra

tion

FREE

FREE

FREE

FREE

FREE

FREE

BOUND

BOUND

Ct, Cp

Cp

Ct

Ct

Cp

(a) (b) (c)

Plasma

Plasma

Plasma

TissuesTissues

Tissues

FIGURE 4.3 Influence of plasma protein binding and tissue binding on a drug’s distributionbetween the plasma and tissues. The histograms show the total plasma (Cp) and tissue (Ct) concen-trations of a drug partitioned into free and bound drug. At equilibrium, the free drug concentration isthe same in the plasma and tissues. If a drug does not bind either to plasma proteins or tissue macro-molecules, the total concentrations in the tissues and plasma will be the same (a). Plasma proteinbinding concentrates drug in the plasma (b) and tissue binding concentrates drug in the tissues (c).

4.2.3 Assessment of the Extent of Drug Distribution:Apparent Volume of Distribution

Once distribution has gone to completion, the ratio of the total tissue concentration to thetotal plasma concentration remains constant. The actual tissue concentration (and the ratio)will vary from tissue to tissue, depending on the relative effects of tissue and plasma proteinbinding. It is not possible to measure individual tissue concentrations, and it is convenientto consider an overall average tissue concentration (Ct). The ratio of Ct to Cp will varyfrom drug to drug.

It is important to find a way to express a drug’s distribution characteristics using a numberor distribution parameter that can easily be estimated clinically. The ratio discussed above(Ct/Cp) expresses distribution but cannot be measured easily. The requirements for thedistribution parameter are:

� It assesses or reflects the relative distribution of the drug between the plasma and therest of the body once distribution is complete.

� It must use the plasma concentration of the drug, as this is usually the only drugconcentration that can be measured routinely.

Consider two drugs, A and B. Assume that A cannot distribute to any great extentbeyond the plasma, perhaps because it binds extensively to the plasma proteins (the Ct/Cp

Page 86: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

66 DRUG DISTRIBUTION

ratio is small). Assume that drug B distributes extensively to the tissues, perhaps becauseof extensive tissue binding (the Ct/Cp ratio is high). Assume that doses of each wereadministered, and that at some time after distribution occurred, their plasma concentrationswere compared at a time when there were equal amounts of each drug in the body.

Let Ab be the amount of drug in the body. Consider the following ratio for each drug:

amount of drug in the body at any time

plasma concentration at the same time:

Ab

Cp(4.1)

For drug A,

Ab

Cp:

constant

high→ low (4.2)

For drug B,

Ab

Cp:

constant

low→ high (4.3)

The ratio would be smaller for drug A than for drug B:

Ratio = Ab

Cpunits:

mg

mg/L= L (4.4)

Thus, the value of this ratio is a measure of a drug’s extent of distribution. A large valueindicates extensive distribution, and a small value indicates limited distribution. The de-nominator and numerator of equation (4.4) can be measured: The plasma concentration canbe measured at any time after a dose, and there are certain times when the amount of drugin the body is known, such as immediately after an intravenous bolus injection. The ratiohas units of volume and is known as the apparent volume of distribution (Vd):

V d = Ab

Cp(4.5)

The box around equation (4.5) signifies that it is an important equation that should bememorized. The word apparent is usually dropped and the parameter is known simply asthe volume of distribution. Table 4.1 shows the calculations of Vd for the example drugsabove based on the amount of drug in the body and the corresponding plasma concentration.The plasma concentration of each was determined at a time when the amount of drug inthe body was known.

It is important to appreciate that the volume of distribution is simply a ratio that hasunits of volume. It is not a physiological volume and, despite its name, it is not the volumeinto which a drug distributes. The fact that drug A has a Vd value of 20 L (Table 4.1) doesnot mean that it distributes into a volume of 20 L, which is greater than extracellular fluidand less than the total body water. In theory it could do so if, for example, it did not bindto plasma proteins or tissue macromolecules and could not access most of the intracellular

Page 87: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 67

TABLE 4.1 Calculation of Vd for Drugs A and B

Drug A Drug B

Ab (mg) at time t 100 100Cp (mg/L) at time t 5 0.1Vd = Ab/Vd (L) 20 1000

fluid. On the other hand, if it binds to plasma proteins, it could distribute throughout thetotal body water (40 L) and have a Vd of 20 L.

The value of Vd depends in part on the volume into which the drug distributes, but it isalso dependent on a drug’s tissue and plasma protein binding characteristics. This can bemade clear by an example.

Example 4.1 Consider three beakers, each filled with 1 L of water (Figure 4.4).

1000 mL1000 mL

Beaker 2 Beaker 3 Beaker 1

Volume of Water: 1 L 1 L 1 LAmount of Drug: 100 mg 100 mg 100 mgConcentration

10 mg/Lof Sample: 100 mg/L = 190 mg/L

1000 mL

Charcoal Adsorbs90% of Drug

Charcoal Above Semi-permeable Membrane Adsorbs 90% of Drug

semi-permeablemembrane

90 mg/0.5 L (adsorbed) +10 mg/L

FIGURE 4.4 Apparent volume of three beakers. Each beaker contains 1 L of water and 100 mgof a soluble drug. Beaker 2 contains charcoal on the bottom that adsorbs 90% of the drug. Beaker 3has charcoal suspended in the upper 500 mL that adsorbs 90% of drug. The charcoal in beaker 3 isprevented from accessing the lower portion of the beaker by a semipermeable membrane.

� Each contains 100 mg of a water-soluble drug.� Beaker 2 contains charcoal on the bottom which adsorbs 90% of the drug.� Beaker 3 has charcoal suspended in the upper half of the liquid. The charcoal cannot

access the lower 500 mL because it cannot penetrate a semipermeable membrane thatseparates the two halves of the liquid. The charcoal adsorbs 90% of the drug in theupper liquid.

Page 88: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

68 DRUG DISTRIBUTION

� It is not possible to measure the volume of liquid in either beaker. They have tobe estimated based on the drug concentration of a sample taken from the top of eachbeaker. The estimated volume is referred to as the apparent volume (based on samplingof the drug concentration in the solution).

Solution Table E4.1 shows the calculation of the apparent volume of fluid in each beakerbased on the concentration of drug sampled in each. Beaker 2 has charcoal in the bottomthat absorbs 90% of the drug. Beaker 3 has charcoal suspended in the upper 500 mL thatadsorbs 90% of drug. The remaining 10% of the drug dissolves in the full 1 L. It can beseen that when the charcoal draws drug away from the sampling fluid (beaker 2), it causesthe apparent volume to be greater than the actual volume. Conversely, when the charcoalconcentrates the drug in the sampling fluid (beaker 3), it causes the apparent volume to beless than the actual volume.

TABLE E4.1 Calculation of the Apparent Volume of Fluid Based on the Amount of DrugPresent in the System and Its Concentration in the Fluid

Beaker 1 Beaker 2 Beaker 3

Volume of water 1 L 1 L 1 LAmount of drug 100 mg 100 mg 100 mgConcentration of

drug in sample100 mg/L

= 100 mg/L10 mg/L

= 10 mg/L90 mg/0.5 L + 10 mg/L

= 190 mg/LApparent volumea 100 mg/100 mg/L

= 1 L100 mg/10 mg/L

= 10 L100 mg/190 mg/L

= 0.53 L

aAmount of drug in beaker/concentration of sample.

In the body, drug concentrations are sampled from the plasma. Binding of drugs to thetissue macromolecules draws drug from the plasma and causes the volume of distributionto be greater than the actual volume into which a drug distributes. Conversely, plasmaprotein binding causes drug to concentrate in the sampling volume (plasma). This resultsin an apparent volume that is less than the actual distribution volume. Under the unusualcircumstances that a drug does not bind to plasma proteins or tissue macromolecules, itsvolume of distribution will be equal to the volume into which it distributes. This is thecase for ethanol, which distributes throughout the total body water, doesn’t bind to plasmaproteins or tissue macromolecules, and has a volume of distribution of around 40 L.

Because of its dependence on physiological volumes, the volume of distribution isdependent on body size and is most conveniently expressed on a per kilogram of bodyweight basis. This value is then multiplied by body weight to obtain a person’s volume ofdistribution. A 35-kg child will thus have a volume of distribution of a drug half that ofa typical 70-kg standard adult male. The volume of distribution expressed in this way isassumed to be a constant for a drug, assuming normal conditions and health. Its value maybe affected by conditions that affect either body volumes (e.g., dehydration, overhydration,the presence of ascites), plasma protein binding, or tissue binding.

Table 4.2 shows the values of the volume of distribution of some drugs. Some commonlyencountered volumes are presented to provide some perspective. A consideration of someof the values in the table clearly illustrates that the volume of distribution cannot possibly beequal to the volume into which a drug distributes. Chloroquine has a volume of distribution

Page 89: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 69

TABLE 4.2 Volume of Distribution of Select Drugs

Drug/Physiological Volume Vd (L/70 kg) Example of Volume

Plasma 3Heparin 4.2 Gallon of milkWarfarin 8Extracellular fluid 15 Water cooler vesselAminoglycosides 20Total body water 40Phenytoin 45 Approximately a “half-barrel” kegAtenolol 65Diazepam 77Digoxin 500 Large refrigeratorFelodipine 700Nortriptyline 1,260Amiodarone 4,600Chloroquine 14,000 Dumpster/aboveground pool

approximately equal to that of an average dumpster or aboveground swimming pool. Adrug’s volume of distribution exceeds physiological volumes because of tissue binding,which draws a large fraction of the drug from the plasma and results in an average tissueconcentration that exceeds the plasma concentration. The volume of distribution could beconsidered to be the hypothetical volume into which the drug distributes if the concentrationof drug throughout the volume was the same and equal to the plasma concentration.

4.2.3.1 Fraction of Drug in the Plasma and TissuesThe value of a drug’s volume of distribution can be used to estimate the fraction of the drugin the body that is physically present in either the plasma or the tissues. The drug in thebody (Ab) may be partitioned into drug in the plasma (Ap) and drug outside the plasma orin the tissues (At):

Ab = Ap + At (4.6)

The fraction of the drug in the plasma,

fraction in plasma = Ap

Ab(4.7)

The amount of drug in the plasma is the product of the plasma concentration and the volumeof the plasma.

The amount of drug in the body is the product of the volume of distribution and theplasma concentration [see equation (4.5)]:

fraction in plasma = Vp · Cp

Vd · Cp= Vp

Vd(4.8)

Page 90: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

70 DRUG DISTRIBUTION

TABLE 4.3 Relative Distribution of Drugs Between thePlasma and Tissues for Different Values of Vd

Vd (L/70 kg) Drug in Plasma (%)a Drug in Tissues (%)

12 25 7521 14 8642 7 93

300 1 9912,000 0.025 99.98

aAssumes that Vp = 3 L for a standard 70-kg person.

In a standard 70-kg adult male, Vp = 3 L:

fraction in plasma = 3

Vd(4.9)

The fraction of the drug in the body located in the tissues:

fraction in tissue = 1 − fraction in plasma

= 1 − Vp

Vd= 1 − 3

Vd

(4.10)

Table 4.3 shows the relative distribution of drugs between the tissues and plasma fordifferent values of volume of distribution.

4.2.3.2 Influence of Tissue and Plasma Protein BindingAs expressed in equation (4.6), drug in the body is located in either the plasma or thetissues. The amount of drug in either of these spaces is the product of the concentration ofdrug and the volume of the space. Equation (4.6) can be rewritten

Cp · Vd = Cp · Vp + Ct · Vt (4.11)

where Vt is the volume outside the plasma into which the drug distributes. Drug diffusion isdriven by the concentration gradient created by the unbound drug in the plasma and tissues.The unbound drug concentrations are expressed as

Cpu = Cp · fu

Ctu = Ct · fut

(4.12)

where Cpu and Ctu are the unbound drug concentrations in the plasma and tissue, respec-tively, and fu and fut are the fractions unbound in the plasma and tissues, respectively.

When distribution is complete, the unbound drug concentrations in the tissue and plasmaare equal:

Cpu = Ctu (4.13)

Substituting for the expressions of Cpu and Ctu given in equation (4.12) into equation (4.13)yields

Cp · fu = Ct · fut (4.14)

Page 91: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 71

TABLE 4.4 Compounds That Do Not Bind to PlasmaProteins or Tissue Macromolecules

Compound Vd (L) in 70-kg Male Distribution Volume

Evans blue 3 PlasmaBr− 15 Extracellular fluidAntipyrene 40 Total body waterEthanol 40 Total body water

Rearranging gives

Ct = Cp · fu

fut(4.15)

Substituting the expression of Ct given in equation (4.15) into equation (4.11), we have

Cp · Vd = Cp · Vp + VtCp · fu

fut

Vd = Vp + Vtfu

fut

(4.16)

Equation (4.16) shows that a drug’s volume of distribution is dependent on both the volumeinto which a drug distributes and on tissue and plasma protein binding. It also shows thatincreased tissue binding (fut ↓) or decreased plasma protein binding (fu ↑) will result in anincrease in the volume of distribution.

Equation (4.16) can also be used to show that if a drug binds to neither the plasmaproteins (fu = 1) nor the tissues (fut = 1), its volume of distribution will be equal tothat of the volume into which the drug distributes. Table 4.4 lists some compounds thathave volumes of distribution that approximate the volumes into which they distribute.The volume of distribution of these substances can be used to estimate the respectivephysiological volumes.

Summary of Volume of Distribution

1. Vd is a ratio that reflects a drug’s relative distribution between the plasma and therest of the body.

2. It is dependent on the volume into which a drug distributes and a drug’s bindingcharacteristics.

3. It is a constant for a drug under normal conditions.

4. Conditions that alter body volume may affect its value.

5. Altered tissue and/or protein binding may alter its value.

6. It provides information about a drug’s distribution pattern. Large values indicateextensive distribution of a drug to the tissues.

7. It can be used to calculate the amount of drug in the body if a drug’s plasmaconcentration is known.

Page 92: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

72 DRUG DISTRIBUTION

4.2.4 Plasma Protein Binding

A very large number of therapeutic drugs bind to certain sites on the proteins in plasmato form drug–protein complexes. The binding process occurs very rapidly, it is completelyreversible [see equation (4.18)], and equilibrium is quickly established between the boundand unbound forms of a drug. If the unbound or free drug concentration falls due todistribution or drug elimination, bound drug dissociates rapidly to restore equilibrium.Clinically, although the total drug concentration is measured routinely, pharmacologicaland toxicological activity is thought to reside with the free unbound drug (Cpu). It is onlythis component of the drug that is thought to be able to diffuse across membranes to thedrug’s site of action and to interact with the receptor. Binding is usually expressed usingthe parameter fraction unbound (fu), and the unbound pharmacologically active componentcan be calculated:

Cpu = Cp · fu (4.17)

Albumin, �1-acid glycoprotein (AAG), and the lipoproteins are the plasma proteins prin-cipally involved in the binding of drugs. Some characteristics of these plasma proteins areprovided in Table 4.5. Albumin is the most abundant and has a concentration of about40 g/L. Many drugs bind to albumin, particularly weak acids and neutral drugs.

�1-Acid glycoprotein is present in lower concentration than albumin and binds primarilyneutral and basic drugs. It is referred to as an acute-phase reactant protein because its con-centration increases in response to a variety of unrelated stressful conditions, such as cancer,inflammation, and acute myocardial infarction. The lipoproteins consist of a heterogeneousgroup of very large molecular mass proteins, including the high-density lipoproteins (HDLs)and low-density lipoproteins (LDLs). Their concentrations vary widely within the popula-tion, depending on diet and genetic factors, and they tend to bind neutral and basic lipophilicdrugs, including cyclosporine and propranolol. Other specialized proteins may be involvedin the binding of a small number of other drugs. For example, corticosteroid-binding

TABLE 4.5 Common Proteins Involved in Drug Binding in Plasma

Typical AverageConcentration

ProteinAverage

MW (Da) g/L �MTypes of Drugs

That BindExamples of

Drugs That Bind

Albumin 66,000 ∼40 600 Acid, neutral,some basic

Diflunisal,phenytoin,salicylic acid,valproic acid,warfarin

�1-Acidglycoprotein

43,000 ∼1 23 Basic, someacid andneutral

Alfentanil,meperidine,saquinavir,verapamil

Lipoproteins 200,000–3,000,000

— Widevariation

Neutral, basic Amiodarone,cyclosporine

HDL ∼1.50LDL ∼3.00

Page 93: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 73

globulin or transcortin is important in the plasma protein binding of corticosteroids such asprednisolone.

Given that the unbound concentration is the clinically important fraction and that it isthe total concentration that is routinely measured, it is important to know how and whenthe unbound fraction may change for a drug. If fu were to vary widely, it would have to beconsidered every time that a plasma concentration was interpreted. On the other hand, if itremained constant, it would not have to be considered because the unbound concentrationwould always be proportional to the total concentration.

4.2.4.1 Factors Controlling BindingThe binding of drugs to plasma proteins is an example of a capacity-limited process that isgoverned by the law of mass action. This is a very important type of process that will beencountered many times in pharmacokinetics and pharmacodynamics. The interaction ofthe drug with the protein is given by the law of mass action:

[D] + [P]k1−→←−k2

[DP] (4.18)

where [D] is the concentration of free drug, [P] the concentration of free protein bindingsites, [DP] the concentration of the drug–protein complex, and k1 and k2 are the rateconstants for the forward and backward processes, respectively.

The process is referred to as capacity limited because there are only a finite numberof binding sites on a protein: Binding is limited by the total capacity of the proteins.Substituting the more familiar symbols in equation (4.18) gives us

Cpu + (PT − Cpb)k1−→←−k2

Cpb (4.19)

where Cpu is the unbound drug concentration, PT the total concentration of protein-bindingsites, and Cpb the concentration of the drug–protein complex or the concentration ofbound drug.

Equating the rates of the forward and backward reactions, which are equal at equilibrium,and rearranging the expression yields

Cpb = PT · Cpu

Kd + Cpu(4.20)

where Kd is the dissociation constant equal to k2/k1, which is a reciprocal measure of thedrug’s affinity for the protein. As Kd increases, affinity decreases, and vice versa. Note fromequation (4.20) that when Cpb = PT/2, Cpu = Kd.

Figure 4.5 shows the typical relationship between the product of a capacity-limitedprocess (in this case the bound drug: Cpb) and the concentration driving the process (freedrug concentration: Cpu). At low concentrations, binding increases in direct proportion toan increase in the free drug (fu remains constant as Cpu increases). As the free drug con-centration increases further, some saturation of the proteins occurs, and proportionally lessdrug can bind (fu will increase as Cpu increases). Eventually, at high drug concentrations,all the binding sites on the protein are taken and binding cannot increase further.

Page 94: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

74 DRUG DISTRIBUTION

Cp b

Cpu

Cpb, max

Cpb, max/2

Kd

In this area, Cpb increases in direct proportionto increases in Cpu : fu remains constant.Therapeutic concentrations of most drugs are lessthan their Kd. As a result, fu is constant overthe range of therapeutic plasma concentrations.

All the binding sites on theprotein are taken. Bindingcannot increase further.

FIGURE 4.5 Binding of drugs to plasma proteins: a capacity-limited process. The graph showsthe relationship between the bound drug concentration (Cpb) and the free drug concentration (Cpu).At low free drug concentrations, there are many free binding sites, and binding can increase in directproportion to increases in the free drug concentration (fu is constant). But as some saturation of thebinding sites occurs, proportionally less drug can bind (fu increases). Eventually, at very high drugconcentrations, all the binding sites are taken, and the concentration bound remains constant at itsmaximum value (Cpb, max).

Affinity The affinity of the drug for the protein is the main determinant of fu. In equation(4.20), affinity is expressed by Kd, which is a reciprocal form of affinity. As affinityincreases, Kd gets smaller. Drugs with small Kd values bind extensively, whereas those withlarge Kd values will not bind extensively.

Influence of Drug Concentration on fu As shown in Figure 4.5, the therapeutic plasmaconcentrations of most drugs are much less than their Kd values. As a result, over therapeuticconcentrations, binding is able to increase in proportion to increases in the total concen-tration: fu remains constant over therapeutic plasma concentrations. There are, however, afew drugs that have therapeutic plasma concentrations that are around the range of their Kd

values. These drugs, which tend to be drugs that have very high therapeutic plasma con-centrations, include valproic acid (therapeutic concentrations range from 40 to 100 mg/L)and salicylates (100 to 400 mg/L), both of which bind to albumin, and disopyramide (2 to8 mg/L), which binds to AAG. The binding of these drugs uses a substantial amount ofprotein, and as a result they display concentration-dependent binding. As the drug con-centration increases, some degree of saturation is observed, and the fraction unbound getslarger. At concentrations of 40 and 130 mg/L, valproic acid is about 10% and 20% free,respectively. Disopyramide is about 20% free at 2 mg/L and about 50% free at 8 mg/L.Variations in the degree of binding over therapeutic plasma concentrations affects thedose–response relationship of these drugs, and complicates their clinical use.

Influence of Protein Concentration on fu As predicted by the law of mass action andequation (4.20), changes in the protein concentration will produce changes in the degreeof binding. Factors that increase the protein concentration will increase binding (decreasefu). Conversely, factors that decrease the protein concentration will decrease binding. Avariety of conditions can reduce albumin concentration, including liver disease, age, preg-nancy, burns, and other trauma. In the case of AAG, increases in the concentration aremore common. Physiological stress caused by myocardial infarction, cancer, and surgery

Page 95: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 75

can lead to four- to fivefold increases in the AAG concentration. Lipoprotein concen-trations vary widely in the population. They can decrease as a result of diet and ther-apy with HMG-CoA reductase inhibitors (statins), and increase due to alcoholism anddiabetes mellitus.

Displacement The binding of one drug may displace a second drug from its binding site.This displacement occurs because two drugs compete for a limited number of binding siteson the protein. Not surprisingly, displacers tend to be those drugs that achieve high concen-trations in the plasma, use up a lot of protein, and display concentration-dependent binding.Examples of displacing drugs include valproic acid, phenylbutazone, and salicylic acid.

Renal and Hepatic Disease The binding of drugs to albumin is often decreased in patientswith severe renal disease. This appears to be the result of both decreased albumin levels andthe accumulation of compounds that are normally eliminated, which may alter the affinityof drugs for albumin and/or compete for binding sites. The binding of several acidic drugs,including phenytoin and valproic acid, is reduced in severe renal disease. Plasma proteinbinding may also be reduced in hepatic disease. It is likely that the reduced albumin andAAG concentrations, particularly those observed in chronic liver disease, explain a largepart of this observation.

In summary, the degree to which drugs bind to the plasma protein is generally constantthroughout the range of therapeutic concentrations of most drugs. Binding will changeif the protein concentration changes and will change if a drug is displaced by either aconcomitant medication or by compounds that may accumulate in renal disease. Examplesof the binding characteristics of some drugs are shown in Table 4.6.

4.2.4.2 Clinical Consequences of Changes in Plasma Protein BindingChanges in fu as a result of altered protein concentration or displacement will result in achange in the fraction of the total drug that is unbound. Two issues need to be addressedwhen considering the clinical consequences of this: the potential changes in the unbound

TABLE 4.6 Binding Characteristics of Some Example Drugs

Drug Percent Bound fu

Amiodarone �99 �0.01Amoxicllin 18 0.82Carbamazepine 74 0.26Diazepam 98.7 0.013Digoxin 25 0.75Felodipine �99 �0.01Gentamicin �10 �0.90Ibuprofen �99 �0.01Imipramine 90 10Lovastatin �95 �0.05Methotrexate 46 0.54Propranolol 87 0.13Ritonavir 98 0.02Tamoxifen �98 �0.02

Page 96: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

76 DRUG DISTRIBUTION

drug concentration at the site of action, and the interpretation and evaluation of the routinelymeasured total plasma concentrations.

Changes in Unbound Concentration When binding decreases, the pharmacologicallyactive unbound component increases, and in theory, the response or toxicity could increase.However, the clinical consequences of altered plasma protein binding are minimized bytwo factors: (1) increased elimination and (2) little change in drug concentrations outsidethe plasma.

increased elimination In many cases, only the unbound drug is accessible to the organsof elimination. This is known as restrictive elimination because elimination is restrictedby protein binding and is limited to the unbound drug. For drugs that display restrictiveclearance, the increase in the unbound concentration that occurs when binding decreasesresults in an increase in elimination of the drug. The increase in elimination is usuallyproportional to the increase in unbound concentration. As a result, the unbound drugconcentration in the plasma eventually falls to exactly the same value as that before thechange in binding. In other words, the increase in the unbound concentration is canceledout by increased elimination.

Example 4.2 A drug is 90% bound to the plasma proteins and has a resting total concen-tration of 10 mg/L:

Cp = 10 mg/L f u = 0.1 Cpu = 1 mg/L

Note that the unbound pharmacologically active concentration is 1 mg/L.A second drug is coadministered and displaces the first drug. Only 80% is now bound:

Cp = 10 mg/L f u = 0.2 Cpu = 2 mg/L

The displacement has caused a doubling of the unbound pharmacologically active con-centration, but elimination of the drug now increases proportionally to this increase. Thedoubling of elimination results in a halving of the resting Cp, which decreases from 10 mg/Lto 5 mg/L:

Cp = 5 mg/L f u = 0.2 Cpu = 1 mg/L

Note that the unbound pharmacologically active concentration is equal to its original valuebefore the displacement.

The time it takes for the unbound concentration to return to its normal level is determinedby the rate of elimination of the drug (the elimination half-life) (this is explained inSection 11.3.5). If the drug is eliminated rapidly (short half-life), the unbound concentrationreturns to its original level quickly. If the drug is eliminated slowly, it takes a long time forthe unbound concentration to return to its original level. The time it takes to return can beimportant for drugs that have a narrow therapeutic index.

changes in drug concentration outside the plasma The plasma comprises a rel-atively small physiological volume (3 L). This is shown clearly in Figure 4.2, in whichthe main physiological volumes are drawn to scale. Even when plasma protein binding is

Page 97: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

EXTENT OF DRUG DISTRIBUTION 77

extensive, the fraction of the drug in the body that is located in the plasma is much lessthan that in the tissues. As a result, when the fraction bound increases, the extra drug thatdistributes to the tissues is often very small in comparison to the amount of drug alreadypresent. This is particularly the case for drugs that have large volumes of distribution, wherethe majority of the drug in the body is in the tissues and only a very small fraction residesin the plasma.

Example 4.3 Consider a drug with Vd = 60 L and fu = 0.10. For this drug, 3/60 × 100 =5% of the drug in the body is in the plasma, and 95% of the drug is in the tissues. If thedrug were displaced and fu increased to 0.2, Vd would be predicted to increase to 117 Land the fraction of drug in the plasma would decrease to about 2.5%. At the same time, thefraction in the tissues would increase by only about 2.5%, to 97.5%.

Thus, displacement of drugs from their plasma protein binding sites often has little effecton the concentration of drug outside the plasma.

warfarin The anticoagulant warfarin has a very narrow therapeutic range and there arevery serious clinical consequences of being outside the range. Subtherapeutic concentra-tions put patients at risk for blood clots and stroke. High concentrations predispose patientsto dangerous bleeding episodes. It is very important that therapeutic plasma concentrationsbe achieved at all times. Warfarin binds extensively to the plasma proteins (∼99%) and isdisplaced by several drugs, including diflunisal and phenylbutazone. The clinical signifi-cance of changes in the protein binding of warfarin is controversial and there is evidencethat the interaction with phenylbutazone has its roots in a reduction in the metabolism ofwarfarin rather than a displacement (1). In theory, any displacement will lead to only a tran-sient increase in the fraction unbound. However, warfarin has two characteristics that couldmake displacement more significant than normal. First, warfarin’s volume of distribution(8 L) is among the smallest of all therapeutic drugs. As a result, about 37.5% of the drug inthe body is in the plasma, and changes in the unbound plasma concentration may produceclinically important changes in the unbound concentration outside the plasma. Second, theactive S-isomer of warfarin is metabolized by cytochrome P450 2C9 (CYP2C9), whichdisplays genetic polymorphism, and within the population there are individuals who have amutant type of CYP2C9 that is associated with an impaired ability to metabolize warfarin.After a displacement in these patients, it will take a very long time for the unbound con-centration to return to the resting level. During this prolonged period, the patient may beexposed to potentially dangerously high concentrations of unbound warfarin. As a result, adisplacement in these patients may have clinical consequences.

Interpreting Cp In clinical practice, drug therapy may be monitored by ensuring thatplasma concentrations lie within the therapeutic range. The therapeutic range of a drugis expressed most conveniently in terms of concentration routinely measured, the totalplasma concentration (Cp). But since the unbound concentration is the pharmacologicallyactive component, the therapeutic range should more correctly be expressed in terms ofthis unbound concentration.

For example, the therapeutic range of phenytoin is usually expressed as 10 to 20 mg/Ltotal plasma concentration. This is based on an optimum unbound (pharmacologicallyactive) concentration range of 1 to 2 mg/L and the assumption of normal binding (fu =0.1). If binding is altered in a patient due to altered protein concentration or renal disease,

Page 98: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

78 DRUG DISTRIBUTION

TABLE 4.7 Phenytoin’s Therapeutic Range in Patients withNormal and Reduced Binding

Normal Binding Reduced Binding

Therapeutic rangebased on Cpu (mg/L)

1–2 1–2

fu 0.1 0.2Therapeutic range

based on Cp (mg/L):Cp = Cpu/fu

10–20 5–10

the therapeutic range based on the unbound concentration will not be affected, but therange based on total concentration will be different. Table 4.7 shows the therapeutic rangeof phenytoin based on total plasma concentration for a normal patient (fu = 0.1) and apatient with reduced binding (fu = 0.2). When evaluating a plasma concentration (totaldrug concentration) and assessing whether it is within the therapeutic range, it is importantto be aware of any potential changes in protein binding.

Example 4.4 G.Y. is a 45-year-old male who has been taking 400 mg of phenytoin twicedaily for several years. He has recently developed serious hepatic disease, and his albuminconcentration is abnormally low. The reduced albumin concentration is predicted to reducethe binding of phenytoin to the plasma proteins. Based on G.Y.’s albumin concentration,fu is estimated to be 0.2. During a routine clinic visit, G.Y.’s phenytoin concentration (totalconcentration) is found to be 8 mg/L. Is this within the therapeutic range, or should thedose be increased to get plasma concentrations in the range 10 to 20 mg/L?

Solution The therapeutic range of phenytoin is 1 to 2 mg/L based on the unboundconcentration. In patients with normal binding (fu = 0.1) this is equivalent to 10 to 20 mg/Ltotal phenytoin. Since G.Y. has altered binding, the range 10 to 20 mg is not applicable.G.Y.’s fu value is 0.2 and the therapeutic range based on total phenytoin may be calculated:

Cp = Cpu

fuif Cpu = 1, Cp = 1

0.2= 5 if Cpu = 2, Cp = 2

0.2= 10

The therapeutic range based on total phenytoin is 5 to 10 mg/L. This patient’s phenytoinlevel (8 mg/L) is within the therapeutic range.

Formulas have been developed for some drugs that will convert a measured plasmaconcentration of a drug to the value that it would be if the protein concentration werenormal. For example, if a phenytoin plasma concentration is measured in a patient withlow albumin, it can be converted to the value that would be expected with normal albuminusing the formula (2)

Cpnormal = Cpobserved

0.2Alb + 0.1(4.21)

where Cpnormal is the plasma concentration in the presence of a normal albumin concen-tration and Cpmeasured is the measured plasma concentration in a patient with an albumin

Page 99: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

RATE OF DRUG DISTRIBUTION 79

concentration of Alb g/dL. When the plasma concentration is converted in this way, theusual therapeutic range of 10 to 20 mg/L may be used.

In summary, decreased protein binding usually results in only a very small increase inthe tissue distribution of a drug. Furthermore, any changes in the unbound concentrationshould only be temporary, due to the resulting increase in elimination, which in theoryshould return the unbound concentration to its normal level. It is important to consideraltered binding when interpreting plasma concentrations. A drug’s therapeutic range basedon total plasma concentrations will change if the fraction unbound changes.

4.3 RATE OF DRUG DISTRIBUTION

The factors that determine the extent of drug distribution have now been addressed. Thesecond part of the discussion of drug distribution addresses the time it takes for a drugto distribute in the tissues. Figure 4.1 shows the plasma concentration and the typicaltissue concentration profile after the administration of a drug by intravenous injection. Itcan be seen that during the distribution phase, the tissue concentration increases as thedrug distributes to the tissue. Eventually, a type of equilibrium is reached, and followingthis, in the postdistribution phase, the tissue concentration falls in parallel with the plasmaconcentration. How long does the distribution phase last?

Drug distribution is a two-stage process that consists of:

1. Delivery of the drug to the tissue by the blood

2. Diffusion or uptake of drug from the blood to the tissue

The overall rate of distribution is controlled by the slowest of these steps. The deliveryof drug to the tissue is controlled by the specific blood flow to a given tissue. This isexpressed as tissue perfusion, the volume of blood delivered per unit time (mL/min) perunit of tissue (g). Table 4.8 shows the perfusion of some tissues. Once at the tissue site,uptake or distribution from the blood is driven largely by the passive diffusion of drugacross the epithelial membrane of the capillaries. Because most capillary membranes arevery loose, drugs can usually diffuse from the plasma very easily. Consequently, in mostcases, drug distribution is perfusion controlled. The rate of drug distribution will vary from

TABLE 4.8 Approximate Blood Flow and Perfusion Ratesfor Several Tissues in a Standard 70-kg Male

TissueBlood Flow(mL/min)

Perfusion Rate(mL/min/100 g tissue)

Lung 5400 400Kidney 1230 350Liver 1550 85Heart 250 84Brain 750 55Skeletal muscle 600 2Skin 400 5Fat 250 3

Page 100: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

80 DRUG DISTRIBUTION

one tissue to another, and generally, drugs will distribute fastest to the tissues that have thehigher perfusion rates.

4.3.1 Perfusion-Controlled Drug Distribution

Drug is presented to the tissues in the arterial blood, and any uptake of drug by the tissue willresult in a lower concentration of drug leaving the tissue in the venous blood (Figure 4.6).The amount of drug delivered to the tissue per unit time or rate of presentation of a drug toa tissue is given by

rate of presentation = Q · Ca (4.22)

where Ca is the drug concentration in the arterial blood and Q is the blood flow to thetissue:

rate drug leaves the tissue = Q · Cv (4.23)

where Cv is the drug concentration in the venous blood:

rate of uptake = Q · (Ca − Cv) (4.24)

When drug uptake is perfusion controlled, the tissue presents no barrier for drug uptake,and the initial rate of uptake will equal the rate of presentation:

initial rate of uptake = Q · Ca (4.25)

Thus, it is a first-order process. The value of Ca will change continuously as distributionproceeds throughout the body and as drug is eliminated. When the distribution phase ina tissue is complete, the concentration of drug in the tissue will be in equilibrium withthe concentration leaving the tissue (venous blood). The ratio of these concentrations isexpressed using the tissue blood partition coefficient (Kp):

Kp = Ct

Cv(4.26)

Ca Cv

Ct

Tissue

VtQ Q

FIGURE 4.6 Diagrammatic representation of the delivery of drug in the blood to a tissue. Drugis delivered to the tissue in the arterial blood with a concentration of Ca. The concentration in theemergent blood is Cv. The drug concentration in the tissue is Ct. The blood flow to the tissue is Qand the volume of the tissue is Vt. Thus, the tissue perfusion is Q/Vt.

Page 101: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

RATE OF DRUG DISTRIBUTION 81

where Ct is the tissue concentration. The value of Kp will depend on the binding and therelative affinity of a drug for the blood and tissues. Tissue binding will promote a largevalue of Kp, whereas extensive binding to the plasma proteins will promote a small Kp.

Once the initial distribution phase is complete, the amount of drug in the tissue (At) atany time is

At = Ct · Vt (4.27)

But Ct = Kp · Cv [see equation (4.26)]:

At = Kp · Cv · Vt (4.28)

where At is the amount of drug in the tissue.Equation (4.25) demonstrated that distribution is a first-order process and that the rate of

distribution may be expressed using the first-order rate constant for distribution (kd). Thephysiological determinants of the rate constant for distribution are most easily identified byconsidering the redistribution process, which is governed by the same physiological factorsand has the same rate constant as those for distribution.

If the drug concentration in arterial blood suddenly became zero; the

rate of redistribution = kd · At (4.29)

where kd is the first-order rate constant for distribution. Substituting for At from equation(4.28) into equation (4.29) yields

rate of redistribution = kd · Kp · Cv · Vt (4.30)

But the rate of redistribution is equal to the rate at which the drug leaves the tissue:

rate of redistribution = Q · Cv (4.31)

Thus,

Q · Cv = kd · Cv · Vt · Kp

kd = Q/Vt

Kp

(4.32)

The first-order rate constant for distribution is equal to tissue perfusion divided by thetissue : blood partition coefficient and the corresponding distribution half-life is

t1/2,d = 0.693

kdor t1/2,d = 0.693Kp

Q/Vt(4.33)

As with any first-order process (see Appendix B), it will take about 4 distribution half-livesfor distribution to go to completion in a tissue. Thus, the actual time for this to occur willdepend on the tissue : blood partition coefficient and the tissue perfusion (Q/Vt).

Page 102: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

82 DRUG DISTRIBUTION

These expressions illustrate some important points for perfusion-controlled distribution:

1. The time it takes for distribution to occur is dependent on tissue perfusion. Generally,drugs distribute to well-perfused tissues such as the lungs and major organs fasterthan they do to poorly perfused tissues such as resting muscle and skin.

2. The duration of the distribution phase is also dependent on Kp. If a drug has a highKp value, it may take a long time to achieve equilibrium even if the tissue perfusionis relatively high. If on the other hand, a drug has a high Kp value in a tissue with lowperfusion, it will require an extended period of drug exposure to reach equilibrium.An example of this is the distribution of lipophilic drugs to fat tissue.

3. The amount of drug in tissue at equilibrium depends on Kp (the affinity of the drugfor the tissue) and on the size of the tissue [equation (4.28)]. A drug may concentratein a tissue (high Kp), but if the tissue is physically small, the total amount of drugpresent in the tissue will be low. The distribution of a drug to such a tissue may nothave a strong impact on the plasma concentration of the drug.

4. Redistribution of a drug from the tissues back to the blood is controlled by exactlythe same principles. Thus, redistribution takes less time when the Kp value is low andthe perfusion is high, and will take a long time when the Kp is high and the perfusionis low. For example, highly lipophilic drugs can partition into fat, where they achievevery high concentrations (the Kp value is very high). Fat is poorly perfused, and asa result it takes a long time to achieve equilibrium (assuming continued exposureto the compound; otherwise, equilibrium may never be achieved). Once the drug iswithdrawn, redistribution will be slow and the compound may persist in the fat foran extended period of time. Later, the drug in the fat may be the only drug in thebody, and its redistribution may control the drug’s plasma concentration and the rateof elimination during this period.

4.3.2 Diffusion-Controlled Drug Distribution

The epithelial junctions in some tissues, such as the brain, placenta, and testes, are verytightly knit, and the diffusion of more polar and/or large drugs may proceed slowly. Asa result, drug distribution in these tissues may be diffusion controlled. In this case, drugdistribution will proceed more slowly for polar drugs than for more lipophilic drugs. It mustbe pointed out that not all drug distribution to these sites is diffusion controlled. For example,small lipophilic drugs such as the intravenous anesthetics can easily pass membranes bythe transcellular route and display perfusion-controlled distribution to the brain.

Diffusion-controlled distribution may be expressed by Fick’s law [equation (2.1)]:

rate of uptake = Pm · SAm · (Cpu − Ctu) (4.34)

where Pm is the permeability of the drug through the membrane (cm/h), SAm the surfacearea of the membrane (cm2), Cpu the unbound drug concentration in the plasma (mg/mL),and Ctu the unbound concentration in the tissue (mg/mL).

Initially, the drug concentration in the tissue is very low, Cpu ��� Ctu, so the equationmay be written

rate of uptake = Pm · SAm · Cpu (4.35)

Page 103: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

DISTRIBUTION OF DRUGS TO THE CENTRAL NERVOUS SYSTEM 83

From equation (4.35) it can be seen that under these circumstances, the rate of diffusionapproximates a first-order process.

4.4 DISTRIBUTION OF DRUGS TO THE CENTRAL NERVOUS SYSTEM

Drug distribution to the central nervous system (CNS) is limited by the blood–brain barrier(BBB) and the blood–cerebrospinal fluid (CSF) barrier. These two gatekeepers that guarddrug access to the brain severely hamper the development of drugs for the treatment of suchdiseases of the CNS as cancer, neurological conditions, mood disorders, and infections. As aresult, far fewer drugs are available for the treatment of conditions of the CNS than for non-CNS conditions. It is estimated that about 98% of newly developed centrally acting drugsfail during development, due to their inability to penetrate the BBB and the blood–CSFbarrier (3). Consequently, much research is directed to trying to increase drug access to theCNS. The use of a combination of levodopa and carbidopa for the treatment of Parkinson’sdisease provides an old but creative example of how the BBB cannot only be circumvented,but also used for therapeutic advantage (Figure 4.7).

Parkinson’s disease is characterized by reduced dopamine levels in the substantia nigra.A logical treatment would be the administration of dopamine, but dopamine does not pene-trate the BBB. Instead, Parkinson’s disease is treated using a dopamine precursor, levodopa,which is able to penetrate the BBB. Once inside the CNS it undergoes decarboxylation by

PeripheralCirculation

Central NervousSystem

DOPAMINE

Adverse Effects

LEVODOPA

DOPAMINE

LEVODOPA

DOPAMINE

CARBIDOPA

(-)

No Effect

No Effect

dopa decarboxylasedopa decarboxylase

FIGURE 4.7 Use of the combination of levodopa and carbidopa in the treatment of Parkinson’sdisease. Dopamine, which cannot penetrate the blood–brain barrier (BBB), is administered as itsprecursor levodopa, which can penetrate the BBB. Levodopa is converted to dopamine by dopadecarboxylase. In the peripheral system dopamine produces unwanted adverse effects. Carbidopainhibits dopa decarboxylase but is not able to penetrate the BBB. Thus, it inhibits the formation ofdopamine in the periphery but not in the central nervous system (CNS). Combining carbidopa withlevodopa reduces peripheral side effects to dopamine and increases the amount of dopamine availableto the CNS.

Page 104: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

84 DRUG DISTRIBUTION

dopa decarboxylase to dopamine (Figure 4.7). Dopa decarboxylase is present throughoutthe body, and the formation of dopamine outside the CNS is associated with two prob-lems. First, peripheral dopamine causes unpleasant adverse effects, such as nausea andvomiting. Second, the peripheral conversion reduces the amount of levodopa available tothe CNS. These problems have been overcome by the coadministration of carbidopa, aninhibitor of dopa decarboxylase that cannot penetrate the BBB (Figure 4.7). By includingit in the formulation with levodopa, side effects are reduced as a result of reduced pe-ripheral exposure to dopamine, and the amount of levodopa that is available to the brainis increased.

The BBB is made up of a structural component and a biological component or trans-porter system (4). The structural component consists of the capillary endothelial cells,joined together by tight junctions which prevent paracellular passage for all but extremelysmall molecules (Figure 4.8). This essentially restricts membrane diffusion of drugs to thetranscellular route, which in turn limits diffusion to small lipophilic drug molecules andprevents polar molecules from diffusing into the brain. The biological component consistsprimarily of uptake and efflux transporter systems (Figure 4.8). Drug-metabolizing en-zymes have also been found to be part of the BBB, but their role in controlling drug accessto the brain is not completely understood. Uptake transporters allow essential nutrients togain entry to the CNS and are probably critical for the transport of polar drugs. OAPT1A2has been found on the apical side of the capillary endothelial cells of the BBB (Figure 4.8)and appears to function by transporting drugs from the blood into the brain. Levofloxacinand methotrexate are OATP1A2 substrates, and it has been suggested that the transporterat the BBB may be an important factor in their CNS toxicity (5).

The efflux transporters P-gp, MRP4, and BCRP are also expressed on the apical sideof the capillary endothelial cells (Figure 4.8) (5), where they are believed to extrude drugs

Brain

Blood

Out InOut Out

Tight junctions

E E EUApical Side

Basolateral Side

BCRP P-gp

OAPT1A2

MRP4

FIGURE 4.8 Drug transporters at the blood–brain barrier. Efflux transporters such as BCRP, P-gp,and MRP4 are expressed on the apical side and reduce the CNS exposure of their substrates. TheOATP1A2 uptake transporter is also expressed on the apical side and increases the CNS exposure ofits substrates.

Page 105: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

DISTRIBUTION OF DRUGS TO THE CENTRAL NERVOUS SYSTEM 85

that pass the capillary membrane and pump them back in the systemic circulation. Therole of P-gp in limiting access of drugs to the central nervous system has been demon-strated primarily in studies in mice deficient in P-gp (knockout mice) and other breeds ofanimals that are naturally deficient in P-gp (e.g., collie dogs). These studies have demon-strated greater CNS penetration of many P-gp substrates (see Table 2.3) in the absence ofP-gp. Studies in humans are difficult to perform, but evidence supports a similar role ofP-gp in the human brain. For example, the opioid loperamide normally has no centraleffects and it is available over the counter for the treatment of diarrhea. It is a substratefor P-gp, which is believed to limit its access to the CNS. In support of this theory, coad-ministration of quinidine, an inhibitor of P-gp, results in the emergence of central effectsfrom loperamide (6). Additionally, cerebrospinal fluid concentrations of ritonavir, whichis also a substrate for P-gp, have been shown to increase when it was coadministered witha P-gp inhibitor (ketoconazole). Additional evidence of the involvement of P-gp in vivohas been obtained from positron-emission tomography (PET) studies. For example, PETstudies demonstrated a greater CNS penetration of 11C-labeled verapamil when P-gp wasinhibited by cyclosporine (4). There appears to be overlap in the substrate specificity ofP-gp and BCRP. For example, the CNS uptake of topotecan appears to be limited by bothP-gp and BCRP (5). P-gp and other transporters have been found in the epithelium of thechoroid plexus, which makes up the blood–CSF barrier, but at this time there is limitedinformation on the clinical impact of these transporters.

The development of a new drug intended to treat a condition of the CNS may be halted ifit is found to be a substrate for P-gp. Inhibition of the efflux transporters at the BBB presentsa way to bypass the BBB and potentially increase the number of drugs available to treatconditions of the CNS. Studies in animals have found that the brain uptake of paclitaxel,docetaxel, and imatinib was increased when they were administered with inhibitors of P-gpand BCRP such as cyclosporine and elacridar (4,7). The effectiveness and safety of thisstrategy are being evaluated in humans.

The rate of drug uptake to the CNS is important for intravenous anesthetic inductionagents such as thiopental, propofol, and ketamine. These drugs are all small lipophilicmolecules that easily penetrate the BBB and exert their effects within 0.5 to 2 minutes fol-lowing administration. The magnitude and duration of effect of these drugs is determinedby drug distribution rather than drug elimination (8). In the seconds after an intravenousinjection of thiopental, for example, the drug travels to the heart, through the pulmonarycirculation, and is then pumped through the systemic circulation. This first-pass concentra-tion is very high, and at the BBB the small highly lipophilic drug distributes rapidly to theCNS, where the high perfusion and small volume of the organ result in a very high initialconcentration and a rapid onset of action. As the drug distributes to other, less well-perfusedtissues, the plasma concentration quickly falls and thiopental redistributes from the brainalong its concentration gradient back into the plasma. As the concentration in the CNS falls,the effect of the drug is terminated. Response to intravenous anesthetics is highly dependenton blood volume, cardiac output, and cerebral blood flow. Reduced blood volume and/orreduced cardiac output increases response to these agents by increasing the fraction of thedose that is taken up by the brain. Thus, smaller doses are required in patients in septicshock and in the elderly. Increased cerebral blood flow was found to increase the depthbut not the duration of anesthesia (9). The use of standard doses of thiopental on severelyhypovolemic victims of the Pearl Harbor attack resulted in so many fatalities that it wasreferred to as “an ideal form of euthanasia” (8,10).

Page 106: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

86 DRUG DISTRIBUTION

PROBLEMS

4.1 The values of the volume of distribution of three drugs (A, B, and C) are givenbelow. For each drug, discuss the information provided by the value of this parameter.Address specifically the potential volume into which the drugs distribute, the relativedistribution of the drugs between plasma and tissues, and the binding to the tissuesand the plasma proteins.

Drug A: 0.5 L/kg or 35 L in a 70-kg person

Drug B: 0.143 L/kg or 10 L in a 70-kg person

Drug C: 14.3 L/kg or 1000 L in a 70-kg person

4.2 Theophylline has a volume of distribution of 0.50 L/kg.

(a) A 40-kg child has a theophylline plasma concentration of 10 mg/L. How muchdrug is in the child’s body?

(b) A therapeutic plasma concentration of 12 mg/L theophylline is desired in a110-kg male patient. How much drug will be in his body at this plasma concen-tration?

4.3 A drug binds extensively to the albumin. When it is administered with valproic acid,it is displaced from its protein binding site and the fraction bound in the plasma (fu)increases. How will this affect the volume of distribution?

4.4 Amiodarone has a volume of distribution of 4600 L. If the plasma concentration is1 mg/L:

(a) How much drug is in the body?

(b) How much drug is in the plasma? (Assume that the volume of plasma is 3 L.)

(c) How much drug is in the tissues?

(d) What percentage of the drug in the body is in the tissues?

4.5 Warfarin has a volume of distribution of 8 L. If the plasma concentration is1 mg/L:

(a) How much drug is in the body?

(b) How much drug is in the plasma? (Assume that the volume of plasma is 3 L.)

(c) How much drug is in the tissues?

(d) What percentage of the drug in the body is in the tissues?

4.6 R.S. is a pregnant woman who has been stabilized on phenytoin for 15 years. She isin her third trimester and her phenytoin plasma concentration is found to be 7.0 mg/L.The therapeutic range of phenytoin is 10 to 20 mg/L when the plasma protein bindingis normal (fu = 0.1). R.S.’s albumin level is measured and found to be low, and her fuvalue for phenytoin is estimated to be 0.15. Is the measured phenytoin concentrationtherapeutic, toxic, or subtherapeutic?

4.7 The volume of distribution and protein binding of the three drugs introduced inChapter 3 are listed in Table P4.7. Use this information to summarize their distributioncharacteristics.

Page 107: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc04 JWBS062-Rosenbaum February 17, 2011 8:13 Printer Name: Yet to Come

REFERENCES 87

TABLE P4.7

Drug Vd (L/kg) fu

Lipoamide 4 0.05Nosolatol 2 0.6Disolvprazole 1 �0.95

4.8 Nosolatol binds extensively in skeletal muscle, and the equilibrium tissue/blood ratiois 3. Assume perfusion, controlled distribution. Given that the blood perfusion tomuscle is 0.02 ml/min/g:

(a) What is the distribution half-life for nosolatol in muscle?

(b) How long will it take for the drug to distribute to the muscle?

4.9 The tissue to equilibrium tissue/blood ratio of disolvprazole in heart, kidney, andlungs is 1. Assume perfusion, controlled distribution. Given that the perfusion ofthese tissues is 0.84, 3.5, and 4 ml/min/g, respectively, calculate the time it takes fordistribution to go to completion in these tissues.

REFERENCES

1. Sands, C. D., Chan, E. S., and Welty, T. E. (2002) Revisiting the significance of warfarin protein-binding displacement interactions, Ann Pharmacother 36, 1642–1644.

2. Winter, M. E., and Tozer, T. N. (2006) Phenytoin, In Applied Pharmacokinetics and Pharma-codyamics (Burton, M. E., Shaw, L. M., Schentag, J. J., and Evans, W. E., Eds.) 4th ed., LippincottWilliams and Wilkins, Baltimore.

3. Nicolazzo, J. A., and Katneni, K. (2009) Drug transport across the blood–brain barrier and theimpact of breast cancer resistance protein (ABCG2), Curr Top Med Chem 9, 130–147.

4. Eyal, S., Hsiao, P., and Unadkat, J. D. (2009) Drug interactions at the blood–brain barrier: factor fantasy? Pharmacol Ther 123, 80–104.

5. Urquhart, B. L., and Kim, R. B. (2009) Blood–brain barrier transporters and response to CNS-active drugs, Eur J Clin Pharmacol 65, 1063–1070.

6. Ho, R. H., and Kim, R. B. (2005) Transporters and drug therapy: implications for drug dispositionand disease, Clin Pharmacol Ther 78, 260–277.

7. Breedveld, P., Beijnen, J. H., and Schellens, J. H. (2006) Use of P-glycoprotein and BCRPinhibitors to improve oral bioavailability and CNS penetration of anticancer drugs, Trends Phar-macol Sci 27, 17–24.

8. Henthorn, T. K., Krejcie, T. C., and Avram, M. J. (2008) Early drug distribution: a generallyneglected aspect of pharmacokinetics of particular relevance to intravenously administered anes-thetic agents, Clin Pharmacol Ther 84, 18–22.

9. Ludbrook, G. L., and Upton, R. N. (1997) A physiological model of induction of anaesthesiawith propofol in sheep: 2. Model analysis and implications for dose requirements, Br J Anaesth79, 505–513.

10. Halford, F. J. (1943) A critique of intravenous anesthesia in war surgery, Anesthesiology 21,40–45.

Page 108: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

5DRUG ELIMINATIONAND CLEARANCE

5.1 Introduction5.1.1 First-Order Elimination5.1.2 Determinants of the Elimination Rate Constant and the Half-Life

5.2 Clearance5.2.1 Definition and Determinants of Clearance5.2.2 Total Clearance, Renal Clearance, and Hepatic Clearance5.2.3 Relationships Among Clearance, Volume of Distribution, Elimination Rate Constant,

and Half-Life5.2.4 Primary and Secondary Parameters

5.3 Renal Clearance5.3.1 Glomerular Filtration5.3.2 Tubular Secretion5.3.3 Tubular Reabsorption5.3.4 Putting Meaning into the Value of Renal Clearance

5.4 Hepatic Clearance5.4.1 Phase I and Phase II Metabolism5.4.2 The Cytochrome P450 Enzyme System5.4.3 Glucuronidation5.4.4 Drug–Drug Interactions5.4.5 Hepatic Drug Transporters5.4.6 Kinetics of Drug Metabolism5.4.7 Hepatic Clearance

5.4.7.1 High Extraction: Nonrestrictive Clearance5.4.7.2 Low Extraction: Restrictive Clearance5.4.7.3 Blood Hepatic Clearance and Plasma Hepatic Clearance

5.5 Measurement of Clearances5.5.1 Total Body Clearance5.5.2 Renal Clearance5.5.3 Fraction of the Drug Excreted Unchanged

Problems

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

88

Page 109: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

INTRODUCTION 89

Objectives

The material in this chapter will enable the reader to:

1. Identify the major routes of drug elimination

2. Apply first-order kinetics to drug elimination

3. Understand how clearance is used to express drug elimination

4. Understand the relationships among clearance, volume of distribution, and the rateof elimination

5. Understand the factors that control renal clearance: glomerular filtration, tubularsecretion, and tubular reabsorption

6. Identify the major enzymes involved in drug metabolism

7. Identify the characteristics of metabolism-based drug–drug interactions

8. Know the role of the major drug transporters in hepatic clearance

9. Understand the factors that control hepatic clearance

10. Understand restrictive and nonrestrictive hepatic clearance

11. Distinguish the effects of modifiers of drug metabolism on the pharmacokinetics ofnonrestrictively and restrictively cleared drugs

12. Determine total body and renal clearance from clinical data

Note: This chapter assumes that the reader possesses a knowledge and understanding of thecharacteristics of first-order processes. Readers who are not familiar with this topic shouldreview this material, which is presented in Appendix B.

5.1 INTRODUCTION

Drug elimination is a general term that incorporates all the processes that may be involvedin removing a parent drug from the body. Renal excretion and metabolism (which occursprimarily in the liver) are the major processes of drug elimination. Collectively, they areresponsible for the elimination of over 90% of drugs (1) (Figure 5.1). The excretion of theparent drug into the bile, the third-most-prevalent process, is involved in the elimination ofless than 10% of parent drugs. Other forms of elimination, such as the excretion of parentdrug in the sweat, or exhalation of the drug by the lungs, may exist, but they generallyconstitute very minor pathways.

Renal elimination involves the transfer or excretion of the parent drug from the bloodto the renal tubule, from where it is subsequently eliminated in the urine. Metabolism,which takes place primarily in the liver, involves the conversion of the parent drug toanother molecular species (metabolite) through the action of an enzyme. Metabolites mayundergo further metabolism, renal excretion, and/or biliary excretion. It is important tonote that when the parent drug is altered chemically and converted to a metabolite, thedrug is considered to be eliminated, and that the subsequent fate of the metabolite is notpart of the pharmacokinetic profile of the parent drug. If the metabolite possesses sometherapeutic and/or toxic activity, a separate study of its pharmacokinetics would be ofclinical importance.

Page 110: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

90 DRUG ELIMINATION AND CLEARANCE

Metabolism

BiliaryExcretion

RenalExcretion

FIGURE 5.1 Routes of elimination of the top 200 drugs.

5.1.1 First-Order Elimination

Renal excretion and metabolism are first-order processes for over 90% of all drugs. Elim-ination that is not first-order results in nonlinear pharmacokinetics, which is discussedin Chapter 15. As first-order processes, the rates of excretion and metabolism may beexpressed

rate of excretion = kr · Ab (5.1)

rate of metabolism = km · Ab (5.2)

where kr and km are the first-order rate constants (units, time−1) for renal excretion andmetabolism, respectively, and Ab is the amount of drug in the body.

The rate of overall elimination, which is the sum of all component processes, may beexpressed

rate of elimination = rate of (excretion + metabolism + other processes)

= (kr · Ab) + (km · Ab) + (kother · Ab) = (kr + km + kother) · Ab

−dAb

dt= k · Ab (5.3)

where kother is the sum of the first-order rate constants for any processes other than renalexcretion or metabolism, and k, the sum of all the rate constants, is known as the over-all elimination rate constant. Because most drugs are eliminated by renal excretion ormetabolism, k ≈ kr + km.

Elimination of a drug can also be expressed in terms of its elimination half-life (t1/2)(see Appendix B):

t1/2 = 0.693

k(5.4)

The half-life and the elimination rate constant are both measures of the speed with whicha drug is eliminated. One is the reciprocal form of the other. Under normal circumstancesthey are constants for a drug and their values can be obtained from the literature. A drug

Page 111: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

CLEARANCE 91

that has a large elimination rate constant will have a short half-life and will be eliminatedrapidly. Conversely, a drug that has a small elimination rate constant will have a long half-life and will be eliminated slowly. Clinically, the half-life is used more frequently becauseit has more meaning. For example, if a drug has a half-life of 7 h, it is clear that the plasmaconcentration and the amount of drug in the body will fall by half every 7 h. An eliminationrate constant of 0.1 h−1 provides the same information but has much less direct practicalmeaning.

5.1.2 Determinants of the Elimination Rate Constant and the Half-Life

The half-life and the elimination rate constant are simply parameters of a first-order process.It is important to understand how they are related to physiological processes in the body andthe disposition characteristics of a drug. The half-life and the elimination rate constant areknown as secondary or derived parameters, because they are derived from, or determinedby, two primary drug parameters: the primary parameter for elimination (clearance) andthe primary parameter for distribution (volume of distribution).

Clearance is a measure of the efficiency with which the liver and/or kidney can extract adrug from the plasma and eliminate it from the body. High values of clearance will promotelarge elimination rate constants. However, for the liver and kidney to have the opportunityto eliminate a drug, the drug must be present in the plasma. As a result, a drug’s volumeof distribution, which reflects the relative distribution of a drug between the plasma andthe rest of the body, is also an important determinant of the elimination rate constant andhalf-life. If a drug has a large volume of distribution, a large fraction of the drug in the bodywill be located in the tissues and will not be accessible to the organs of elimination. Thiswill impede the body’s efforts to remove the drug and will promote a long half-life.

� A large clearance and a small volume of distribution will promote rapid eliminationand a short half-life.

� A small clearance and a large volume of distribution will promote slow eliminationand a long half-life.

5.2 CLEARANCE

Clearance is the most important of all the pharmacokinetic parameters. As we show inChapter 11, it controls the average plasma concentration achieved from a particular rate ofdrug administration. If a patient is suspected to have altered clearance for a drug, the dosemay have to be adjusted appropriately or the drug may have to be avoided altogether.

5.2.1 Definition and Determinants of Clearance

Clearance is the primary or fundamental pharmacokinetic parameter for elimination. Itexpresses the collective ability of the organs of elimination to remove drug from plasma.Because drugs are eliminated primarily by renal excretion and/or hepatic metabolism, totalclearance is made up primarily of renal clearance and hepatic clearance, which expressthe ability of the kidney and liver, respectively, to eliminate drug presented to them in theblood. A drug’s clearance is a constant under normal circumstances but can be altered by

Page 112: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

92 DRUG ELIMINATION AND CLEARANCE

diseases of the liver and/or kidney, concomitant medications, and other factors that affectrenal and/or hepatic function.

Clearance, by definition, is the constant of proportionality between the rate of eliminationand the plasma concentration:

rate of elimination = Cl · Cp (5.5)

Although the processes of renal and hepatic clearance are very different, both eliminationprocesses involve the extraction of drug from the blood as it flows through the organ. Asa result, the efficiency of the elimination process is expressed in terms of the volume ofblood cleared of drug. Specifically, the elimination is quantified in terms of the equivalentvolume of the blood that would be cleared completely to match the extraction observed.Take, for example, a solution that flows through an extraction unit at a rate of 1 L/h. If theconcentration entering is 10 mg/L and the concentration leaving after extraction is 5 mg/L,5 mg/L is extracted. This degree of extraction is equivalent to clearing completely 50% ofthe fluid entering. The clearance is 0.5 × 1 L/h = 0.5 L/h. Further, it can be seen that therate of elimination in the example above is Cl · C or 0.5 L/h × 10 mg/L = 5 mg/h.

Clearance is a function of both the blood flow to the organ and the efficiency withwhich the processes within the liver or kidney can eliminate a drug. Its characteristicsand determinants are most conveniently presented using a hypothetical extraction unit torepresent an organ of elimination (Figure 5.2). A solution of drug flows through this unit ata rate of Q(L/h). The concentration of drug in the incoming solution is Ca(mg/L) and theconcentration leaving is Cv (mg/L). As a result of the elimination/extraction, Ca � Cv.

The rate of presentation of the drug to the extraction unit is expressed as

rate of presentation = Ca (mg/L) · Q(L/h) = Ca · Q(mg/h) (5.6)

The rate of which the drug leaves the extraction unit is

rate of exit = Cv (mg/L) · Q (L/h) = Cv · Q (mg/h) (5.7)

Ca Cv

Q Q

D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D DD D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D

FIGURE 5.2 Extraction unit. Drug (D) in solution is extracted as it flows through the unit. Theconcentration of drug entering the extraction unit is Ca (mg/L) and the concentration of drug leavingthe unit is Cv (mg/L). The solution flows through at a rate of Q (L/h).

Page 113: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

CLEARANCE 93

The rate of extraction or elimination is calculated as

rate of extraction = rate of presentation − rate of exit

= (Ca − Cv) · Q(mg/h)(5.8)

The fraction of drug entering that is extracted, the extraction ratio,

E = (Ca − Cv) · Q

Ca · Q= Ca − Cv

Ca(5.9)

The fraction extracted, or fraction of incoming drug that is removed, E, is a measureof the unit’s ability to extract a given drug. It is independent of Ca and is a constant(elimination is first-order) that reflects the ability of the unit to extract drug that is presentedto it.

If E = 1, 100% of the incoming drug is extracted or eliminated; that is, the unitpossesses exceptional ability to extract this particular drug. Thus, clearance in thiscase is Q L/h (all the solution entering is completely cleared of drug). Mathematically,we have

Cl = 1 · Q(L/h) (5.10)

If E = 0.5, 50% of drug entering the unit is removed; that is, the unit possesses moderateability to extract this particular drug. As the solution flows through the unit, the concen-tration of the drug would fall by 50%. Reducing the concentration by 50% is equivalentto removing drug completely from half of the solution as it flows through the extractionunit:

Cl = 0.5Q (5.11)

So if E = 0.3, 30% of the incoming drug is eliminated. This is equivalent to clearing 30%of the solution of drug:

Cl = 0.3Q (5.12)

More generally,

Cl = E · Q (5.13)

Thus, clearance is a function of the ability of the organ to eliminate a given drug (E) andthe blood flow (Q) to the organ. Clearance can never exceed the blood flow to the organ.To make these principles clearer, they can be illustrated further by an example.

Example 5.1 Consider an extraction unit, where Ca = 200 mg/L, Cv = 140 mg/L,and Q = 2 L/h (Figure E5.1). Table E5.1 shows the step-by-step determination of ex-traction ratio, clearance, and rate of elimination for this example and for the generalcase.

Page 114: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

94 DRUG ELIMINATION AND CLEARANCE

200 mg/L 140 mg/L

2 L/h 2 L/h

D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D DD D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D

FIGURE E5.1 Extraction unit. Drug (D) in solution is extracted as it flows through the unit. Theconcentration of drug entering the extraction unit is 200 mg/L and the concentration of drug leavingthe unit is 140 mg/L. The fluid flows through at a rate of 2 L/h.

TABLE E5.1 Step-by-Step Derivation of Expressions for Clearance and the Rate ofElimination

Extraction Unit General Expressiona

Rate of presentation of drug 200 × 2 = 400 mg/h Q · CaRate drug exits 140 × 2 = 280 mg/h Q · Cv

Rate of extraction 400 − 280 = 120 mg (Ca − Cv) · QFraction extracted: extraction ratio (E) 120/400 = 0.3 or 30% (Ca − Cv)/CaClearance (volume of fluid cleared

completely per unit time)2 L/h × 0.3 = 0.6 L/h Q · E

Rate of extraction or elimination 0.6 L/h × 200 mg/L= 120 mg/h

Cl · Ca

aCa and Cv are the concentrations entering and leaving the extraction unit, respectively, Q is the blood flow, E isthe fraction of the drug entering in the solution that is extracted, and Cl is the clearance of the extraction unit.

5.2.2 Total Clearance, Renal Clearance, and Hepatic Clearance

Total clearance (Cl), which is referred to as total body clearance or systemic clearance, issum of all the component clearances:

Cl = Clr + Clh + Clother (5.14)

where Cl is the total body clearance, Clr the renal clearance, Clh the hepatic clearance,and Clother represents any other form of clearance. As we demonstrate later in the chap-ter, total body clearance and renal clearance, are easily measured, and values for spe-cific drugs are usually widely available. In contrast, other forms of clearance, includinghepatic clearance, are much more difficult to quantify. As a result, clearance is oftenexpressed

Cl = Clr + Clnr (5.15)

Page 115: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

CLEARANCE 95

where Clnr, which is the sum of all nonrenal forms of clearance, is the difference betweenmeasured total body clearance and renal clearance.

As discussed previously, hepatic clearance is the major form of nonrenal clearance, andas a result,

Clnr ≈ Clh (5.16)

and the expression for total clearance is often presented as

Cl = Clr + Clh (5.17)

In summary:

1. Clearance is the primary parameter for elimination and expresses the ability of thekidneys and liver to remove drug from the systemic circulation.

2. Clearance is expressed in terms of the equivalent volume of blood or plasma com-pletely cleared of drug per unit time at it passes through the organ.

3. Clearance is dependent on the blood flow to the organ (Q) and on the ability of theorgan to extract the drug from the bloodstream and eliminate it (E).

4. Clearance is the constant of proportionality between the rate of drug elimination andthe plasma concentration.

5. Clearances are additive, and total body clearance is often expressed as the sum of itstwo main component clearances: renal and hepatic clearances.

5.2.3 Relationships Among Clearance, Volume of Distribution, Elimination RateConstant, and Half-Life

Renal excretion and hepatic metabolism are first-order processes, and the rate of eliminationcan be expressed using first-order kinetics:

−dAb

dt= k · Ab (mg/L) (5.18)

But elimination can also be expressed using clearance (Cl):

−dAb

dt= Cl · Cp (mg/L) (5.19)

Combining equations (5.18) and (5.19) gives us

k · Ab = Cl · Cp (5.20)

Page 116: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

96 DRUG ELIMINATION AND CLEARANCE

But Ab = Cp · Vd:

k · Cp · Vd = Cl · Cp

k = Cl

Vd

(5.21)

Since t1/2 = 0.693/k

t1/2 = 0.693Vd

Cl(5.22)

Equations (5.21) and (5.22) show the relationship between the two parameters for the rateof drug elimination and the parameters for elimination (clearance) and distribution (volumeof distribution). The boxes around equations (5.21) and (5.22) indicate they are importantand should be memorized.

5.2.4 Primary and Secondary Parameters

It is very important to distinguish between the dependent (secondary parameters) andindependent (primary parameters) in equations (5.21) and (5.22). Clearance and volume ofdistribution are the primary pharmacokinetics parameters for elimination and distribution,respectively. Primary parameters are independent parameters. Thus, clearance will notchange if distribution changes, and volume of distribution will not change if eliminationchanges. In contrast, the elimination rate constant and the half-life are secondary or derivedparameters. They are dependent on the primary parameters of clearance and volume ofdistribution, and their value will change with changes in elimination (Cl) and/or distribution(Vd). As derived parameters, the elimination rate constant and half-life cannot changeindependent of clearance and volume of distribution, and cannot change either of theseprimary parameters.

It is also important to appreciate that clearance is not a measure of the rate of drugelimination. It is one of the two factors (Vd is the other) that determine how quickly orslowly a drug is eliminated from the body. Many drugs that have high values of clearance areeliminated rapidly (short half-lives). For example, buspirone, didanosine, metoprolol, andmorphine all have high clearances and short half-lives in the region of 1 to 2 h. Similarly,many drugs that have low clearances are eliminated slowly; for example, phenobarbital hasa very low clearance and a half-life of around 4 days. But a drug’s volume of distributionis also a factor. For example, diltiazem and felodipine have similar values for clearance(around 800 mL/min), but felodipine’s elimination half-life is about four times largerthan diltiazem’s because its volume of distribution is about four times larger than thatof dilitazem. Also, chloroquine and amiodarone both have moderate clearances but haveexceptionally long half-lives (∼one month) because they both have very large volumes ofdistribution (∼14,000 and 4,500 L, respectively). In both cases the majority of the drug inthe body is located in the tissues and is inaccessible to the organs of elimination.

Page 117: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

RENAL CLEARANCE 97

GlomerularFiltration

Tubular Secretion Tubular

Reabsorption

Glomerulus

Loop of Henle

Proximal Tubule

Distal Tubule

To Collecting Duct,Bladder and Urine

Renal Tubule

FIGURE 5.3 Simplified diagrammatic representation of the nephron. Three processes in the kidneyparticipate in drug excretion: glomerular filtration in the glomerulus; tubular secretion, which takesplace primarily in the proximal renal tubule; and tubular reabsorption, which occurs primarily in thedistal renal tubule.

5.3 RENAL CLEARANCE

In the kidney the parent drug may be eliminated by excretion into the urine. A drug’srenal clearance is a measure of the efficacy with which the kidney can accomplish thisprocess. The value of renal clearance is determined by blood flow to the part of the kidneyinvolved in drug excretion and on the ability of the kidney to excrete the drug. The abilityof the kidney to excrete the drug is a function of renal physiology and the physicochemicalproperties of the drug.

The nephron is the functioning unit of the kidney, and each kidney possesses about 1to 1.5 million of these. A simplified diagram of the nephron is shown in Figure 5.3. In theglomerulus, plasma water is filtered (glomerular filtration) into the renal tubule, the contentsof which eventually drain into the bladder. However, as the filtrate passes through the tubule,components such as water and dissolved substances, including drugs, may move back andforth across the renal tubule membrane between the blood and the lumen of the tubule.The movement of compounds from the capillaries surrounding the tubules (peritubularcapillaries) into the tubule is referred to as tubular secretion. The movement in the oppositedirection, from the tubules back into the blood, is referred to as tubular reabsorption. Duringtransit through the tubule, much of the plasma water is reabsorbed back into the bloodstream.

5.3.1 Glomerular Filtration

About 20% of the renal blood supply (about 1.2 L/min) is directed to the glomerulus. Inthe glomerulus the blood is subjected to hydrostatic pressure, which forces plasma waterand small solutes, including most drugs, through the capillary membrane and into the renaltubule. The glomerular capillaries are extremely permeable and permit the free passageof neutral molecules below 4 nm in diameter. The filtration of compounds with diametersbetween 4 and 8 nm is inversely proportional to their size. Compounds greater than 8 nm are

Page 118: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

98 DRUG ELIMINATION AND CLEARANCE

excluded completely. The glomerular capillary wall appears to possess a negative charge,which repels negatively charged molecules. As a result, the permeability of anions is lessthan that of neutral molecules. Plasma proteins, including albumin, which has a negativecharge and a diameter of about 7 nm, do not undergo any appreciable filtration (2). Smallproteins such as insulin (MW ∼ 5000 Da) are able to undergo filtration.

The normal glomerular filtration rate (GFR) in the standard 70-kg young adult male isabout 120 to 130 mL/min per 1.73 m2 (about 7.5 L/h or 180 L/day), and it declines withage. If a drug does not bind to plasma proteins and it is small enough to be filtered in theglomerulus, its clearance by glomerular filtration is equal to the glomerular filtration rate(at this point, the filtrate, and any drug it contains, is eliminated and removed from thecirculation). It represents the volume of plasma completely cleared of drug:

ClGF = GFR

where ClGF is clearance by glomerular filtration.However, many drugs bind to the plasma proteins, and bound drug will not be filtered.

For example, if a drug is bound completely (100%), its clearance by glomerular filtrationwill be zero. If, on the other hand, a drug is 70% bound to the plasma proteins (fu = 0.3),only 30% of the drug in the plasma will be filtered and

ClGF = fu · GFR

= 0.3 × 120 = 36 mL/min

In summary:

ClGF = fu · GFR (5.23)

5.3.2 Tubular Secretion

Drug elimination can be augmented further by tubular secretion, a process that results inthe movement of drug from the blood in the peritubular capillaries that surround the tubuleinto the lumen of the tubule. In many cases, this process is accomplished by the action oftransporters, which appear to be concentrated in the proximal tubular cells. Several reviewsof the role of transporters in renal excretion have been written (3–5). A summary of thismaterial is presented below.

Uptake transporters (OAT1, OAT3, and OCT2) are present on the basolateral side of therenal tubular membrane, and efflux transporters (P-gp, MRP2, and MRP4) are present onthe apical side (Figure 5.4). The two transporter systems can work together to facilitatetubular secretion and the excretion of drugs. The uptake transporters initiate the process andmove the drug from the blood into the renal tubular membrane. The efflux transporters canthen conclude tubular secretion by carrying the drug into the tubule. The tubular secretion ofboth fexofenadine, which is a substrate for renal OAT3 and P-gp (6, 7), and that of adefovir,which is a substrate for OAT1 and MRP4, appears to be brought about in this manner.Substrates of the renal OAT transporters include �-lactam antibiotics, diuretics (furosemide,bendroflumethiazide), nonsteroidal antiinflammatory drugs (e.g., aspirin, indomethacin),antiviral drugs (e.g., acyclovir, cidofovir, zidovudine), and methotrexate. There is someoverlap in the substrate specificity of the OATs, but OAT1 appears to be more important

Page 119: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

RENAL CLEARANCE 99

OAT1OCT2

MRP4

BLOOD

MRP2Pgp

OAT3

RENAL TUBULE

Renal Tubule Cell

Apical Side

BasolateralSide U U

E E E

U

FIGURE 5.4 Drug transporters in the human renal tubule. The uptake transporters (U) organiccation transporter (OCT2) and organic anion transporters (OAT1 and OAT3) are found on the basolat-eral side of the renal tubular membrane. These transporters promote excretion by transporting drugsfrom the blood into the tubular cells. The efflux transporters (E), permeability glycoprotein (P-gp)and multidrug resistance–associated protein family (MRP2 and MRP4) can conclude the process bytransporting drugs from the tubular cells into the tubule.

for small molecular mass drugs such as adefovir, and OAT3 appears to be more importantfor larger drugs such as the penicillin G and for sulfate and glucoronide conjugates such asestradiol glucoronide. OAT3 can also carry positively charged molecules, such as the H2-receptor antagonists (famotidine), and molecules with both positive and negative charges,such as fexofenadine. The high drug concentrations in the renal tubular cells produced byuptake transporters have been implicated in promoting the renal toxicity of drugs, such asadefovir, cidofovir, and cephaloridine. This raises the possibility that the renal toxicity ofthese drugs could be reduced by the coadministration of inhibitors of the OAT transporters,such as probenecid.

Substrates for OCT2 include the H2 antagonists (cimetidine, famotidine), cisplatin, andmetformin, which is almost completely eliminated by renal excretion. OCT2-mediateduptake of cisplatin is also believed to be an important factor in its renal toxicity. In supportof this, cimetidine, an OCT2 inhibitor, was found to reduce the severity of the toxicity.

Modification of activity of the uptake transporters by concomitant medications is animportant source of drug–drug interactions for drugs that are primarily cleared renally.Probenecid, which inhibits OAT, reduces the clearance of many drugs, including the peni-cillins, the quinolone antibiotics, and fexofenadine (7). This interaction has been used toadvantage to prolong the half-life of penicllins. In the case of methotrexate, inhibition ofOAT by either probenecid or penicillin is associated with increased methotrexate-inducedbone marrow suppression. The coadministration of metformin with cimetidine, which isboth a substrate and an inhibitor of OCT2, decreases the renal clearance of metformin.

The role of efflux transporters in drug excretion is less clear at this time. P-gp, MRP2,and MRP4 are all present in the renal tubular membrane, and it is probable that theycontribute to the excretion of their substrate drugs. Digoxin is often used as a modelsubstrate for P-gp because it does not undergo any metabolism by CYP3A4. As discussedin Chapter 3, digoxin is also a substrate for intestinal P-gp. In theory, inhibition of P-gp

Page 120: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

100 DRUG ELIMINATION AND CLEARANCE

could increase the absorption of digoxin in the intestine and/or decrease its eliminationin the kidney. Both would increase the serum concentrations of digoxin. Rifampin, whichinduces P-gp, was found to have a greater effect on the intestinal efflux of digoxin than onthe renal efflux. In other studies, the relative contribution of altered renal and/or intestinalP-gp on digoxin’s pharmacokinetics is not clear. From a clinical standpoint, modifiers ofP-gp should be used cautiously with digoxin. Irrespective of the mechanism, inhibitors andinducers may increase and decrease, respectively, the body’s exposure to digoxin. MRP2in the renal tubule appears to be involved in the elimination of methroxeate, and studies inmice suggest that MRP4 is involved in the excretion of hydrocholorothiazide, furosemide,some cephaloridins (ceftizoxime and cefazolin), adefovir, and tenofovir.

5.3.3 Tubular Reabsorption

As the filtrate moves through the proximal tubule and the loop of Henle, water is reabsorbedback into the systemic circulation. When the filtrate reaches the distal tubule, about 80% ofthe filtered water has been reabsorbed. As a result, the concentration of drugs in the filtratebecomes higher than that in the blood in the surrounding capillaries, and drugs diffuse alongtheir concentration gradient back into the systemic circulation. The extent of this tubularreabsorption is controlled by:

1. The drug’s lipophilicity. Lipophilic drugs will readily pass through the tubular mem-brane and be reabsorbed back into the circulation. This highlights the importance ofhepatic metabolism in producing more polar, less lipophilic molecules that are muchless susceptible to tubular reabsorption.

2. pH. Most drugs are weak acids or bases and exist in solution in an equilibriumbetween their ionized and nonionized forms. The pH of the filtrate and the drug’s pKa

control the equilibrium. Since only the nonionized form is able to diffuse through thetubular membrane, urinary pH could, in theory, influence tubular reabsorption. Analkaline pH will favor the ionized form of weak acids, and inhibit their reabsorption:

HA � A− + H+

. . . . . . >...OH−

(5.24)

(The dotted lines represent the direction in which the equilibrium change.)An acidic pH will favor the ionized form of weak bases and inhibit their reabsorp-

tion:

B + H+ � BH+

- - - - - ->

H+

(5.25)

(The dashed line shows the direction of change of the equilibrium.)The pH can be modified by several compounds. Potassium citrate and sodium

bicarbonate increase urinary pH, and ammonium chloride and the thiazide diureticsdecrease pH. In practice, however, variation in urinary pH affects the renal clearance

Page 121: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

RENAL CLEARANCE 101

of only a limited number of drugs because of the constraint placed on possible pHchanges. For example, ammonium chloride treatment produces pH values in the range5 to 6, and sodium bicarbonate treatment leads to pH values in the range 7 to 8. Thislimited pH range will only influence the ionization of weak acids and bases withmidrange pKa values. Amphetamine and phenobarbital are examples of drugs withmidrange pKa values. The tubular reabsorption of amphetamine (weak base) can bedecreased (and its clearance increased) by acidifying the filtrate using ammoniumchloride. Conversely, the tubular reabsorption of phenobarbital (weak base) can bedecreased (and its clearance increased) by making the filtrate more alkaline usingpotassium citrate or sodium bicarbonate.

3. Filtrate/urine flow. The faster the filtrate flows through the tubules, the more lim-ited the opportunity for reabsorption. Thus, tubular reabsorption can be decreased,and renal clearance increased, by the consumption of large amounts of water andcoadministration of diuretics, including ethanol and caffeine.

5.3.4 Putting Meaning into the Value of Renal Clearance

A drug’s overall renal clearance is a function of glomerular filtration, tubular secretion, andtubular reabsorption. It may be expressed as

Clr = ClGF + ClTS − TR or Clr = GFR · fu + ClTS − TR (5.26)

where ClTS is clearance by tubular secretion and TR represents tubular reabsorption.

Example 5.2 Consider a hypothetical drug that does not bind to the plasma proteins(fu = 1) and has a renal clearance of 50 mL/min (Figure E5.2). Use this information to gaininsight into the processes involved in its renal clearance.

ClGF = fu•GFR=120 mL/min

ClTS TR

Glomerulus

Loop of Henle

Proximal TubuleDistal Tubule

Clr = 50 mL/min

Clr = Cl GF + ClTS – TRTR – ClTS = 120 – 50 = 70 mL/min

FIGURE E5.2 Diagrammatic representation of renal clearance of a hypothetical drug. The drug’sclearance by glomerular filtration is 120 mL/min, but its ultimate renal clearance is 50 mL/min. Thus,tubular reabsoprtion must exceed any clearance by tubular secretion by 70 mL/min.

Page 122: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

102 DRUG ELIMINATION AND CLEARANCE

TABLE 5.1 Interpretation of the Renal Clearance of Some Drugs

Drug Clr (mL/min) fu ClGF (mL/min) Comments

Atenolol 170 0.95 114 Some tabular secretionCiprofloxacin 500 0.6 72 Significant tabular secretionMethotrexate 120 0.5 60 Some tabular secretion;

OAT family involved

Solution Assuming that GFR = 120 mL/min, the clearance by glomerular filtration is

ClGF = GFR · fu= 120 × 1 = 120 mL/min

Substituting into equation (5.26) yields

50 = 120 + ClTS − TR

−70 = ClTS − TR

Although it is not possible to know exactly how much tubular secretion and/or reabsorptionoccurs, it is clear from equation (5.26) and Figure E5.2 that tubular reabsorption mustexceed any tubular secretion by 70 mL/min.

Example 5.3 A second drug, which is 20% bound to proteins, has a renal clearance of300 mL/min. What are the relative values of active secretion and tubular reabsorption?

Solution Substituting into equation (5.26) gives us

300 = 120 × 0.8 + ClTS − TR204 = ClTS − TR

For this drug, tubular secretion must exceed any tubular reabsorption by 204 mL/min.Table 5.1 provides the interpretation of the renal clearance of some example drugs.

5.4 HEPATIC CLEARANCE

Lipophilic drugs cannot be eliminated by renal excretion because they undergo tubularreabsorption in the distal tubule. They may undergo glomerular filtration, and this excretioncan be augmented by drug transporters in the proximal tubule, but when they reach thedistal tubule, the concentration gradient between the tubule and the blood, combined withtheir good membrane permeability properties, will result in extensive, if not complete,reabsorption of these molecules. The primary purpose of hepatic metabolism is to createmore hydrophilic molecules that will not undergo tubular reabsorption and thus can beeliminated from the body in the urine. Most drugs are lipophilic in nature and are elimi-nated by metabolism or biotransformation. Over 70% of the 200 drugs most prescribed inthe United States are eliminated by metabolism (1) (Figure 5.1). In the majority of cases,metabolites have greater water solubility than that of the parent drug, but occasionally,metabolites are less soluble. For example, some of the metabolites of the sulfonamides

Page 123: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 103

possess poor water solubility and can crystallize out in the renal tubule, which can leadto serious kidney damage and blood in the urine. Patients taking sulfonamides are advisedto drink a lot of water and to avoid taking any compounds that acidify the urine, such asmethenamine. Although drug metabolism may be referred to as “drug detoxifaction,” this ismisleading because drug metabolites can be pharmacologically active and/or toxic. Thereare many examples of prodrugs, whose pharmacological activity resides with their metabo-lite, including codeine, prednisone, and tamoxifen. Acetaminophen is a classic exampleof a drug that produces toxic metabolites. Normally, acetaminophen’s toxic metabolite isinactivated by glutathione in the liver. In the event of acetaminophen overdose, the amountof the highly reactive toxic metabolite exceeds the capacity of hepatic glutathione, and itreacts with hepatic macromolecules to produce necrosis.

5.4.1 Phase I and Phase II Metabolism

Although many tissues contain some drug-metabolizing enzymes and have the ability tometabolize drugs, the liver has the greatest abundance and greatest variety of these en-zymes. As a result, the liver is the major organ of drug metabolism. It should be noted thatenzymes in other locations can affect a drug’s pharmacokinetic characteristics. Notably,as discussed in Chapter 3, the enzymes in the intestinal enterocytes can cause significantpresystemic extraction and drastically reduce the oral bioavailability of some drugs. Thevarious enzymatic reactions that are involved in hepatic metabolism are categorized aseither phase I or phase II processes. Phase I reactions generally result in small chemicalmodifications of the drug molecule. They often involve oxidation, such as the addition ofa hydroxyl group or the removal of a methyl group, although some reductions can occur.Although several families of enzymes, such as flavin monooxygenase, alcohol dehydroge-nase, adlehyde dehydrogenase, and xanthine oxidase, participate in phase I reactions, thecytochrome P450 (CYP) enzyme system is by far the most important. It is estimated thatthis enzyme system is responsible for the elimination of around 70% of the drugs eliminatedby metabolism (1) (Figure 5.5). In phase II metabolism, the parent drug or the product ofphase I metabolism is conjugated with a polar function, such as a glucoronide, sulfate, orglutathione molecule. The UDP-glucuronosyltransferases (UGTs) are the most importantof the phase II enzymes and are estimated to be responsible for the elimination of around10% of the drugs eliminated by metabolism (1) (Figure 5.5). The parent drug, a phase Imetabolite or more often a phase II metabolite, may be excreted into the bile.

CYPUGT

Other

FIGURE 5.5 Relative contribution of different enzymes involved in the metabolism of thetop 200 drugs. Here CYP is the cytochrome P450 enzyme system and UGT represents UDP-glucuronosyltransferases.

Page 124: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

104 DRUG ELIMINATION AND CLEARANCE

5.4.2 The Cytochrome P450 Enzyme System

The CYP enzyme system is responsible for the elimination of over 50% of drugs usedclinically (1), and as such plays a very important role in drug elimination. This functionis carried out primarily by nine specific enzymes or isoforms within the CYP superfamily.The classification system for the families and subfamilies is based on the similarity of theiramino acid sequencing. These isoforms and their relative abundance in the human liver areshown in Table 5.2. CYP 3A4 is the most abundant and is involved in the metabolism ofthe greatest number of drugs. It is estimated to be involved in the metabolism of almost50% of therapeutic drugs. Many drugs are metabolized by several enzymes, which makesit difficult to rank the enzymes in terms of their involvement in drug elimination. However,about 80% of the oxidative phase I reactions are thought to be mediated by four CYPenzymes: CYP3A4, CYP2D6, CYP2C9, and CYP2C19.

Note that the relative importance of an individual isoform in metabolizing drugs doesnot necessarily parallel its relative abundance. CYP2D6 constitutes less than 5% of totalhepatic CYP, yet is responsible for the metabolism of around 20% of the drugs metabolizedby the CYP system. In contrast, CYP2E1 is relatively abundant but plays little role inthe metabolism of drugs. This isozyme is induced by ethanol and acetone, which may beproduced by diabetic patients, and is thought to be important in producing toxic metabolites,including that of acetaminophen. The activity of some of the individual enzymes is undergenetic control (Table 5.2). Some people within the population may have extremely lowactivity of some of these enzymes, whereas others may have exceptionally high activity.Genetic polymorphism in CYP2D6 and CYP 2C9 is particularly problematic because theymetabolize a large number of drugs. Genetic polymorphism of CYP2C9, for example, is oneof the important factors responsible for the wide variation in dose requirements of warfarin.Patients with low CYP2C9 activity often require lower doses. Tamoxifen is converted toits active antiestrogen metabolite, endoxifen, by CYP2D6, and women with a geneticallydetermined low activity of CYP2D6 have been found to have a greater risk of breast cancerrelapse than do women with normal CYP2D6 levels. CYP3A5, a minor CYP3A isoform, is

TABLE 5.2 Characteristics of the MajorHepatic CYP Enzymesa

CYPEnzyme

RelativeAbundance

(%)

ClinicallySignificant

Polymorphism

3A4 �35 No2D6 �5 Yes2C9 �15 Yes2E1 ∼15 No1A2 �10 No2A6 ∼10 Yes2C19 �5 Yes2C8 ∼5 Yes2B6 �5 Yes

Source: Ref. (8).aThe table shows the relative abundance of the various CYPisoforms and shows which display genetic polymorphism.

Page 125: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 105

only expressed in about 20% of human livers. There are ethnic differences in its expression,and it is more common in Africans. When present, it can make up 17–50% of CYP3A.Generally, it appears to have similar properties to CYP3A4 but some differences have beenobserved. For example, individuals with CYP3A5 may have increased susceptibility tocertain cancers and have higher a clearance of tacrolimus (19).

5.4.3 Glucuronidation

It can be seen from Figure 5.5 that around 10% of drugs are metabolized by the UGTs toglucuronide metabolites. In addition, a large number of the products of phase I metabolismare also metabolized along this pathway. As drug companies strive to develop drugs thatare resistant to the action of the CYP enzymes, it is likely that this route will become evenmore important in the future (10).

Examples of drugs that are metabolized by the UGTs include morphine, zidovudine,olanzapine, valproic acid, and codeine (1). There are several isoforms of UGT, includingUGT1A1, 1A4, 1A6, 1A9, 2B7, and 2B15, with UGT2B7 having the largest number ofdrug substrates. There appears to be wide interindividual variability in the activity of thedifferent isoforms. This could predispose certain individuals to either toxic or subtherapeuticconcentrations of the substrates of the UGTs. Genetic factors and induction by alcohol andsmoking have been identified as factors contributing to their variability (10).

5.4.4 Drug–Drug Interactions

Changes in the hepatic metabolism of one drug (subject drug) as a result of the concomitantadministration of a second medication (perpetrator drug) is the most common cause ofdrug–drug interactions. Perpetrator drugs can act as either inhibitors or inducers of thespecific enzymes involved in drug metabolism. Inhibitors reduce the activity of an enzyme,which in turn can reduce the metabolism of the enzyme’s substrate drugs, and lead toincreased concentrations of these drugs. Inducers increase the amount of an enzyme, whichcan result in a more rapid metabolism of the enzyme’s substrate drugs and decreased plasmaconcentrations. Not surprisingly, given the importance of the CYP, perpetrator drugs thatalter the activity of the CYP enzymes are most frequently involved in causing drug–druginteractions.

Metabolism-based drug–drug interactions can result in very serious, sometimes life-threatening clinical consequences. Over a five-year period (1998–2003) five drugs werewithdrawn from the market because of CYP-based interactions. These included the antihis-tamines terfenadine and astemizole and the gastrointestinal prokinetic agent cisapride. Allthree of these agents are inhibitors of the cardiac human ether-a-go-go related gene (hERG)potassium ion channel, and as a result can cause torsades de pointes (QT prolongation)and serious cardiac arrhythmias. All are eliminated primarily by CYP3A4 metabolism,and inhibitors of CYP3A4, such as ketoconazole, itraconazole, and erythromycin, pre-disposed patients to life-threatening arrhythmias and sudden death. The calcium channelblocker mibefradil was also withdrawn during this period. It is a perpetrator drug thatcaused potent inhibition of several CYP enzymes, including CYP3A4. It caused seriouslife-threatening interactions with many drugs with which it was frequently coadminis-tered, such as �-blockers and dihydropyridine calcium channel blockers. Although seriousdrug–drug interactions can occur with many drugs, they are particularly dangerous fordrugs that have narrow therapeutic ranges, such as warfarin, theophylline, digoxin, and

Page 126: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

106 DRUG ELIMINATION AND CLEARANCE

TABLE 5.3 Substrates, Inhibitors, and Inducers of the Major CYP Enzymes Involved in theMetabolism of Drugs

Enzyme Substrates Inhibitors Inducers

CYP3A4 Benzodiazepines,immunosuppressants,statins (exceptpravastatin androsuvastatin), Ca2+

channel blockers

Macrolides (notazithromycin), azoleantifungals, proteaseinhibitors, cimetidine(weak)

Rifampin,phenobarbital,phenytoin,St. John’s wort

CYP2D6 Debrisoquine, metoprolol,desimipramine,codeine, tamoxifen,flecainide, paroxetine

Quinidine, paroxetine,fluoxetine, ritonavir,bupropion

Not inducible

CYP2C9 S-Warfarin, phenytoin,losartan, fluvastatin

Fluconazole,amiodarone

Rifampin, phenytoin,carbamazepine

CYP1A2 Theophylline, clozapine,tizanidine

Fluvoxamine,fluroquinolones

Smoking, omeprazole

CYP2C19 Proton pump inhibitors,S-mephenytoin,diazepam

Omeprazole,fluvoxamine,fluconazole

Rifampin

CYP2B6 Bupropion, efavirenz,methadone

Ticlopidine, clopidogrel Rifampin,phenobarbital

CYP2C8 Cerivastatin, paclitaxel Gemfibrozil Rifampin

Source: Refs. (8,9).

phenytoin, where inhibitors and inducers can easily alter the delicate balance betweendose and therapeutic plasma concentrations and propel plasma concentrations into toxic orsubtherapeutic ranges. Modifiers of drug metabolism can also have serious clinical conse-quences for prodrugs that rely on metabolism to form the pharmacologically active species.In this case inhibitors will tend to cause subtherapeutic levels of the active drug. For exam-ple, inhibitors of CYP2D6, which converts tamoxifen to the active compound endoxifen,can reduce the effectiveness of tamoxifen and predispose patients to an increased risk ofbreast cancer relapse.

A very large number of therapeutic drugs can modify the activity of the CYP enzymesand cause adverse drug interactions. It is beyond the scope of this book to list all potentialdrug–drug interactions. Table 5.3 provides some examples of substrates, inhibitors, andinducers of the major CYP isoforms, but more comprehensive lists can be found in theliterature (8,11,12) and on Web sites [e.g., (9)]. Drugs that alter the activity of other enzymesinvolved in drug metabolism [e.g., UGTs (1)] can also cause drug–drug interactions.

Many inhibitors act through a competitive mechanism and reduce the available activesites on an enzyme. Inhibition depends on the physical presence of the competitive inhibitor,which has a higher affinity for the enzyme than that of the subject drug. Thus, inhibition isusually apparent as soon as the inhibitor is introduced, although in some cases, the effectmay require the inhibitor concentration to build up to reach steady-state levels, which takesabout 3 to 5 elimination half-lives. Similarly, the inhibitory effect will persist as long asthere is sufficient inhibitor present at the enzyme site. As a result, the effect may last for upto 3 to 5 elimination half-lives (time to eliminate the drug completely) after the inhibitor has

Page 127: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 107

been withdrawn. For drugs with long half-lives, such as amiodarone (t1/2 ≈ 1 month) thiswill lead to prolonged periods (several months) of reduced enzyme activity after therapywith the inhibitor has been stopped. Some inhibitors act by binding irreversibly to theenzyme and destroying it. In many situations the inhibitor drug is converted by the enzymeto a reactive metabolite that subsequently binds covalently to the enzyme. This is knownas mechanism-based inhibition, and examples of inhibitors that act in this way include themacrolide antibiotics clarithromycin and erythromycin, diltiazem, ritonavir, and tamoxifen.The inhibition caused by these agents will persist until a new enzyme is synthesized, whichis dependent on the rate constant or the half-life for enzyme degradation (see Chapter17). The range of the CYP3A4 degradation half-life has been reported to be 14 to 99 h(13), which would suggest that inhibition would persist for several days after the inhibitorhas been eliminated. For example, one week after the administration of erythromycin, itsinhibitory effect was still apparent on the elimination of alfentanil.

Inducers of the enzymes act by stimulating the synthesis of additional enzyme and/orby inhibiting enzyme degradation. As a result, the full effect of these agents is usuallynot apparent immediately, and it may take between a few days to a few weeks for themaximum effect to be observed. The onset of the effect is dependent on the turnover timeof the enzymes and on the time it takes the inducers to reach steady state. Inducers withshort half-lives, such as rifampin, reach steady state more quickly and have faster onsetsof action than do inducers with long half-lives, such as phenobarbital. It will also take anextended period for the enzyme activity to return to normal once an inducer is withdrawnfrom therapy.

Inhibitors and inducers do not alter the pharmacokinetics of all substrates of the affectedenzyme. The susceptibility of a drug to interactions is dependent on several factors, includ-ing the contribution of the affected enzyme to the overall elimination of the drug and theopportunity for other enzymes and pathways of metabolism to compensate for the affectedpathway. Additionally, the perpetrator drug and the subject drug may bind to different activesites on the enzyme. Thus, it is not possible to predict drug interactions based solely onthe enzymes involved in the metabolism of a drug. Several software packages are availablethat allow health care professionals to screen for drug–drug interactions in patients takingmore than one medication.

5.4.5 Hepatic Drug Transporters

The uptake transporters OATP1B1, OATP1B3, OATP2B1, OAT2, and OCT1, and effluxtransporters P-gp, MRPs, and BCRP are all found in the liver, where they participate in thehepatic clearance of drugs (Figure 5.6). The uptake transporters are expressed primarilyon the sinusoidal (blood or basolateral) side of the hepatocyte membrane, where theycan initiate the first step of hepatic clearance by promoting the uptake of drugs into thehepatocyte. The efflux transporters, which reside primarily on the canalicular membrane,are able to conclude hepatic clearance by secreting unchanged drugs, drug metabolites, andconjugates of glucoronide, sulfate, and glutathione into the bile. The action of the varioushepatic transporters is sometimes referred to as phase III metabolism.

The OATPs, mainly OATP1B1, facilitate the uptake of HMG-CoA reductase inhibitors(statins) into the hepatocyte, which is the site of both action and elimination for thesedrugs (14). The uptake of the hydrophilic pravastatin is thought to be the main determinantof its hepatic clearance. Low OATP1B1 activity is also associated with increased plasmaconcentrations of pravastatin, atorvastatin, rosuvastatin, and, to a lesser extent, fluvastatin.

Page 128: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

108 DRUG ELIMINATION AND CLEARANCE

OATP1B1OATP1B3OATP2B1 OAT2 OCT1

MPR3MRP4

P-gp

MRP2

BCRP

BSEP

BLOOD

BILE

U U U

E

E

E

E

E

FIGURE 5.6 Drug transporters in the liver. The diagram shows a hepatocyte. The uptake trans-porters (U), organic anion transporting polypeptide (OATP), organic cation transporter (OCT), andorganic anion transporter (OAT) are found on the basolateral side of the hepatocyte. The efflux trans-porters (E), permeability glycoprotein (P-gp), multidrug resistance–associated protein family (MRP),breast cancer resistance protein (BCRP), and bile salt export pump (BSEP) are found mainly onthe canalicular membrane. Two efflux transporters (MRP3 and MRP4) are found on the basolateralmembrane.

Patients with genetically determined low activity of OATP1B1 were found to have in-creased plasma concentrations of simvastatin acid and to be at increased risk of developingmyopathy (15,16). The OCT1 carrier appears to be important for the uptake of variouscations, including cimetidine, desimipramine, and the hydrophilic antidiabetic drug met-formin (4,14). The uptake facilitates metformin’s access to its site of action in the liverbut does not play a role in metformin’s elimination, as it is eliminated primarily renally.Several protease inhibitors, including ritonavir, are inhibitors of both OCT1 and OCT2(important in the kidney) and could impair the uptake of substrates of these transportersand cause drug interactions (17). Substrates for the OAT transporter include endogenousand exogenous anions, such as para-aminohippurate, nonsteroidal antiinflammatory drugs,and methotrexate (14).

The efflux transporters, located primarily on the canalicular (luminal) membrane ofthe hepatocyte, include BCRP, P-gp and MRP2 (Figure 5.6). Substrates for the effluxtransporters include pravastatin, irinotecan, its active metabolite SN38, and the glucoronideof SN38. Some efflux transporters, including MRP3 and MRP4, are located on the sinusoidalmembrane, where they transport drugs and metabolites back into the bloodstream. Althoughlittle is known about the involvement of BSEP in the elimination of drugs, it is importantfor the biliary secretion of bile salts, and its inhibition can lead to the hepatic accumulation

Page 129: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 109

of bile salts and cholestasis. A number of compounds associated with liver toxicity andcholestasis are inhibitors of BSEP. These include cyclosporine, glyburide, troglitazone,sulindac, and nefazodone (14).

5.4.6 Kinetics of Drug Metabolism

Drug metabolism is an enzymatic process which is initiated when the drug binds to theenzyme in a reversible manner. The enzyme converts the bound drug to a metabolite, andthe metabolite and the enzyme are released. The process is summarized as follows:

D + Ek1−→←−k2

DEk3−→ M + E

where D is the drug concentration, E the free enzyme concentration, DE the concentrationof the drug–enzyme complex, M the metabolite concentration, k1 and k2 the forward andbackward rate constants for the drug–enzyme interaction, and k3 the rate constant for theformation of the metabolite.

Metabolism is an example of a capacity limited process and the rate or velocity (V) ofthe process is given by the Michaelis–Menton equation, which can be expressed as follows:

V = Vmax · Cp

Km + Cp(5.27)

where Vmax is the maximum rate of the process that occurs when the enzyme is saturated, Cpthe plasma concentration of the drug that drives the process, and Km the Michaelis–Mentonconstant, which is a dissociation constant and is equal to (k2 + k3)/k1. Note that when V =Vmax/2:

Vmax

2= Vmax · Cp

Km + Cp

Km + Cp = 2Cp (5.28)

Km = Cp

According to the Michaelis–Menton equation (5.27), the relationship between the drugconcentration and the rate of the process is defined by a hyperbolic curve (Figure 5.7).This curve is explained by the fact that there is only a finite amount of enzyme. At lowdrug concentrations, the free enzyme concentration is high and the rate of metabolism canincrease in proportion to increases in the drug concentration (first-order) (Figure 5.7). Asthe drug concentration increases further, some degree of saturation occurs, and the rate ofmetabolism can no longer increase in proportion to increases in the drug concentrations.Eventually, all the enzyme is occupied by the drug and the rate of metabolism is constant(zero order) and at its maximum level (Figure 5.7).

Thus, at the extremes of low and high drug concentrations, the rate of metabolism is firstand zero order, respectively. This can be expressed mathematically as follows:

� At low drug concentrations: Cp � Km, Km + Cp ≈ Km, and the denominator inequation (5.27) simplifies to

V = Vmax · Cp

Km= k · Cp (5.29)

Page 130: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

110 DRUG ELIMINATION AND CLEARANCE

Rate ofMetabolism (V )

Cp

Vmax

Vmax/2

Km

Cp

V

When Cp << Km the rateof metabolism is first order

When Cp >> Km, theenzyme is saturated,the rate is constant(zero order)

FIGURE 5.7 Plot of the rate of metabolism against plasma concentration of the drug. TheMichaelis–Menton constant (Km) corresponds to the plasma concentration where the rate is halfthe maximum rate (Vmax). At low concentrations (Cp � Km) the rate is first-order. At high concen-trations (Cp � Km) the enzymes are saturated and the process is constant (zero order), and equal toVmax. Between these extremes, the rate is of mixed order and nonlinear.

where k is a constant equal to Vmax/Km. Thus, when Cp � Km, the rate of an enzymaticprocess is first-order (Figure 5.7).

� At high drug concentrations: Cp � Km, Km + Cp ≈ Cp, and the denominator inequation (5.27) simplifies to

V = Vmax · Cp

Cp= Vmax (5.30)

At high concentrations, the enzymes are saturated and the process, which occurs at itsmaximum possible rate (Vmax), is zero order (Figure 5.7). Between these two extremesthe rate is a nonlinear, mixed-order process.

The therapeutic concentrations of most drugs are well below their Km values. As a result,the enzymatic process for most therapeutic drugs follows first-order kinetics (Figure 5.7).Consideration of equation (5.29), and the previous definition of clearance as the constantof proportionality between the rate of elimination and the plasma concentration [equation(5.5)], demonstrates that hepatic clearance for drugs that follow first-order metabolism isgiven by

Clh = Vmax

Km(5.31)

A small number of drugs, however, have therapeutic plasma concentrations that approachor can exceed their Km value. For example, the average Km value for phenytoin, which hasa therapeutic range of 10 to 20 mg/L, is 4 mg/L. Drugs such as phenytoin display nonlinearpharmacokinetics, which we discuss in Chapter 15.

Page 131: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 111

5.4.7 Hepatic Clearance

The model for hepatic clearance (Figure 5.8) is very similar to the extraction unit modelshown in Figure 5.2 and discussed earlier in the chapter. As discussed previously, clearanceis a function of blood flow to an organ and the ability of the organ to extract and eliminatethe drug that is presented to it. Thus, hepatic clearance may be expressed

Clbh = Q · E (5.32)

where Clbh is hepatic blood clearance, Q is hepatic blood flow, and E is the hepaticextraction ratio.

Hepatic blood flow (about 1.3 L/min) represents the maximum value of hepatic clearance.A drug’s hepatic clearance would achieve this value if all the drug entering the liver wasmetabolized (E = 1). According to the venous equilibrium model (18), which is the mostcommonly used model for hepatic clearance, the extraction ratio, or the ability of the liverto extract and metabolize a drug, is determined by:

� The extent to which a drug binds to the plasma proteins because only free drugparticipates in the concentration gradient that drives drug uptake into the hepatocyte.Binding is expressed using the fraction unbound (fu).

� The inherent or intrinsic ability of the liver enzymes to metabolize a drug. This isknown as intrinsic clearance (Clint) and is essentially hepatic clearance in the absenceof any other limiting factors. It is dependent on the affinity of a drug for the enzyme(s)and the amount of enzyme present.

� The hepatic blood flow. The faster the blood flows through the liver, the faster a drugpasses through and the less the opportunity for metabolism.

Ca Cv

LIVER

Q (1.3 L/min) Q (1.3 L/min)

D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D DD D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D D

Rate of Presentation = Q •Ca Rate of Exit = Q •Cv

Rate of Elimination = Q • (Ca - Cv)Fraction of Incoming Drug Extracted (E ) = (Ca - Cv)/CaHepatic Clearance = Q •E

D " M

D " M D " M

D " M

FIGURE 5.8 Diagrammatic representation of hepatic clearance. Hepatic blood flow (Q) is1.3 L/min. The concentration of drug in the arterial blood is Ca and the concentration leavingthe liver is Cv. Drug (D) passing through the liver may be converted to metabolites (M) by hepaticenzymes.

Page 132: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

112 DRUG ELIMINATION AND CLEARANCE

Overall:

E = fu · Clint

Q + fu · Clint(5.33)

where Clint is intrinsic clearance, fu the fraction of the drug in the plasma that is not boundto the plasma proteins, and Q the hepatic blood flow. Substituting for E in equation (5.32)yields

Clbh = Q · fu · Clint

Q + fu · Clint(5.34)

In equation (5.34) all the parameters appear in both the numerator and the denominator. Asa result, it is not easy to see how the individual parameters may affect overall clearance.Fortunately, the equation can be simplified based on the fact that most drugs have either ahigh intrinsic clearance and a high extraction ratio (�0.7) or poor intrinsic clearance and alow extraction ratio (�0.3).

5.4.7.1 High Extraction: Nonrestrictive ClearanceIf a drug has a very high intrinsic clearance, the liver enzymes are able to metabolize thedrug avidly. Under these circumstances, hepatic blood flow is much less than the productof intrinsic clearance and the fraction unbound. Thus, Q � fu · Clint, Q + fu · Clint ≈fu · Clint, and the numerator in equation (5.34) simplifies to

Clbh = Q · fu · Clint

fu · Clint= Q (5.35)

The hepatic clearance of these drugs is not limited or restricted by processes in theliver. The liver possesses an exceptional ability to metabolize these drugs and is able tometabolize all that is presented to it in the blood. As a result, hepatic clearance is limitedonly by the delivery of drug to the liver (hepatic blood flow). If hepatic blood flow increasesor decreases, the hepatic clearance will increase and decrease, respectively. Because theliver has excess capability to metabolize the drug it receives, small changes in the activity ofthe drug-metabolizing enzymes or intrinsic clearance (Clint) will have no impact on overallclearance. Also, protein binding has no impact on clearance because any protein-bounddrug is rapidly stripped from its binding site and metabolized as rapidly as is free drug.

The relationship between the hepatic extraction ratio and hepatic bioavailability wasdiscussed in Chapter 3, where it was shown that

Fh = 1 − E (5.36)

where Fh is the hepatic bioavailability or the fraction of the drug entering the liver thatescapes extraction. It follows that high extraction drugs will have poor oral bioavailabilitydue to extensive first-pass hepatic metabolism. Based on equations (5.35) and (5.36), it canbe shown that

Fh = Q

Q + Clint · fu(5.37)

Page 133: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

HEPATIC CLEARANCE 113

TABLE 5.4 Summary of the Effect of Inhibitors of Drug Metabolismor Increased Plasma Protein Binding on the Pharmacokinetics ofNonrestrictively and Restrictively Cleared Drugsa

Nonrestrictive Cl Restrictive Cl

IV PO IV PO

F N.A. ↑ N.A. ↔Cmax N.A. ↑ N.A. ↔AUC ↔ ↑ ↑ ↑Cl ↔ ↔ ↓ ↓t1/2 ↔ ↔ ↑ ↑aN.A. not applicable; ↔, minimal (if any) change.

From equation (5.37) it can be seen that the hepatic bioavailability of these drugs, in contrastto their hepatic clearance, is sensitive to changes in intrinsic clearance and protein bindingas well as hepatic blood flow.

In summary, the clearance of high-extraction drugs is not influenced by changes in in-trinsic clearance and protein binding but is sensitive to changes in hepatic blood flow. Thesedrugs have high first-pass extraction and poor bioavailability, which is, however, sensitiveto changes in hepatic blood flow, protein binding, and intrinsic clearance. Of these threedeterminants of hepatic clearance, intrinsic clearance is most frequently affected clinically.Inhibitors and inducers of drug metabolism decrease and increase intrinsic clearance, re-spectively. The effects of a reduction in intrinsic clearance on the pharmacokinetics ofnonrestrictively cleared drugs is summarized in Table 5.4. Note these effects will also beobserved by an increase in binding, i.e., reduction in fu. For example, a compound thatinhibited the CYP isoform responsible for the metabolism of diltiazem would not affectdiltiazem’s clearance or its half-life and would not produce clinically important changes indiltiazem’s pharmacokinetics after intravenous administration. The inhibitor would, how-ever, affect the pharmacokinetics of oral diltiazem. It would decrease first-pass extractionand increase bioavailability. This would increase the maximum plasma concentration (Cmax)after an oral dose and increase the area under the plasma concentration-time curve (AUC).

Note that inducers of metabolism and decreased binding (increased fu) alter the sameparameters as those shown in Table 5.4, but the direction of the change is the opposite. Forexample, the displacement of a drug from its plasma protein binding site (increase in fu)would not affect the clearance or half-life of a nonrestrictively cleared drug, but it woulddecrease its bioavailability, Cmax, and AUC after oral administration. Examples of drugsthat undergo nonrestrictive clearance are shown in Table 5.5.

TABLE 5.5 Examples of Nonrestrictively andRestrictively Cleared Drugs

Nonrestrictive RestrictiveClearance (E �0.7) Clearance (�0.3)

Diltiazem DiazepamMeperidine PhenytoinMorphine TheophyllinePropranolol Valproic acidVerapamil Warfarin

Page 134: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

114 DRUG ELIMINATION AND CLEARANCE

5.4.7.2 Low Extraction: Restrictive ClearanceIf a drug has a very low intrinsic clearance, the liver enzymes possess only a poor ornegligible ability to metabolize it. Under these circumstances, equation (5.34) simplifies asfollows: Hepatic blood flow is much greater than the product of intrinsic clearance and thefraction unbound. Thus, Q � fu · Clint, Q + fu · Clint ≈ Q, and the numerator in equation(5.34) simplifies to

Clbh = Q · fu · Clint

Q= fu · Clint (5.38)

The hepatic clearance of these drugs is limited or restricted by the activity of the drug-metabolizing enzymes and by the extent to which the drugs bind to the plasma proteins.The liver is presented with much more drug than it can metabolize. As a result, changesin the liver blood flow (delivery) will not affect the clearance of these drugs, providedthat hepatic blood flow is not reduced to such as extent that it affects the oxygenation andfunction of the hepatocytes. Because hepatic extraction is low, drugs in this class havecomplete hepatic bioavailability. Thus, the effect of changes in hepatic blood flow, proteinbinding, and intrinsic clearance is the same for oral and intravenous administration. Thepharmacokinetics of these drugs are influenced by changes in protein binding and intrinsicclearance.

The effects of a reduction in intrinsic clearance on the pharmacokinetics of restrictivelycleared drugs is summarized in Table 5.4. Note that these effects will also be observed by anincrease in binding (a reduction in fu). Enzyme inhibitors and increased binding will reducethe clearance and increase both the half-life and the AUC of these drugs. Since absorptionis not affected, there will be minimal changes in Cmax, and the changes associated withoral administration will be the same as those after intravenous administration. Note thatinducers of metabolism and decreased binding (increased fu) alter the same parameters asthose shown in Table 5.4, but the direction of the change is the opposite. For example,a displacement of a drug from its plasma protein binding site would increase hepaticclearance and decrease both the elimination half-life and the AUC of a restrictively cleareddrug. Table 5.5 provides some examples of drugs that display restrictive clearance.

5.4.7.3 Blood Hepatic Clearance and Plasma Hepatic ClearanceClearance can be expressed in terms of the blood clearance (volume of blood cleared perunit time) or plasma clearance (volume of plasma cleared per unit time). Blood clearanceand plasma clearance are the constants of proportionality between the rate of metabolism,and the blood and plasma concentrations, respectively:

rate of metabolism = Clbh · Cb (5.39)

rate of metabolism = Clh · Cp (5.40)

where Clh is the hepatic plasma clearance.A drug is presented to the liver in the blood. As such, the liver has the potential to clear

the entire amount of drug in the blood (the drug in the plasma and the drug in the cellularelements, such as the red blood cells). Thus, the extraction ratio is based on blood clearance(Clbh). It is the fraction of all the drug in the blood (not just the plasma) that is extracted.

Clinically, because plasma concentrations, not blood concentrations, are measured rou-tinely, hepatic plasma clearance is used almost universally. It is the clearance that is

Page 135: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

MEASUREMENT OF CLEARANCES 115

TABLE 5.6 Determination of the Extraction Ratio from Blood Clearancea

Drug 1 Drug 2

Clh 800 800Cp/Cb 0.2 1.2Clbh 160 960E 0.12 0.74Type of clearance Restrictive Nonrestrictive

aClh and Clbh are hepatic clearances based on plasma and blood, respectively. Thecalculation of E assumes a hepatic blood flow of 1.3 L/min.

determined in clinical studies, the clearance reported in the literature, and the clearancethat is used to calculate dosing regimens. It is the clearance inferred when the term hepaticclearance is used without qualification.

The relationship between plasma and blood clearance can be derived easily, based onthe expression for the rate of metabolism shown in equations (5.39) and (5.40):

rate of metabolism = Clh · Cp = Clbh · Cb

Clbh = Clh · Cp

Cb

(5.41)

Thus, the relationship between plasma and blood clearance depends on how a drug dis-tributes between plasma and the cellular elements of the blood. If a drug distributes evenlythroughout the blood, the plasma and blood concentrations will be about the same, andplasma and blood clearances will also be about the same. If a drug is unable to penetratethe cells of the blood and is concentrated in the plasma, blood clearance will be greaterthan plasma clearance. Conversely, if a drug concentrates in red blood cells, the bloodconcentration of the drug will be higher than that of the plasma and according to equation(5.41), clearance expressed in terms of the blood will be lower than clearance expressedin terms of the plasma. The plasma/blood concentration ratio for most drugs lies betweenabout 0.3 and 2. For example, the ratio for midazolam, alprazolam, and cylcosporine is0.54, 0.85, and 1.36, respectively.

Clinically, blood clearance has little use. It is, however, important to use blood clearanceto estimate an extraction ratio from a drug’s hepatic clearance. For example, considertwo hypothetical drugs (drugs 1 and 2), each of which has a hepatic clearance (plasmaclearance) of 800 mL/min. Given that hepatic blood flow is about 1.3 L/min, it might betempting to assume they both have high extraction ratios and that both undergo nonrestrictiveclearance. However, as can be seen in Table 5.6, when their respective blood clearancesare determined, drug 1, which concentrates in the cellular elements of blood, in fact has asmall blood clearance and extraction ratio.

5.5 MEASUREMENT OF CLEARANCES

5.5.1 Total Body Clearance

Total body clearance can easily be determined from plasma concentration-time data. Fig-ure 5.9 shows a typical plasma concentration–time profile obtained after administration of

Page 136: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

116 DRUG ELIMINATION AND CLEARANCE

Cp

Timedt

FIGURE 5.9 Plot of plasma concentration against time. The amount of drug eliminated during thevery small period dt is equal to the area under the curve over that period (the shaded area).

an intravenous dose of a drug. The solid circles indicate the measured data points. Considera very small time frame, dt (Figure 5.9). Note that dt has been magnified in the figure forclarity.

The rate of elimination during the period dt is

−dAb

dt= Cl · Cp (5.42)

The amount of drug eliminated during the period dt is

−dAb = Cl · Cp · dt (5.43)

But Cp · dt is the area under the plasma concentration–time curve (AUC) for the period dt:

−dAb = Cl · AUCdt (5.44)

By extension, the amount of drug eliminated from time zero to infinity is the sum of all thesmall areas from time zero to infinity:

Amount eliminated by infinity = Cl ·∞∑

0

AUCdt = Cl · AUC∞0 (5.45)

But the effective dose is eliminated by infinity:

DIV = Cl · AUC∞0 (5.46)

Rearranging yields

Cl = DIV

AUC(5.47)

Page 137: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

MEASUREMENT OF CLEARANCES 117

In the event that a different route of administration is used and/or a salt of a drug isadministered, a more general expression is

Cl = S · F · D

AUC∞0

(5.48)

where S is the salt factor (see Section 6.2) and F is the bioavailability. The box aroundequation (5.48) signifies that it is a very important and useful equation in pharmacokinetics.Familiarity with the equation is strongly recommended.

Thus, clearance is calculated by measuring the AUC. A drug is administered to a subjectand plasma samples are taken at various times and analyzed for the parent drug. A graph ofplasma concentration versus time is constructed. The AUC is most commonly calculatedusing the trapezoidal rule, which uses the measured plasma concentration–time data pointsto divide the curve into a series of trapezoids. The area of each trapezoid is calculated andthe total area is determined from the sum of the area of each trapezoid. The measurementof the AUC using the trapezoidal rule is described in Appendix C, where how to set up anExcel worksheet to determine the AUC and calculate clearance is also described.

5.5.2 Renal Clearance

Several methods are available to determine a drug’s renal clearance. They are all based onthe relationship between the excretion rate and the plasma concentration:

dAu

dt= Clr · Cp (5.49)

where Au is the amount of drug in the urine. One of the most straightforward methods forthe determination of renal clearance is to collect urine samples over successive periods oftime and measure the average rate of excretion of a drug during each period. As shownin equation (5.49), renal clearance is the constant of proportionality between the rate ofexcretion and the plasma concentration. Thus, a plot of the rate of excretion against the cor-responding plasma concentration yields a straight line with a slope equal to renal clearance(Figure 5.10).

Experimentally:

� Urine is collected over successive time periods after a dose.� The urine is analyzed for unchanged drug and the average rate of excretion over each

time period is calculated.� During a collection period, the plasma concentration changes continuously as drug

is excreted. Since excretion is a first-order process, the rate of excretion will alsochange continuously during the collection period. The excretion rate calculated aboverepresents the average excretion rate over a collection period and will correspondmost closely to the plasma concentration at the midpoint of the collection period. Thisplasma concentration is determined.

� The rate of excretion for each time period is then plotted against the plasma con-centration at the midpoint of the collection period. The slope of the line is the renalclearance (Figure 5.10).

Page 138: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

118 DRUG ELIMINATION AND CLEARANCE

Rat

e of

Exc

reti

on, d

Au

/dt

Slope = Clr

Cp

FIGURE 5.10 Determination of renal clearance. Renal clearance is the slope of a plot of the rateof excretion against plasma concentration (Cp).

Example 5.4 The ficticious drug disolvprazole is almost completely eliminated by renalexcretion. A 10-mg dose was administered intravenously to a healthy subject. Urine sampleswere collected over various periods and the plasma concentration was measured at themidpoint of each collection period. The data are given in Table E5.4A. Determine the renalclearance of disolvprazole.

Solution The average rate of excretion over a collection period is calculated as follows:

average rate of excretion = volume of urine · urinary concentration

duration of collection period

For the first collection period in Table E5.4A:

average rate of excretion = 200 mL × 15 �g/mL

1 h= 3000 �g/h or 3 mg/h

The plasma concentration that most closely corresponds to the time when the excretion rateis 3 mg/h is the plasma concentration at the midpoint of the period (Cp at 0.5 h).

TABLE E5.4A

Urine Data Plasma Data

CollectionPeriod

Volume ofUrine (mL)

Urinary Concentration(�g/mL)

Time (h) (Midpoint of UrineCollection Period) Cp (�g/L)

0–1 200 15 0.5 2401–3 180 19.4 2 1423–5 140 12.8 4 715–10 400 3.5 7.5 21

Page 139: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

MEASUREMENT OF CLEARANCES 119

TABLE E5.4B

Urine Data Plasma Data

CollectionPeriod (h)

Rate of Excretion(�g/h) Time (h)

Cp(�g/L)

0–1 3000 0.5 2401–3 1746 2 1423–5 896 4 715–10 280 7.5 21

0

500

1000

1500

2000

2500

3000

3500

0 50 100 150 200 250 300

Exc

reti

on R

ate

(µg/

hr)

Cp (µg/L)

Slope = 12.4 L/h

FIGURE E5.4 Plot of rate of average excretion of disolvprazole against plasma concentration.

The remaining average excretion rates calculated and the data tabulated prior to plottingare listed in Table E5.4B.

The average rate of excretion is plotted against the plasma concentration (Figure E5.4)and yields a slope of 12.4 L/h. Thus, the renal clearance of disolvprazole is 12.4 L/h.

Single-Point Determination of Renal Clearance Under steady-state conditions when theplasma concentration of a drug is constant, renal clearance will be constant and it can beestimated using a simpler single-point determination:

� The steady-state plasma concentration of the drug is determined.� One large urine sample rather than individual urine samples is collected over an

extended period of time (3 to 5 elimination half-lives of the drug).� The average rate of excretion over the entire collection period is calculated.� Renal clearance is calculated by dividing the excretion rate determined above by the

steady-state plasma concentration.

This method should be used only under steady-state conditions, when the plasma con-centration is constant. If the plasma concentration undergoes wide changes during the studyperiod, the rate of excretion will change constantly, and by averaging the rate over an ex-tended period, a bias value of renal clearance will be obtained. This is the method usedto calculate the clearance of creatinine, an endogenous compound produced from musclemetabolism. Under normal circumstances the concentration of creatinine in the blood is

Page 140: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

120 DRUG ELIMINATION AND CLEARANCE

constant. It does not bind to proteins, and undergoes minimal tubular secretion and tubularreabsorption. As a result, its renal clearance approximates the glomerular filtration rate [seeequation (5.26)], and it is used to assess renal function. Because creatinine is eliminatedexclusively by the renal route, its total body clearance is equal to its renal clearance.

Example 5.5 Creatinine clearance is to be determined in a 55-year-old 65-kg femalepatient. Her urine was collected over a 24-h period, and the urinary concentration ofcreatinine determined. The patient’s serum concentration of creatinine was measured at thebeginning of the study. The data are as follows:

Collection period: 0–24 h

Volume of urine collected: 1050 mL

Urinary creatinine concentration: 1.14 mg/mL

Serum creatinine concentration: 1.0 mg/dL

Determine creatinine’s clearance rate.

Solution

Amount of drug excreted in urine: 1197 mg

Average rate of drug excretion: 1197/24 mg/h = 49.9 mg/h

Renal clearance:

excretion rate/Srcr = 49.9 mg/h/(10 mg/L)= 4.99 L/h= 83.1 mL/min

Note: Because it is assumed that serum creatinine is constant, the time of its determinationis not important.

5.5.3 Fraction of the Drug Excreted Unchanged

Recall that the rate of excretion of a drug is

dAu

dt= Clr · Cp (5.50)

where Au is the amount of drug in the urine. Rearranging to obtain an expression for theamount of drug excreted in period dt gives

dAu = Clr · Cp · dt (5.51)

But Cp · dt is the area under the plasma concentration–time curve (AUC) for the period dt:

dAu = Clr · AUCdt (5.52)

Page 141: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

PROBLEMS 121

By extension, the amount of drug excreted from time zero to infinity is the sum of all theareas from time zero to infinity:

Amount excreted by infinity = Clr ·∞∑

0

AUCdt = Clr · AUC∞0 (5.53)

The amount of drug excreted in the urine by infinity is Au∞:

Au∞ = Clr · AUC∞0 (5.54)

Rearranging equation (5.48) gives

S · F · D = Cl · AUC∞0 (5.55)

Dividing equation (5.54) by equation (5.55) yields

fe = Au∞

S · F · D= Clr

Cl(5.56)

where fe is the fraction of the dose excreted unchanged.Equation (5.56) shows that the fraction of the dose excreted unchanged is also the

fraction of total body clearance made up of renal clearance. Thus, if 10% of a dose isexcreted unchanged, renal clearance is 10% of total body clearance. Also, since total bodyclearance is usually the sum of renal and hepatic clearance, 1 − fe will be the fraction oftotal body clearance that comprises hepatic clearance. Continuing the example above, fe =0.1, so 1 − fe = 0.9. Hepatic clearance is 90% of total clearance.

Note that equation (5.54) can be re-arranged to provide an alternative approach to theassessment of renal clearance. Rearranging the equation

Clr = AUC∞0

Au∞ (5.57)

Thus, renal clearance can be determined from the area under the plasma concentration–time curve divided by the amount of drug ultimately excreted unchanged in the urine.

PROBLEMS

5.1 Clearance is defined as a constant for a drug, but as a biological parameter it isexpected to vary somewhat from patient to patient. Beyond this, what factors mightcause a patient to have a clearance value that is either much greater than or muchless than the population average value for a drug?

5.2 Gentamicin is eliminated almost completely in the kidneys (fe � 0.9), and its elim-ination half-life is about 2 to 3 h. Its elimination half-life is determined in a patientand found to be more than double the average value in the population. Suggest somepotential explanations.

Page 142: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

122 DRUG ELIMINATION AND CLEARANCE

5.3 A new drug under development has just undergone phase I trials to determine itshuman pharmacokinetics. These studies have obtained the following information:

Cl = 1000 mL/min Vd = 25 L/kg fu = 0.85

fe = 0.05 Cp/Cb = 1 log D7.4 = 2

The drug is also a potent competitive inhibitor of CYP2D6. Discuss the drug’spharmacokinetics with particular reference to:

(a) Type of clearance: renal/hepatic, restrictive/nonrestrictive

(b) Expected bioavailability

(c) Distribution characteristics

(d) Elimination half-life

(e) Susceptibility to drug interactions from modifiers of drug metabolism

(f) How long would it take after discontinuation of therapy for its inhibition ofCYP2D6 to cease?

5.4 A drug has a hepatic clearance (plasma) of 900 mL/min. It concentrates in red bloodcells, and its Cp/Cb ratio is 0.3. Assume that the hepatic blood flow is 1.3 L/min.

(a) What is the drug’s extraction ratio?

(b) Does this drug undergo restrictive or nonrestrictive clearance?

5.5 Separate clinical studies were conducted on lipoamide, nosolatol, and disolvprazole.Each drug was administered as an intravenous dose to different groups of healthyvolunteers. Blood samples were obtained at various times after the dose. Plasmasamples were prepared and frozen until they could be analyzed for unchanged drug.The data from a single patient for each drug is provided in Table P5.5. Use thedirections provided in Appendix C to create an Excel worksheet to determine theAUC and clearance of lipoamide, nosolatol, and disolvprazole in these subjects.

TABLE P5.5

Lipoamide Nosolatol Disolvprazole

T (h) Cp (�g/L) T (h) Cp (�g/L) T (h) Cp (�g/L)

0 23.8 0 476 0 2860.5 22.1 0.5 462 0.2 2661 20.5 1 448 0.4 2491.5 19 1.5 435 0.8 2162 17.6 2 422 1 2023 15.2 2.5 409 1.2 1885 11.3 5 352 1.6 1637.5 7.82 8 294 2 142

10 5.39 12 231 3 10015 2.57 15 193 5 5020 1.22 20 143 8 1824 0.68 24 112 12 4

Page 143: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

PROBLEMS 123

The doses of lipoamide, nosolatol, and disolvprazole were 10, 100, and 10 mg,respectively.

5.6 A 36-year-old man who is 5 feet 11 inches tall is receiving a constant infusionof a drug. The steady-state plasma concentration is constant at 6 mcg/L. Over a24-h period 1200 mL of urine is collected. The drug concentration in the urine is1.44 �g/mL. Calculate the drug’s renal clearance in this patient. The drug does notbind to plasma proteins. Discuss the processes involved in the renal excretion ofthis drug.

5.7 A drug that binds to the plasma proteins (fu = 0.7) has a total body clearance of500 mL/min, and 75% of a dose is excreted unchanged.

(a) Calculate the drug’s renal clearance.

(b) Calculate the drug’s nonrenal clearance.

(c) Discuss possible processes in the kidney involved in its excretion.

(d) How may its excretion be modified?

5.8 Urine was collected over a 24-h period for a 32-year-old male patient. Serum cre-atinine was measured at the end of the collection period. Using the data given,calculate the creatinine clearance in this man. (Note: Under normal circumstances,serum creatinine is a constant value.) Assume that creatinine undergoes only renalclearance.

Serum creatinine: 1.1 mg/dL

Volume of urine collected: 1500 mL

Urine creatinine concentration: 140 mg/dL

5.9 A 10-mg intravenous dose of disolvprazole was administered to a 70-kg male patientwith slight renal impairment. Urine was collected at various times over a 12-h period,and the plasma concentration was measured at the midpoint of each collection period.The data are given in Table P5.9.

TABLE P5.9

Urine Data Plasma Data

CollectionPeriod (h)

AmountExcreted (mg)

Time (h) (Midpoint ofUrine Collection Period)

Cp(�g/L)

0–1 2.5 0.5 2751–3 3.25 2 1773–9 3.48 6 589–12 0.39 10.5 15

5.10 Lipoamide (fe � 0.01) and nosolatol (fe �0.01) are both subject to drug interactionswhen they are administered with ketoconazole. Use the clearance values calculatedin Problem 5.5 to determine the type of hepatic clearance that these drugs display.Predict how ketoconazole may affect their pharmacokinetics after intravenous andoral administration. Complete Table P5.10.

Page 144: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

124 DRUG ELIMINATION AND CLEARANCE

TABLE P5.10

Lipoamide Nosolatol

Type of hepatic clearance

IV PO IV PO

AUC

F

Cl

t1/2

Note: Answers may be checked in Chapter 10, where data are provided for analysisof drug interaction studies on these two drugs.

5.11 Discuss how the pharmacokinetics of lipoamide, nosolatol, and disolvprazole maybe affected by:

(a) Renal disease

(b) Hepatic disease

REFERENCES

1. Williams, J. A., Hyland, R., Jones, B. C., Smith, D. A., Hurst, S., Goosen, T. C., Peterkin, V.,Koup, J. R., and Ball, S. E. (2004) Drug–drug interactions for UDP-glucuronosyltransferasesubstrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC)ratios, Drug Metab Dispos 32, 1201–1208.

2. Ganong, W. F. (1985) Review of Medical Physiology, 13th ed., Appleton & Lange, East Norwalk,CT.

3. Ho, R. H., and Kim, R. B. (2005) Transporters and drug therapy: implications for drug dispositionand disease, Clin Pharmacol Ther 78, 260–277.

4. Kusuhara, H., and Sugiyama, Y. (2009) In vitro–in vivo extrapolation of transporter-mediatedclearance in the liver and kidney, Drug Metab Pharmacokinet 24, 37–52.

5. Choi, M. K., and Song, I. S. (2008) Organic cation transporters and their pharmacokinetic andpharmacodynamic consequences, Drug Metab Pharmacokinet 23, 243–253.

6. Yasui-Furukori, N., Uno, T., Sugawara, K., and Tateishi, T. (2005) Different effects of threetransporting inhibitors, verapamil, cimetidine, and probenecid, on fexofenadine pharmacokinet-ics, Clin Pharmacol Ther 77, 17–23.

7. Liu, S., Beringer, P. M., Hidayat, L., Rao, A. P., Louie, S., Burckart, G. J., and Shapiro, B. (2008)Probenecid, but not cystic fibrosis, alters the total and renal clearance of fexofenadine, J ClinPharmacol 48, 957–965.

8. Pelkonen, O., Turpeinen, M., Hakkola, J., Honkakoski, P., Hukkanen, J., and Raunio, H. (2008)Inhibition and induction of human cytochrome P450 enzymes: current status, Arch Toxicol 82,667–715.

9. Division of Clinical Pharmacology, Indiana University (2010) P450 Drug Interaction Table.

10. Court, M. H. (2010) Interindividual variability in hepatic drug glucuronidation: studies into therole of age, sex, enzyme inducers, and genetic polymorphism using the human liver bank as amodel system, Drug Metab Rev 42, 202–217.

Page 145: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc05 JWBS062-Rosenbaum February 16, 2011 7:42 Printer Name: Yet to Come

REFERENCES 125

11. Rendic, S. (2002) Summary of information on human CYP enzymes: human P450 metabolismdata, Drug Metab Rev 34, 83–448.

12. Rendic, S., and Guengerich, F. P. (2010) Update information on drug metabolism systems—2009:II. Summary of information on the effects of diseases and environmental factors on humancytochrome P450 (CYP) enzymes and transporters, Curr Drug Metab 11, 4–84.

13. Zhou, Z. W., and Zhou, S. F. (2009) Application of mechanism-based CYP inhibition for pre-dicting drug–drug interactions, Expert Opin Drug Metab Toxicol 5, 579–605.

14. Funk, C. (2008) The role of hepatic transporters in drug elimination, Expert Opin Drug MetabToxicol 4, 363–379.

15. Link, E., Parish, S., Armitage, J., Bowman, L., Heath, S., Matsuda, F., Gut, I., Lathrop, M., andCollins, R. (2008) SLCO1B1 variants and statin-induced myopathy: a genomewide study, N EnglJ Med 359, 789–799.

16. Pasanen, M. K., Neuvonen, M., Neuvonen, P. J., and Niemi, M. (2006) SLCO1B1 polymor-phism markedly affects the pharmacokinetics of simvastatin acid, Pharmacogenet Genomics 16,873–879.

17. Jung, N., Lehmann, C., Rubbert, A., Knispel, M., Hartmann, P., van Lunzen, J., Stellbrink, H. J.,Faetkenheuer, G., and Taubert, D. (2008) Relevance of the organic cation transporters 1 and 2for antiretroviral drug therapy in human immunodeficiency virus infection, Drug Metab Dispos36, 1616–1623.

18. Wilkinson, G. R., and Shand, D. G. (1975) Commentary: a physiological approach to hepaticdrug clearance, Clin Pharmacol Ther 18, 377–390.

19. Daly, A. K. (2006) Significance of the minor cytochrome P450 3A isoforms, Clin Pharmacokinet45, 13–31.

Page 146: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

6COMPARTMENTAL MODELSIN PHARMACOKINETICS

6.1 Introduction

6.2 Expressions for Component Parts of the Dose–Plasma Concentration Relationship6.2.1 Effective Dose6.2.2 Rate of Drug Absorption6.2.3 Rate of Drug Elimination6.2.4 Rate of Drug Distribution

6.3 Putting Everything Together: Compartments and Models6.3.1 One-Compartment Model6.3.2 Two-Compartment Model6.3.3 Three-Compartment Model

6.4 Examples of Complete Compartment Models6.4.1 Intravenous Bolus Injection in a One-Compartment Model with First-Order

Elimination6.4.2 Intravenous Bolus Injection in a Two-Compartment Model with First-Order

Elimination6.4.3 First-Order Absorption in a Two-Compartment Model with First-Order Elimination

6.5 Use of Compartmental Models to Study Metabolite Pharmacokinetics

6.6 Selecting and Applying Models

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand how compartment models can be used to simplify drug disposition

2. Apply the compartmental concept to develop pharmacokinetic models

3. Write equations to express how the amount of drug in a compartment will changewith time.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

126

Page 147: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

EXPRESSIONS FOR ADME 127

6.1 INTRODUCTION

Pharmacokinetics is the study of the time course of drug concentrations in the body. Usually,the plasma concentrations are the main focus of attention, and, as discussed in Chapter 1,a major goal is to express the time course of the plasma concentrations mathematically.

The plasma concentration of a drug can be viewed as the response over time to the doseof a drug that has been administered. The concentration at any time is controlled by the sizeof the dose and the processes of drug absorption, distribution, metabolism, and excretion(ADME), which may all be under way at the same time. Thus, the mathematical equationfor the time course of the plasma concentrations must incorporate the dose and expressionsfor the rates of each of these processes. Drug ADME were discussed in detail in the initialchapters of the book, where expressions for each of their rates were presented. Compart-mental models allow the dose and the individual processes of ADME to be combined in alogical, straightforward manner to create simple models of a complex physiological system.Mathematical expressions for the effective dose and rate of each of the processes in ADMEare first reviewed in this chapter.

6.2 EXPRESSIONS FOR COMPONENT PARTS OF THE DOSE–PLASMACONCENTRATION RELATIONSHIP

6.2.1 Effective Dose

In pharmacokinetic equations, the dose is referred to as a constant because its value isconstant for a given administration. The effective dose is defined as the amount of parentdrug that reaches the systemic circulation. This may differ from the dose administered fortwo reasons:

1. Salt factor. Many drugs are administered as salts. As such, the dose will consist ofpure drug and its conjugate acid or base. For example, phenytoin sodium consistsof 92% phenytoin and 8% sodium. Quinidine sulfate consists of 82% quinidine and18% sulfate. To account for the fact that only a portion (usually a large portion) of adose administered is pure drug, the dose is adjusted using the salt factor (S), definedas the fraction of the salt that is made up of pure drug. Thus, phenytoin sodium andquinidine sulfate have salt factors of 0.92 and 0.82, respectively.

2. Bioavailability factor. When drugs are administered by any route other than directsystemic administration (e.g., intravenous route), a portion of the dose may be lostprior to reaching the systemic circulation. In the case of orally administered drugs,some of the dose can be lost at several points during absorption, including destructionin the gastrointestinal fluid, poor membrane penetration, efflux and/or metabolism inthe enterocyte, and hepatic first-pass extraction. The fraction of the parent drug thatreaches the systemic circulation is the drug’s bioavailability (F).

Overall, the dose reaching the systemic circulation or the effective dose is given by theexpression

effective dose = S · F · D (6.1)

where S is the salt factor and F is the bioavailability or fraction of the dose administered(D) that reaches the systemic circulation.

Page 148: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

128 COMPARTMENTAL MODELS IN PHARMACOKINETICS

Example 6.1 Theophylline is to be administered as its salt aminophylline (S = 0.8) in asustained-release preparation (F = 0.9). What is the effective dose from a 400-mg tablet?

Solution

Effective dose = S · F · D

= 0.8 × 0.9 × 400 mg

= 288 mg

Example 6.2 A drug is administered orally as its hydrochloride salt (S = 0.95). It issusceptible to acid hydrolysis in the stomach and metabolism by CYP3A4 in the enterocytesand the liver. On average, only about 25% of a dose reaches the systemic circulation. Whatis the effective dose from a 5-mg tablet?

Solution

Effective dose = S · F · D

= 0.95 × 0.25 × 5 = 1.19 mg

6.2.2 Rate of Drug Absorption

The process of drug absorption occurs for all routes of administration except for the directsystemic routes, such as intravenous administration. As discussed in Chapter 3, absorptionis usually a first-order process. For orally administered drugs, the rate of absorption is afunction of the amount of drug in the gastrointestinal tract (AGI), and the first-order rateconstant for absorption (ka) and can be expressed

rate of absorption = ka · AGI (6.2)

Absorption may begin as soon as the drug reaches and dissolves in the gastrointestinalfluid. The rate of drug absorption is greatest initially, when there is a lot of drug in thegastrointestinal tract. As drug is absorbed, the amount in the gastrointestinal tract decreasesand the rate of absorption decreases proportionally. Eventually, the entire dose is absorbedand the absorption process ceases. At this point, drug absorption no longer has any influenceon the plasma concentration–time profile. The time for absorption can be estimated as 3 to5 absorption half-lives (where absorption t1/2 = 0.693/ka).

The key pharmacokinetic parameters for absorption are the first-order rate constant forabsorption (ka), and the fraction of the dose that reaches the systemic circulation (F).Because the absorption of a drug can be influenced by the manufacturing process andthe excipients contained in the dosage form, both ka and F are dependent on the specificbrand of the dosage form. Thus, in contrast to other pharmacokinetic parameters, theyare not necessarily constant for a drug. For example, the rate and extent of absorption ofcyclosporine differ in the Noeral preparation from those in the Sandimmune preparation.These parameters (F and ka) can also be affected by anything that influences drug absorption,such as gastrointestinal motility and concomitant medications.

Other models that can be used for the rate of absorption include zero-order absorption,two parallel first-order processes, and a combination of zero- and first-order processes. Ifnecessary, an absorption lag time can be incorporated into the absorption model.

Page 149: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

EXPRESSIONS FOR ADME 129

6.2.3 Rate of Drug Elimination

As soon as drug is present in the plasma, the organs of elimination try to eliminate the drug.Elimination is a first-order process and is dependent on the plasma concentration of the drugand the amount of drug in the body. The rate is higher with higher plasma concentrationsand lower with smaller plasma concentrations.

Elimination can be expressed

rate of elimination = k · Ab (6.3)

where k is the overall elimination rate constant and Ab is the amount of drug in the body.Elimination can also be expressed using clearance:

rate of elimination = Cl · Cp (6.4)

where Cl is the total body clearance.In the equations above, clearance is the primary pharmacokinetic parameter for elimina-

tion, and the overall elimination rate constant is the secondary or dependent pharmacokineticparameter representing the rate of elimination. The elimination rate constant is dependenton the primary parameters of clearance and volume of distribution.

6.2.4 Rate of Drug Distribution

Once in the bloodstream, a drug is able to distribute to the tissues in the body. Some tissueswill take up more drug than others, and there may be certain tissues that the drug cannotaccess at all. As distribution proceeds, the tissue/plasma concentration ratio increasesin each tissue. Eventually, a form of equilibrium is achieved, the distribution phase iscomplete, and the tissue/plasma concentration ratio remains constant. Thereafter the tissueconcentrations and plasma concentration parallel each other. In Chapter 4 it was shown thatthe initial distribution of drugs is generally a first-order process and that tissue perfusion isan important determinant of the first-order rate constant for distribution. Thus, tissues takeup drugs at different rates, depending on their perfusion rates. The physiological approachto pharmacokinetic modeling considers different tissues separately and uses individualtissue perfusion rates to develop models that can then be used to estimate individual tissueconcentrations. A simpler approach is adopted, however, in compartmental modeling, wheretissues are grouped together based on the rate at which they take up the drug. The groups oftissues constitute a compartment. Thus, a compartment is an imaginary unit that consistsof a group of tissues that display similar rates of drug uptake.

There are always some tissues, usually the well-perfused tissues, where drug uptakeis extremely rapid. These tissues are grouped together. There may be another group oftissues, which take up the drug with similar uptake rates but where the rate is slower thanthat of the well-perfused tissues. There may be a third group where uptake is exceedinglyslow, perhaps because the drug partitions slowly into the tissue(s). In practice it has beenfound that the pharmacokinetics of almost all drugs can be described adequately using nomore than three compartments; many can be described using two compartments; and whenpharmacokinetics are applied to specific clinical situations (e.g., to individualize a dose fora patient), the one-compartment model can usually provide a sufficient degree of accuracyto predict the dose/plasma concentration relationship.

Page 150: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

130 COMPARTMENTAL MODELS IN PHARMACOKINETICS

6.3 PUTTING EVERYTHING TOGETHER:COMPARTMENTS AND MODELS

A compartment is an imaginary unit that is used to represent a group of tissues with similarrates of drug distribution. The specific tissues that make up a compartment are unknown,and the number of compartments selected for a particular drug is based on the behavior ofthe plasma concentrations observed over time. A compartment is a homogeneous unit: Thedrug concentration is uniform throughout at all times.

As mentioned earlier, between one and three compartments are needed to producethe typical types of plasma concentration–time profiles observed clinically. One of thecompartments in any model (the only one in a one-compartment model) is the centralcompartment, which always consists of the plasma and tissues that take up the drug rapidly.The concentration of drug in the central compartment is always equal to concentrationroutinely measured in vivo, the plasma concentration. One or two additional compartmentsmay have to be added if, based on the behavior of the plasma concentrations, it appearsthat a significant amount of drug is distributing to some tissues at a slower rate. The organsof drug elimination are well-perfused tissues. Thus, elimination is usually, although notalways, assumed to occur from the central compartment. The fundamental characteristicsof each of the three compartment models are presented below.

6.3.1 One-Compartment Model

The body is viewed as a single compartment (Figure 6.1). All the tissues where a druggoes have very rapid rates of drug uptake. The distribution of the drug to the tissues isso rapid that there is no evidence of it when plasma concentrations are observed overtime. When a drug is administered intravenously, even during the initial period after theinjection, the plasma concentrations appear only to be influenced by drug eliminationand fall monoexponentially (Figure 6.1b). There is no evidence of a distribution phase.

V1=VdA1=AbCp

CentralCompartment

Ek

Ln(Cp)

Time

Straight line relationship betweenLnCp and time indicative of a single first order elimination process

(a) (b)

FIGURE 6.1 The one-compartment model. (a) The one-compartment model consists only of acentral compartment. Distribution to those tissues that the drug can access occurs rapidly and appearsto be an instantaneous process. The compartment is characterized by a volume (V1), the amount ofdrug it contains (A1), and the drug concentration, which is equal to the plasma concentration (Cp). Inthe one-compartment model, A1 is equal to the amount of drug in the body (Ab) and V1 is equal to thedrug’s volume of distribution (Vd). The first-order rate constant for elimination (E) is k. (b) Semilogplot Cp against time after intravenous administration.

Page 151: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

PUTTING EVERYTHING TOGETHER: COMPARTMENTS AND MODELS 131

Thus, for all intents and purposes, drug distribution can be considered to be instantaneous.The compartment is characterized by a volume, the amount of drug it contains, and theconcentration of the drug. In the one-compartment model these quantities are given thesymbols V1, A1, and C1, respectively. However, since the single compartment of the one-compartment model is equivalent to the central compartment, the drug concentration isequal to the plasma concentration (C1 = Cp). In the special case of a one-compartmentmodel, A1 is equal to the amount of drug in the body (Ab). Also V1, which is A1/Cp in theone-compartment model, is the drug’s volume of distribution (Vd).

6.3.2 Two-Compartment Model

The body is viewed as two compartments: the central and peripheral compartments (Fig-ure 6.2). The central compartment consists of the plasma and tissues that take up the drugso rapidly that distribution can be considered to be instantaneous. Other tissues in thebody take up the drug at a similar but slower rate than that for the tissues of the centralcompartment. These tissues constitute the peripheral compartment. The volume, amount,and concentration symbols for the peripheral compartment are qualified by the number 2(e.g., A2 is the amount of drug in the peripheral compartment) (Figure 6.2). Distributionto the peripheral compartment is modeled as a first-order process driven by the amountof drug in the central compartment, and redistribution of the drug from the peripheralcompartment back to the central compartment is also modeled as a first-order process,in this case driven by the amount of drug in the peripheral compartment. The rate con-stants for distribution and redistribution are appropriately labeled k12 and k21, respectively(Figure 6.2). After an intravenous dose, a steep initial fall in the plasma concentration isseen as drug distributes to the peripheral compartment (Figure 6.3a). Many drugs displaytwo-compartmental pharmacokinetics.

6.3.3 Three-Compartment Model

The three-compartment model is an extension of the two-compartment model, where asizable amount of the drug distributes to certain very poorly perfused tissues, such as

V1

A1

Cp

V2

A2

C2k12

k21

CentralCompartment

PeripheralCompartment

k10

FIGURE 6.2 The two-compartment model. Drug distribution occurs very rapidly in the tissuesthat make up the central compartment, but the distribution of a significant amount of the drug to othertissues occurs at a noticeably slower rate. The latter tissues make up the peripheral compartment. Thevolumes (V), amounts (A), and concentrations (C) in each compartment are qualified by 1 and 2 forthe central and peripheral compartment, respectively. Drug concentration in the central compartmentis equal to the plasma concentration. The rate constants for distribution (D) and redistribution (R) arek12 and k21, respectively. The first-order rate constant for elimination (E) is k10.

Page 152: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

132 COMPARTMENTAL MODELS IN PHARMACOKINETICS

Ln(Cp)

Time

Distribution to PeripheralCompartment

Elimination Phase

Redistribution from the Deep Tissue Compartment

Ln(Cp)

Time

(a) (b)

Distribution to PeripheralCompartment

Elimination Phase

FIGURE 6.3 Plasma concentration–time profiles of the two (a) and three-compartment (b) models.The main determinants of the different sections of the profiles are shown. After an intravenous injectiondistribution to the tissues of the central compartment is essentially instantaneous and is not visibleon the profile. Distribution to the peripheral compartment is associated with the steep initial fallin the plasma concentration. This is followed by the elimination phase, where a drug’s eliminationcharacteristics control the slope of the fall. In the three-compartment model a third phase is observedat later times, when the drug in the deep tissue compartment comprises a large fraction of the totaldrug in the body. At this time the redistribution of drug from the deep tissue compartment into theplasma controls the fall in the plasma concentration.

fat and bone, at an extremely slow rate. These tissues make up a third compartment,the deep tissue compartment. The volume, amount, and concentration symbols associatedwith the deep tissue compartment are qualified by the number 3 (Figure 6.4) and thefirst-order distribution and redistribution rate constants are labeled k13 and k31, respectively(Figure 6.4). The three-compartment model has three groups of tissues. The group of tissuesthat comprise the central compartment take up the drug very rapidly. There is no evidenceof this distribution on the plasma concentration–time profile. Tissues in the peripheralcompartment take up the drug more slowly and their distribution is associated with thesteep initial fall in the plasma concentration, the distribution phase (Figure 6.4). Followingthe distribution phase, the fall in the plasma concentrations is controlled by elimination (theelimination phase, Figure 6.3b). The existence of a third group of tissues, the deep tissuecompartment, only becomes apparent after a large fraction of the dose has been eliminatedand plasma concentrations are very low. The initial distribution of drug to tissues in the thirdcompartment is so slow that it has no discernible influence on the plasma concentrations.The redistribution of drug from this compartment is also slow. But at later times, thedrug in this compartment constitutes a very large fraction of the drug in the body, andits redistribution into the plasma controls the fall in plasma concentrations (Figure 6.3b).Digoxin and the aminoglycosides are examples of drugs that display three-compartmentalpharmacokinetics. From the model mapped in Figure 6.4, differential equations can readilybe written for the rate of change of the amounts in each compartment. The solution of thedifferential equations and the clinical application of three-compartmental pharmacokineticscan get quite complex.

Selection of the most appropriate model for a given drug is driven by the characteristicsof the plasma concentration–time profile. This, in turn, is dependent on the distributioncharacteristics of the drug and the timing of the plasma samples.

Page 153: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

EXAMPLES OF COMPLETE COMPARTMENT MODELS 133

V1A1Cp

V2A2C2

k12

k10

k21

V3A3C3

k31

k13Deep Tissue

CompartmentCentral

CompartmentPeripheral

Compartment

FIGURE 6.4 The three-compartment model. Compared to the two-compartment model, the three-compartment model has an additional compartment that is generally made up of very poorly perfusedtissues, such as fat or bone, where distribution proceeds at an extremely slow rate. These tissuescomprise the deep tissue compartment. The volumes (V), amounts (A), and concentrations (C) ineach compartment are qualified by 1, 2, and 3 for the central, peripheral, and deep tissue compartment,respectively. Drug concentration in the central compartment is equal to the plasma concentration.The rate constants for distribution (D) and redistribution (R) to the peripheral compartment are k12

and k21, respectively. The rate constants for distribution (D′) and redistribution (R′) to the deep tissuecompartment are k13 and k31, respectively. The first-order rate constant for elimination (E) is k10.

6.4 EXAMPLES OF COMPLETE COMPARTMENT MODELS

The three basic models discussed above can be used to develop models appropriate for anytype of drug input or route of administration. Once the type of drug input has been added toa model, equations for the rate of change of the amount of drug in any of the compartmentscan readily be written. Applying calculus to develop explicit solutions to these expressions,however, can be quite complex. Some examples of complete models and the equationsderived from them are provided below.

6.4.1 Intravenous Bolus Injection in a One-Compartment Modelwith First-Order Elimination

The model for an intravenous injection in a one-compartment model is shown in Figure 6.5.At time zero, the entire dose is placed into the body:

Ab0 = S · F · D (6.5)

where Ab0 is the amount of drug in the body at time zero. Note that F = 1 for IVadministration. The uptake of drug to those tissues where the drug distributes is so rapidthat it can be considered to be instantaneous. Thus, at time zero the drug is distributeduniformly throughout the homogeneous compartment and

Cp0 = Ab0

V d= S · F · D

V d(6.6)

Page 154: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

134 COMPARTMENTAL MODELS IN PHARMACOKINETICS

Initial Conditions: t = 0, Ab = Ab0 = S•F•D k•Ab

Cl•Cp

Vd

Cp

Ab

FIGURE 6.5 Model for an intravenous injection in a one-compartment model. The amount of drugin the compartment is equal to the amount of drug in the body (Ab), the concentration of drug in thecompartment is equal to the plasma concentration (Cp), and the volume of the compartment (V1) isequal to the drug’s volume of distribution (Vd). Elimination (E) is a first-order process and can beexpressed using the first order elimination rate constant (k) or clearance (Cl). The effective dose isthe product of the dose (D), the salt factor (S), and the bioavailability (F). Note that F = 1 for IVadministration.

The only process that will influence the plasma concentration is elimination, a first-orderprocess that can be expressed

rate of elimination − dAb

dt= k · Ab (6.7)

or

rate of elimination − dAb

dt= Cl · Cp (6.8)

6.4.2 Intravenous Bolus Injection in a Two-Compartment Modelwith First-Order Elimination

The model for an intravenous injection in a two-compartment model is shown in Figure 6.6.At time zero, the entire dose is placed into the body. Distribution throughout the tissuesthat comprise the central compartment is so rapid that it can be considered instantaneous,but distribution to the tissues of the peripheral compartment is slower. At time zero,

A10 = S · F · D and A20 = 0 (6.9)

where A10 and A20 are the initial amounts in the central and peripheral compartments,respectively. Note that F = 1 for IV administration.

By definition, the concentration of drug throughout a compartment is uniform, and bydefinition, the concentration of drug in the central compartment is Cp. Thus, at time zero,

Cp = A1

V1= S · F · D

V1(6.10)

Drug elimination is usually modeled from the central compartment (Figure 6.6). However, ifappropriate in a specific situation, elimination could be modeled to arise from the peripheral

Page 155: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

EXAMPLES OF COMPLETE COMPARTMENT MODELS 135

V1

A1

Cp

V2

A2

C2

CentralCompartment

PeripheralCompartment

Initial Conditions: t = 0,A1 = S•F•D, A2 = 0

A1•k10

A1•k12

A2•k21

FIGURE 6.6 Model for an intravenous injection in a two-compartment model. Elimination (E) isa first-order process (rate constant, k10) that occurs from the central compartment. Distribution (D)and redistribution (R) are also first-order processes with rate constants of k12 and k21, respectively.Note that F = 1 for IV administration.

compartment. Elimination in the two-compartment model is driven by the amount of drugin the central compartment (A1), and the elimination rate constant is k10 (Figure 6.6).

The amount of drug in the central compartment (A1) is the initial focus of a mathematicalexpression based on the model. Once an expression has been obtained, it can be convertedto plasma concentration using the volume of the central compartment (V1). The rate ofchange of A1 with time =

d A1

dt= rate of inputs − rate of outputs

= rate of redistribution − (rate of elimination + rate of distribution)

= (k21 · A2) − (k10 · A1 + k12 · A1) (6.11)

The rate of change of A2 with time is

d A2

dt= rate of inputs − rate of outputs

= rate of distribution − rate of redistribution

= (k12 · A1) − (k21 · A2) (6.12)

6.4.3 First-Order Absorption in a Two-Compartment Modelwith First-Order Elimination

The model for first-order absorption in a two-compartment model is similar to the modeldescribed above except that it incorporates drug absorption, which is represented by first-order drug input into the central compartment (Figure 6.7). The initial conditions of thesystem are also different:

A10 = 0 and AGI0 = SFD

Page 156: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

136 COMPARTMENTAL MODELS IN PHARMACOKINETICS

V1A1Cp

V2A2C2

Initial Conditions: t = 0,AGI = S•F•D, A1 = 0, A2 = 0

A1•k10A1•k12

A2•k21

AGI•ka

Outside the BodyAGI The Gastrointestinal

Tract

Inside the Body

FIGURE 6.7 Model for first-order absorption in a two-compartment model. The symbols are thesame as those defined in Figure 6.5. The gastrointestinal tract, which is outside the body, is symbolizedas a compartment, and AGI is the amount of drug is contains. The rate of absorption (A) is a first-orderprocess controlled by AGI and the first-order rate constant for absorption (ka). The bioavailabilityfactor (F) is the fraction of the dose that reaches the systemic circulation. S, F, and D are as definedin Figure 6.6.

where AGI0 is the amount of drug in the gastrointestinal tract at time zero. Note that theinitial amount in the gastrointestinal tract is considered to be the effective dose. The amountof drug [S · D · (1 − F)] that never reaches the systemic circulation is not included in theinitial amount in the gastrointestinal tract.

An equation for the rate of change of the amount of drug in the central compartment caneasily be obtained:

dA1

dt= rate of inputs − rate of outputs

= (rate of absorption + rate of redistribution)

− (rate of elimination + rate of distribution)

= [(ka · AGI) + (k21 · A2)] + [(k10 · A1) + (k12 · A1)] (6.13)

Similar expressions can be derived for the way in which the amounts in the gastrointestinaltract and peripheral compartment change over time.

6.5 USE OF COMPARTMENTAL MODELS TO STUDYMETABOLITE PHARMACOKINETICS

In addition to simplifying drug distribution, compartment models can also be used tostudy the pharmacokinetics of a drug’s metabolite. The distribution of the metabolite

Page 157: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

SELECTING AND APPLYING MODELS 137

V1AbCp

VmAmCm

Initial Conditions: t = 0, Ab = S•F•D, Am = 0

Ab•(1–fm)•k Ab•( fm•k) Am•km

Parent Drug Metabolite

FIGURE 6.8 Compartment model for a drug and its metabolite after the intravenous administrationof the parent drug. The parent drug is assumed to follow one-compartmental pharmacokinetics.Elimination (E) is a first-order process with a rate constant of k. A fraction of the parent drug (fm)is converted to the metabolite with a rate constant equal to k · fm. The rate constant for the otherelimination processes for the parent drug is k · (1 − fm). The metabolite is also assumed to followone-compartmental pharmacokinetics. The plasma concentration of the metabolite is Cm, its volumeof distribution is Vm, and the amount of metabolite in the compartment at any time is Am. Themetabolite is eliminated (Em) by a first-order process with a rate constant of km. S, F, and D for theparent drug are as defined in Figure 6.6.

can be modeled using either a single- or a multiple-compartment model. The metabolitecompartment(s) can then be attached to the compartmental model for the parent drug(Figure 6.8). Metabolites are formed primarily by a first-order process in the liver. Since theliver is usually part of the central compartment, the central compartment of the metaboliteis usually attached to the central compartment of the parent drug. Figure 6.8 shows anintegrated drug–metabolite model in which the distribution of both the parent drug andmetabolite are represented as one-compartment models. The model applies to intravenousadministration of the parent drug.

Equations can be developed for the rate of change of the amount of drug and metabolite:

dAb

dt= −k · Ab

dAm

dt= [(k · f m) · Ab] − (km · Am)

(6.14)

where Ab and Am are the amounts of parent drug and metabolite, respectively, k and km arethe overall elimination rate constants of the parent drug and metabolite, respectively, andfm is the fraction of the parent drug converted to the metabolite. Note that the rate constantfor the formation of the metabolite is k · fm.

6.6 SELECTING AND APPLYING MODELS

Models are selected for a given drug by comparing the behavior of the model to thebehavior of the data observed. Specifically, model-predicted plasma concentrations arecompared to actual plasma concentrations measured at various times after a dose. Thisprocess is performed using robust mathematical and statistical procedures that providenumerical values of how well the data fit a model. Various models are tested and the

Page 158: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc06 JWBS062-Rosenbaum February 16, 2011 7:46 Printer Name: Yet to Come

138 COMPARTMENTAL MODELS IN PHARMACOKINETICS

simplest model that predicts the observed data adequately is selected. Once a satisfactorymodel has been found, it can be used to summarize a drug’s properties and to estimate themodel parameters (i.e., clearance, volume of distribution, etc.). The model can also be usedto perform simulations to observe drug behavior and plasma concentrations in situationsthat have not yet been studied. For example, plasma concentrations can be simulated usingvarious types of drug administration; parameters derived from single doses can be usedto simulate steady-state conditions after multiple doses of a drug; and different doses anddosing intervals can be used to try to determine optimum regimens to target desired plasmaconcentrations.

PROBLEMS

6.1 Draw a two-compartment model for a drug that is to be administered orally and isthought to undergo constant zero-order absorption (rate = k0). Assume that absorptionand elimination are into and out of the central compartment, respectively. Write anexpression for how the amount of drug in the central and peripheral compartmentschanges with time. What are the initial conditions of the system?

6.2 A drug follows one-compartment pharmacokinetics with first-order elimination withan overall elimination rate constant k. About 80% of the drug is excreted unchangedin the urine and 20% is metabolized to a metabolite. The metabolite also followsone-compartmental pharmacokinetics and has an overall elimination rate constantof km. About 70% of the metabolite is excreted unchanged into the urine and 30%is converted back to the parent drug. Draw an integrated parent drug–metabolitecompartmental model for intravenous administration of the drug. Write expressionsfor the rate of change of the parent drug (Ab) and metabolite (Am). What are the initialconditions of the system?

RECOMMENDED READING

1. Bourne, D. (1995) Mathematical Modeling of Pharmacokinetic Data, Technomic, Lancaster, PA.

2. Gibaldi, M., and Perrier, D. (1982) Pharmacokinetics, 2nd ed., Marcel Dekker, New York.

3. Wagner, J. G. (1993) Pharmacokinetics for the Pharmaceutical Scientist, Technomic, Lancaster,PA.

Page 159: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

7PHARMACOKINETICS OF ANINTRAVENOUS BOLUS INJECTIONIN A ONE-COMPARTMENT MODEL

7.1 Introduction

7.2 One-Compartment Model

7.3 Pharmacokinetic Equations7.3.1 Basic Equation7.3.2 Half-Life7.3.3 Time to Eliminate a Dose

7.4 Simulation Exercise

7.5 Application of the Model7.5.1 Predicting Plasma Concentrations7.5.2 Duration of Action7.5.3 Value of a Dose to Give a Desired Initial Plasma Concentration7.5.4 Intravenous Loading Dose

7.6 Determination of Pharmacokinetic Parameters Experimentally7.6.1 Study Design for the Determination of Parameters7.6.2 Pharmacokinetic Analysis

7.7 Pharmacokinetic Analysis in Clinical Practice

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand the derivation of a general equation that describes how a drug’s plasmaconcentration at any time is related to the dose and the drug’s pharmacokineticparameters

2. Apply the equation to determine plasma concentrations at any time after a dose andto determine doses necessary to achieve specific plasma concentrations

3. Understand how a drug’s pharmacokinetic parameters influence the plasmaconcentration–time profile

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

139

Page 160: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

140 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

4. Analyze plasma concentration–time data to obtain a drug’s pharmacokinetic param-eters

5. Apply the model to clinical situations

7.1 INTRODUCTION

The intravenous injection of a drug into a peripheral vein is the most common form of directsystemic drug administration. This route can provide immediate therapeutic plasma con-centrations, and because it has 100% bioavailability, the intravenous route produces muchmore predictable concentrations than those for other routes. Consequently, intravenous ad-ministration is used when an immediate effect is desired and/or it is important that the dosebe administered with a high degree of precision. Poor bioavailability from other routesis another reason that drugs may be given intravenously. Many drugs are administeredintravenously, including antiarrhythmic drugs, narcotic analgesics, certain antibiotics, andanticancer drugs.

There are two main types of intravenous administration: the intravenous bolus injectionand the intravenous infusion. The intravenous bolus injection involves administration ofthe entire dose at one time. Because a rapid injection can produce undesirable high plasmaconcentrations immediately after the injection, the injection is often given over a periodof one minute or more. For an intravenous infusion, the administration period is extendedover a much more prolonged period, during which the drug is administered at a constantrate. Relatively short administration periods of around 0.5 to about 1 to 2 h can be usedto give intermittent doses of a drug at regular intervals (e.g., every 8 or 12 h). This isreferred to as an intermittent infusion. Alternatively, in hospitalized patients, an infusionmay be continued over an extended period of up to several days. During this period thepatient receives the drug at a constant rate. This is referred to as a continuous infusion. Thepharmacokinetics of intravenous infusions is discussed in subsequent chapters.

The intravenous injection of a single dose of a drug provides the simplest pharma-cokinetic profile because there is no absorption and ongoing drug input. For simplicity itwill be assumed that the entire dose is injected instantaneously, even though, clinically, asdiscussed above, the dose is usually administered over a period of a minute or more.

7.2 ONE-COMPARTMENT MODEL

The one-compartment model, the simplest of the pharmacokinetic models, applies whena drug distributes very rapidly throughout its total distribution volume. As a result, all thetissues that take up the drug achieve equilibrium with the plasma extremely quickly. Indeed,distribution is so rapid that there is no evidence of it on the plasma concentration–timeprofile. Thus, after an intravenous injection of a drug, plasma concentrations appear tobe influenced only by first-order elimination, and the plasma concentration–time profilepresents as a smooth monoexponential fall on a linear scale, and as a straight line on asemilogarithmic scale (Figure 7.1).

When a drug is administered (t = 0), the initial amount of drug in the body (Ab0) isequal to the effective dose (S · F · D). Drug distribution from the plasma to the tissues isessentially instantaneous, and the initial plasma concentration (Cp0) is equal to Ab0/Vd, or

Page 161: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

ONE-COMPARTMENT MODEL 141

time

ln C

p

Cp

time

(a) (b)

FIGURE 7.1 Linear (a) and semilogarithmic (b) plots of Cp against time after intravenous admin-istration in the one-compartment model.

S · F · D/Vd. The body is represented by a single imaginary compartment. By definition thecompartment is homogeneous, and at any given time the drug concentration throughout isexactly the same. By definition the concentration of drug in the compartment is equal tothe plasma concentration (Cp). The amount of drug in the compartment (A1) is equal to theamount of drug in the body (Ab). The volume of the compartment (V1) is equal to

V1 = A1

C1= Ab

Cp

But by definition,

Ab

Cp= Vd

Thus for a one-compartment model,

V1 = Vd

To help understand how compartments are used in pharmacokinetics, it is useful to bear inmind how the compartmental characteristics are related to true body characteristics. Theserelationships are summarized in Table 7.1.

TABLE 7.1 Comparison of True Body Characteristics and Those of the Single Compartment

Characteristic Body Compartment

Ab Amount of drug in the body Amount of drug in the compartmentCp Plasma concentration Compartment concentrationDrug concentration Varies from tissue to tissue Constant throughoutVd Ratio: Ab/Cp Volume of the compartment

Page 162: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

142 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

7.3 PHARMACOKINETIC EQUATIONS

7.3.1 Basic Equation

The basic model for an intravenous injection is shown in Figure 7.2. This model can beused to write expressions for how the amount of drug in the body changes over time. Oncean expression for amount of drug in the body has been derived, it can converted to anexpression for the plasma concentrations using the drug’s volume of distribution (Cp =Ab/Vd). Thus, the starting point,

dAb

dt= rate of inputs – rate of outputs

dAb

dt= 0 − k · Ab (7.1)

Integrate equation (7.1) from zero to infinity:

Ab = Ab0 · e−kt (7.2)

Recall that the function e−kt decays from 1 to zero over time (see Appendix A). During thisperiod, Ab decays from Ab0 to zero. The speed of decay is governed by k. The larger thevalue of k, the faster is the decay. Recall Ab0 = S · F · D,

Ab = S · F · D · e−kt (7.3)

and Cp = Ab/Vd,

Cp = S · F · D

Vd· e−kt or Cp = Cp0 · e−kt (7.4)

where Cp0 = S · F · D/Vd.

Initial Conditions: t = 0,Ab = Ab0 = S•F•DCp = Cp0 = S•F•D/Vd k•Ab

Cl•Cp

VdCpAb

FIGURE 7.2 Model for intravenous injection in a one-compartment model. S is the salt factor,F the bioavailability, D the dose, Ab the amount of drug in the body, Cp the plasma concentration,Vd the apparent volume of distribution (Ab/Cp), Cl the clearance, and k the overall elimination rateconstant. Note that F = 1 after intravenous administration.

Page 163: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

PHARMACOKINETIC EQUATIONS 143

Also, since k = Cl/Vd [see equation (5.21)], all of the equations above could be rewrittensubstituting Cl/Vd for k. For example,

Cp = S · F · D

Vd· e−(Cl/Vd) · t (7.5)

In summary, after an intravenous injection in a one-compartment model, Cp and Ab, whichare always directly proportional to each other in a one-compartment model, decay fromtheir initial values in a first-order manner:

� Ab decays over time from S · F · D to zero.� Cp decays from S · F · D/Vd to zero.� The rate of decay is controlled by k; the larger k is, the more rapid is the decay.

7.3.2 Half-Life

The half-life is the time required for the plasma concentration or the amount ofdrug in the body to fall by 50%. Elimination is a first-order process, and as shown inAppendix B,

t1/2 = 0.693

k

7.3.3 Time to Eliminate a Dose

The plasma concentration and the amount of drug in the body are affected only by elim-ination, which is a first-order process. As a result, the number of half-lives required toeliminate any fraction of the original dose are the same as those needed to complete anyfraction of a first-order process (see Appendix B). These are shown in Table 7.2.

TABLE 7.2 Number of Half-Livesto Eliminate a Certain Fraction ofthe Original Dose

Fraction ofDose Eliminated

Number ofHalf-Lives

0.10 ∼ 16

a

0.20 ∼ 13

a

0.50 10.90 3.30.95 4.40.99 6.6

aApproximate values.

Page 164: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

144 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

7.4 SIMULATION EXERCISE

Open the model “IV Bolus Injection in a One-Compartment Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter7

Default settings for the model are dose = 100 mg; Cl = 4.6 L/h, and Vd = 20 L.

1. Review the objectives and the “Model Summary” page.

2. Explore the model.

3. Go to the “Cp–Time Profile” page. Perform a simulation using a default dose of100 mg and observe the shape of the Cp–time profile on the linear and semilog-arithmic scales.Observe:� The Cp falls monoexponentially. It is influenced only by first-order elimination:

Cp = Cp0 · e−kt.� A linear relationship exists between ln Cp and time: ln Cp = ln Cp0 − kt.

These plots are observed for drugs that distribute in a very rapid, essentiallyinstantaneous manner.

� Cp0 = 5 mg/L = 100 mg20 L = S·F ·D

Vd . Cp0 is inversely proportional to Vd, t1/2 = 3 h,

and k = 0.693/t1/2 = 0.231 h−1.

4. Go to the “Effect of Dose” page. Use doses of 50, 100, and 200 mg to observehow dose influences Cp0, Cp at any time, the slope of the fall in Cp, and t1/2.Summarize the answers in Table SE7.4.Observe:� Cp0 is proportional to the dose. Cp at any time is proportional to the dose. The

slope and t1/2 are not influenced by the dose.

5. Go to the “Effect of Clearance” page. Use Cl values of 2.3, 4.6, and 9.2 L/h toobserve how clearance influences Cp0, Cp at any time; slope of fall in Cp, andt1/2. Summarize the answers in Table SE7.4.Observe:� Cl does not influence Cp0, but as Cl increases the slope of the fall in Cp be-

comes steeper (k increases and t1/2 decreases). Thus, increases in clearanceresult in lower plasma concentrations at all times, apart from that at time zero.

TABLE SE7.4 Summary of the Effects of Dose, Clearance, and Volume ofDistribution on the Plasma Concentration–Time Profile

Cp0 Cp Slope Half-Life

Effect of dose

Effect of Cl

Effect of Vd

Page 165: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

APPLICATION OF THE MODEL 145

� Clearance is a constant for a particular drug, assuming normal healthy func-tion. It should be noted, however, that as a biological parameter it will varysomewhat within the population. The values reported in textbooks and in theliterature represent population average values. Beyond normal biological vari-ability, clearance will change if the function of one or both of the major organsof elimination change. Thus, clearance is often changed in renal and/or hepaticdisease. Concomitant medications can also alter the clearance of some drugsby inhibiting or inducing the enzymes involved in their metabolism. Changes inthe activity of uptake and/or efflux transporters in the liver and kidney can alsoalter clearance.

6. Go to the “Effect of Volume of Distribution” page. Use Vd values of 10, 20, and40 L to observe how the volume of distribution influences Cp0, Cp at any time,the slope of the fall in Cp, and t1/2. Summarize the answers in Table SE7.4.Observe:� As Vd increases, Cp0 gets smaller and the slope gets less steep (k decreases

and t1/2 increases; t1/2 is proportional to Vd). Essentially, increases in Vd com-press Cp over time and result in much less change or fluctuation in Cp.

� Again, the volume of distribution is a constant for a drug but in common withother biological parameters, its value will vary somewhat within the popula-tion. Beyond the normal variability, the volume of distribution can be altered byfactors that change either body volumes or alter tissue/plasma protein bind-ing. These include, hydration, dehydration, diseases such as congestive heartfailure, hepatic disease, and renal disease, and concomitant medications thatdisplace drugs from their binding sites volume of distribution.

7.5 APPLICATION OF THE MODEL

7.5.1 Predicting Plasma Concentrations

If a drug’s pharmacokinetic parameters are known, the plasma concentration can be esti-mated at any time after any intravenous dose.

Example 7.1 A 20-mg dose of a drug (S = 1) was administered as an intravenous bolusinjection. The drug has the following pharmacokinetic parameters: k = 0.1 h−1 and Vd =20 L.

(a) Calculate Cp0:

Cp0 = S · F · D

VdS = 1 (given) and F = 1 for IV administration

= 20

20

= 1 mg/L

(b) Calculate the plasma concentration at 3 h.

Page 166: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

146 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

Solution Note that half-life is very useful for getting rough estimates of the answersto pharmacokinetic calculations and for checking calculations and should be calculated assoon as possible.

t1/2 = 0.693

k

= 0.693

0.1 h−1

= 6.93 h

Thus, plasma concentrations will fall by 50% every 6.9 h. At 3 h, Cp should be about 75%its initial value, or about 0.75 mg/L:

Cp = S · F · D

Vd· e−kt

= 20

20mg/L · e−0.13

= 1 × 0.74

= 0.74 mg/L

7.5.2 Duration of Action

The duration of action of a drug may be considered to be the length of time the plasmaconcentration spends above the MEC. Its determination is best illustrated by example.

Example 7.2 Continuing with the drug used in Example 7.1, if the therapeutic range isbetween 5 and 0.3 mg/L, how long are the plasma concentrations in the therapeutic range?

Solution A small diagram is useful for this type of problem (Figure E7.2). Cp0 =1 mg/L. Thus, at time zero the plasma concentration is in the therapeutic range. The plasma

Cp

(mg/

L)

Time (h)

5

1

0.3

MTC

MEC

Durationof Action

FIGURE E7.2 Therapeutic range superimposed over the plasma concentration–time profile.

Page 167: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

APPLICATION OF THE MODEL 147

concentration will remain therapeutic until it falls to the MEC (0.3 mg/L). At what timedoes this occur?

Cp = Cp0 · e−kt

0.3 = 1 · e−0.1t

ln 0.3 = −0.1t

−1.204 = −0.1t

t = 12.04 h

Thus, the drug is in therapeutic range for 12 h. (Use the drug’s t1/2 value to check theanswer.)

7.5.3 Value of a Dose to Give a Desired Initial Plasma Concentration

The equation for the initial plasma concentration can be used to determine the value of adose to give a certain desired plasma concentration (Cpdes).

Example 7.3 The initial Cp of 1 mg/L is unsatisfactory. Calculate a dose to provide aninitial plasma concentration of 5 mg/L.

Solution

Cp0 = S · F · D

Vd

Let Cp0 = Cpdes and rearrange:

dose = Cpdes · Vd

S · F(7.6)

= 5 × 20

1 × 1= 100 mg

7.5.4 Intravenous Loading Dose

Most patients receive long-term treatment with a drug rather than a single isolated dose.Figure 7.3 shows the typical average plasma concentration observed over extended drugtherapy. This profile is typical of that of an intravenous infusion, where no fluctuation in theplasma concentration is observed. It can be seen that the plasma concentration graduallyincreases during the course of therapy and eventually reaches a plateau. The plateau isknown as the steady state, and the plasma concentration at this time is known as thesteady-state plasma concentration (Cpss). Dosing regimens are designed so that the plasmaconcentration at steady state is therapeutic. Often, the plasma concentrations leading upto steady state are subtherapeutic, and drug only becomes effective once steady state isachieved. In Chapter 9 we show that it takes about 3 to 5 elimination half-lives to get tosteady state. If a drug has a very long half-life and/or if it is clinically important to achieve

Page 168: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

148 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

0

20

40

60

80

100

0 2 4 6 8 10 12

Cp

(% S

tead

y St

ate)

Time Since Start of Therapy (Elimination t1/2 )

Cpss

FIGURE 7.3 Graph of typical plasma concentration–time profile associated with extended drugadministration.

therapeutic plasma concentrations immediately, it may be necessary to administer a loadingdose that will achieve the steady state immediately.

Equation (7.6) can be used to calculate the value of the loading dose. The value of Cpss

is equal to Cpdes. The general expression of the formula to calculate a loading dose, DL is

DL = Cpss · Vd

S · F(7.7)

Note that formula (7.7) contains the bioavailability factor even though F = 1 for intravenousadministration. This formula is frequently used for other routes of administration, and byalways including the bioavailability factor in the formula, there is less chance that it willbe ignored when it is needed.

Example 7.4 Calculate the value of an intravenous loading dose of phenytoin sodium(S = 0.92) for a 80-kg patient. A plasma concentration of 12 mg/L phenytoin is desired.The volume of distribution of phenytoin is 0.65 L/kg.

Solution

Vd = 0.65 L/kg × 80 kg = 52 L

DL = Cpss · Vd

S · F= 12 × 52

0.92= 678 mg

7.6 DETERMINATION OF PHARMACOKINETICPARAMETERS EXPERIMENTALLY

The expression of an intravenous injection in a one-compartment model consists essentiallyof two pharmacokinetic parameters: the primary parameter for elimination (Cl), and theprimary parameter for distribution (Vd). From these an additional parameter for the rate of

Page 169: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

DETERMINATION OF PHARMACOKINETIC PARAMETERS EXPERIMENTALLY 149

drug elimination is derived (k or a reciprocal form, t1/2). All the parameters of the modelcan be determined either from the two primary parameters or from one primary and onesecondary parameter. For example, k and t1/2 can be determined if Cl and Vd are known.Also, Cl can be determined if Vd and k are known. The pharmacokinetic parameters aredetermined by fitting plasma concentration–time data to the pharmacokinetic model.

7.6.1 Study Design for the Determination of Parameters

The plasma concentration–time data needed to model a drug’s pharmacokinetics mustbe obtained from human subjects. The protocol or procedure for any study that involveshumans must be approved by the institutional review board of the institution where thestudy is to be conducted and/or where the scientists conducting the study are employed.Additionally, anyone who volunteers to participate in a clinical study must sign an informedconsent form. The specific structure of the study will depend on the goal of the study andon the pharmacokinetic characteristics of the drug predicted for the population under study.The study will take the following general form:

1. An intravenous dose is administered to about 10 to 12 volunteers.

2. Plasma samples (10 to 20) are collected at various times after the dose for a periodof at least 3 elimination half-lives (at this time, 90% of the dose will have beeneliminated).

3. Plasma samples are prepared and frozen until they can be analyzed to obtain theconcentration of parent drug. The concentration of metabolites may also be measuredif the pharmacokinetics of metabolites are to be studied.

4. The plasma concentration–time data from each person are subject to pharmacokineticanalysis as described below. The parameters are determined.

5. The individual parameters for each subject are combined to determine the meanvalues for the study population and to determine their variability (variance or standarddeviation).

7.6.2 Pharmacokinetic Analysis

All the mathematical expressions that relate a drug’s pharmacokinetic parameters to theplasma concentration are nonlinear [see equations (7.4) and (7.5)]. Computer software isnow readily available that can perform nonlinear regression analysis and fit the plasma con-centrations directly to pharmacokinetic models. However, prior to the availability of thissoftware it was necessary to linearize the expression for the plasma concentration so that or-dinary linear regression analysis could be used to determine the parameter values. The latterprocedure is presented here. In order to focus on the procedure itself, most data used in thisbook for these examples and problems are “perfect” rather than “real.” Thus, the data shouldfit the models perfectly. In this book we do not discuss statistical procedures such as least-squares regression analysis for obtaining the best fit for real data that are scattered aroundthe model-predicted data. These procedures can be found in any basic textbook on statistics.

1. The plasma concentration–time data are observed on linear and semilogarithmic plotsto ensure that the data fit the model and that there are no data points that appear to bevastly different from the others (outliers).

Page 170: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

150 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

ln C

p

Time

Cp = Cp0 = S•F•D/Vd

Slope = ln(Cp1/Cp2)/(t1 – t2) k = – slope

FIGURE 7.4 Determination of k and Vd from semilog plot of plasma concentration against time.

2. The elimination rate constant (k) is determined from the slope of the plot of ln Cpand time (Figure 7.4). Recall from equation (7.4) that

Cp = Cp0 · e−kt

Taking the natural logarithm gives

ln Cp = ln Cp0 − kt (7.8)

This is the equation of a straight line (Figure 7.4):

slope = ln (Cp1/Cp2)

t1 − t2

k = −slope

= ln (Cp1/Cp2)

t2 − t1

(7.9)

3. The half-life is determined:

t1/2 = 0.693/k

4. The volume of distribution (Vd) is determined from the relationship

Vd = Ab

Cp

The only time the amount of drug in the body (Ab) is known is at time zero, when Abis equal to the effective dose (S · F · D). Thus,

Vd = S · F · D

Cp0

The value of Cp0 is obtained from the intercept of the ln Cp–time plot (Figure 7.4).

Page 171: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

DETERMINATION OF PHARMACOKINETIC PARAMETERS EXPERIMENTALLY 151

5. The clearance can be determined in several ways:

(a) It can be calculated from k and Vd:

Cl = k · Vd

(b) It can be determined from the AUC. It was shown previously (see Chapter 5) that

Cl = DIV

AUC(7.10)

The AUC can be determined using the trapezoidal rule, but more simply for theintravenous injection in a one-compartment model, the AUC can be calculated as

AUC =∫ ∞

0Cp · dt =

∫ ∞

0Cp0 · e−kt · dt = Cp0

k(7.11)

Substituting for AUC in equation (7.10) yields

Cl = DIV

AUC= DIV · k

Cp0(7.12)

Example 7.5 A single intravenous bolus injection of a drug (50 mg) was administered to10 normal subjects. Plasma samples were taken at various intervals and were analyzed forunchanged drug. The data from one subject are listed in Table E7.5. Determine the drug’sclearance, volume of distribution, and elimination half-life.

This problem can be solved either by hand using semilogarithmic graph paper or bycreating a special Excel worksheet for the analysis. Instructions to create an Excel worksheetto conduct the analysis of the data are given in Appendix C.

Solution Create a worksheet as described in Appendix C.

1. Plot Cp against time on both the linear and semilogarithmic scales to visualize the dataand to ensure that the data appear to fit the one-compartmental model (Figure E7.5).

TABLE E7.5

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0.1 2.45 3 1.370.2 2.40 5 0.920.5 2.26 7 0.611 2.04 10 0.331.5 1.85 12 0.222 1.67 15 0.12

Page 172: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

152 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

0

1.5

3

0 5 10 15 20Time (h)

-3

-2

-1

0

1

2

0 5 10 15 20

Time (h)

(a) (b)

Cp

(mg/

L)

ln C

p

FIGURE E7.5 Plots of plasma concentration against time on the linear (a) and semilogarithmicscales (b).

2. Calculate the elimination rate constant. The slope function in Excel is applied to theln Cp–time data. All the data should be used to determine the slope in Excel.

Slope = −0.20 h−1

k = −slope

= 0.20 h−1

3. Half-life:

t1/2 = 0.693

k

= 0.693

0.2

= 3.47 h

4. Volume of distribution: Cp0 is found using the intercept function in Excel:

intercept = ln Cp0 = 0.916

Cp0 = e0.916 = 2.5 mg/L

Vd = dose

Cp0

= 50 mg

2.5 mg/L

= 20 L

5. Clearance:

Cl = k · Vd

= 0.2 × 20 = 4 L/h

Page 173: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

PHARMACOKINETIC ANALYSIS IN CLINICAL PRACTICE 153

or

Cl = doseIV

AUC

AUC = Cp0/k, so

Cl = doseIV

Cp0/k

= 50 × 0.2

2.5= 4 L/h

6. The plasma concentration 6 h after a 100-mg IV dose of this drug is found as

Cp = D

Vd· e−kt

= 100

20· e−0.2 × 6 = 1.51 mg/L

7. The plasma concentration 10 h after this, that is, 16 h after the dose, is determinedusing

Cp = Cp0 · e−kt

where Cp0 = 1.51 mg/L, t = 10 h or Cp0 = 10020 mg/L, t = 16 h

Cp = 0.204 mg/L

7.7 PHARMACOKINETIC ANALYSIS IN CLINICAL PRACTICE

As discussed earlier, the primary pharmacokinetic parameters (and by extension the sec-ondary parameters) are considered to be constants for a drug. However, as biological param-eters they will display some person-to-person, or interindividual, variability. Furthermore,within a person, some day-to-day intraindividual variability in the parameters will be ob-served. Intraindividual variability is usually much smaller than interindividual variability.Population average values of the parameters can be obtained from the literature. Since thepharmacokinetics parameters control the dose–plasma concentration relationship, interindi-vidual variability in the pharmacokinetic parameters will result in interindividual variabilityin the plasma concentrations produced by a standard dose. For many drugs the margin ofsafety is large enough that interindividual variability is not associated with adverse clinicaleffects. However, for drugs that have narrow therapeutic ranges, interindividual variabilitycan result in the standard dose producing subtherapeutic plasma concentrations in somepatients, toxic concentrations in others, and therapeutic plasma concentrations only in somepatients. Drugs that have both wide interindividual variability in their pharmacokinetic pa-rameters and narrow therapeutic ranges include digoxin, lithium, warfarin, phenytoin, theaminoglycosides, and the immunosuppressants (i.e., cyclosporine, tacrolimus, etc.). Whenstandard doses of these drugs are used, the plasma concentrations should be assessed in

Page 174: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

154 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

individual patients. If plasma concentrations are not therapeutic, doses must be individ-ualized, based ideally on a patient’s individual pharmacokinetic parameters. In a clinicalsetting it is not possible to collect a large number of blood samples for pharmacokineticanalysis. Instead, a patient’s pharmacokinetic parameters must frequently be determinedfrom only two plasma concentrations.

Example 7.6 A patient is to be given an 80-mg IV dose of gentamicin every 8 h. A peakand a trough of 6 and 0.5 mg/L, respectively, are desired after the first dose. Two plasmaconcentrations obtained after the first dose are shown in Table E7.6. Determine if the peakand trough after this dose meets the goal. If not, suggest a more appropriate dose.

Solution

k = ln(4.5/1.1)

4= 0.352 h−1

t1/2 = 0.693

0.352= 1.97 h

A peak and a trough from the dose occur at t = 0 and 8 h, respectively:

Cp = Cp0 · e−kt

(a) Cp0:

1.1 = Cp0 · e−0.352 × 6

Cp0 = 9.1 mg/L

(b) Cp at 8 h and Cp8:

Cp = 9.1e−0.352 × 8

Cp8 = 0.54 mg/L

The volume of distribution:

Vd = dose

Cp0= 80 mg

9 mg/L

= 8.8 L

Thus, the dose did not achieve the therapeutic goal. A more appropriate dose must becalculated.

TABLE E7.6 Plasma Concentrationsof Gentamicin After a 80-mgIntravenous Dose

Time After Dose (h) Cp (mg/L)

2 4.56 1.1

Page 175: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

PROBLEMS 155

We want Cp0 = 6 mg/L:

Cp0 = S · F · D

Vd

S · F · D = Cp0 · Vd

= 6 mg/L × 8.8 L

= 52.8 mg

A trough of 0.5 mg/L is required. Thus, the time when Cp falls to 0.5 mg/L will be thedosing interval, or the time when the next dose should be given.

Cp = Cp0 · e−kt

0.5 = 6e−0.352t

t = 7.1 h

PROBLEMS

7.1 A drug (S = 1) has the therapeutic range 3.5 to 1.0 mg/L. It has a Vd of 120 L and aCl of 20 L/h.

(a) Recommend an intravenous dose of the free base to give an initial plasma con-centration of 3 mg/L.

(b) How long do plasma concentrations remain in the therapeutic range?

(c) If the lower plasma concentration must not to fall below 1.5 mg/L, when shouldthe second dose be administered?

(d) If the drug were given to a patient suffering from malnutrition in whom the Vd isestimated to be 160 L, what changes in the drug’s pharmacokinetic profile wouldyou expect?

7.2 A 35-year-old female patient (weight 60 kg), who has been taking theophylline forseveral years is being treated in the emergency room for an asthma attack. Hertheophylline plasma concentration is found to be 5 mg/L. Calculate the value of anintravenous loading dose of aminophylline (S = 0.8) that will bring the plasma con-centration up to 15 mg/L. In this patient, theophylline’s pharmacokinetic parametersare estimated to be as follows: Cl = 0.04 L/h/kg and Vd = 0.5 L/kg.

7.3 Procainamide is an antiarrhythmic used in the treatment of ventricular tachyarrhyth-mias. Determine the value of an intravenous loading dose of procainamide hydrochlo-ride (S = 0.87) that would achieve immediate plasma concentrations of 6 mg/L in a70-kg man. Procainamide’s Vd is 2 L/kg.

7.4 A loading dose of digoxin is to be administered to a 55-kg woman who has a creatinineclearance (ClCR) of 75 mL/min. Digoxin’s volume of distribution is dependent on renalfunction. Specifically,

Vd(L) = 3.8 × weight(kg) + 3.1ClCR(mL/min)

Page 176: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

156 PHARMACOKINETICS OF AN INTRAVENOUS BOLUS INJECTION

Determine the value of an oral loading dose for digoxin (F = 0.7) that will achievea desired serum digoxin of 0.8 �g/L. Use the equations for an intravenous bolusinjection.

7.5 A loading dose of phenobarbital sodium (S = 0.9) was administered to a 60-kg femalepatient. The initial plasma concentration was estimated to be 20 mg/L. The clearanceand volume of distribution of phenobarbital are 4 ml/h/kg and 0.6 L/kg, respectively.How long will it take for the plasma concentration to fall to 15 mg/L?

7.6 A patient with pneumonia due to Klebsiella pneumonia is being treated with gen-tamicin. Therapeutic drug monitoring is being performed in order to have peaks andtroughs of 20 and �0.5 mg/L, respectively, after the first dose. An initial regimenof 400 mg every 24 h was selected. The first intravenous dose was administered andplasma samples taken 1 and 7 h later had a gentamicin concentration of 12.5 and 1.51mg/L, respectively. Assume that the dose was given by intravenous bolus injection.

(a) Calculate the peak and trough plasma concentrations after this first dose. Are thetroughs and peaks acceptable?

(b) If not, what dose would have produced a peak of 20 mg/L? Would this dose haveproduced an acceptable trough?

7.7 After the intravenous administration of a new drug (dose = 100 mg, S = 1) plasmaconcentrations of 8 and 3 mg/L were found at 1 and 7 h after the dose, respectively.

(a) Estimate the drug’s elimination rate constant.

(b) Estimate the drug’s half-life.

(c) Estimate the AUC from zero to infinity.

(d) Estimate the drug’s clearance.

(e) Estimate the drug’s volume of distribution.

7.8 The problem set in Table P5.5 provided plasma concentration–time data for lipoamide,nosolatol, and disolvprazole after an intravenous bolus injection. The data are repeatedin Table P7.8.

TABLE P7.8

Lipoamide Nosolatol Disolvprazole

T (h) Cp (�g/L) T (h) Cp (�g/L) T (h) Cp (�g/L)

0 23.8 0 476 0 2860.5 22.1 0.5 462 0.2 2661 20.5 1 448 0.4 2491.5 19 1.5 435 0.8 2162 17.6 2 422 1 2023 15.2 2.5 409 1.2 1885 11.3 5 352 1.6 1637.5 7.82 8 294 2 142

10 5.39 12 231 3 10015 2.57 15 193 5 5020 1.22 20 143 8 1824 0.68 24 112 12 4

Page 177: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc07 JWBS062-Rosenbaum February 16, 2011 13:18 Printer Name: Yet to Come

RECOMMENDED READING 157

Doses administered: 10, 100, and 10 mg for lipoamide, nosolatol, and disolvprazole,respectively. Create an Excel worksheet using the directions provided in Appendix Cand analyze the data given to determine the following pharmacokinetic parameters foreach drug: elimination rate constant, volume of distribution, clearance, and eliminationhalf-life.

RECOMMENDED READING

1. Winter, M. E. (2010) Basic Clinical Pharmacokinetics, 5th ed., Lippincott Williams & Wilkins,Baltimore.

Page 178: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

8PHARMACOKINETICS OF ANINTRAVENOUS BOLUS INJECTIONIN A TWO-COMPARTMENT MODEL

8.1 Introduction

8.2 Tissue and Compartmental Distribution of a Drug8.2.1 Drug Distribution to the Tissues8.2.2 Compartmental Distribution of a Drug

8.3 Basic Equation8.3.1 Distribution: A, �, and the Distribution t1/2

8.3.2 Elimination: B, �, and the Beta t1/2

8.4 Relationship Between Macro and Micro Rate Constants

8.5 Primary Pharmacokinetic Parameters8.5.1 Clearance8.5.2 Distribution Clearance8.5.3 Volume of Distribution

8.5.3.1 Volume of the Central Compartment8.5.3.2 Volume of Distribution in the Distribution Phase8.5.3.3 Volume of Distribution at Steady State

8.6 Simulation Exercise

8.7 Determination of the Pharmacokinetic Parameters of the Two-Compartment Model8.7.1 Determination of Intercepts and Macro Rate Constants

8.7.1.1 Determination of B and �8.7.1.2 Determination of A and �

8.7.2 Determination of the Micro Rate Constants: k12, k21, and k10

8.7.3 Determination of the Primary Pharmacokinetic Parameters

8.8 Clinical Application of the Two-Compartment Model8.8.1 Measurement of the Elimination Half-Life in the Postdistribution Phase8.8.2 Determination of the Loading Dose8.8.3 Evaluation of a Dose: Monitoring Plasma Concentrations and Patient Response

Problems

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

158

Page 179: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

TISSUE AND COMPARTMENTAL DISTRIBUTION OF A DRUG 159

Objectives

The material in this chapter will enable the reader to:

1. Understand the physiological basis for the two-compartment model and the charac-teristics of the Cp versus time profile after intravenous administration

2. Identify the various ways in which the model can be parameterized

3. Identify and understand the difference between the three volumes of distribution ofthe model

4. Understand the relationships among the various parameters

5. Use feathering to estimate the model parameters

6. Understand how the model is applied clinically to determine an elimination half-life,estimate loading doses, and evaluate drug therapy

8.1 INTRODUCTION

The distribution of many drugs follows two-compartment pharmacokinetics. This is ap-parent because a steep fall in the plasma concentration is observed in the early periodafter the administration of an intravenous dose (Figure 8.1). As a result, the plasmaconcentration–time curve is biphasic, a feature that is most apparent on the semilogarithmicscale (Figure 8.1). The simple one-compartment model, in which plasma concentration fallsmonoexponentially under the sole influence of first-order elimination, cannot be used topredict this behavior. Two-compartment characteristics are the result of a slower distribu-tion of the drug to some tissues in the body. This distribution pattern is presented below,and it is followed by a discussion of how it is represented in the two-compartment model.

8.2 TISSUE AND COMPARTMENTAL DISTRIBUTION OF A DRUG

8.2.1 Drug Distribution to the Tissues

For drugs that display two-compartmental pharmacokinetics, there appear to be two typesof tissues with respect to their rate of uptake of drug. One group of tissues, like those ofthe one-compartment model, take up the drug extremely quickly. The uptake is so rapid

0.1

1

10

0 5 10 15 20 25 300

1

2

3

4

5

6

0 5 10 15 20 25 30

Cp

Time

Cp

(a)

Distribution Phase

Elimination PhaseElimination Phase

Time(b)

Distribution Phase

FIGURE 8.1 Graph of plasma concentration against time on a linear (a) and a semilogarithmic scale(b) for a drug that displays two-compartmental characteristics after an intravenous bolus injection.

Page 180: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

160 TWO-COMPARTMENTAL PHARMACOKINETICS

0.1

1

10

0 10 20 30

DrugConcentration

Time

Tissue of the Peripheral Compartment

Tissue of the Central Compartment

Plasma

FIGURE 8.2 Time course of drug concentrations in the plasma and two hypothetical tissues. Notethat drug distribution to the tissue of the central compartment (dashed line) appears to be instantaneousand is not visible on the graph. Distribution to the tissue of the peripheral compartment (dotted line)proceeds more slowly and is associated with a sharp fall in concentrations in the plasma (solid line)and tissue of the central compartment.

that there is no evidence of the distribution based on blood samples taken during theearly period after injection. The tissues where this occurs are usually the well-perfusedtissues, and the tissue–plasma ratio in these tissues achieves its equilibrium value ex-tremely rapidly and remains constant. Distribution to some other tissues, usually the poorlyperfused tissues, occurs more gradually. It is the distribution of the drug to these tis-sues that is associated with the pronounced initial fall in the plasma concentration. Thisperiod is known as the distribution phase (Figure 8.1). Note that drug elimination alsooccurs during this phase, but it is drug distribution that dominates the fall in the plasmaconcentration.

In these tissues, where distribution proceeds more slowly, the tissue–plasma concentra-tion ratio increases throughout the distribution phase. Eventually, a type of equilibrium isachieved between the plasma and the poorly perfused tissues. At this time, the distributionphase is complete and the tissue/plasma concentration ratio remains constant. First-orderdrug elimination then drives the fall in plasma concentration, which as a result falls in asmooth monoexponential manner. This is known as the elimination phase. Figure 8.2 showshow drug concentrations change in hypothetical tissues in both the central and peripheralcompartments relative to the plasma concentration. Note that the concentration of drug inboth groups of tissues will vary from tissue to tissue. The concentration of drug in a tissuein either compartment could be greater than, less than, or equal to the plasma concentration,depending on factors such as binding and drug transporters.

8.2.2 Compartmental Distribution of a Drug

According to the pharmacokinetic model, the body is assumed to consist of two hypo-thetical compartments (Figure 8.3). The central compartment consists of the plasma andthose tissues where drug distribution proceeds very rapidly. By definition the concentration

Page 181: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

TISSUE AND COMPARTMENTAL DISTRIBUTION OF A DRUG 161

V1A1Cp

V2A2C2

CentralCompartment

PeripheralCompartment

Initial Conditions: t = 0, A1 = S•F•D, A2 = 0

A2•k21

A1•k12

Cp•Cl or A1•k10

FIGURE 8.3 Two-compartment model with intravenous bolus injection. Ab, A1, and A2 are theamount of drug in the body, the central compartment, and the peripheral compartment, respectively;Cp is the plasma concentration of the drug and the concentration in the central compartment; C2 isthe drug concentrations in the peripheral compartment; V1 and V2 are the respective volumes of thecentral and peripheral compartments; k10 is the first-order elimination (E) rate constant; k12 and k21

are the rate constants for distribution (D) and redistribution (R), respectively; Cl is clearance; and theeffective dose is given by the product of S (salt factor), F (bioavailability factor), and D (the doseadministered). Note that for intravenous administration, F = 1.

throughout this compartment is equal to the plasma concentration (see Chapter 6). Drugelimination is usually assumed to occur from the central compartment because the organs ofelimination are well perfused and are usually part of the central compartment. The second,peripheral compartment consists of those tissues where distribution proceeds more slowly.By definition it is homogeneous and the drug concentration throughout is uniform at alltimes. Movement between the compartments is assumed to be first-order driven by theamount of drug in a compartment. The movement of drug from the central to the peripheralcompartment is referred to as distribution (rate constant k12) and the movement of drugback from the peripheral to the central compartment is referred to as redistribution (rateconstant k21). When an intravenous dose is administered, the drug is assumed to undergoinstantaneous distribution throughout the central compartment. At this time, the entire doseis present in the central compartment and is distributed throughout the compartment in acompletely uniform manner; that is, at any time the drug concentration throughout is thesame (Figure 8.4). At time zero, no drug is present in the peripheral compartment. Theconcentration gradient between compartments then causes a net movement of drug into theperipheral compartment. The concentration in the peripheral compartment increases andthe concentration in the central compartment decreases (due to distribution and elimination)(Figure 8.4). Eventually, an equilibrium (pseudo) is achieved, at which time the concen-tration in the central compartment equals that of the peripheral compartment (Figure 8.4).At this time the concentration in the central compartment decreases due to elimination. Asthe concentration in the central compartment falls there is a net movement of drug fromthe peripheral compartment back into the central compartment. The concentration of drugin the peripheral compartment does not correspond to the concentration of drug in anyparticular tissue that is part of the peripheral compartment. Instead, it represents an aver-aged concentration of all the tissues that comprise the peripheral compartment. The time

Page 182: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

162 TWO-COMPARTMENTAL PHARMACOKINETICS

CentralCompartment

PeripheralCompartmentTime

0 C2 = 0

0 +

0 ++

0 +++

Distribution phase is complete and the drug concentration in the compartments is the same. It is not a true equilibrium because it is continuously destroyed by elimination.

EliminationRedistribution

Cp > C2

Cp > C2

Cp = C2

Distribution

FIGURE 8.4 Relative concentrations of drug in the central (Cp) and peripheral (C2) compartmentsat different times after intravenous injection in a two-compartment model.

course of the distribution process for the two compartments is demonstrated in an interactivevideo found in the Model, “IV Bolus Injection in a Two-Compartment Model,” at

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter8

8.3 BASIC EQUATION

Based on the model shown in Figure 8.3, it is possible to write equations for the rate ofchange of the amount of drug in the two compartments:

rate of change of the amount of drug in any compartment = rate of inputs − rate of outputs

Page 183: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

BASIC EQUATION 163

For the central compartment,

dA1

dt= k21 A2 − k12 A1 − k10 A1 (8.1)

For the peripheral compartment,

dA2

dt= k12 A1 − k21 A2 (8.2)

When these equations are integrated and solved for Cp, the following biexponential solutionis obtained:

Cp = A · e−�t + B · e−�t (8.3)

where � is the hybrid rate constant for distribution, � the hybrid rate constant for elimination,and A and B are the intercepts of the two exponential functions. Thus, according to equation(8.3), the plasma concentration–time profile consists of two exponential terms, A · e−�t andB · e−�t. According to convention, � is always larger than �, and in the vast majority ofsituations, A · e−�t represents drug distribution and B · e−�t, drug elimination.

8.3.1 Distribution: A, � , and the Distribution t1/2

The function A · e−�t describes the distribution characteristics of a drug during the distri-bution phase. Note the parameter A, which is an intercept, is dependent on the dose as wellas a drug’s pharmacokinetic parameters. The rate constant � is the hybrid rate constantfor distribution because although it expresses distribution, it is neither k12 nor k21. It isreferred to as a macro rate constant, as it is a complex function of all three of the first-orderrate constants (k10, k12, k21) [see equation (8.13)]. The latter first-order rate constants arereferred to as the micro rate contants. The half-life determined from � is known as thedistribution half-life:

t1/2,� = 0.693

�(8.4)

As distribution is a first-order process, it takes 1 t1/2,� for distribution to go to 50% com-pletion and around 3 to 5 t1/2,� for the distribution phase to go to completion.

8.3.2 Elimination: B, �, and the Beta t1/2

The function B · e−�t describes the characteristics of the plasma concentration–time profilein the elimination phase. The parameter B, like A, is an intercept that is dependent on thedose as well as a drug’s pharmacokinetic parameters. The rate constant � is the hybridrate constant for elimination and is distinct from the true elimination rate constant k10. Itexpresses the manner in which the plasma concentration and the amount of drug in the bodyfall due to drug elimination during the elimination phase. It is also a macro rate constantthat is dependent on all three of the micro rate constants (k10, k12, k21) [see equation (8.14)].

Page 184: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

164 TWO-COMPARTMENTAL PHARMACOKINETICS

The half-life determined from � is referred to as the elimination, biological, or dispositionhalf-life:

t1/2,� = 0.693

�(8.5)

Once the distribution phase is complete, the beta half-life is used to determine how longit will take to eliminate a drug from the body. Since elimination is a first-order process, itwill take 1 t1/2,� to eliminate 50% of a dose and around 3 to 5 t1/2,� to eliminate the dose.

Clinically, � or, more specifically, t1/2,� is an important parameter and may be measuredin individual patients to optimise therapy. In contrast, � is rarely determined clinically,but it is important to know its approximate value so that the approximate duration of thedistribution phase can be determined. This is because when blood samples are collectedto determine the t1/2,�, it is important to avoid taking samples in the distribution phase.For example, the elimination half-lives of aminoglycosides such as tobramycin are oftenmeasured in patients. These drugs have an t1/2,� of about 5 min. Thus, it is important towait at least 20 min after an injection before taking blood samples to estimate the t1/2,�. Incase of digoxin, which also displays two-compartment pharmacokinetics, the t1/2,� is about1.5 h. Thus, the distribution phase for digoxin lasts about 6 h.

8.4 RELATIONSHIP BETWEEN MACRO AND MICRO RATE CONSTANTS

The macro rate constants (� and �) and intercepts (A and B) are complex functions of thefirst-order micro rate constants (k10, k12, k10). The relationship between these parameters isas follows:

� + � = k12 + k21 + k10 (8.6)

� · � = k21 · k10 (8.7)

A = S · DIV · (� − k21)

V1 · (� − �)(8.8)

B = S · DIV · (k21 −�)

V1 · (� − �)(8.9)

where S is the salt factor and DIV is the value of the intravenous dose. It can be seenin equations (8.8) and (8.9) that A and B are directly proportional to the dose. When theparameters of the two-compartment model are determined experimentally, A, B, �, and �are determined first. These are then used to determine the value of the micro rate constantsusing the computational formula based on equations (8.6) to (8.9):

k21 = A · � + B · �

A + B(8.10)

k10 = � · �

k21(8.11)

k12 = � + � − k10 − k21 (8.12)

Page 185: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

PRIMARY PHARMACOKINETIC PARAMETERS 165

Explicit equations for � and � have also been derived, which allow � and � to be calculatedfrom the micro rate constants:

� = 0.5[(k10 + k12 + k21) +

√(k12 + k21 + k10)2 − (4 × k21 × k10)

](8.13)

� = 0.5[(k10 + k12 + k21) −

√(k12 + k21 + k10)2 − (4 × k21 × k10)

](8.14)

8.5 PRIMARY PHARMACOKINETIC PARAMETERS

The pharmacokinetic parameters of the model shown in Figure 8.3 consists of first-orderrate constants (k10, k12, and k21) and volumes (V1 and V2). As was the case for theone-compartment model, the rate constants are derived, or dependent pharmacokineticparameters with little direct physiological meaning. The two-compartment model has atotal of four primary pharmacokinetic parameters: clearance (Cl), distribution clearance(Cld), volume of the central compartment (V1), and volume of the peripheral compartment(V2). A description of these primary parameters and their relationship to the volume ofdistribution and the dependent parameters (k10, k12, k21) is presented below.

8.5.1 Clearance

Clearance is consistent with previous definitions: It is a measure of the ability of the organsof elimination to remove drug from the plasma, and it is a constant of proportionalitybetween the rate of elimination at any time and the corresponding plasma concentration.The rate of elimination of a drug can be expressed using either the elimination rate constant(k10) or clearance:

−dAb

dt= k10 · A1 (8.15)

−dAb

dt= Cl · Cp (8.16)

Equating equations (8.15) and (8.16), and given that Cp = A1/V1, the equation can berearranged to yield

k10 = Cl

V1(8.17)

Clearance can be determined in the usual way from the area under the curve (AUC):

Cl = S · F · D

AUC(8.18)

For the two-compartment model the AUC can be calculated:

AUC =∫ ∞

0Cp · dt =

∫ ∞

0A · e−�t + B · e−�t · dt (8.19)

Page 186: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

166 TWO-COMPARTMENTAL PHARMACOKINETICS

AUC = A

�+ B

�(8.20)

8.5.2 Distribution Clearance

The Distribution clearance (Cld) is a measure of the ability of a drug to pass into and outof the tissues of the peripheral compartment. It is determined by the permeability of thedrug across the capillary membrane in these tissues as well as the blood flow to the tissues.It can be shown that

Cld = Qb(1 − e−P/Qb ) (8.21)

where P is the permeability and Qb is the blood flow. It can be seen in equation (8.21) thatas the permeability increases, the distribution clearance becomes limited by blood flow.Thus, when a drug is able to diffuse across a membrane with ease, as is the case for mostcapillary membranes, the distribution clearance will be dependent on the blood flow to atissue and will be high in those tissues with large blood flows. The distribution clearance isthe constant of proportionality between the rate of distribution and the rate of redistributionand the concentration driving the process.

Drug distribution from the central compartment to the peripheral compartment can beexpressed using either the first-order rate constant for distribution (k12) or the distributionclearance:

rate of distribution = A1 · k12(8.22)

rate of distribution = Cld · Cp

Equating the two expressions in equation (8.22), and substituting for Cp (Cp = A1/V1),it can be written

k12 = Cld

V1(8.23)

The rate constant for distribution to the peripheral compartment is derived from the twoprimary parameters of distribution clearance and the volume of the central compartment.

Drug redistribution from the peripheral compartment back into the central compart-ment can also be expressed using either distribution clearance or the rate constant forredistribution:

rate of redistribution = Cld · C2(8.24)

rate of redistribution = A2 · k21

Equating the two expressions in equation (8.24) and substituting for C2 (C2 = A2/V2), itcan be shown as

k21 = Cld

V2(8.25)

Page 187: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

PRIMARY PHARMACOKINETIC PARAMETERS 167

The rate constant for redistribution from the peripheral compartment is derived from thetwo primary parameters of distribution clearance and the volume of the peripheral compart-ment. Thus, distribution clearance is a primary pharmacokinetic parameter that determinesthe values of the rate constants for distribution and redistribution. The rate constants fordistribution and redistribution are also determined by V1 and V2, respectively.

When there is equilibrium between the two compartments, the rate of distribution = therate of redistribution. Thus,

A1 · k12 = A2 · k21

k12

k21= A2

A1

(8.26)

Substituting for k12 and k21 from equations (8.23) and (8.25) yields

V2

V1= A2

A1(8.27)

Thus, both the ratio k12/k21 [equation (8.26)] and the ratio V2/V1 [equation (8.27)] aremeasures of the relative distribution of the drug between the two compartments. A largeratio indicates that a large fraction of the drug in the body resides in the peripheralcompartment.

8.5.3 Volume of Distribution

The two-compartment model contains two volume terms (V1 and V2), but it is not clearhow these volumes are related to the volume of distribution. Recall that the volume ofdistribution of a drug is a ratio of the amount of drug in the body at any time to the plasmaconcentration at that time. In a two-compartment model the volume of distribution changesafter the administration of a dose, and at different times one of three volumes of distributionmay hold: V1, V�, and Vdss. The definition and characteristics of these three volumes arediscussed below.

8.5.3.1 Volume of the Central CompartmentThe volume of the central compartment (V1) is the volume of distribution at time zeroimmediately after intravenous administration of a drug. At this time the entire dose iscontained within the central compartment: A1 = dose; A2 = 0. Thus,

Vd = Ab

Cp= A1

Cp= V1 (8.28)

The plasma concentration at t = 0 (Cp0) can be expressed

Cp0 = A · e−�t + B · e−�t = A + B (8.29)

Page 188: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

168 TWO-COMPARTMENTAL PHARMACOKINETICS

Thus, V1 may be determined:

V1 = doseIV

Cp0= doseIV

A + B(8.30)

8.5.3.2 Volume of Distribution in the Distribution PhaseAt time zero the entire dose is contained within the central compartment and Vd = V1.The drug then gradually distributes to the peripheral compartment. As the physical volumethrough which the drug distributes increases, the volume of distribution increases. Thiscan be appreciated by considering the relative rates of fall of the plasma concentration andthe total amount of drug in the body during the distribution period. During this period thefall in the plasma concentration is dominated by distribution, but some drug eliminationwill occur. In contrast, elimination alone causes the amount of drug in the body to de-crease. Consequently, the fall in the plasma concentration is much greater than the fall inthe amount of drug in the body and the volume of distribution (Vd = Ab/Cp) increases(Figure 8.5). Once the distribution phase has been completed, the plasma concentrationand the amount of drug in the body fall in parallel, the volume of distribution is constantand is called V�. Thus, V� represents the volume of distribution in the postdistribution orelimination phase.

The disadvantage of V� is that it is dependent on elimination and as a result is nota true primary, independent parameter. In the postdistribution or elimination phase, trueequilibrium between the two compartments does not exist. A momentary equilibrium isestablished and then destroyed continuously by drug elimination. If true equilibrium wasestablished, Vd = V1 + V2. As a result of elimination, the drug concentration in thecentral compartment (Cp) falls to a greater extent than the amount of drug in the body(Ab). As a result, V� (Ab/Cp) is larger than V1 + V2. Furthermore, the greater the amountof elimination, the greater is the difference between V� and V1 + V2. The system tendstoward true equilibrium, and V� tends toward V1 + V2 as elimination tends toward zero. Atrue equilibrium between the two compartments can also be established if drug eliminationfrom the central compartment is matched exactly by drug administration into the centralcompartment (Figure 8.6).

Vd

V1

Vd = Ab/CpDuring the distribution phase, Cp falls (distribution and elimination)more rapidly than Ab (elimination).As a result, Vd increases.

Time After Dose

FIGURE 8.5 Graph showing how the volume of distribution changes with time after an intravenousinjection in a two-compartment model.

Page 189: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

PRIMARY PHARMACOKINETIC PARAMETERS 169

Elimination

Drug Inputto MatchElimination

Reduce Elimination toZero

Distribution

Redistribution

CentralCompartment

PeripheralCompartment

FIGURE 8.6 Ways to establish true equilibrium between the central and peripheral compartments.True equilibrium between compartments can be obtained either by exactly matching the rate ofelimination with drug input or by reducing elimination to zero.

The value of V� is usually close to the value of V1 + V2. It is easily calculated andis a useful volume of distribution, but its dependence on elimination can limit its use. Forexample, it would not be an appropriate parameter to use in a clinical study designed toevaluate if distribution was altered in a situation in which clearance may be altered. It wouldnot be possible to determine if any changes in V� were due to altered clearance or altereddistribution.

Determination of V� During the elimination phase, Vd = V� and the rate of drug elimi-nation can be expressed as

rate of elimination = � · Ab (8.31)

But elimination can also be expressed in terms of clearance:

rate of elimination = Cl · Cp (8.32)

equating equations (8.31) and (8.32). Substituting Ab/V� for Cp, and substituting S · F ·D/AUC for Cl, the equation can be rearranged:

V� = S · F · D

� · AUC(8.33)

The AUC is calculated as described previously in equation (8.20).

8.5.3.3 Volume of Distribution at Steady StateAt steady state the loss of drug from elimination is matched exactly by the gain of drugfrom administration. True equilibrium exists between the compartments, and the volumeof distribution is equal to V1 + V2. This volume of distribution is known as the volumeof distribution at steady state (Vdss). It is a true primary pharmacokinetic parameter thatreflects only distribution and is not influenced by elimination.

At steady state, rate of distribution = rate of redistribution:

A1 · k12 = A2 · k21 (8.34)

Page 190: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

170 TWO-COMPARTMENTAL PHARMACOKINETICS

TABLE 8.1 Various Ways of Parameterizing One- andTwo-Compartment Models

Rate Constants Physiological Parameters

IntravenousOne-compartment k, Vd Cl, VdTwo-compartment k10, k12, k21, V1, and V2

a Cl, Cld, V1, V2b

aSince one of these parameters can be calculated if the other four are known, thisparameterization consists of four, not five, parameters.bVdss = V1 + V2.

Thus,

A2 = A1 · k12

k21(8.35)

Vdss = Ab

Cp= A1 + A2

Cp= A1 + A2

A1/V1(8.36)

Substituting A2 from equation (8.35) into equation (8.36) yields

Vdss = V1 · k12 + k21

k21(8.37)

In summary, the model of an intravenous injection in a two-compartment model has fourprimary parameters: clearance, distribution clearance, volume of the central compartment,and volume of the peripheral compartment (or Vdss, which is equal to V1 + V2). This modelcan be parameterized in terms of these primary pharmacokinetic parameters. Alternatively,the model can be described using the derived parameters or micro rate constants: k10, k12,k21, and the volumes. The various ways of parameterizing the model are summarized inTable 8.1, which for comparison includes the parameterization of the one-compartmentmodel.

8.6 SIMULATION EXERCISE

Open the model “IV Bolus Injection in a Two-Compartment Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter8

The default parameters of the model are dose = 1000 mg, S = 1, Cl = 4 L/h, Cld =30L/h, V 1 = 10 L, and V 2 = 40 L.

1. Review the objectives and model summary.

2. Explore the model and the various pharmacokinetic parameters.

Page 191: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

SIMULATION EXERCISE 171

3. Exercise 1: Cp–time profile. The left graph shows the drug concentration inplasma and in the hypothetical peripheral compartment on a linear scale. Theright graph shows a plasma concentration–time profile on the semilogarithmicscale. Choose a dose.Observe:� The nature of the Cp–time profile is biphasic.� The steep initial fall corresponds to the distribution of drug to the peripheral

compartment (distribution phase). When it is over, the fall of ln Cp with time islinear.

� The peak concentration in the peripheral compartment corresponds to the endof the distribution phase.

� In the postdistribution or elimination phase, the drug concentration in the hypo-thetical peripheral compartment is equal to the plasma concentration.

� In the elimination phase, the peripheral compartment concentration and theplasma concentration fall in parallel as drug is eliminated from the body.

4. Exercise 2: Effect of dose. Graphs of plasma concentration against time areshown on the linear and semilogarithmic scales. The t1/2,� is also shown in adisplay window. Without clearing the graphs between doses, give doses of 50,100, and 150 mg.Observe:� Drag the mouse along the curves of the plots to display the values of Cp and

time. It can be seen that Cp at any time is proportional to the dose.� From the plot of ln Cp against time, it can be seen that the slope of the terminal

linear portion of the graph does not change with dose. The elimination half-lifedoes not change with dose.

5. Prominence of two-compartment characteristics Drug. The distribution of manydrugs to some of the tissues in the body, particularly the poorly perfused tissues,is not instantaneous. Yet a two compartment model isn’t always needed for thesedrugs. Sometimes, the simpler one compartment model will provide an acceptabledegree of accuracy. The prominence of two-compartmental pharmacokinetics,and the error that would result if a one compartmental model was used, dependson a number of factors. First, the amount of drug involved in the slower distributionis important. If this distribution involves only a relatively small amount of the totaldrug in the body, it will have little impact on the initial plasma concentrations.The amount of elimination that occurs during the distribution phase is anotherimportant factor. In contrast to the one compartment model, where distributionis instantaneous, some elimination will occur during distribution to the peripheralcompartment. The greater the amount of drug elimination during the distributionphase, the greater the prominence of two compartmental characteristics.

Exercise 3: Elimination during the distribution phase. During the early period afteran injection, distribution to the peripheral compartment and elimination competefor the drug in the central compartment. The rate of distribution is controlledby k12, which is equal to Cld/V 1. The rate of elimination is controlled by k10,which is equal to Cl/V 1. Thus, the relative values of Cld and Cl control which

Page 192: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

172 TWO-COMPARTMENTAL PHARMACOKINETICS

is the dominant process. As Cl increases relative to Cld, the amount of drugeliminated during the distribution phase increases, and the prominence of thetwo-compartmental characteristics increase.

Go to the “Elimination, Cl and Cld” interface page. The graph on the left isa plot of Ln(Cp) against time, and can be used to evaluate the duration of thedistribution period. The graph on the right shows the percentage of the dose(1000mg) eliminated at any time. The values of � and � are also displayed� With the default value of Cld (30 L/hr) simulate with Cl values of 10, 20 and

40 L/hr. Comment on how Cl effects the two-compartmental characteristics,the amount of drug eliminated during the distribution phase, � and �.

� Clear the graphs. With Cl set to 4 L/hr, simulate with values of Cld of 30, 15and 7 L/hr. Comment on the effects on the two-compartmental characteristics,the amount of drug eliminated during the distribution phase, � and �.

� Clear the graphs. Now make Cld very low (1L/hr) and observe the effects of Clvalues of 10, 20 and 40L/hr. Note as above, the prominence of the two com-partmental characteristics increases as Cl increases, but note the changes areassociated with minimal change in � and instead bring about almost propor-tional changes in �. This is because in the early period after the dose, therate of elimination is greater than the rate of distribution and dominates thefall in the plasma concentrations. At later times when most of the dose hasbeen eliminated, the gentler fall in the plasma concentration is due to drugredistributing from the peripheral compartment. This is known as a flip-flopmodel because the normal arrangement of the phases is reversed: A and �

reflect elimination and B and � reflect distribution. This situation is much lesscommon in a two compartment model but is observed in a three compartmentmodel (Section 6.3.3) where a deep-tissue compartment is incorporated intothe model. The pharmacokinetics of both gentamicin and digoxin are mostaccurately modeled using a three compartment model. The tissues of the cen-tral compartment take up the drug in an essentially instantaneous manner;the tissues of the peripheral compartment take up the drug more slowly, andthis distribution is associated with a steep initial decline in plasma concentra-tions; the tissues of the deep tissue compartment take up the drug extremelyslowly. This distribution is slower than elimination, and at later times redistri-bution from the deep tissue compartment controls the terminal slope of theLnCp versus time plot (Figure 6.3b). In the case of gentamicin, this latter phaseis only apparent many hours after a dose when plasma concentrations arevery low.

6. Exercise 4: The fraction of the drug that distributes to the peripheral compartment.Observe how the amount of drug that distributes to the peripheral compartmentinfluences the prominence of two compartmental characteristics. Note, the frac-tion of the overall drug that distributes to the peripheral compartment is reflectedby the relative values of V 1 and V 2, and recall k12 is Cld/V 1, and k21 is Cld/V 2.Thus V 2/V 1 is equal to k12/k21.� Simulate with values of V2 of 10, 40, 80 and 160 L. Comment on the effects on

the difference between the initial Cp, and Cp at the beginning of the distributionphase. Also comment on the effects on �.

Page 193: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

DETERMINATION OF THE PHARMACOKINETIC PARAMETERS 173

8.7 DETERMINATION OF THE PHARMACOKINETIC PARAMETERS OFTHE TWO-COMPARTMENT MODEL

The parameters of the two-compartment model are determined using a study protocolsimilar to the one discussed for a one-compartment model. Thus, an intravenous dose isadministered to subjects, and plasma concentrations of the drug obtained at various timesafter the dose are subject to pharmacokinetic analysis. In the case of a two-compartmentmodel, it is important to obtain sufficient blood samples during the distribution phase tocharacterize the two-compartmental characteristics.

The plot of plasma concentration against time for a two-compartment model is describedby a biexponential equation and cannot be converted to a straight line by convertingplasma concentrations to the logarithmic domain. Thus, simple linear regression cannotbe used to estimate the parameters of the two-compartment model. Today, a wide varietyof commercial software packages, such as WinNonlin (Pharsight, Mountain View, CA),are available that can perform nonlinear regression analysis. These products use nonlinearregression analysis to model the plasma concentration–time data directly to obtain estimatesof the pharmacokinetic parameters (i.e., they do not need the plasma concentrations to beconverted to the logarithmic domain). Prior to the general availability of nonlinear regressionsoftware, the plasma concentration–time data had to be linearized using a process calledcurve stripping (also known as the method of residuals or feathering). This method is stilluseful today when initial estimates of the parameters are needed for computer analysis. Theprocess takes the basic equation for the two-compartment model:

Cp = A · e−�t + B · e−�t

and isolates the two-component exponential functions to allow A and � and B and � tobe determined from straight lines obtained from semilogarithmic plots. The micro rateconstants and the primary pharmacokinetic parameters are then calculated from these fourparameters. This method is described in detail below.

8.7.1 Determination of Intercepts and Macro Rate Constants

8.7.1.1 Determination of B and �The equation for the overall plasma concentration–time is given as

Cp = A · e−�t + B · e−�t (8.38)

Recall that the hybrid rate constant for distribution (�) is almost always larger than thehybrid rate constant for elimination (�). As a result, e−�t decays to zero (at about 3 to 5 �t1/2) before e−�t. At this point the equation becomes monoexponential:

Cp = B · e−�t (8.39)

Taking logarithms of equation (8.39) converts it to the equation of a straight line. Let Cpall along this line (from time zero to infinity) = Cp′:

Cp′ = B · e−�t (8.40)

Page 194: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

174 TWO-COMPARTMENTAL PHARMACOKINETICS

ln C

pB

−β

Time

Cp′ = B•e–βt

FIGURE 8.7 Determination of B and � from a semilogarithmic plot of plasma concentrationagainst time.

During the elimination phase, Cp′ = Cp, but during the distribution phase, Cp′ � Cp.Estimates of B and � can be determined from the intercept and slope, respectively, of asemilogarithmic plot of Cp′ against time (Figure 8.7):

ln Cp′ = ln B − � · t (8.41)

The drug’s half-life during the elimination phase can be calculated:

t1/2,� = 0.693

�(8.42)

8.7.1.2 Determination of A and �Parameters A and � are determined using curve stripping to separate the two exponentialcomponents of equation (8.38). Recall that during the distribution phase, Cp′ � Cp. Ifduring this period, Cp′ is subtracted from Cp, we obtain

Cp − Cp′ = A · e−�t − B · e−�t − B · e−�t (8.43)

or

Cp − Cp′ = A · e−�t (8.44)

Thus, A · e−�t is isolated, and taking logarithms yields

ln(Cp − Cp′) = ln A − � · t (8.45)

Thus, a plot of ln(Cp − Cp′) yields a straight line of slope (−�) and intercept A.The plot of ln(Cp − Cp′) is constructed as follows:

1. The times of the data points in the distribution phase are noted.

2. From the back-extrapolated part of the plot of ln Cp′ against time, the correspondingCp′ values of these times are noted.

Page 195: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

DETERMINATION OF THE PHARMACOKINETIC PARAMETERS 175

ln C

p

Time

1. Plot ln Cp against time. Note the linear terminal linear portion.2. Let Cp along this line and its back-extrapolated portion = Cp′: ln Cp′ = ln B – βt. The intercept of this line is B and its slope = –β. 2. Values of Cp′ ( ) that correspond to the time of given data ( ) are read off the line.3. Values of Cp – Cp′ ( ) are calculated and ln(Cp – Cp′) is plotted against time to create a new line: ln(Cp – Cp′) = ln A – αt Intercept = A; slope = –α

ln C

pA

B−α −β

Cp = A•e–αt + B•e–βt

Cp – Cp′ = A•e–αt

Time

– =

Cp′ = B•e–βt

FIGURE 8.8 Method of residuals to determine A and �.

3. These values of Cp′ are subtracted from their corresponding given data points toobtain the values of Cp − Cp′.

4. Cp − Cp′ is plotted against time on a semilogarithmic scale.

The process is summarized in Figure 8.8.

8.7.2 Determination of the Micro Rate Constants: k12, k21, and k10

The micro rate constants k10, k21, and k12 can be calculated from the computation formulaspresented earlier (8.10) to (8.12).

8.7.3 Determination of the Primary Pharmacokinetic Parameters

The primary pharmacokinetic parameters of the two-compartment model can be determinedusing formulas presented previously:

� V1 from equation (8.30)� V� from equation (8.33)� Vdss from equation (8.37)� Clearance from equation (8.18)� Distribution clearance from equation (8.23) or (8.25)

Page 196: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

176 TWO-COMPARTMENTAL PHARMACOKINETICS

TABLE E8.1

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0.1 7.95 2 1.070.2 6.38 3 0.890.4 4.25 4 0.760.7 2.57 6 0.561 1.79 8 0.421.5 1.27 12 0.23

Example 8.1 The data in Table E8.1 simulated for a 100-mg intravenous bolus injectionin a two-compartment model. Analyze the data to determine:

(a) The intercepts and macro rate constants: A, B, �, and �

(b) The micro rate constants: k10, k12, and k21

(c) The primary pharmacokinetic parameters: Cl, Cld, V1, V�, and Vdss

Solution A completed Excel worksheet created for this analysis is shown in Appendix C(Figure C.6). The answers are as follows:

(a) A = 8.47 mg/L; B = 1.38 mg/L; � = 2.62 h−1; � = 0.149 h−1

(b) k10 = 0.789 h−1; k12 = 1.49 h−1; k21 = 0.495 h−1

(c) Cl = 8.02 L/h; Cld = 15.1 L/h; V1 = 10.2 L; V� = 53.8 L; Vdss = 40.7 L

8.8 CLINICAL APPLICATION OF THE TWO-COMPARTMENT MODEL

When pharmacokinetics is used to optimize drug therapy in individual patients, it is rarelypossible to obtain enough plasma samples to be able to use the two-compartment model,and as a result, the simpler one-compartment model is frequently used. It has been foundthat provided that certain precautions are taken, the one-compartment model providesan acceptable degree of accuracy when applied to drugs that display two-compartmentalpharmacokinetics.

8.8.1 Measurement of the Elimination Half-Life in the Postdistribution Phase

For drugs that display wide interpatient variability in clearance and/or the volume ofdistribution, it is often necessary to determine the drug’s elimination half-life in individualpatients. Usually, the half-life is determined from only two blood samples, and if a drugdisplays two-compartmental pharmacokinetics, it is important to wait until the conclusionof the distribution phase before taking blood samples to measure the elimination half-life.If sample(s) are taken in the distribution phase, the estimate of the half-life will be biased(Figure 8.9).

For example, the half-life of the aminoglycoside antibiotics is frequently determined inindividual patients because they display wide interindividual variability in their clearance.These drugs are administered as short infusions over a period of about 30 min to 1 h to treatvery serious, often life-threatening conditions. They can also produce very serious toxicity,including renal toxicity, which is usually reversible, and ototoxicity, which often is not

Page 197: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

CLINICAL APPLICATION OF THE TWO-COMPARTMENT MODEL 177

0.1

1

10

0 5 10 15 20

Cp

Time

Data point in distribution phase

Biased estimate of β and β half-life

Correct estimate of β and β half-life

FIGURE 8.9 Data points used to estimate the beta elimination half-life. Biased results are obtainedwhen plasma concentrations in the distribution phase are used to calculate t1/2,�.

reversible and may leave the patient with residual hearing loss. To maximize the therapeuticresponse of the drugs, and to minimize toxicity, aminoglycoside levels are usually monitoredin individual patients, and the patient’s individual half-life is determined to help guidetherapy. Because the aminoglycosides display two-compartmental characteristics, and one-compartmental pharmacokinetics are used to guide dosage adjustment, it is important toavoid the distribution phase when sampling for determination of the half-life. It is usual towait about 30 min after the end of a 30-min infusion before taking blood samples.

8.8.2 Determination of the Loading Dose

The two-compartment model has three volumes of distribution, each of which applies atdifferent times. V1 is the smallest and applies only at time zero, when the entire dose is inthe central compartment. The values of Vdss and V� are quite similar. The former appliesat steady state and the latter in the postdistribution phase. These two volumes provide verysimilar estimates of a loading dose. A loading dose calculated from V1 is much smaller,however.

When V1 is used to calculate a loading dose, the plasma concentration at time zerowill achieve the desired concentration, but it will fall rapidly as drug distributes to theperipheral compartment (Figure 8.10). When either of the other two volumes are used,the initial plasma concentration will overshoot the desired concentration but will quicklyapproach the target, depending on the duration of the distribution phase (Figure 8.10).

The compartmental location of the site of action or toxicity is an important factor indetermining which volume it is appropriate to use. If the site of action or toxicity is inthe peripheral compartment, the high initial plasma concentrations plasma concentrationsassociated with using Vdss or V� should not, within reason, lead to adverse effects. If,on the other hand, the site of action or toxicity is in the central compartment, the highinitial concentration could cause toxicity. In this case, a loading dose based on V1 couldbe used. But as the drug distributes to the peripheral compartment, plasma concentrationswill rapidly become subtherapeutic. Alternatively, a total loading dose based on Vdss or V�

could be estimated and split into a few smaller units that could be administered in severalminute intervals, depending on the speed of drug distribution and the patient’s response.

Page 198: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

178 TWO-COMPARTMENTAL PHARMACOKINETICS

Cp

Cpdes

Loading dose based on V1hits target but rapidly declines

Loading dose based on Vdss or Vβexceeds target and may produce toxicity

Time

FIGURE 8.10 Plasma concentrations produced by different loading doses. Loading doses cal-culated using V1 achieve the desired plasma concentration (Cpdes) immediately, but rapidly decline(dotted line). The initial concentration produced by a loading dose calculated using Vdss or V� exceedsthe target and may produce toxicity (solid line).

Lidocaine and procainamide are examples of drugs with sites of action/toxicity in thecentral compartment.

Example 8.2 Lidocaine is an antiarrhythmic drug that is used in the treatment of prema-ture ventricular contractions. A 70-kg male patient is to receive an intravenous infusion oflidocaine to maintain a plasma concentration of 2 mg/L. Calculate a loading dose of lido-caine hydrochloride (S = 0.87) to achieve this plasma concentration immediately. Lidocainehas the following volumes: V1 = 0.5 L/kg and Vdss = 1.3 L/kg.

Solution The site of action and toxicity of lidocaine is in the central compartment, andthe initial loading dose is often based on the volume of the central compartment (V1):

DL = Cpss · Vd

S · F

DL = 2 mg/L × 0.5 L/kg × 70 kg

0.87 × 1

= 80 mg

To avoid toxicity, the loading dose would be administered over a period of about 2 to 3 min.The plasma concentration will fall rapidly as the drug distributes throughout the tissues ofthe peripheral compartment. To maintain the plasma concentration at a therapeutic level,it is often necessary to give additional injections which are usually about half the initialloading dose.

Digoxin’s site of action and toxicity is in the peripheral compartment, and loading dosesof digoxin are based on the larger Vdss or V�. However, because digoxin has a narrowtherapeutic range, the total dose is still split into smaller units, which are given in 6-h

Page 199: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

CLINICAL APPLICATION OF THE TWO-COMPARTMENT MODEL 179

intervals. The patient is monitored between the units, and subsequent doses are withheld ifnecessary.

8.8.3 Evaluation of a Dose: Monitoring Plasma Concentrationsand Patient Response

In the two-compartment model the tissues of the central compartment are always in equi-librium with the plasma. This is not the case for the tissues of the peripheral compartment.Immediately after an intravenous dose, the plasma concentration is at its highest and theperipheral compartment concentration is zero. During the course of the distribution phase,the concentration in the tissues of the peripheral compartment increases and approachesequilibrium with the plasma (Figure 8.11). If the site of action of the drug is in the pe-ripheral compartment, the plasma concentration will reflect concentrations at the site ofaction only during the postdistribution phase, and the therapeutic range of these drugs willapply only at this time. This concept is particularly important for digoxin because it hasan exceptionally long distribution phase (over 4 h after an intravenous dose and over 6 hafter an oral dose) and its site of action in the peripheral compartment. Because digoxinhas a narrow therapeutic range and variable pharmacokinetics, serum levels are monitoredto check the adequacy of the dose. It is important that digoxin’s serum levels be evaluatedonly at least 6 h after an intravenous dose and about 8 h after an oral dose, as it is only atthese times that the plasma concentration (serum concentration) reflects the concentrationat the site of action.

Dru

g C

once

ntra

tion

Time

MTC

Initial high Cp values above the MTC occur during the distributionphase. During this period Cp is not reflective of the drug concentrationin tissues of the peripheral compartment.

Tissue within the peripheral compartment. Oncedistribution is complete, the value of Cp reflectsthe value of the drug concentration in this tissue

Plasma

FIGURE 8.11 Time course of drug concentrations in the plasma (Cp) and a hypothetical tissueof the peripheral compartment after an intravenous dose. At time zero, Cp is at its maximum valueand the concentration in the tissue is zero. During the distribution phase, Cp decreases and the tissueconcentration increases. During this period Cp is not reflective of the concentration in the peripheralcompartment. Once distribution is complete, the ratio of the two concentrations remains constant. Itis only at this time that the Cp reflects drug concentration in the peripheral compartment. For drugswhose site of action is in the peripheral compartment, it is only at this time that the therapeutic rangeholds and Cp can be used to predict the effect of the drug.

Page 200: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

180 TWO-COMPARTMENTAL PHARMACOKINETICS

PROBLEMS

8.1 Plasma concentration–time data collected after the intravenous administration of adose (100 mg) of a drug (S = 1) was best fit by a two-compartment model. Thefollowing equation was obtained:

Cp = 8.47e−2.62t + 1.38e−0.15t

where Cp is in mg/L and time in hours. Calculate the following parameters for thisdrug: k10, k12, k21, V1, V�, Vdss, Cl, and Cld.

8.2 How long would the distribution phase last for the drug presented in Problem 8.1?

8.3 If a 50-mg dose of the drug discussed in Problem 8.1 was administered, estimate:

(a) Cp at 0.5 h

(b) Cp at 6 h

8.4 The drug discussed in Problem 8.1 was administered to a patient who was suspected tohave altered clearance. Three plasma samples were collected after the administrationof a 100-mg IV dose. The samples were analyzed for unchanged drug and the dataare shown in Table P8.4.

TABLE P8.4

T (h) Cp (mg/L)

0.5 2.202.0 0.82

12 0.19

Calculate t1/2,�.

8.5 A study was conducted to see how renal impairment might affect a drug’s disposition(fe = 0.95). The drug’s pharmacokinetic parameters were estimated in a group ofpatients with normal renal function and in a group that had impaired renal function.Table P8.5 summarizes the results. From this information:

TABLE P8.5

Renal Function

Parameter Normal Less Than 20% Normal

Cl (L/h) 16.8 ± 0.87 2.2 ± 1.2b

V� (L) 46 ± 5.6 36 ± 6.3b

Vdss (L) 37 ± 5.5 34 ± 6.3t1/2, � (h) 1.80 ± 0.65 9.67 ± 2.7b

aMean parameter values ± the standard deviation.bOf statistical significance.Cl is clearance, V� is the volume of distribution in thepost-distribution phase, Vdss is the steady-state volume ofdistribution and t1/2� is the elimination half-life.

Page 201: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc08 JWBS062-Rosenbaum February 3, 2011 7:34 Printer Name: Yet to Come

RECOMMENDED READING 181

(a) Do you conclude that renal impairment affects this drug’s elimination?

(b) Do you conclude that renal impairment affects this drug’s elimination distribu-tion?

(c) Discuss how renal impairment affects each parameter.

8.6 A 72-year-old female patient with congestive heart failure has been taking digoxin250 �g for several years. On a routine visit to the community health center shediscusses her medication with her health team. She says she finds it easy be rememberto take her digoxin, as it is a once-daily regimen and she always takes it with herbreakfast at 8:00 a.m. Her physician would like to maintain her serum digoxin in therange 0.5 to 1.5 �g/L. A blood sample is taken just before she leaves at 9:30 a.m. Thefollowing day the results come back from the lab and reveal a serum digoxin level of2.6 �g/L. Is this patient at risk for digoxin toxicity?

8.7 A 100-mg dose of a drug was administered intravenously. Plasma samples weretaken at various times after the dose and analyzed for unchanged drug. The dataare listed in Table P8.7. The data indicate that the drug follows two-compartmentalpharmacokinetics. Set up an Excel worksheet and:

(a) Plot Cp against time on the linear and semilogarithmic scales.

(b) Determine the intercepts and macro rate constants: A, B, �, and �.

(c) Determine the micro rate constants: k10, k12, and k21.

(d) Determine the primary pharmacokinetic parameters: Cl, Cld, V1, V�, and Vdss

[use the formula Vdss = V1 (k12 + k21)/k21].

TABLE P8.7

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0.1 7.12 1.5 0.880.2 5.15 2 0.840.4 2.88 3 0.80.5 2.24 5 0.740.7 1.51 7 0.681 1.07 12 0.56

RECOMMENDED READING

1. Wagner, J. (1993) Pharmacokinetics for the Pharmaceutical Scientist, Technomic, Lancaster, PA.

2. Jambhekar, S. S., and Breen, P. J. (2009) Basic Pharmacokinetics, Pharmaceutical Press, London.

Page 202: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

9PHARMACOKINETICS OFEXTRAVASCULAR DRUGADMINISTRATION

9.1 Introduction

9.2 Model for First-Order Absorption in a One-Compartment Model9.2.1 Model and Equations9.2.2 Determination of the Model Parameters

9.2.2.1 First-Order Elimination Rate Constant9.2.2.2 Elimination Half-Life9.2.2.3 First-Order Absorption Rate Constant9.2.2.4 Volume of Distribution9.2.2.5 Clearance

9.2.3 Absorption Lag Time9.2.4 Flip-Flop Model and Sustained-Release Preparations9.2.5 Determinants of Tmax and Cmax

9.2.5.1 Tmax

9.2.5.2 Cmax

9.3 Bioavailability9.3.1 Bioavailability Parameters

9.3.1.1 Rate of Drug Absorption9.3.1.2 Extent of Drug Absorption

9.3.2 Absolute Bioavailability9.3.3 Relative Bioavailability9.3.4 Bioequivalence9.3.5 Example Bioavailability Analysis

9.4 Simulation Exercise

Problems

Objectives

The material in this chapter will enable the reader to:

1. Apply a pharmacokinetic model to understand the derivation of a general equationthat describes plasma concentration of a drug at any time after the administration ofan oral dose

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

182

Page 203: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

INTRODUCTION 183

2. Understand the meaning and significance of the absorption parameters

3. Understand the factors that control the time it takes to achieve therapeutic plasmaconcentrations

4. Understand the factors that control the value of the peak plasma concentration

5. Apply the model to determine the major pharmacokinetic parameters from plasmaconcentration–time data obtained after the oral administration of a drug

6. Define absolute and relative bioavailability

7. Understand how bioavailability is assessed

9.1 INTRODUCTION

Extravascular drug administration refers to any route of drug administration where the drugis not administered directly into the systemic circulation. Generally, drugs administered byextravascular routes rely on the systemic circulation to deliver them to their site of action.Thus, access of the drug to the systemic circulation, or absorption, is a critical pharmacoki-netic characteristic of extravascular administration. The typical plasma concentration–timeprofile observed after extravascular drug administration is shown in Figure 9.1. After drugadministration the plasma concentration increases gradually as a result of absorption, apeak is achieved, and then plasma concentrations fall. The pharmacokinetics of absorptionare presented in this chapter, with the major emphasis being placed on oral drug adminis-tration. However, almost all of the general principles discussed in this chapter apply equallyto other forms of extravascular drug absorption.

The administration of drugs by the oral route is noninvasive and very convenient forpatients. As a result, it is the most common, and in most situations, the preferred route ofdrug administration. Several oral dosage forms are available to accommodate the needs of avariety of patients. Solid dosage forms, such as tablets and capsules, are the most common,

Cp

Time

Cmax

Tmax

A

B

C

D

FIGURE 9.1 Typical plasma concentration–time profile after extravascular drug administration.The curve can be considered to have four areas: A, B, C and D.These will be discussed in the text.Point B corresponds to the peak plasma concentration. The value of the peak is Cmax and the timeof the peak is Tmax.

Page 204: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

184 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

but liquid dosage forms, including syrups, suspensions, and emulsions, are available togroups of patients, such as children and the elderly, who may have difficulty swallowingsolid dosage forms. Despite the popularity of oral dosage forms, they cannot be used in allsituations. Some of the major reasons that preclude the oral route include the destructionof a drug by components of the gastrointestinal fluid and/or its inability to pass throughthe intestinal membrane; extensive presystemic extraction; an immediate drug action isrequired; the dose must be administered with great accuracy; or the patient is unconscious,uncooperative, or nauseous.

In view of the widespread use of the oral route, it is important to appreciate the uniquepharmacokinetic characteristics of oral administration and understand how these may af-fect drug response. The absorption process brings two additional parameters into the phar-macokinetic model: the bioavailability factor (F) and a parameter for the rate of drugabsorption, the first-order absorption rate constant (ka). Unlike clearance and volume ofdistribution, these two parameters are properties not only of the drug itself but also of thedosage form, and can vary from one brand of a drug to another.

In this chapter we focus on presenting a pharmacokinetic model for orally administereddrugs and discuss how the various model parameters affect the plasma concentration–timeprofile. The determination of the model parameters and the assessment of bioavailability areaddressed. In clinical practice, the absorption parameters (ka and F) cannot be determinedfrom the limited (1 to 2) samples available from patients. Consequently, the equations fororal administration are not frequently used clinically to individualize doses for patients.

9.2 MODEL FOR FIRST-ORDER ABSORPTION IN AONE-COMPARTMENT MODEL

9.2.1 Model and Equations

As discussed in Chapter 2, the absorption of drugs from the gastrointestinal tract oftenfollows first-order kinetics. As a result, the pharmacokinetic model can be created simplyby adding first-order absorption into the central compartment of the one-compartmentmodel (Figure 9.2). The gastrointestinal tract is represented by a compartment in Figure9.2. However, since it is outside the body, the body is still modeled as a single compartment.The amount of drug in the gastrointestinal is influenced only by first-order drug absorption:

dAGI

dt= −ka · AGI (9.1)

where AGI is the amount of drug in the gastrointestinal tract and ka is the first-orderabsorption rate constant. Integrating, we have

AGI = AGI,0 · e−ka t (9.2)

where AGI,0 is the initial amount of drug in the gastrointestinal tract, and is equal to theeffective dose (S · F · D):

AGI = S · F · D · e−ka t (9.3)

Page 205: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

MODEL FOR FIRST-ORDER ABSORPTION IN A ONE-COMPARTMENT MODEL 185

VdAbCp

Initial Conditions: t = 0 A = SFD, Ab = 0

E

Cl*Cp or Ab*k

AA *ka

Outside the BodyA The gastrointestinal

tract

Inside the Body

GI

GI

GI

FIGURE 9.2 Pharmacokinetic model for first-order absorption in a one-compartment model. Aband AGI are the amounts of drug in the body and gastrointestinal tract, respectively; k and ka arethe first-order rate constants for elimination (E) and absorption (A), respectively; Cp is the plasmaconcentration and the concentration of drug in the compartment. The compartment has a volume ofVd, the drug’s volume of distribution; Cl is the clearance; and the effective dose is the product theproduct of S (salt factor), F (bioavailability factor), and D (the dose administered).

Equation (9.3) shows that the amount of drug in the gastrointestinal tract starts off at S · F · Dand decays to zero by infinity. The speed of the decay is dependent on the first-order rateconstant for absorption.

The amount of drug in the body at any time will depend on the relative rates of drugabsorption and elimination:

rate of change of the amount of drug in the body = rate of inputs − rate of outputs

dAb

dt= ka · AGI − k · Ab (9.4)

Consideration of equation (9.4) provides an explanation for the shape of the plasmaconcentration–time curve (Figure 9.1). Immediately after drug administration (area A),plasma concentrations increase because the rate of absorption is greater than the rate ofelimination. The amount of drug in the gastrointestinal tract is at its maximum, so therate of absorption is also maximum. In contrast, initially the amount of drug in the bodyis small, so the rate of elimination is low. As the absorption process continues, drugis depleted from the gastrointestinal tract, so the rate of absorption decreases. At thesame time, the amount of drug in the body increases, so the rate of elimination increases.At the peak (B), the rate of absorption is momentarily equal to the rate of elimination. Afterthis time the rate of elimination exceeds the rate of absorption and plasma concentrationsfall (area C). Eventually, all the drug is depleted from the intestinal tract and drugabsorption stops. At this time (area D) the plasma concentration is influenced only byelimination.

Page 206: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

186 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

Equation (9.3) can be substituted into equation (9.4), which can then be integrated toyield

Ab = S · F · D · ka

ka − k· (e−kt − e−ka t ) (9.5)

But Cp = Ab/Vd:

Cp = S · F · D · ka

Vd · (ka − k)· (e−kt − e−ka t ) (9.6)

Thus, the plasma concentration at any time after an oral dose is described by a biexponentialequation.

9.2.2 Determination of the Model Parameters

The pharmacokinetic model for extravascular administration has four fundamental pa-rameters. Two of the parameters, clearance and volume of distribution, are disposition(elimination and distribution) parameters that are not dependent on the nature of the ex-travascular dosage form and drug absorption. The other two parameters, the bioavailabilityfactor (F) and the first-order rate constant for absorption (ka) are functions of both the drugand dosage form. Thus, these parameters can vary from one type and brand of dosage formto another. As always, the one-compartment model has the derived parameter for the rateof elimination (the elimination rate constant and the half-life).

Experimentally the parameters of the model are determined by following a protocolsimilar to those presented in earlier chapters. Oral doses are administered to a group ofindividuals, and plasma concentrations are determined at various times after the dose. Itis important to obtain several plasma concentration samples around the peak in order tocharacterize this feature adequately. Data from each person are subject to pharmacokineticanalysis to determine the pharmacokinetic parameters. Each parameter is then averagedacross the group to calculate the mean and standard deviation. The basic equation for oralabsorption [equation (9.6)] is biexponential and cannot be made linear by transforming thedata to the logarithm scale. Generally, pharmacokinetic analysis is conducted using com-puter software that can perform nonlinear regression analysis to find the model parametersthat will most closely match the data observed. However, as was the case with the two-compartment model, the parameters can be obtained by linearizing the data through curvestripping. A discussion of this method helps to demonstrate the importance and influenceof each parameter of the model. Additionally, it is a useful procedure to employ if initialestimates of the parameters are needed for computer analysis.

9.2.2.1 First-Order Elimination Rate ConstantAt some time after drug administration, the entire dose will have been absorbed and theplasma concentration will decline in a monoexponential manner, due only to first-orderdrug elimination. Mathematically:

� For extravascular administration, ka is usually greater than k.� As a result, e−kat becomes equal to zero before e−kt.

Page 207: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

MODEL FOR FIRST-ORDER ABSORPTION IN A ONE-COMPARTMENT MODEL 187

� When this occurs the equation for the plasma concentration (9.6) reduces to

Cp = S · F · D · ka

Vd · (ka − k)· (e−kt − 0)

Cp = S · F · D · ka

Vd · (ka − k)· e−kt

(9.7)

Taking logarithms gives

ln Cp = lnS · F · D · ka

Vd · (ka − k)− kt (9.8)

This is the equation of a straight line. Thus, at later times, the plot of ln(Cp) againsttime becomes straight with a slope of the equal to the negative value elimination rateconstant (−k) and an intercept of ln[S · F · D · ka/Vd (ka − k)] (Figure 9.3). This periodis referred to as the terminal elimination phase.

9.2.2.2 Elimination Half-LifeAs always, the elimination half-life is calculated from the elimination rate constant:

t1/2 = 0.693

k(9.9)

9.2.2.3 First-Order Absorption Rate ConstantThe period before the elimination phase is referred to as the absorption phase. During thisperiod the plasma concentration is under the influence of both absorption and elimination,and the equation is biexponential. Curve stripping is used to separate the two exponentialfunctions during this phase.

lnS•F•D•ka

Vd•(ka – k)

Slope = –k

ln C

p

Time

FIGURE 9.3 Plot of logarithm of plasma concentration against time. At later times a lin-ear relationship is observed. The slope of the line is −k and the intercept is ln S · F · D · ka/[Vd (ka − k)].

Page 208: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

188 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

To make the equations less cumbersome, let

I = S · F · D · ka

Vd · (ka − k)(9.10)

The full expression for Cp [equation (9.6)] is given by

Cp = I · (e−kt − e−ka t ) (9.11)

Let the plasma concentration during the elimination phase and its back-extrapolated com-ponent be Cp′. Thus,

Cp′ = I · e−kt (9.12)

In the elimination phase, Cp′ = Cp, but in the absorption phase, Cp′ � Cp. Subtractingequation (9.11) from equation (9.12) yields

Cp′ − Cp = I · e−kt − I · (e−kt − e−ka t )

Cp′ − Cp = I · e−ka t (9.13)

Taking logarithms gives us

ln(Cp′ − Cp) = ln I − ka · t (9.14)

This is the equation of a straight line of slope (−ka) and intercept lnI. It is referred to as thefeathered line.

The plot of ln(Cp′ − Cp) is constructed as follows:

1. Note the time of the given data points in the absorption phase (open circles in Figure9.4).

2. From the back-extrapolated part of the plot of ln Cp′ against time, read off thecorresponding values of Cp′ at these times (solid circles in Figure 9.4)

3. Subtract the given data points from these values of Cp′ to obtain the values of Cp′ −Cp.

4. Plot Cp′ − Cp against time on the semilogarithmic scale (diamonds in Figure 9.4).

5. Notice that the two straight lines intercept at the same place on the Cp-axis.

This procedure is performed most conveniently using either semilogarithmic (base 10logarithms) graph paper or using an Excel worksheet. The creation of an Excel worksheetis described in Appendix C.

The absorption half-life can be determined from the first-order rate constant for absorp-tion:

t1/2,absorption = 0.693

ka(9.15)

Page 209: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

MODEL FOR FIRST-ORDER ABSORPTION IN A ONE-COMPARTMENT MODEL 189

ln I

Slope = –k

ln Cp′ ( )ln Cp ( )

ln (Cp′ – Cp) ( )

Time

Slope = –ka

FIGURE 9.4 Plot of logarithm of Cp (solid line), Cp′ (upper dashed line), and Cp′ − Cp (broaddashed line) against time. The values of Cp′ (closed circles) that correspond to the time of the givenvalues of Cp (open circles) are noted. ln(Cp′ − Cp) (diamonds) is plotted against time to obtain astraight line of slope −ka and intercept ln I, where I is S · F · D · ka/[Vd (ka − k)].

This parameter is useful to determine the approximate time for absorption to be completed(3 to 5 absorption t1/2 values).

9.2.2.4 Volume of DistributionThe absolute values of the primary pharmacokinetic parameters, volume of distribution, andclearance cannot be determined from oral data alone because the bioavailability factor (F)is unknown. The volume of distribution relative to F (Vd/F) can, however, be determinedfrom the intercept of the plots of ln Cp′ and ln(Cp′ − Cp) against time (Figure 9.4). Theintercept is lnI, where

I = S · F · D · ka

Vd · (ka − k)(9.16)

Thus,

Vd

F= S · D · ka

I · (ka − k)(9.17)

9.2.2.5 ClearanceClearance may be calculated from the first-order rate constant and the volume of distribution.If only oral data are available, F cannot be determined and only clearance relative to thebioavailability (Cl/F) can be estimated. This clearance (Cl/F) is called apparent clearanceor oral clearance, and can be calculated from k and Vd/F:

Cl

F= k · Vd

F(9.18)

Page 210: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

190 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

Oral clearance can also be calculated from the area under the plasma concentration–timecurve (AUC) from zero to infinity:

Cl

F= S · D

AUC(9.19)

Then AUC can be calculated:

AUC =∫ ∞

0Cp · dt = S · F · D · ka

Vd · (ka − k)·(

1

k− 1

ka

)= I ·

(1

k− 1

ka

)(9.20)

Alternatively, the AUC can be estimated from the trapezoidal rule.

Example 9.1 Parameter Determination from Plasma Concentration–Time Data. Theplasma concentration–time data in Table E9.1A were obtained after the administration ofa 100-mg oral dose to a healthy volunteer. Analyze the data and determine (a) k, (b) ka,(c) Vd/F, and (d) Cl/F by creating an Excel worksheet as described in Appendix C. Assumethat S = 1.

Solution A detailed solution and associated plots are shown in Appendix C, Figure C.8.

(a) From the straight line from the last three data points on the semilogarithmic scale:

slope = −0.23 h−1

k = 0.23 h−1

ln I = 1.77

I = 5.85 mg/L where I = S · F · D · ka

Vd · (ka − k)

(b) The earlier values along the back-extrapolated portion of this terminal eliminationline are calculated using

Cp′ = I · e−kt

where at later times, Cp = Cp′, but at earlier times, Cp′ � Cp. The values of Cp′ atthe times of the early given data points are calculated in Table E9.1B. The values of

TABLE E9.1A

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0 0 2 3.430.6 2.74 2.6 3.120.8 3.13 3 2.891 3.37 4 2.331.4 3.55 7 1.171.8 3.5 12 0.37

Page 211: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

MODEL FOR FIRST-ORDER ABSORPTION IN A ONE-COMPARTMENT MODEL 191

TABLE E9.1B

T (h)Cp (mg/L):

Given ValuesCp′ (mg/L)

= 5.85e−0.23t Cp′ − Cp

0 0 5.85 5.850.6 2.74 5.10 2.360.8 3.13 4.87 1.741 3.37 4.65 1.281.4 3.5 4.24 0.691.8 3.55 3.87 0.372 3.43 3.69 0.262.6 3.12 3.22 0.10

Cp′ − Cp are determined and the values of ka and the intercept are determined fromthe line of ln(Cp′ − Cp) against time (Figure 9.4):

slope = −1.57 h−1

ka = 1.57 h−1

ln I = 1.80

I = 6.07 mg/L = S · F · D · ka

Vd · (ka − k)

(c) The value of Vd is determined. Since intravenous data are not available for this drug,pure Vd cannot be determined, only Vd/F:

S = 1Vd

F= D · ka

I · (ka − k)

= 20.0 L

(d) Cl/F can be calculated as follows:

1. From k and Vd/F:

Cl

F= Vd · k

F= 20.0 × 0.23

= 4.61 L/h

2. From the AUC:

AUC = S · F · D · ka

Vd · (ka − k)·(

1

k− 1

ka

)= 21.71 mg · h/L

Cl

F= D

AUC= 4.61 L/h

Page 212: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

192 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

ln C

once

ntra

tion

Timet0

FIGURE 9.5 Semilogarithmic plot of plasma concentration against time. The terminal linear lineand the feathered line do not intercept at the y-axis. Their point of intersection corresponds to theabsorption lag time (t0).

9.2.3 Absorption Lag Time

The absorption of a drug from the oral route may not occur immediately. Absorption can bedelayed by several factors, including slow disintegration, poor dissolution, delayed stomachemptying, or a coating that delays drug release. Some controlled-release dosage forms aredesigned specifically to incorporate a delay in absorption. In all these cases an absorptionlag time or delay can be incorporated into the pharmacokinetic model. Experimentally, theabsorption lag time is apparent in clinical data if the back-extrapolated terminal eliminationline and the feathered absorption line intercept at a point to the right of the y-axis. The timeat which they intercept corresponds to the absorption lag time (Figure 9.5). The absorptionlag time simply shifts the curve to the right and is accommodated in equations by subtractingthe lag time from the absolute time in the basic equation (9.6):

Cp = S · F · D · ka

Vd · (ka − k)· (

e−k(t−t0) − e−ka (t−t0))

(9.21)

where t0 is the absorption lag time.

9.2.4 Flip-Flop Model and Sustained-Release Preparations

Recall the mathematical procedure to separate the two exponential components of the fullequation for the plasma concentration (9.6) assumed that the rate constant for absorptionwas greater than the rate constant for elimination. As a result,

e−ka t → 0 before e−kt

This is not always the case.Drugs that have large elimination rate constants (short half-lives) are eliminated rapidly

and often need to be administered very frequently to maintain therapeutic plasma con-centrations. To improve patient adherence, these drugs are commonly formulated intoprolonged-release products that slow down absorption and permit less frequent dosing. The

Page 213: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

MODEL FOR FIRST-ORDER ABSORPTION IN A ONE-COMPARTMENT MODEL 193

formulation modification made to reduce the rate of absorption results in a reduction inthe absorption rate constant (ka). Under these conditions the elimination rate constant maybecome greater than the absorption rate constant. As a result,

e−kt → 0 before e−ka t

Under these conditions the basic formula for Cp (9.6) at later times reduces to

Cp = S · F · D · ka

Vd · (ka − k)· (

0 − e−ka t)

Cp = − S · F · D · ka

Vd · (ka − k)· e−ka t

Cp = S · F · D · ka

Vd · (k − ka)· e−ka t

(9.22)

Taking logarithms yields

ln Cp = lnS · F · D · ka

Vd · (k − ka)− kat (9.23)

In this situation the terminal fall in Cp is controlled not by k but by ka. The slope of thecorresponding feathered line is controlled by elimination and equal to −k (Figure 9.6).

This is referred to as a flip-flop model and is frequently the case for sustained-releasepreparations where drugs have very large values of k and are formulated in a manner tocreate small values of ka. A flip-flop model may be identified by comparing the value of theelimination rate constant or half-life to values obtained after intravenous administration. Aflip-flop model can also be identified if the dosage form is administered to patients withaltered clearance. The altered clearance will be apparent in changes of the slope of thefeathered line and not the terminal line.

–k

–ka

In C

p

Time

FIGURE 9.6 Semilogarithmic plot of plasma concentration against time for the flip-flop model.When the elimination rate constant (k) is greater than the absorption rate constant (ka), the negativeslope of the terminal line is ka and the negative slope of the feathered line is k.

Page 214: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

194 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

9.2.5 Determinants of Tmax and Cmax

The presence of a peak in the plasma concentration–time profile is characteristic of ex-travascular administration (Figure 9.1). The peak can be summarized by the value of thepeak plasma concentration (Cmax) and the time at which it occurs (Tmax) (Figure 9.1). Thesetwo metrics can have an important influence on drug response. The time of the peak cancontrol the onset of action of the drug, and Cmax may determine if a dose is subtherapeutic,therapeutic, or toxic. The values of Cmax and Tmax can also easily be measured directlyfrom the data, and they frequently play an important role in evaluating the results of clin-ical pharmacokinetic studies, including bioavailability studies and drug–drug interactionstudies. To understand the factors that control Cmax and Tmax, equations must be derivedfor these two parameters.

9.2.5.1 Tmax

When the plasma concentration reaches its peak, t = Tmax. At the peak,

dCp

dt= 0

The basic equation for Cp [equation (9.6)] is differentiated and set to zero. Time in theequation is set to Tmax. The equation is rearranged to yield

Tmax = ln(ka/k)

ka − k(9.24)

From equation (9.24) it can be seen that the value of Tmax is a function of the first-orderelimination rate constant and the first-order absorption rate constant.

Under conditions where elimination remains constant, Tmax becomes a function of onlythe absorption rate constant (ka). Bioavailability studies are performed to evaluate a drug’sabsorption properties among different formulations. These studies are designed to minimizevariability in elimination, and under these conditions Tmax can be used to assess the rate ofdrug absorption. Because ka is present in both the numerator and denominator of equation(9.24), it is difficult to predict how ka will affect Tmax. Figure 9.7b shows that as ka increases,

0

2.5

5

0

2.5

5

0 0.5 1 1.5

Tm

ax (

h)

Tm

ax (

h)

k (h–1) ka (h–1)

(a) (b)

0 0.5 1 1.5 2

FIGURE 9.7 Dependency of time of peak plasma concentration (Tmax) on the elimination rateconstant (k) (a) and the absorption rate constant (ka) (b) Tmax was calculated for values of k from 0.05to 1.2 h−1 with ka fixed to 0.6 h−1. Tmax was also calculated for values of ka from 0.1 to 1.8 h−1 withk fixed at 0.4 h−1.

Page 215: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

BIOAVAILABILITY 195

Tmax decreases. This is in keeping with common sense: If ka increases, the rate of absorptionincreases and it will take less time to reach the peak (Tmax decreases).

Under conditions where elimination does not remain constant, Tmax will change ifeither the rate of absorption and/or the rate of elimination changes. Figure 9.7a shows therelationship between k and Tmax. It can be seen that as k increases, Tmax decreases. This alsomakes sense based on the knowledge that after a dose the rate of absorption decreases andthe rate of elimination increases, and the peak occurs when the rate of elimination equalsthe rate of absorption. This will occur fastest when the elimination rate constant is high.

9.2.5.2 Cmax

The peak plasma concentration (Cmax) is the plasma concentration at time Tmax. Thus, anexpression for Cmax may be obtained by substituting the expression for Tmax [equation(9.24)] into equation (9.6) and rearranging to solve for Cmax:

Cmax = S · F · D

Vd· e−kTmax (9.25)

Equation (9.25) demonstrates that Cmax is dependent on all the pharmacokinetic parametersof a drug. It is directly proportional to the effective dose, dependent on the volume ofdistribution, and through its dependency on Tmax, the rate of absorption and elimination.

Bioavailability studies are designed to minimize variability in the disposition parameters(clearance, volume of distribution, and the elimination rate constant). Under these condi-tions Cmax will only be a function of F and ka. It is directly proportional to F: If F doubles,Cmax doubles. This is also in keeping with common sense: If F increases, the effective doseof the drug increases and thus Cmax increases. An inverse relationship exists between Cmax

and Tmax. For example, if the rate of absorption increases, absorption is more rapid, thetime to peak decreases, and the value of the peak increases.

The value of Cmax is frequently evaluated in drug–drug interaction studies. In thesestudies the clearance of a drug and/or the volume of distribution of a drug may varybetween the control and test conditions. Under these circumstances, changes in Cmax maybe brought about by changes in the clearance, volume of distribution, the rate of drugabsorption, and/or the extent of absorption.

9.3 BIOAVAILABILITY

The bioavailability of a dosage form is the rate, and extent to which, the drug reaches thesystemic circulation. It is a very important characteristic of an oral dosage form: The rate ofabsorption controls the speed with which therapeutic plasma concentrations are achieved,and more important, the extent of drug absorption controls the effective dose of a drug. Asdiscussed previously, bioavailability is a property not only of the drug but also of the dosageform. It is important that it remain constant among different batches of a product. It is alsoimportant that it be constant among a drug’s brand name product and generic equivalents,as these products may be used interchangably in patients.

9.3.1 Bioavailability Parameters

9.3.1.1 Rate of Drug AbsorptionFrom a consideration of the pharmacokinetic model and equations associated with oralabsorption, the most obvious way to assess the rate of absorption would appear to be

Page 216: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

196 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

the measurement of the first-order rate constant for absorption (ka). However, it is fre-quently very difficult to obtain precise estimates of this parameter. Many drugs displaytwo-compartment characteristics, and early after drug administration, drug absorption, dis-tribution, and elimination occur simultaneously, and it can be very difficult to separatethese processes to measure the absorption rate constant. Additionally, the measurement ofka assumes that absorption is a single first-order process. But the absorption of some drugsmay be more complex and may involve multiple first- and/or zero-order processes. As aresult, two other metrics are used to assess the rate of absorption: Tmax [equation (9.24)]and Cmax [equation (9.25)]. It was shown previously that when a drug is administered underconditions where disposition (distribution and elimination) is unlikely to vary, Tmax andCmax are measures of the rate of drug absorption. These metrics are obtained directly fromthe plasma concentration–time data. Thus, Cmax and Tmax are simply the highest recordedplasma concentration and its corresponding time.

9.3.1.2 Extent of Drug AbsorptionThe extent of drug absorption is evaluated through assessment of the bioavailability factor(F) and Cmax, which is directly proportional to F [equation (9.25)] and can be read directlyfrom the data. The bioavailability factor is the fraction of the dose that is able to gain accessto the systemic circulation. It is assessed by means of the AUC, which is a measure of thebody’s exposure to a drug. Recall that

AUC = S · F · D

Cl(9.26)

Equation (9.26) demonstrates that the AUC is directly proportional to the effective dose ofdrug and inversely proportional to its clearance. The AUC is a measure of drug exposure,and for a given drug (clearance is constant) the AUC is directly proportional to the effectivedose:

AUC ∝ F · D (9.27)

or

AUC

D∝ F (9.28)

Assuming a constant value of S, the AUC per unit dose of a drug is directly proportionalto F.

In summary, bioavailability is assessed by measuring AUC, Cmax, and Tmax. AUC andCmax are measures of the extent of drug absorption (F), and Cmax and Tmax are measuresof the rate of drug absorption. Figure 9.8 shows the plasma concentration–time profile ofthree formulations of equal doses of a drug. The relative bioavailability characteristics ofthe three formulations are summarized as

FA = FB � FC

kaA � kaB = kaC

Page 217: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

BIOAVAILABILITY 197

0

1

2

3

4

0 5 10 15 20

Cp

(mg/

L)

Time (h)

A

B

C

AUCA = 21.6 mg•h/L

AUCB = 21.6 mg•h/L

AUCC = 13.0 mg•h/L

Tmax,B,CTmax,A

FIGURE 9.8 Plasma concentration–time profile of three different formulations of equal doses ofa drug. The rate of absorption from formulation B is the same as from formulation C. The rate ofabsorption from A is greater than that from B and C. The extent of absorption of A and B are thesame and greater than that from formulation C.

Thus,

AUCA = AUCB � AUCC (Figure 9.8)

Tmax,A � Tmax,B = Tmax,C (Figure 9.8)

Cmax,A � Cmax,B � Cmax,C (Figure 9.8)

9.3.2 Absolute Bioavailability

The actual value of F is referred to as the absolute bioavailability. It can only be determinedby comparing the AUC from an oral dosage form (PO) to the AUC after intravenousadministration (IV):

FPO

FIV= AUCPO

/DPO

AUCIV/

DIV(9.29)

But FIV = 1,

FPO = AUCPO/

DPO

AUCIV/

DIV(9.30)

Thus, F for an oral dosage form is the AUC per unit dose divided by the AUC per unit dosewhen the drug is given IV. A more computationally friendly version of equation (9.30) is

FPO = AUCPO · DIV

AUCIV · DPO(9.31)

Page 218: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

198 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

9.3.3 Relative Bioavailability

The bioavailability of an oral dosage form may also be determined through comparison toa standard oral preparation. This may be an oral solution, which as it is further along inthe absorption process, often has the highest bioavailability of all the oral dosage forms ofa drug. Alternatively, the bioavailability may be compared to a standard oral preparationor to one that has optimal bioavailability characteristics. These are examples of relativebioavailability. The relative bioavailability of a test product is

FT

FS= AUCT /DT

AUCS/DSor

AUCT · DS

AUCS · DT

where the subscript S stands for standard and T for test.

9.3.4 Bioequivalence

Bioequivalence is a special type of relative bioavailability. Two or more products areclassified as bioequivalent if a clinical study has demonstrated that the products haveessentially the same bioavailability. To demonstrate the bioequivalence of two oral products,their AUC and Cmax ratios are calculated in a group of subjects in a crossover study. The90% confidence interval of the average ratios for the group must lie within the range 80 to125%. Bioequivalence studies must be performed during drug development to demonstratethe equivalency of the various batches and trial formulations that are used in clinical studies.Drug companies must perform bioequivalence studies whenever major formulation changesare made. Bioequivalence studies are also conducted routinely by drug manufacturers whowish to market a generic form of an innovator or brand-name product. In addition to thedemonstration of bioequivalence, generic products must also be pharmaceutical equivalentsof the innovator product. That is, they must be the same type of dosage form (tablets,capsules, etc.), they must contain the same dose of the drug(s), and they must have the samechemical form (e.g., salt).

9.3.5 Example Bioavailability Analysis

An example of a bioavailability analysis can be found in the problems at the end ofChapter 10.

9.4 SIMULATION EXERCISE

Open the model “First-Order Absorption in a One-Compartment Model,” the link

http://www.ur i.edu/phar macy/faculty/rosenbaum/basicmodels.html#chapter9

Default settings for the model are dose = 100 mg, Cl = 4.6 L/h, Vd = 20 L, F = 1 andka = 0.6 h−1.

1. Review the objectives, the “Model Summary” page, and explore the model.

Page 219: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

PROBLEMS 199

2. Go to the “Transfer of Drug” page. Give a dose (100 mg) and observe howthe amount of drug changes over time in the compartments representing thegastrointestinal tract, the body, and the drug eliminated. Based on the amountof drug that has been eliminated at the end of the simulation, it is possible toobtain a minimum value of bioavailability (F ). By 12 h, 90 mg of drug has beeneliminated, so F must be at least 0.9. Since drug is still present in the body at12 h, the actual value of F will be greater than 0.9.

3. Go to the “Cp–Time Profile” page.Give the default dose (100mg) and observe theplasma concentration on the regular linear scale and the semilogarithmic scale.Note that at later times on the semilogarithmic scale, a straight-line relationshipis observed between ln Cp and time.

4. Compare the profiles after doses of 50, 100, and 200 mg. Note:� Cp at any time is proportional to dose.� Tmax is independent of dose.� AUC increases in proportion with dose.� The slope of the linear terminal slope of the plot of ln Cp is constant withdose—each line is parallel.

� The t1/2 remains constant as the dose changes.5. Go to the “Influence of ka and F” page.

6. Observe the effects of increases in ka:� Tmax gets smaller as ka increases.� Cmax increases as ka increases.� AUC is not affected.

7. Observe the effects of increases in F :� Tmax is not affected.� Cmax increases in proportion with increases in F .� AUC increases in proportion with increases in F .

8. Go to the “Flip-Flop Model” page. With the slow release (SR) switch off (down),give doses with different values of clearance. Note that the terminal slope of theplot of ln Cp versus time changes. Flip the SR switch on (up: green) and repeatthe exercise. Note that in this case the terminal slope of the plot of ln Cp versustime does not change with clearance. The lines are all parallel. This is becausethe slope of this line is controlled by ka, not by k .

PROBLEMS

9.1 A 75-mg dose of a drug was administered as an oral solution to nine healthy volunteers.Plasma samples were collected at various times after administration and were analyzedfor the parent drug. After at least 10 elimination half-lives, the study was repeated buton this occasions the drug (75 mg) was administered as an oral tablet. The results fromone of the subjects are given in Table P9.1. Analyze the data using a one-compartmentmodel with first-order absorption. Determine Cl/F, Vd/F, k, t1/2, and ka for eachformulation. Comment on any differences between the two formulations.

Page 220: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc09 JWBS062-Rosenbaum February 3, 2011 7:26 Printer Name: Yet to Come

200 PHARMACOKINETICS OF EXTRAVASCULAR DRUG ADMINISTRATION

TABLE P9.1

Solution Tablet

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0 0 0 01 0.78 1 0.462 1.21 2 0.753 1.42 3 0.914 1.5 4 15 1.49 5 1.036 1.43 6 1.027 1.35 7 0.989 1.14 10 0.8

12 0.84 16 0.4518 0.43 18 0.3624 0.21 24 0.18

9.2 The bioavailability of four different formulations (A, B, C, and D) of a drug are beingcompared. The results of a bioavailability study are provided in Table P9.2. How dothe rate and extent of drug absorption compare among the formulations?

TABLE P9.2

Formulation AUC (mg · h/L) Cmax (mg/L) Tmax (h)

A 21.4 1.68 3.4B 21.4 1.37 5.3C 16.1 1.26 3.2D 10.7 0.69 5.2

RECOMMENDED READING

1. Shargel, L., Wu-Pong, S., and Yu, A. B. C. (2004) Applied Biopharmaceutics and Pharmacoki-netics, 5th ed., McGraw-Hill, New York.

2. Jambhekar, S. S., and Breen, P. J. (2009) Basic Pharmacokinetics, Pharmaceutical Press, London.

Page 221: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

10INTRODUCTION TONONCOMPARTMENTAL ANALYSIS

10.1 Introduction

10.2 Mean Residence Time

10.3 Determination of Other Important Pharmacokinetic Parameters

10.4 Different Routes of Administration

10.5 Application of Noncompartmental Analysis to Clinical Studies

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand how the simpler approach of noncompartmental analysis can be used toestimate a drug’s pharmacokinetic parameters

2. Use noncompartmental analysis to determine the mean residence time after intra-venous administration

3. Use noncompartmental analysis to evaluate the results of a clinical study

10.1 INTRODUCTION

Clinical pharmacokinetic studies performed during drug development, and after a drug hasbeen marketed, are frequently designed to study how specific conditions or patient char-acteristics may affect a drug’s pharmacokinetics. The purpose of these studies is usuallyto try to identify special populations that may have altered dose requirements. The mag-nitude of any changes observed in one or more pharmacokinetic parameter(s) are used todevelop more appropriate dosing recommendations. For example, if food is found to reducea drug’s bioavailability, the labeling may be modified to recommend that the drug be takenon an empty stomach. If a perpetrator drug is found to reduce the clearance of a subjectdrug, the labeling of the subject drug may be modified to counterindicate the concomitant

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

201

Page 222: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

202 INTRODUCTION TO NONCOMPARTMENTAL ANALYSIS

administration of the two drugs. Alternatively, the labeling may recommend a dosage re-duction for the subject drug if it is taken with the perpetrator drug. Other common focusesof these studies include, but are not limited to, the effect of food on the bioavailability ofa drug, how a drug’s pharmacokinetics may be influenced by renal disease and/or hepaticdisease; age; genetics; and the activity of drug transporters.

Many of these studies are conducted using orally administered drugs, and their focus is toidentify and quantify any effects of the condition under study on the main pharmacokineticparameters of a drug: clearance, volume of distribution, bioavailability, and half-life. Ratherthan determining the pharmacokinetic parameters by trying to fit the data to compartmentalmodels, a simpler approach known as noncompartmental analysis (NCA) is used. NCAoffers several benefits over compartmental analysis. These include:

1. Fewer plasma samples may be required than in multicompartmental analysis.

2. The timing of the samples is not as critical as it is for multicompartmental analysis.

3. The modeling process is more straightforward and requires less experience and skillon the part of the modeler.

4. It avoids a problem frequently encountered with the compartmental approach, wherea drug displays one-compartmental properties in some subjects, and multi (two oreven three)-compartmental properties in other participants.

Although NCA can get quite complex, its application to the types of studies discussedabove is very straightforward. The starting point of the discussion on NCA is the meanresidence time of a drug.

10.2 MEAN RESIDENCE TIME

After an intravenous dose of a drug, the time that an individual drug molecule spends in thebody will vary enormously: Some drug molecules will be eliminated almost instantaneously,whereas others may reside in the body much longer. A few molecules may still be in thebody several weeks after the dose. The mean residence time (MRT) is defined as theaverage time spent in the body by a drug molecule. The determination of the MRT requiresno assumptions about the number of compartments involved in the drug’s disposition, andthe derivation of its formula is best understood by using an example of a situation that maybe encountered in everyday life.

Example 10.1 Suppose that a hotel is interested in determining the average length of stayof guests who attend a professional meeting at the hotel. Over the six days of the meeting,they track 20 guests. The results are shown in Table E10.1.

Four guests stayed for 1 day. This group provided a total of 4 days’ stay.

Five guests stayed for 2 days. This group provided a total of 10 days’ stay.

Six guests stayed for 3 days. This group provided a total of 18 days’ stay.

Two guests stayed for 4 days. This group provided a total of 8 days’ stay.

Two guests stayed for 5 days. This group provided a total of 10 days’ stay

One guest stayed for 6 days. This group provided a total of 6 days’ stay.

Page 223: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

MEAN RESIDENCE TIME 203

TABLE E10.1 Length of Stay of 20 Guests in a Hotel

Duration of Stay(days) (a)

Number ofGuests (b)

Total Length of Stay ofGroup (days) (a · b)

1 4 1 × 4 = 42 5 2 × 5 = 103 6 3 × 6 = 184 2 4 × 2 = 85 2 5 × 2 = 106 1 6 × 1 = 6Overall 20 56

Overall, the 20 guests stayed a total of (4 + 10 + 18 + 8 + 10 + 6) = 56 days. The meanresidence time is the total number of days spent divided by the total number of guests:56/20 = 2.8 days.

A drug’s MRT, which can be considered to be the mean time spent in the body by asmall mass of drug, is determined in the same way. A drug is administered intravenously.Assuming first-order elimination, the amount of drug eliminated per unit time is

dAe

dt= k · Ab (10.1)

where Ae is the amount of drug eliminated, k is the elimination rate constant, and Ab is theamount of drug in the body.

During the period dt, the small mass or amount of drug eliminated is given by

dAe = k · Ab · dt (10.2)

During the period dt, the amount of drug eliminated is equal to k · Ab · dt (equivalent to b inTable E10.1). This amount of drug has a residence time of t (equivalent to a in Table E10.1).The total amount having this residence time is (a · b), or

k · Ab · dt · t (10.3)

The sum of all the residence times from the time of administration (t = 0) until all the drughas been eliminated (t = ∞) is

∞∑

0

k · Ab · dt · t (10.4)

or

k∞∑

0

Ab · t · dt (10.5)

Page 224: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

204 INTRODUCTION TO NONCOMPARTMENTAL ANALYSIS

The mean residence time (MRT) is the sum of all the residence times divided by the totalamount of drug originally present in the body, the effective dose (S · F · D):

MRT = k∑∞

0 Ab · dt · t

S · F · D(10.6)

Assuming that Cp and Ab are always proportional and Ab = Cp · Vd, we have

MRT = k∑∞

0 Cp · Vd · dt · t

S · F · D(10.7)

Rearranging yields

MRT =∑∞

0 Cp · t · dt

S · F · D/k · Vd(10.8)

But Cl = k · Vd, so

MRT =∑∞

0 Cp · t · dt

S · F · D/Cl(10.9)

But S · F · D/Cl = AUC, so

MRT =∑∞

0 Cp · t · dt

AUC(10.10)

The denominator in equation (10.10) is the area under the plasma concentration–time curvefrom time zero to infinity (AUC) (Figure 10.1a). The numerator is the area under the curveof the plot of Cp · t versus time from zero to infinity (Figure 10.1b). This is referred to asthe area under the first moment curve (AUMC):

MRT = AUMC

AUC(10.11)

Thus, MRT is calculated from the area under the curve (from time zero to ∞) for the plotof plasma concentration against time (Figure 10.1) (AUC), and the area under the curve(from time 0 to ∞) for the plot of the product of plasma concentration and time againsttime (AUMC) (Figure 10.1). Both the AUC and the AUMC can be determined using thetrapezoidal rule (see Appendix C). Note that in NCA the terminal elimination rate constantis given the symbol �. Recall that for the AUC calculation, the area from the last data pointto infinity is Cplast/�. Note that for the AUMC, the area from the last data point to infinity is(Cplast · tlast/�) + (Cplast/�2). Thus, the MRT can be determined without subjecting the datato compartmental analysis.

Page 225: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

DETERMINATION OF OTHER IMPORTANT PHARMACOKINETIC PARAMETERS 205

0

1

2

3

4

5

0 5 10 15 200

1

2

3

0 2 4 6 8 10 12 14 16

(a) (b)

AUC AUMCCp

(mg/

L)

Cp•

t (m

g•h/

L)

Time (h) Time (h)

FIGURE 10.1 Graphs of plasma concentration against time (a) and the product of plasma concen-tration and time against time (b). The AUC is the area under the Cp versus time curve and the AUMCis the area under the Cp · t versus time curve.

10.3 DETERMINATION OF OTHER IMPORTANTPHARMACOKINETIC PARAMETERS

The other important pharmacokinetic parameters, including the mean elimination rateconstant, the mean elimination half-life, and the volume of distribution at steady state, canbe determined as follows. The mean elimination rate constant

k = 1

MRT(10.12)

the elimination half-life

t1/2 = 0.693

k(10.13)

the clearance

Cl = S · F · D

AUC(10.14)

and the volume of distribution at steady state,

Vdss = Cl

k= S · F · D/AUC

1/MRT= S · F · D · MRT

AUC(10.15)

Substituting for MRT from equation (10.12) yields

Vdss = S · F · D · AUMC

AUC2 (10.16)

Example 10.2 In Chapter 7 a data set was presented and subject to compartmental analysisusing the model for an intravenous injection on a one-compartment model. The data areshown in Table E10.2A and will now be subjected to NCA.

Page 226: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

206 INTRODUCTION TO NONCOMPARTMENTAL ANALYSIS

TABLE E10.2A Plasma Concentration–Time Data SimulatedAfter a 50-mg Intravenous Dose in a One-Compartment Model

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0 2.5 3 1.370.1 2.45 5 0.920.2 2.40 7 0.610.5 2.26 10 0.331 2.04 12 0.221.5 1.85 15 0.122 1.67

Solution The parameters are determined using NCA as follows:

1. Terminal elimination rate constant (�). This is determined from the slope of ln Cpversus time for the last three data points:

� = 0.20 h−1

2. Terminal elimination half-life (t1/2,�)

t1/2,� = 0.693

0.2= 3.46 h

3. Determination of AUC∞0 . This is calculated using the trapezoidal rule (Appendix C)

as outlined in Table E10.2B.

TABLE E10.2B Determination of the AUC and the AUMC

AUC Calculation AUMC Calculation

Time(h)

Cp(mg/L)

AUC Segmenta

(mg · h/L)Time(h)

Cp · t(mg · h/L)

AUMC Segmenta

(mg · h2/L)

0 2.50 0.25 0 0.00 0.010.1 2.45 0.24 0.1 0.25 0.040.2 2.40 0.70 0.2 0.48 0.240.5 2.26 1.08 0.5 1.13 0.791 2.05 0.97 1 2.05 1.211.5 1.85 0.88 1.5 2.78 1.532 1.68 1.52 2 3.35 3.733 1.37 2.29 3 4.12 8.725 0.92 1.54 5 4.60 8.917 0.62 1.43 7 4.31 11.54

10 0.34 0.57 10 3.38 6.1012 0.23 0.53 12 2.72 6.8815 0.12 15 1.86

aThe area of each segment was determined using the trapezoidal rule as described in Appendix C.

AUC150 = 12.00 mg · h/L

AUC∞15 = 0.124

0.2= 0.62 mg · h/L

AUC∞0 = 12.62 mg · h/L

Page 227: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

DIFFERENT ROUTES OF ADMINISTRATION 207

4. Determination of AUMC∞0 . This is calculated using the trapezoidal rule as outlined

in Table E10.2B.

AUMC150 = 49.70 mg · h2/L

AUMC∞15 = 1.86

0.2+ 0.124

0.04= 12.4 mg · h2/L

AUMC∞0 = 62.1 mg · h2/L

5. Determination of MRT

MRT = AUMC

AUC= 62.1

12.6= 4.92 h

6. Determination of mean elimination rate constant

k = 1

MRT= 1

4.92= 0.2 h−1

Note that in this example, because Cp falls in a monoexponentially the mean elimi-nation rate constant is the same as the terminal elimination rate constant (�).

7. Determination of clearance

Cl = S · F · D

AUC= 50

12.6= 3.97 L/h

8. Determination of steady-state volume of distribution

Vdss = S · F · D · AUMC

AUC2 = 50 × 62.1

12.62= 19.47 L

10.4 DIFFERENT ROUTES OF ADMINISTRATION

With a route of drug administration that does not involve drug absorption, such as anintravenous bolus injection, the MRT reflects the average time it takes for a drug moleculeto pass through the body. When drug input is noninstantaneous, such as oral administration,the drug molecule’s journey is extended by the absorption process. In this situation, thetime (after administration) it takes a drug molecules to pass through the body is the sum ofthe absorption time and the residence time. This overall time is known as the transit time.The mean transit time (MTT) is the sum of mean absorption time (MAT) and the MRT:

MTT = MRT + MAT

For noninstantaneous routes,

MTT = AUMC

AUC(10.17)

MAT is the difference between the MRT obtained after intravenous administration and theMTT after oral administration and reflects the mean time for absorption.

Page 228: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

208 INTRODUCTION TO NONCOMPARTMENTAL ANALYSIS

The dependency on both absorption and elimination of MTT after oral administrationlimits its use in noncompartmental analysis. The focus of many clinical studies, such asdrug interaction studies, is to observe how clearance and the rate of elimination (terminalelimination rate constant and t1/2) may be affected. In many cases, intravenous data are notavailable, which makes it impossible to determine the MRT from the MTT. As a result, thevolume of distribution at steady state cannot be determined [see equation (10.15)]. Thesestudies generally calculate the volume of distribution based on the terminal elimination rateconstant. This volume is equivalent to V� in the two-compartment model.

10.5 APPLICATION OF NONCOMPARTMENTAL ANALYSISTO CLINICAL STUDIES

As discussed in the introduction, NCA is often used to analyze the results of clinicalstudies designed to quantify the effects, if any, of specific conditions on the drug’s phar-macokinetics. The results of such studies will be translated into dosage recommendationsor contraindication warnings in the drug’s labeling. Typically, the crossover design is usedfor these clinical studies. Thus, in one leg of the study, the drug is administered to a subjectin the absence of the factor (control leg). Plasma concentration–time data are collected andNCA is used to estimate the pharmacokinetic parameters. In the other leg of the study,the drug is administered to the same subject in the presence of the factor (test leg). Thesequencing of the legs is usually randomized among participants. Thus, half of the groupmembers participate in the control leg first, while the other half participate in the test legfirst. The two legs are separated by a suitable washout period (over 7 elimination half-lives).Any changes in the pharmacokinetic parameters are assessed. The study must be performedin several subjects to provide the statistical power needed to perform tests of statistical sig-nificance. Typically, somewhere between 10 and 20 subjects are required, depending on thevariability of the pharmacokinetic parameters and the magnitude of any expected changes.

Example 10.3 Drug–Drug Interaction Study During the course of this book the phar-macokinetic characteristics of the fictitious drug lipoamide have evolved. Lipoamide iscleared almost exclusively by metabolism by cytochrome 2C9 (CYP 2C9). It is a high-extraction drug and undergoes nonrestrictive hepatic clearance. Fluconazole is a stronginhibitor of CYP2C9 and alters the pharmacokinetics of several CYP2C9 substrates, in-cluding phenytoin warfarin and losartan. The results of a hypothetical drug–drug interactionstudy conducted to evaluate the effects of fluconazole on lipoamide are presented. A ran-domized crossover study with a two-week interval between the phases was conducted in12 healthy volunteers. In each phase the subjects received 200 mg of either fluconazole (testleg) or placebo (control leg) once daily for 5 days. On day 5 each subject received 120 mgof lipoamide before breakfast. Blood samples were taken at various times and analyzed forunchanged lipoamide. The results from one subject are presented in Table E10.3A.

Directions to create an Excel worksheet to analyze these data using NCA are provided inAppendix C Section 5. Note that the purpose of this exercise is to provide experience withNCA. As a result, the analysis will be performed on the data from one subject. Conclusionsabout altered pharmacokinetics cannot be based on the results of one subject. Assume thatthe analysis of data from the other subjects demonstrated that the effects observed on Cmax

and AUC are statistically significant.Plots of plasma concentration against time for the two phases are shown in Figure E10.3,

where it can be seen that coadministration of fluconazole resulted in higher plasma

Page 229: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

APPLICATION OF NONCOMPARTMENTAL ANALYSIS TO CLINICAL STUDIES 209

TABLE E10.3A Plasma Concentration–Time Data Obtained After the Oral Administrationof Lipoamide (120 mg) to a Healthy Volunteer Concurrently with Placebo (Control) andFluconazole (Test)

Time(h)

ControlCp (�g/L)

TestCp (�g/L)

Time(h)

ControlCp (�g/L)

TestCp (�g/L)

0.0 0.0 0.0 1.2 52.0 160.90.2 38.1 117.8 2.0 46.3 143.20.4 50.6 156.7 4.0 34.4 106.40.6 54.1 167.4 10.0 14.1 43.60.8 54.3 168.0 15.0 6.7 20.71.0 53.3 165.0 24.0 1.8 5.4

0

30

90

60

120

150

180

0 5 10 15 20

Lipoamide plus fluconazole

Lipoamide alone

Cp

(mg/

L)

Time (h)

FIGURE E10.3 Plasma concentration of lipoamide administered with placebo (dashed line) andwith fluconazole (solid line).

concentrations of lipoamide. A summary of lipoamide’s pharmacokinetic parameters inthe absence and presence of fluconazole is provided in Table E10.3B.

The data show that lipoamide’s oral clearance decreases and Cmax increases with con-comitant fluconazole. The decrease in oral clearance (Cl/F) could result from either adecrease in clearance (Cl) or an increase in the bioavailability (F). Any changes in theclearance would alter the drug’s half-life. Since the half-life of lipoamide was not affected,one can assume that the reduction in oral clearance is primarily the result of increasedbioavailability. In keeping with this theory, the Cmax of lipoamide increased with concomi-tant fluconazole. Furthermore, the increase in Cmax (about threefold) was about the same

TABLE E10.3B Summary of the Effect of Fluconazole on thePharmacokinetics of Lipoamide

Parameter Control (Placebo) Test (Fluconazole)

Cl/F (L/h) 284 91.8a

� (h−1) 0.15 0.15t1/2,� (h) 4.67 4.66Cmax (�g/L) 54.3 168a

Tmax (h) 0.8 0.8

aStatistically significant when combined with the results from the other subjects.

Page 230: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

210 INTRODUCTION TO NONCOMPARTMENTAL ANALYSIS

magnitude as the decrease in oral clearance (about threefold). It is likely that fluconazolereduces the presystemic hepatic extraction of lipoamide. As lipoamide is a high-extractiondrug, it is likely that its hepatic clearance is fairly insensitive to changes in intrinsic clearancebrought about by modifiers of the CYP2C9 enzyme system.

In conclusion, the results demonstrate that the pharmacokinetics of lipoamide are alteredby fluconazole and suggest that decreased first-pass hepatic extraction may be the causeof the interaction. Based on these results, the combination of lipoamide should either beavoided or the dose of lipoamide should be reduced by about a third.

PROBLEMS

10.1 Oral tablets of lipoamide (120 mg), nosolatol (250 mg), and disolvprazole (50 mg)were all subject to a relative bioavailability study to determine if food altered thebioavailability of these drugs. The design of the three studies was similar, and eachconsisted of a randomized crossover study involving 20 healthy volunteers. On oneleg of the study the subjects took the drug with water after an overnight fast (fastleg). On the other occasion, the participants took the drug on the morning of thestudy after a standard U.S. Food and Drug Administration breakfast consisting oftwo eggs, bacon, hash browns, two slices of toast, and an 8-oz serving of milk (fedstudy). The two legs were separated by a period of two weeks. Example data fromone participant from each of the studies are presented in Table P10.1.

TABLE P10.1 Results of Three Separate Relative Bioavailability Studies Designed to Probethe Effect of Food on the Bioavailability of Lipoamide, Nosolatol, and Disolvprazole

Lipoamide Nosolatol Disolvprazole

Fast Fed Fast Fed Fast Fed

Time(h)

Cp(�g/L)

Cp(�g/L)

Time(h)

Cp(�g/L)

Cp(�g/L)

Time(h)

Cp(�g/L)

Cp(�g/L)

0 0 0 0 0 0 0 0 00.2 35.0 20.4 0.2 299 274 0.2 252 93.30.4 48.3 33.2 1 717 822 0.6 473 2130.6 52.7 41.0 1.6 756 949 1 516 2710.8 53.6 45.5 2 752 978 1.6 469 2941 53.0 48.0 2.6 732 982 2.6 345 2581.2 51.9 49.1 3 716 972 3.2 282 2232 46.3 47.2 4 675 928 4 213 1774 34.4 35.8 8 531 733 6 106 92.4

10 14.2 14.7 12 417 576 8 52.9 46.515 6.75 7.02 24 203 280 10 26.3 23.224 1.78 1.85 48 47.8 68.0 12 13.1 11.6

Use NCA to analyze the results and determine how food affects the bioavailability ofthese drugs. Assume that any differences that you find achieve statistical significancewhen combined with the results from the other participants.

10.2 A study was conducted on 10 healthy volunteers to determine if itraconazole altersthe pharmacokinetics of nosolatol. A sequential two-treatment design was used. Onday 1 of the study, participants received an oral dose of nosolatol (250 mg) after an

Page 231: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc10 JWBS062-Rosenbaum February 3, 2011 7:33 Printer Name: Yet to Come

PROBLEMS 211

overnight fast. Blood samples were taken at various times after the dose, and theplasma concentration of the drug was determined. At the conclusion of the first partof the study, participants returned home and were instructed to take 200 mg of oralitraconazole twice daily. On day 6, the subjects returned to the clinic and receivedanother dose of nosolatol (250 mg). Blood samples were withdrawn and analyzedfor nosolatol. The data from a single subject are presented in (Table P10.2).

TABLE P10.2 Plasma Concentrations of Nosolatol at Various Times After an Oral Dose inthe Absence and Presence of Itraconazole

Cp (�g/L) Cp (�g/L)

Time(h)

NosolatolAlone

Nosolatol PlusItraoconazole

Time(h)

NosolatolAlone

Nosolatol PlusItraoconazole

0 0 0 3 716 10330.2 299 406 4 675 9981 717 987 8 531 8651.6 756 1055 12 417 7502 752 1059 24 203 4882.6 732 1046 48 47.8 207

Use NCA to analyze the results and comment on the effect of itraconazole. Assumethat any differences you find achieve statistical significance when combined withthe results from the other participants. Interpret the results based on nosolatol’spharmacokinetic properties summarized in Appendix E.

10.3 A randomized crossover study was conducted on 15 healthy volunteers to determineif probenecid alters the pharmacokinetics of disolvprazole. In one phase of the study,subjects received an oral dose of disolvprazole after an overnight fast. In the secondphase, subjects received disolvprazole (50 mg) in combination with probenecid (1 g).In both phases blood samples were withdrawn over a 12-h period and the plasmawas analyzed for unchanged drug. The two phases were separated by a washoutperiod of one week. The data are shown in Table P10.3. Use NCA to analyze theresults and comment on the effect, if any, of probenecid. Assume that any differencesyou find achieve statistical significance when combined with the results from theother participants. Interpret the results based on disolvprazole’s pharmacokineticproperties summarized in Appendix E.

TABLE P10.3 Plasma Concentrations of Disolvprazole at Various Times After an Oral Dosein the Absence and Presence of Probenecid

Cp (�g/L) Cp (�g/L)

Time(h)

DisolvprazoleAlone

Disolvprazole PlusProbenecid

Time(h)

DisolvprazoleAlone

Disolvprazole PlusProbenecid

0 0 0 3.2 282 4850.2 252 255 4 213 4330.6 473 504 6 106 3241 516 584 8 52.9 2431.6 469 589 10 26.3 1822.6 345 527 12 13.1 137

Page 232: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

11PHARMACOKINETICS OFINTRAVENOUS INFUSION IN AONE-COMPARTMENT MODEL

11.1 Introduction

11.2 Model and Equations11.2.1 Basic Equation11.2.2 Application of the Basic Equation11.2.3 Simulation Exercise: Part 1

11.3 Steady-State Plasma Concentration11.3.1 Equation for Steady-State Plasma Concentrations11.3.2 Application of the Equation11.3.3 Basic Formula Revisited11.3.4 Factors Controlling Steady-State Plasma Concentration11.3.5 Time to Steady State11.3.6 Simulation Exercise: Part 2

11.4 Loading Dose11.4.1 Loading-Dose Equation11.4.2 Simulation Exercise: Part 3

11.5 Termination of Infusion11.5.1 Equations for Termination Before and After Steady State11.5.2 Simulation Exercise: Part 4

11.6 Individualization of Dosing Regimens11.6.1 Initial Doses11.6.2 Monitoring and Individualizing Therapy

11.6.2.1 Proportional Changes in the Rate of Administration11.6.2.2 Estimation of Patient’s Clearance

Problems

Objectives

The material in this chapter will enable the reader to:

1. Have an understanding of the pharmacokinetics of continuous drug administration

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

212

Page 233: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

INTRODUCTION 213

2. Appreciate the determinants of steady-state plasma concentrations, particularly theinfluence of clearance and the rate of drug administration

3. Understand the factors that control the time to reach steady state

4. Develop an appropriate drug administration regimen based on population averagepharmacokinetic parameter values

5. Individualize a dosing regimen for a patient based on the patient’s steady-state plasmaconcentration

11.1 INTRODUCTION

In previous chapters we addressed the pharmacokinetics of single doses. Clinically, theadministration of isolated single doses is limited to only a few situations, such as over-the-counter analgesics and cough and cold remedies. Most drugs used in the treatment ofdiseases are taken over a course of at least several days and sometimes a lifetime. Forlong-term drug treatment, drugs may be administered in many different ways, includingintravenous infusions, multiple intravenous doses, skin patches, and multiple oral doses.

An understanding of the pharmacokinetics and pharmacodynamics of continued drugadministration is needed to design optimum dosing regimens. For example, in Chapter 7we demonstrated how a drug’s pharmacokinetic characteristics could be used to determinethe value of single doses to achieve certain target plasma concentrations. But if a drug’sprimary pharmacokinetics parameters (Cl and Vd) and therapeutic range are known, howcan a dosing regimen be developed to maintain plasma concentrations in the therapeuticrange over an extended period? The answer to this question requires an understanding of thepharmacokinetics of extended drug administration. Chronic drug administration involvestwo phenomenan that have not been addressed previously: accumulation and fluctuation.

Accumulation refers to a gradual buildup of drug concentrations with successive doses.It occurs whenever a dose is administered at a time when drug from a previous doseis still in the body. It is important to understand the properties of accumulation. Forexample, does accumulation continue for as long as the therapy continues? Alternatively,does accumulation gradually attenuate and eventually stop?

Multiple doses are administered in a quantum or pulse-like fashion; doses are given atregular intervals (e.g., 10 mg every 8, 12, or 24 h) over the course of therapy. This pulse-like administration is associated with fluctuation, the rise and fall in plasma concentrationduring a dosing interval.A peak concentration is usually seen at the time of, or slightly after,the administration of a dose. Trough concentrations usually occur at the time the next doseis given. However, in the case of orally administered drugs, the trough may occur slightlylater if there is an absorption lag time.

The simplest way initially to study the pharmacokinetics of chronic drug administration isto consider the characteristics of constant (zero-order) continuous drug input into the body.Under these circumstances no fluctuation in the plasma concentration will be observed.The most common example of zero-order drug administration is the intravenous infusion,where drug is administered directly into a patient’s vein at a constant continuous rate usinga drip or electronic infusion pump. Intravenous infusions offer several advantages: Theyprovide a very accurate means of administering drugs; they can be very convenient if thepatient has an existing indwelling catheter; they eliminate fluctuation and provide a constantplasma concentration, which can be targeted to the middle of the therapeutic range; and the

Page 234: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

214 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

absence of fluctuation reduces the tendency of the peak and trough plasma concentrations toventure into toxic and subtherapeutic areas, respectively. Pharmaceutical scientists strive todevelop controlled release oral dosage forms that provide constant, continuous drug input.The elementary infusion pump or Oros system is an example of an oral dosage form thatappears to provide a zero-order input.

11.2 MODEL AND EQUATIONS

In the one-compartment model, the body is represented by a single imaginary compartment.Recall from Chapter 7 that the concentration in the compartment is assumed to be equalto the plasma concentration, and the volume of the imaginary compartment is equal toits volume of distribution (Vd). Like the distribution of drug throughout the body, thedistribution of drug throughout the compartment is assumed to occur extremely rapidly,in an essentially instantaneous manner. The compartment is homogeneous and at any timethe concentration of drug throughout is constant. As always, elimination is first orderand can be described using clearance (Cl) or the overall elimination rate constant (k),where k = Cl/Vd. Drug administration into the body or compartment occurs at a constantcontinuous rate (zero-order process). The rate (k0) has units of amount per unit time (e.g.,mg/h). In the event that the salt of a drug is used or the equations are applied to extravascularadministration, the rate of administration is qualified by the salt factor and bioavailability,respectively. The model is shown in Figure 11.1.

11.2.1 Basic Equation

The starting point for the development of the basic equation is to consider how the amountof drug in the body changes with time:

rate of change of the amount of drug in the body = rate of inputs − rate of outputs

dAb

dt= S · F · k0 − k · Ab (11.1)

Initial Conditions: t = 0,Ab = Ab0 = 0

k•AbCl•Cp

VdCpAb

S•F•k0

FIGURE 11.1 Intravenous infusion in a one-compartment model. Drug input (I) into the body isconstant and equal to the product of the infusion rate (k0), the salt factor (S), and the bioavailability(F). For intravenous administration, F = 1. Ab is the amount of drug in the body, Vd the drug’s volumeof distribution, and Cp the plasma concentration. Elimination (E) is a first-order process and can beexpressed using clearance (Cl) or the elimination rate constant (k).

Page 235: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

MODEL AND EQUATIONS 215

where S is the salt factor, F the bioavailability, and k0 the constant rate of drug administration.Equation (11.1) is integrated from zero to infinity:

Ab = S · F · k0

k· (1 − e−kt ) (11.2)

with Cp = Ab/Vd,

Cp = S · F · k0

Vd · k· (1 − e−kt ) (11.3)

and with Cl = Vd · k,

Cp = S · F · k0

Cl· (1 − e−kt ) (11.4)

Analysis of the Equation

� Recall (see Appendix A) that 1 - e−kt is the growth factor that starts at zero when t = 0and grows to 1 at infinity.

� Thus, plasma concentrations start at zero and grow to the value of S · F · k0/Cl atinfinity.

� The speed of growth is determined by the value of k (t1/2).� The equation allows the plasma concentration to be estimated at any time during

zero-order drug input.� The shape of the plasma concentration–time profile corresponding to equation (11.4)

is shown in Figure 11.2.

Note in Figure 11.2 that after the start of the infusion, the drug accumulates in the bodyand the plasma concentration increases. The rate of increase in the plasma concentrationand the accumulation become less and less as therapy continues. Eventually, the plasmaconcentration becomes constant and drug accumulation ceases. The latter period is known

0

20

40

60

80

100

0 1 2 3 4 5 6 7 8

Cpss

Time After Start of Therapy, t1/2

Cp

(% s

s)

Steady State

FIGURE 11.2 Plasma concentration–time profile during an intravenous infusion. When the plasmaconcentration remains constant, steady state has been achieved. The plasma concentration at steady-state plasma concentration is Cpss. Note that the unit of time is the elimination half-life.

Page 236: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

216 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

as a steady-state and the corresponding plasma concentration is referred to as the steady-state plasma concentration (Cpss). It can be concluded that during a constant intravenousinfusion, drug accumulation is a self-limiting process that eventually stops at steady state.

11.2.2 Application of the Basic Equation

The basic equation (11.4) enables the plasma concentration to be calculated at any timeduring continuous zero drug input (e.g., from an infusion or an oral preparation that deliversdrug at a constant continuous rate).

Example 11.1 A drug (S = 1, Cl = 2 L/h, Vd = 50 L) is administered as an intravenousinfusion at a rate of 10 mg/h. Calculate the plasma concentration 4 h into the infusion.

Solution Cl = 2 l/h, Vd = 50 L, k = Cl/Vd = 2/50 = 0.04 h−1, and t1/2 = 0.693/k = 17.3 h.

Cp = S · F · k0

Cl(1 − e−kt )

= 10

2(1 − e−0.04×4)

= 0.74 mg/L

11.2.3 Simulation Exercise: Part 1

Open the model “Intravenous Infusion Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter11a

The default model parameters are S = 1, F = 1, infusion rate = 10 mg/h, Cl = 5 L/h, andVd = 20 L.

1. Review the objectives, explore the model, and review the “Model Summary” page.

2. Go to the “Cp–time Profile” page. Perform a simulation with the default rate ofadministration and observe the Cp–time profile.

(a) Simulate using infusion rates of 5, 10, and 20 mg/L. Hold the mouse overeach line to note the values of the steady-state plasma concentration. Addthese to Table SE11.1.

(b) Describe the relationship between Cpss and the infusion rate.

(c) Determine an infusion rate to give a Cpss of 6 mg/L.

TABLE SE11.1 Infusion Rates and the Corresponding Cpss

Simulation Infusion Rate Cpss

1 5 mg/h

2 10 mg/h

3 20 mg/h

4 6 mg/L

Page 237: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

STEADY-STATE PLASMA CONCENTRATION 217

11.3 STEADY-STATE PLASMA CONCENTRATION

At steady state, the plasma concentration becomes constant and independent of time (Figure11.2). The value of the steady-state plasma concentration is a very important focus of drugtreatment. Usually, it is desirable to have a steady-state plasma concentration in the middleof the therapeutic range. To plan this, it is necessary to understand the factors that controlthe steady-state plasma concentration.

11.3.1 Equation for Steady-State Plasma Concentrations

There are two ways to derive the equation for the steady-state plasma concentration.

Derivation 1 Before steady state, the plasma concentration is given by equation (11.4).As t increases, the exponential growth function, 1 − e−kt, tends to 1, and Cp tends to Cpss:

Cpss = S · F · k0

Cl(11.5)

The box around equation (11.5) signifies that it is an important equation, as we demonstratein Section 11.3.4.

Derivation 2 At steady state the plasma concentration is constant. Thus, the rate of drugadministration must equal the rate of drug elimination:

S · F · k0 = Cl · Cpss (11.6)

Equation (11.6) can be rearranged to yield equation (11.5).

11.3.2 Application of the Equation

We look next at two examples that illustrate application of the steady-state plasma concen-tration equation.

Example 11.2 The same drug as that described in Example 11.1 is administered asan intravenous infusion at a rate of 10 mg/h. Calculate the steady-state plasma con-centration. Recall that Cl = 2 L/h, Vd = 50 L, S = 1, k = Cl/Vd = 2/50 = 0.04 h−1, andt1/2 = 0.693/k = 17.3 h.

Solution

Cpss = S · F · k0

Cl= 10

2= 5 mg/L

Example 11.3 A drug (S = 0.9, F = 0.94, Cl = 1.44 L/h, Vd = 0.65 L/kg) has a therapeuticrange of 10 to 20 mg/L. It is formulated into an oral elementary osmotic pump, whichdelivers the drug at a constant rate of 12 mg/h. The preparation is designed for administration

Page 238: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

218 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

every 12 h and is available in units of 250, 300, and 400 mg. Determine a suitable dose toachieve a steady-state plasma concentration of 15 mg/L in a 60-kg woman.

Solution

Cpss = S · F · k0

Cl

k0 = Cl · Cpss

S · F= 1.44 × 15

0.9 × 0.94

= 25.5 mg/h

The unit dosage forms are designed to administer the dose at a constant rate over a 12-hperiod. If a rate of 25.5 mg/h is required, the unit dose must contain 25.5 × 12 = 306 mg.Thus, the 300-mg unit should be given twice daily.

11.3.3 Basic Formula Revisited

The expression for the steady-state plasma concentration given in equation (11.5) may besubstituted into the basic equation for the plasma concentration before steady state, as givenin equation (11.4):

Cp = Cpss · (1 − e−kt ) (11.7)

This equation shows that the plasma concentration grows from zero at time zero (1 −e−kt = 0) to the steady-state plasma concentration at time infinity (1 − e−kt = 1).

11.3.4 Factors Controlling Steady-State Plasma Concentration

Equation (11.5), which shows the factors that control the steady-state plasma concentration,is reproduced below.

Cpss = S · F · k0

Cl

The box surrounding it designates that it is an important equation. This equation, probablythe most important and useful of all pharmacokinetic equations, shows that the terminalplasma concentration achieved by a given rate of drug administration is dependent only onthe effective rate of drug administration and clearance. It also provides insight into when itmay be prudent to modify the usual rate of drug administration.

Specifically, the equation shows the following points:

1. Cpss is directly proportional to the infusion rate (k0). If the infusion rate is doubled,Cpss will double. For example, if a drug is administered at a rate of 5 mg/h andachieves a steady-state plasma concentration of 2 mg/L, doubling the infusion rate to10 mg/h would double the steady-state plasma concentration to 4 mg/L.

Page 239: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

STEADY-STATE PLASMA CONCENTRATION 219

2. Cpss is directly proportional to the drug’s bioavailability (F).

3. Cpss is inversely proportional to the drug’s clearance (Cl). For example, consider adrug normally administered at a rate of 10 mg/h to achieve a desired steady-stateplasma concentration of 2 mg/L. The same rate of administration in an elderly patient,who has a clearance of half the normal value, would result in a steady-state plasmaconcentration twice the desired value—4 mg/L.

4. Cpss is independent of the volume of distribution (Vd). The volume of distribution isabsent from the basic formula for the steady-state plasma concentration. Variabilityin Vd will not influence the steady-state plasma concentration. In the next sectionwe demonstrate that volume of distribution, through its influence on the half-life,influences the time it takes to get to steady state.

5. The equation can be used to estimate a rate of drug administration to achieve a desiredsteady-state plasma concentration and to estimate a steady-state plasma concentrationfrom a given rate of administration.

6. The equation demonstrates that it may be necessary to modify the usual rate of drugadministration in patients who have altered clearance or bioavailability.

11.3.5 Time to Steady State

The basic equation for the plasma concentration during constant continuous drug adminis-tration is given by equation (11.7), which can be rearranged as

Cp

Cpss= 1 − e−kt (11.8)

The left-hand side of equation (11.8) is the fraction of steady state achieved at any time.Thus, 1 − e−kt represents the fraction of steady state achieved at any time. Recall (seeAppendix A) that the function 1 − e−kt is a fraction that starts off at zero and climbs to1 at infinity. The time for the function to achieve certain fractional values depends on thefirst-order rate constant or the half-life. The number of half-lives for the function to achievevarious fractions is presented in Appendix B. The same values can be used to show thenumber of half-lives to achieve certain fractional values of the steady state (Table 11.1).

TABLE 11.1 Time (Half-Lives) to Achieve CertainFractions of Steady State

Time (t1/2) 1 − e−ktFraction of Steady

State (%)

16

a 0.1 1013

a 0.2 20

1 0.5 503.3 0.9 904.3 0.95 956.6 0.99 99

aApproximate value.

Page 240: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

220 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

The time to achieve various fractions of steady state can also be proven through calcu-lation.

Example 11.4 How many half-lives does it take to get to 95% steady state?

Solution At 95% of steady state, Cp = 0.95 Cpss. Let k = 0.693/t1/2. Substituting intoequation (11.8) for Cp and k gives us

0.95Cpss

Cpss= 1 − e−(0.693/t1/2) · t

e−(0.693/t1/2) · t = 0.05

0.693t

t1/2= − ln 0.05 = 3.0

t = 4.33t1/2

In summary, the time to get to steady state is determined by a drug’s half-life. Clinically,it is usual to consider that it takes 3 to 5 half-lives to achieve steady state. It can be seenin Figure 11.2 (the units of time are elimination half-lives) that steady state is achievedby about 5 half-lives. Recall that the half-life is a secondary pharmacokinetic parameter,determined by a drug’s clearance and volume of distribution. Thus, changes in either ofthese primary parameters will alter the time to steady state by virtue of their influence on thehalf-life. For example, increases in the volume of distribution and decreases in clearancewill increase the time it takes to get to steady state.

11.3.6 Simulation Exercise: Part 2

Open the model “Intravenous Infusion Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter11a

Go to the “Effect of Cl and Vd” page.

1. Record the value of Cpss for the three values of Cl (2.5, 5, and 10 L/h) in TableSE11.2 and summarize the relationship between Cl and Cpss.

2. Describe how Cl affects the time to reach steady state.

TABLE SE11.2 Influence of Changes in Clearance and Volume of Distribution on theValue of the Steady-State Plasma Concentration and the Time to Get to Steady State

Simulation Clearance Volume of Distribution Cpss

1 2.5 L/h 20 L (default)

2 5 L/h 20 L (default)

3 10 L/h 20 L (default)

4 5 L/h (default) 10 L

5 5 L/h (default) 20 L

6 5 L/h (default) 40 L

Page 241: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

LOADING DOSE 221

3. Clear the graph and record the value of Cpss for the three values of Vd (10, 20,and 40 L) in Table SE11.2 and summarize the relationship between Vd and Cpss.

4. Describe how Vd affects the time to reach steady state.

5. Would you recommend dosage adjustments in patients with (a) altered Cl; (b)altered Vd?

6. List three situations where a patient may present with an altered Cl, and threesituations where a patient may present with an altered Vd.

11.4 LOADING DOSE

If a drug has a long half-life and/or if it is necessary to achieve therapeutic plasma concen-trations immediately, a loading dose may be given to achieve steady state immediately.

11.4.1 Loading-Dose Equation

A loading dose may be calculated using equation (7.9) with Cpss as the target plasmaconcentration at time zero:

DL = Cpss · Vd

S · F(11.9)

If the loading dose is administered simultaneously with the start of the infusion, the netdrug in the body at any time is the sum of that remaining from the bolus loading dose andthat gained from the infusion:

Cpnet = CpDL + Cpinf (11.10)

where CpDL is the plasma concentration from the loading dose and Cpinf is that from theinfusion; and can be calculated from equations (7.4) and (11.4), respectively,

CpDL = S · F · DL

Vd· e−kt

Cpinf = S · F · k0

Cl· (1 − e−kt )

Substituting for the expressions above into equation (11.10) yields

Cp = S · F · DL

Vd· e−kt + S · F · k0

Cl· (1 − e−kt ) (11.11)

But

S · F · DL

Vd= Cpss

and

S · F · k0

Cl= Cpss

Page 242: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

222 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

0

2

4

6

8

10

12

0 2 4 6 8

Cp

(mg/

L)

Time After Loading Dose and Start of Infusion (h)

Cp from DL

Cp from Infusion

Net Cp

Note: t1/2 = 1 h

FIGURE 11.3 Net plasma concentration of drug from an intravenous infusion and a bolus loadingdose. The rate of the infusion is Cpss · Cl/S · F and the loading dose is Cpss · Vd. Note that the t1/2 valueof this drug is 1 h. It can be seen that by about 6 h the loading dose is almost completely eliminatedand the infusion is at steady state.

Thus,

Cp = Cpss · e−kt + Cpss · (1 − e−kt )

= Cpss (11.12)

In other words, the loss of drug from the bolus is exactly matched by the gain of drug fromthe infusion (Figure 11.3).

Example 11.5 A drug (Cl = 6 L/h, Vd = 150 L, S = 1) is to be administered as an intra-venous infusion. A steady-state plasma concentration of 1.2 mg/L is desired. Calculate:

(a) A suitable infusion rate.

(b) The time it would take to achieve steady-state plasma concentrations.

(c) The value of a loading dose that would immediately achieve the desired steady-stateplasma concentration of 1.2 mg/L.

Solution As with all pharmacokinetic calculations, the half-life should be calculated assoon as possible because it can be used to check calculations and make sure that they arein the correct range. The elimination rate constant should also be calculated, as that is usedfrequently in calculations.

t1/2 = 0.693Vd/Cl = 17.3 h k = 0.04 h−1

(a) The rate of the infusion is found using equation (11.5); Cpss = 1.2 mg/L:

k0 = 1.2 mg/L × 6 L/h

1 × 1= 7.2 mg/h

Page 243: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

TERMINATION OF INFUSION 223

(b) Time to steady stateIt takes 3 to 5 t1/2 to get to steady state. Thus, it takes (3 × 17) – (5 × 17) = 51 to85 h.

(c) Loading dose, Using equation (11.9), we have

DL = 1.2 mg/L × 150 L

1 × 1= 180 mg

The need for a loading depends on two factors:

1. The drug’s half-life and how long it takes to get to steady state

2. How quickly a therapeutic response is required in a given clinical situation

For example, it takes over a week to get to steady state when therapy with digoxinis started. But it is not critical to achieve therapeutic plasma concentration immediately,and a loading dose is not frequently administered. In contrast, it takes only about 6 to8 h to reach steady state with lidocaine. However, this drug is used to treat serious life-threatening ventricular arrhythmias, and as a result it is important to achieve therapeuticplasma concentrations quickly, and a loading dose is usually administered.

11.4.2 Simulation Exercise: Part 3

Open the model “Intravenous Infusion Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter11a

Go to the “Loading Dose” page.

1. Note the value of Cpss achieved using the default settings.

2. Calculate a loading dose to achieve this Cp immediately.

3. Simulate Cp when a loading dose is given.

4. Note the Cp–time profile.

11.5 TERMINATION OF INFUSION

11.5.1 Equations for Termination Before and After Steady State

At some time the infusion will be stopped or terminated. At this time there will be noongoing inputs into the body, and first-order elimination becomes the only process thataffects the plasma concentration. Thus, after termination, the plasma concentration willdecay monoexponentially from its value at the time of termination:

Cp = CpT · e−kt ′(11.13)

where CpT is Cp when the infusion is terminated at time T and t′ is the time elapsed sincetermination.

Page 244: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

224 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

Termination After Steady State When the infusion is stopped after steady state has beenachieved, the plasma concentration at termination is Cpss, and after termination,

Cp = Cpss · e−kt ′ (11.14)

where t′ is the time elapsed since termination.

Termination Before Steady State When the infusion is stopped before steady state hasbeen achieved, the basic equation (11.4) for an infusion has to be used to calculate theplasma concentration at the time of termination:

CpT = S · F · k0

Cl· (1 − e−kT ) (11.15)

where T is the time of termination. After termination, Cp will decay monoexponentiallyfrom this value:

Cp = S · F · k0

Cl· (1 − e−kT ) · e−kt ′

(11.16)

where t′ is the time elapsed since termination.

11.5.2 Simulation Exercise: Part 4

Open the model “Intravenous Infusion Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter11a

Go to the “Termination” page.

1. Simulate Cp without terminating the infusion.

2. Perform several simulations when the infusion is terminated at various times bothbefore and after the achievement of steady state.

3. Comment on the Cp–time profile after termination for both linear and semilogplots.

4. Write an equation for the plasma concentration after termination (Cp′) using CpT

for the plasma concentration at the time of termination and t ′ for the time sincetermination.

11.6 INDIVIDUALIZATION OF DOSING REGIMENS

11.6.1 Initial Doses

Infusion rates or rates at which drugs are administered are designed to achieve steady-stateplasma concentrations in the middle of the therapeutic range. Typical infusion rates arebased on average values of clearance, which, as can be seen from equation (11.5) is theonly parameter (assuming intravenous administration and F = 1) that affects the dose. If adrug has both a narrow therapeutic range and displays a lot of variability in its clearance in

Page 245: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

INDIVIDUALIZATION OF DOSING REGIMENS 225

the population, it may be necessary to either modify an initial dose, based on a suspectedaltered clearance, and/or to monitor steady-state plasma concentrations to ensure that theyare therapeutic. If they are found to be outside the therapeutic range, it will be necessary toalter the dose and individualize it for the patient.

Patient’s Clearance For drugs that have narrow therapeutic ranges it is important toevaluate patients before therapy is started to determine if they possess any characteristicsthat are known to alter a specific drug’s clearance. If as a result of this evaluation they aresuspected to have altered clearance, the dose of the drug can be modified accordingly. Forexample, if the patient is taking a concomitant medication known to induce the enzymesinvolved in elimination of the drug, their clearance may be higher than normal, and a higher-than-normal rate of drug administration may be required. Conversely, if a patient is takinga drug that inhibits the metabolism of the subject drug, or if the patient has a disease (e.g.,hepatic disease, renal disease, congestive heart failure) that is known to reduce the subjectdrug’s clearance, their clearance may be lower than normal and it may be appropriate touse a dose that is lower than normal.

In many situations there is often no information about a patient’s individual pharma-cokinetic parameters, including clearance. Thus, the population average value of clearanceis assumed, and the typical population average infusion rate is used. Alternatively, equation(11.5) can be used to calculate an infusion rate to achieve the desired plasma concentration,based on the population average clearance.

11.6.2 Monitoring and Individualizing Therapy

It is most convenient to wait 3 to 5 elimination half-lives after the start of therapy andmonitor when steady state has been achieved.

11.6.2.1 Proportional Changes in the Rate of AdministrationIt can be seen from equation (11.5) that the steady-state plasma concentration is directlyproportional to the infusion rate. This allows proportional changes to the infusion rate tobe made to achieve different steady-state plasma concentrations.

Example 11.6 A 70-kg patient is receiving an infusion of lidocaine hydrochloride(S = 0.87) (1 mg/min) to achieve a desired steady-state plasma concentration of 4 mg/L.His steady-state plasma concentration is found to be 3 mg/L. Recommend a new infusionrate.

Solution Using proportions, we have

4 mg/L

3 mg/L= k0 mg/min

1 mg/min

k0 = 1.33 mg/h

11.6.2.2 Estimation of Patient’s ClearanceAlternatively, a steady-state plasma concentration from a known rate of drug administrationcan be used to calculate a patient’s clearance. The clearance can be used in equation (11.5) todetermine a new rate of drug administration. This method involves an extra step comparedto the proportional method presented above, but it provides a little more pharmacokinetic

Page 246: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

226 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

information, and the clearance calculated can be used in conjunction with a populationaverage volume of distribution to estimate the patient’s elimination half-life.

Example 11.7 Use the information from Example (11.6) and substitute in equation (11.5):

3.0 mg/L = 0.87 × 1 × 1 mg/min

Cl

Cl = 0.29 L/min

Use the patient’s clearance and the population average volume of distribution (0.88 L/kg)to calculate the half-life:

t1/2 = 0.693 × 0.88 × 70

0.29= 147 min

Estimate a new infusion rate by again using equation (11.5):

4 mg/L = 0.87 × 1 × k0

0.29 L/mink0 = 1.33 mg/min

An infusion of 1.33 mg/min should provide a steady-state plasma concentration of 4 mg/L.

Example 11.8 A drug (S = 1) has the population average parameters Cl = 10.3 L/h andVd = 75 L. The therapeutic range is 0.5 to 1.25 mg/L. An elderly patient is to be treatedwith the drug.

(a) Calculate a suitable intravenous infusion rate to achieve a Cpss value of 1 mg/L.

(b) What is the earliest time that a plasma sample could be withdrawn to ensure that thesteady-state plasma concentration is on target?

(c) Forty-eight hours after the start of an infusion of 10.3 mg/h, a plasma concentrationis measured and found to be 1.5 mg/L. Suggest a more appropriate infusion regimen.

Solution

(a) At this point there is only information on the population average parameters. Thesewill be used to estimate the initial infusion rate:

k = Cl

Vd= 10.3

75= 0.1371 h−1 t1/2 = 0.693

0.137= 5.05 h

Rearrange equation (11.5) to solve for the infusion rate:

k0 = Cl · Cpss

S · F

k0 = 10.3 L/h × 1 mg/L

1 × 1= 10.3 mg/h

Page 247: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

PROBLEMS 227

(b) It will take about 15 to 25 h to get to steady state in a person with a normal half-life.An elderly patient may have an increased half-life, so it may take a little longer.Thus, it would be wise to wait until at least 24 h before checking.

(c) The plasma concentration was obtained at 48 h, which is equivalent to approximately10 (48/5) half-lives of the drug in a normal patient. It is therefore likely that steadystate has been achieved at this time, even in a patient who has a reduced clearance anda correspondingly increased half-life. Thus, it can be assumed that in this patient aninfusion rate of 10.3 mg/h results in a steady-state plasma concentration of 1.5 mg/L.The patient’s individual clearance can be calculated from these data:

Cl = S · F · k0

Cpss

= 10.3

1.5= 6.87 L/h

The patient’s half-life can be estimated using the population average Vd:

t1/2 = 0.693 × 75

6.87= 7.6 h

In this patient, who has reduced clearance, the half-life is 7.6 h. It would be estimated totake 23 to 38 h to achieve steady state. So the plasma concentration sampled at 48 h wouldbe expected to be in steady state.

The patient’s clearance can be used to determine a more appropriate infusion rate toachieve the desired steady-state plasma concentration (1 mg/L):

k0 = Cpss · Cl

S · F

= 1 mg/L × 6.87 L/h = 6.87 mg/h

PROBLEMS

11.1 A drug (Cl = 100 mL/min, Vd = 0.7 L/kg, S = 1) is administered by intravenousinfusion at a rate of 5 mg/h to a 70-kg man.

(a) Calculate the plasma concentration 6 h after the start of the infusion.

(b) What is the steady-state plasma concentration?

(c) Calculate the plasma concentration 4 h after termination when the infusion isterminated 3 h after the start.

(d) Calculate the plasma concentrations 4 h after infusion is terminated 30 h afterthe start.

11.2 Vancomycin is administered as in IV infusion at a rate of 500 mg/h. The infusion isterminated after 1 h. A plasma concentration taken 2 h after the start of the infusionis 20 mg/L. A second plasma concentration taken 10 h after the start of the infusionis 7 mg/L. What is vancomycin’s half-life in this patient?

Page 248: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

228 PHARMACOKINETICS OF INTRAVENOUS INFUSION IN A ONE-COMPARTMENT MODEL

11.3 A drug company has developed an elementary osmotic pump formulation of thehydrochloride salt (S = 0.85) of their �-blocker. The formulation, which is designedto be taken twice daily, contains 96 mg of drug which is released in zero-orderfashion over a 12-h period. The entire dose is released and absorbed across thegastrointestinal membrane and undergoes no significant intestinal extraction. It issusceptible to first-pass hepatic extraction (E = 0.8). The drug has the followingpharmacokinetic parameters: Cl = 1080 mL/min, Vd = 250 L, fe � 0.01. Calculatethe steady-state plasma concentration achieved by this dosage form.

11.4 A 70-kg male patient with premature ventricular contractions is to receive treatmentwith lidocaine hydrochloride (S = 0.87). Calculate a loading dose and the rate of anintravenous infusion that would achieve a desired plasma concentration of 2 mg/L.Lidocaine has the following population average parameters: Cl = 6 mL/kg · min,V1 = 0.5 L/kg, and Vdss = 1.3 L/kg.

11.5 Recommend suitable rates for administration of the fictitious drugs lipoamide, noso-latol, and disolvprazole by intravenous infusion to target steady-state plasma con-centrations of 50, 1250, and 400 �g/L, respectively. Their relevant pharmacokineticparameters are summarized in Table P11.5.

TABLE P11.5 Summary of the Important PharmacokineticParameters of Lipoamide, Nosolatol, and Disolvprazole

Lipoamide Nosolatol Disolvprazole

Cl (L/h) 62 12.6 12Vd (L/70 kg) 420 210 35S 1 1 1

11.6 Theophylline is a broncodilator used in the treatment of bronchial asthma and otherrespiratory diseases. Chemically, it is a xanthene (1,3-dimethylxanthine) bearing aclose structural similarity to caffeine (1,3,7-trimethylxanthine). About 6% of a doseof theophylline is metabolized to caffeine, which is subsequently metabolized touric acid derivatives. Theophylline is eliminated primarily by metabolism (fe = 0.1to 0.2). Owing to a wide interpatient variability in pharmacokinetic parametersof theophylline (particularly clearance), there is a wide interpatient variation inthe blood levels achieved by a given dose. Furthermore, since theophylline has avery narrow therapeutic range (5 to 15 mg/L), a given dose may produce toxicplasma concentrations in some patients and subtherapeutic levels in others. Thus, itis frequently necessary to individualize a person’s dose of theophylline. An initialestimate of theophylline’s clearance in a patient can be obtained based on somedemographic characteristics, such as the presence of certain diseases, concomitantmedications, and age. This, in turn, allows an initial infusion rate to be estimated.However, since both the clearance and the infusion rate are only estimates, it isimportant that the steady-state plasma concentration be monitored and the infusionrate adjusted if necessary.

A.M. is a 80-kg, 5 foot 11 inch, 50-year-old male who is to be treated with theo-phylline for an asthmatic attack. Theophylline’s clearance in A.M. is estimated to be1.64 L/h based on his demographic features: severe obstructive pulmonary disease;

Page 249: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc11 JWBS062-Rosenbaum February 16, 2011 7:55 Printer Name: Yet to Come

PROBLEMS 229

congestive heart failure; smoking a pack of cigarettes a day. The population averagevolume of distribution of theophylline is 0.5 L/kg.

(a) Advise on a suitable infusion rate of aminophylline (S = 0.8) to achieve a plasmaconcentration of 15 mg/L theophylline.

(b) Determine a suitable intravenous loading dose of aminophylline.

(c) What is the earliest time that a plasma sample can be taken to determine if thesteady-state plasma concentration is in the therapeutic range?

(d) An infusion of 30.75 mg/h is used. A steady-state plasma sample is obtainedand reveals a theophylline Cp value of 25 mg/L (A.M.’s clearance was onlyestimated). Recommend a more suitable infusion rate based on this information.

(e) Assume that the new infusion rate is used and that steady state is achieved. Theinfusion is terminated. How long would it take plasma concentrations to fall to3.5 mg/L?

(f) Comment on how steady-state plasma concentrations of theophylline may beaffected by:

(1) Compounds such as cimetidine and the quinolone antibiotics that inhibittheophylline’s intrinsic clearance

(2) Compounds that alter hepatic blood flow

(3) Phenytoin, which induces theophylline’s metabolism

11.7 Treat the patient described in the “Infusion Challenge” simulation model, which canbe found at:http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter11b

Page 250: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

12MULTIPLE INTRAVENOUSBOLUS INJECTIONS IN THEONE-COMPARTMENT MODEL

12.1 Introduction

12.2 Terms and Symbols Used in Multiple-Dosing Equations

12.3 Monoexponential Decay During a Dosing Interval12.3.1 Calculation of Dosing Interval to Give Specific Steady-State Peaks and Troughs

12.4 Basic Pharmacokinetic Equations for Multiple Doses12.4.1 Principle of Superposition12.4.2 Equations That Apply Before Steady State

12.5 Steady State12.5.1 Steady-State Equations12.5.2 Average Plasma Concentration at Steady State12.5.3 Fluctuation12.5.4 Accumulation12.5.5 Time to Reach Steady State12.5.6 Loading Dose

12.6 Basic Formula Revisited

12.7 Pharmacokinetic-Guided Dosing Regimen Design12.7.1 General Considerations for Selection of the Dosing Interval12.7.2 Protocols for Pharmacokinetic-Guided Dosing Regimens

12.7.2.1 Protocol I: Targeting the Average Steady-State Plasma Concentration12.7.2.2 Protocol II: Targeting Specific Steady-State Peaks and Troughs

12.8 Simulation Exercise

Problems

Objectives

The material presented in this chapter will enable the reader to:

1. Identify the characteristics of the plasma concentration profile after multiple intra-venous doses

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

230

Page 251: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

INTRODUCTION 231

2. Understand the derivation of an expression for the plasma concentration at any timeduring therapy with multiple intravenous doses

3. Understand the factors controlling the average steady-state plasma concentration andthe time to steady state

4. Understand the factors controlling fluctuation

5. Understand the factors controlling accumulation

6. Design dosage regimens based on pharmacokinetic principles

12.1 INTRODUCTION

In Chapter 11 we introduced the pharmacokinetics of continuous drug administration anddemonstrated that once therapy begins, drug concentrations build up gradually until steadystate is achieved. At steady state, plasma concentrations remain constant. The value of thesteady-state plasma concentration is controlled by the rate of drug administration and thedrug’s clearance. The time it takes to get to steady state is dependent on the drug’s half-life.

Most drugs are not administered in a constant and continuous fashion, but as multi-ple discrete doses. Generally, the dose is administered in a pulse-like fashion one, two,three, or four times daily. From the perspective of patient convenience—and by extension,adherence—once daily dosing is most desirable, followed in order of decreasing desirabilityby two, three, or four times daily. Most commonly the goal of therapy is to achieve a sus-tained therapeutic response, and the choice of dosing frequency is dependent on the drug’spharmacokinetic and pharmacodynamic properties. The pharmacokinetics of multiple in-travenous doses are presented in this chapter. Although the oral route is most frequently usedfor multiple-dose administration, the pharmacokinetics of this route are complicated by theabsorption process. Consequently, the characteristics of multiple-dose pharmacokineticsare initially studied most conveniently by removing the absorption process and consideringintravenous doses.

The typical plasma concentration–time profile observed after multiple intravenous dosesis shown in Figure 12.1. The units of time are the drug’s elimination half-life, which isalso the dosing interval or frequency of drug administration. The profile has some notablecharacteristics:

� Fluctuation. The profile appears as a series of peaks and troughs, known as fluctuation.The peaks occur at the time a dose is given, and the troughs occur immediately beforethe next dose.

� Accumulation. It can be seen in Figure 12.1 that the troughs and peaks increase witheach dose, known as accumulation. This occurs when a dose is given at a time whendrug from a previous dose is still in the body. Note that there is less accumulation witheach successive dose.

� Steady state. Eventually, accumulation between doses stops altogether. At this time,the peaks and troughs with successive doses are the same; that is, steady state has beenachieved.

� Average Cp. Note that the average plasma concentration throughout therapy (indicatedby the dashed line in Figure 12.1) has the same shape as the plasma concentration–timeprofile of a continuous constant infusion.

Page 252: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

232 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

0

5

10

15

20

25

0 1 2 3 4 5 6 7 8

Cp

(mg/

L)

Time into Therapy (half-lives)

Troughs (t = )

Peaks (t = 0)

Steady State

FIGURE 12.1 Plasma concentration–time profile observed with multiple intravenous injections(solid line) in a one-compartment model. The data were simulated using the following parameters:dose = 64 mg, Cl = 4 L/t1/2, Vd = 5.77 L, and the dosing interval (� ) = 1 t1/2. Fluctuation is observedin the profile. A peak occurs at the time a dose is given (t = 0), and a trough occurs immediately beforethe next dose (t = � ). Note that the value of the peaks and troughs increases with each successivedose. This is because of accumulation. The amount of accumulation gets smaller and smaller witheach dose. At steady state there is no further accumulation. The steady-state peaks and troughs areexactly the same. It takes about 5 half-lives to reach steady state. The average plasma concentration(dashed line) has the same profile as that of a continuous intravenous infusion.

The goal of drug therapy is to devise a dosing regimen that will maintain therapeuticsteady-state plasma concentrations. A multiple-dosing regimen consists of two parts: (1)the dose and (2) the dosing interval, or the frequency with which the doses are repeated.For example, in Chapter 11 it was found that a constant continuous infusion of 3.1 mg/h ofthe fictitious drug lipoamide would provide a therapeutic plasma concentration of 50 �g/L(see Problem 11.5). It has been found that plasma concentrations of lipoamide greater than90 �g/L are associated with a high frequency of side effects. Plasma concentrations below25 �g/L are subtherapeutic. If multiple discrete doses of lipoamide are to be given, whatdose and what dosing interval should be used to ensure that plasma concentrations aretherapeutic and nontoxic at all times? The material presented in this chapter is directed atanswering this question.

12.2 TERMS AND SYMBOLS USED IN MULTIPLE-DOSING EQUATIONS

The formulas for multiple-dosing pharmacokinetics introduce some new symbols. Theseare shown in Figure 12.2 and described below.

� n is the number of the last dose administered.� � (the lowercase Greek letter tau) is the dosing interval. If 50 mg is administered every

8 h, � is 8 h.� t is the time since the last dose. During therapy, t varies continuously from 0 at the

beginning of a dosing interval to � at the end. When the next dose is given, t oncemore becomes 0.

Page 253: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

TERMS AND SYMBOLS USED IN MULTIPLE-DOSING EQUATIONS 233

0

5

10

15

20

25

0 8 16 24 32 40 48 56 64

Cp

(mg/

L)

Time intoTherapy (h)

Cpmax,4

t = 0

Cpmin,2t =

Cpmax,ss

t = 0

Cpmin,ss

n 1 2 3 4 5 6 7 8

Doses

t =

FIGURE 12.2 Symbols used in multiple-dose pharmacokinetics. In this example, the dose isadministered every 8 h. n is the number of the last dose administered, � the dosing interval (8 h), and tthe time since the last dose (in this regimen, t will vary continuously from 0 through 8 h). Cpmin,n andCpmin,ss the trough plasma concentrations (t = � ) before and after steady state, respectively; Cpmax,n

and Cpmax,ss are the peak plasma concentrations (t = 0) before and after steady state, respectively.

� Time into therapy can be calculated from the product of n · � . Specifically, time intotherapy = [(n − 1) · � ] + t. For example let � = 12; then:

3 h after the first dose, t = (0 × 12) + 3 = 3 h

3 h after the second dose, t = (1 × 12) + 3 = 15 h

At the end of the fourth dosing interval, t = (3 × 12) + 12 = 48 h

6 h after the tenth dose, t = (9 × 12) +6 = 114 h

� Peak plasma concentrations, which occur when t = 0, have the symbol Cpmax. Beforesteady state, Cpmax is dependent on the number of the last dose, so it has the symbolCpmax,n. After steady state, the peak is independent of the number of the last dose andhas the symbol Cpmax,ss.

� Trough plasma concentrations, which occur when t = � , have the symbol Cpmin.Before steady state, Cpmin is dependent on the number of the last dose and has thesymbol, Cpmin,n. After steady state, the peak is independent of the number of the lastdose and has the symbol Cpmin,ss.

� The symbols, Cpn and Cpss are used to represent the plasma concentration at any othertime during a non-steady-state and a steady-state dosing interval, respectively.

The derivation of the equations for multiple doses involves a number of assumptions,which include:

1. With the possible exception of the first dose (loading dose), the dose and the dosinginterval are constant throughout the duration of therapy. The dose may be referred toas the maintenance dose.

Page 254: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

234 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

2. The drug’s pharmacokinetic parameters remain constant throughout the entire dura-tion of therapy. This assumes that the drug displays linear pharmacokinetics.

Thus, these equations can only be applied to situations where the assumptions hold. Forexample, the equations should not be used for drugs that display nonlinear pharmacokinet-ics, such as phenytoin (Chapter 15).

12.3 MONOEXPONENTIAL DECAY DURING A DOSING INTERVAL

When a dose is administered, it is assumed that the entire dose enters the systemic circulationat time zero, even though in reality an injection may be given gradually over a period ofa minute or more. For the one-compartment model, the drug is assumed to distribute in arapid, essentially instantaneous manner. Thus, the peak plasma concentration is assumedto occur at time zero. During a dosing interval, t increases from zero at the time of the doseup to its maximum value of � just before the next dose. During this period, the plasmaconcentration is under the influence of only one process: first-order elimination (Figure12.3). During a dosing interval, the plasma concentration decays monoexponentially fromthe peak (Cpmax) to the trough (Cpmin).

Before steady state, during a dosing interval,

Cpn = Cpmax,n · e−kt (12.1)

and at the trough,

Cpmin,n = Cpmax,n · e−k� (12.2)

During the dosing interval Cp decaysmonoexponentially from the peak:

Cpn = Cpmax,n•e–kt

Dose Dose

Cpmin

Cpmax

Cpn

Cp

Time

FIGURE 12.3 Monoexponential decay during a dosing interval. During a dosing interval, plasmaconcentrations are influenced only by first-order elimination. As a result, they decay monoexponen-tially from the peak. The first-order decay equation can be used to express the plasma concentrationduring the interval as a function of the peak concentration and the time after the dose.

Page 255: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

MONOEXPONENTIAL DECAY DURING A DOSING INTERVAL 235

At steady state, during a dosing interval,

Cpss = Cpmax,ss · e−kt (12.3)

and at the trough,

Cpmin,ss = Cpmax,ss · e−k� (12.4)

This basic relationship between the plasma concentration and time during a dosing intervalpermits the simple derivation of an important equation that is used to calculate dosingintervals needed to achieve certain peak and trough plasma concentrations.

12.3.1 Calculation of Dosing Interval to Give Specific Steady-StatePeaks and Troughs

Knowing that the fall in the plasma concentration between doses is simple monoexponentialdecay makes it possible to calculate the value of a dosing interval to give specific peaksand troughs. This is best addressed through an example.

Example 12.1 Assume that a new drug is under development. It has a very narrowtherapeutic range (12 to 25 mg/L). However, since it is being used to treat a serious life-threatening condition for which few other treatments are available, development of this drugis being pursued. The goal is to design a dosing regimen that will result in a steady-statepeak and trough of 20 and 14 mg/L, respectively. The drug’s elimination rate constant hasa population average value of 0.043 h−1. What dosing interval is needed to provide thedesired steady-state peaks and troughs?

Solution The relationship between the steady-state peak and trough is given in equation(12.4). Substituting the desired steady-state trough and peak into this equation gives us

14 = 20e−0.043�

ln14

20= −0.043�

� = 0.357

0.043= 8.3 h

A more useful arrangement of equation (12.4) for use in calculating a dosing interval is

� = −1

kln

Cpmin,ss

Cpmax,ss(12.5)

The box around equation (12.5) indicates that it is an important equation that is in frequentuse clinically.

Page 256: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

236 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

12.4 BASIC PHARMACOKINETIC EQUATIONS FOR MULTIPLE DOSES

12.4.1 Principle of Superposition

When a dose of a drug is administered at a time when drug from a previous dose(s) is stillin the body, it is assumed that the total amount of drug in the body is equal to the amountprovided by the new dose, plus the drug remaining from previous dose(s). This is knownas the principle of superposition, illustrated in Table 12.1 for a drug that is administeredas a 64-mg dose every half-life. At the end of the first dosing interval, half (32 mg) ofthe first dose remains. So the peak amount after the second dose is the dose (64 mg) plusthat remaining from the first dose (32 mg), or 96 mg. The same process can be used tocalculate the next trough amount and the peaks and troughs after successive doses as shownin Table 12.1. Note that after the first dose the body accumulates 32 mg, but there is lessaccumulation with each dose. For example, in Table 12.1 it can be seen that only 2 mg isaccumulated between the fifth and sixth doses. Eventually, no further accumulation occurs,and steady state is achieved. At steady state an entire dose (64 mg) is lost during a dosinginterval, only to be replaced exactly by the next dose.

12.4.2 Equations That Apply Before Steady State

The equations for multiple intravenous bolus injections are derived using exactly the sameprocess as that described above; the full derivation appears in Appendix D. The basicequation that can be used to determine the plasma concentration at any time during multiple-dose therapy is

Cpn = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−kt (12.6)

TABLE 12.1 Maximum (Abmax) and Minimum (Abmin) Amountsof Drug in the Body and Accumulation After Successive Doses of64 mg Are Given Every Half-Life

Dose ABmax (mg) ABmin (mg)AccumulationBetween Peaks

1 64 322 96 48 323 112 56 164 120 60 85 124 62 46 126 63 27 127 63.5 1...Steady state

N 128 64 0N + 1 128 64 0

Page 257: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

BASIC PHARMACOKINETIC EQUATIONS FOR MULTIPLE DOSES 237

Recall that t is the time that has elapsed since the last dose, n the number of the last dose,and � the dosing interval. When t = 0, a peak plasma concentration is observed:

Cpn,max = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )(12.7)

When t = � , a trough is observed:

Cpn,min = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−k� (12.8)

Alternatively, the trough may be expressed as

Cpn,min = Cpn,max · e−k� (12.9)

Example 12.2 Multiple intravenous bolus injections (250 mg) of a drug are administeredevery 8 h. The drug has the following parameters: S = 1, Vd = 30 L, k = 0.1 h−1, and � =8 h. Calculate:

(a) The plasma concentration 3 h after the second dose.

(b) The peak and trough plasma concentrations during this second dosing interval.

Solution(a) Substituting into equation (12.6) yields

Cpn = 250(1 − e−2×0.1×8)

30(1 − e−0.1×8)· e−0.1×3

= 8.96 mg/L

(b) Cpmax,2 can be calculated when t = 0 [equation (12.7)]:

Cpmax,2 = 250(1 − e−2×0.1×8)

30(1 − e−0.1×8)

= 12.1 mg/L

Cpmin,2 can be determined when t = � [equation (12.8) or (12.9)]. Using the latteryields

Cpmin,2 = 12.1e−0.1×8

= 5.4 mg/L

During the second dosing interval, the plasma concentrations at the peak, 3 h intothe dosing interval, and at the trough are 12.1, 8.96, and 5.4 mg/L, respectively.

Page 258: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

238 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

12.5 STEADY STATE

Usually, steady state is the focus of drug therapy and the goal is to achieve steady-stateplasma concentrations that are in the therapeutic range. Consequently, the equations asso-ciated with steady state are very important and are more commonly used clinically than theequations that apply prior to steady state. The specific aspects of steady state that are ofmost interest are the determinants of:

� The steady-state peak and trough plasma concentrations� The average plasma concentration during a steady-state dosing interval� The factors that control the amount of fluctuation observed at steady state (Cpmax,ss

versus Cpmin,ss)� The degree of accumulation that ultimately occurs at steady state� The time to reach steady state

12.5.1 Steady-State Equations

At steady state, the plasma concentration–time profile is exactly the same for all dosingintervals (i.e., the plasma concentrations become independent of n, the number of the lastdose). Recall the basic equation that applies before steady state:

Cpn = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−kt (12.10)

During therapy n increases with each successive dose. As a result, as therapy progressesand steady state is approached, e−nk� tends to zero, and 1 − e−nk� becomes 1 and disappearsfrom the equation. Thus, the basic equation for steady state is expressed

Cpss = S · F · D

Vd · (1 − e−k� )· e−kt (12.11)

The corresponding equation for the peak and trough may be expressed by letting t = 0 and� , respectively:

Cpmax ss = S · F · D

Vd · (1 − e−k� )(12.12)

Cpmin,ss = S · F · D

Vd · (1 − e−k� )· e−k� (12.13)

or

Cpmin,ss = Cpmax,ss · e−k� (12.14)

Page 259: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

STEADY STATE 239

Example 12.3 Next continuing with our example of a drug (Vd = 30 L, k = 0.1 h−1)that is administered intravenously as 250 mg administered every 8 h. Recall that after thesecond dose,

Cpmax = 12.1 mg/L

Cp3h = 8.96 mg/L

Cpmin = 5.4 mg/L

Calculate the maximum and minimum steady-state plasma concentrations achieved by theregimen.

Solution

Cpmax,ss = 250

30(1 − e−0.1×8)= 15.1 mg/L

Cpmin,ss = 250

30(1 − e−0.1×8)· e−0.1×8 = 6.8 mg/L

The steady-state peaks and troughs are 15.1 and 8.8 mg/L, respectively.

These equations can also be used in the reverse direction to determine a dose to producea desired steady-state peak and/or trough.

Example 12.4 Recall that it was determined earlier (Example 12.1) that a dosing intervalof 8.3 h (approximately 8 h) was needed for a drug (Vd = 50 L, k = 0.043 h−1) to achievesteady-state troughs and peaks of 14 and 20 mg/L, respectively. What dose should be used?

Solution Either the formula for Cpmax,ss or that for Cpmin,ss can be used. The only unknownin either case is the dose:

Cpmax,ss = S · F · D

Vd · (1 − e−k�

) where Cpmax,ss20 mg/L

or

Cpmin,ss = S · F · D

Vd · (1 − e−k�

) · e−k� where Cpmin,ss14 mg/L

Assuming that S = 1, F = 1,

20 = S · F · D

50(1 − e−0.043×8

)

D = 290 mg

The intravenous injection of 290 mg of this drug every 8 h should result in a steady-statepeak and trough of 20 and 14 mg/L, respectively.

Page 260: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

240 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

12.5.2 Average Plasma Concentration at Steady State

Frequently, rather than concentrating on the peaks and troughs, the emphasis of multipledosing therapy is to achieve a desired therapeutic average steady-state plasma concentration.For example, digoxin has a very long half-life, and plasma concentrations remain fairlyconsistent during a dosing interval. It is often necessary to determine the dose needed toachieve an average plasma concentration of 1 �g/L. It would be useful to have an expressionthat relates the average steady-state plasma concentration to the dose and dosing interval.

Because the fall in Cp during a dosing interval is monoexponential and nonlinear, theaverage steady-state plasma concentration is not the arithmetic mean of the troughs andpeaks [(Cpmax,ss + Cpmin,ss)/2]. In Figure 12.4 it can be seen that the arithmetic mean ofthe peak and the trough is not representative of the average concentration during the dosinginterval. The plasma concentration is above this value for a much shorter time than it isbelow it. The average plasma concentration during the interval is calculated as the areaunder the steady-state dosing interval divided by the dosing interval:

Cpav,ss =

∫ T

0Cp · dt

�= AUC�

0

�(12.15)

AUC�0 = S · F · D/Cl.

Note that the AUC during a steady-state dosing interval is the same as the AUC fromzero to infinity after a single dose. Substituting into equation (12.15) yields

Cpav,ss = S · F · D

Cl · �(12.16)

Cpmin,ss

Cpmax,ss

arithmetic mean of Cpmax,ss and Cpmin,ss

time above arithmetic mean

time below arithmetic mean

*0

Cp•dt S•F•DCpav,ss

T

Cl= =∫

Cp

Time

FIGURE 12.4 Average steady-state plasma concentration. Plasma concentrations fall monoexpo-nentially during a dosing interval. As a result, the arithmetic mean of the peak and trough is notrepresentative of the average plasma concentration. The average plasma concentration is the areaunder the curve during a steady-state dosing interval divided by � .

Page 261: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

STEADY STATE 241

It can be observed by rearranging formula (12.16) that the average steady-state plasmaconcentration occurs at the time when the rate of drug administration (S · F · D/T) is equalto the rate of drug elimination (Cp · Cl).

Note that D/T represents the average rate of drug administration over the dosing interval(e.g., 160 mg every 8 h represents an average of 20 mg/h). Equation (12.16) can also bewritten using Ra:

Cpav,ss = S · F · Ra

Cl(12.17)

where Ra = D/� and represents the average rate of drug administration.If equation (12.17) is compared to the equation for the steady-state plasma concentration

that results from a constant, continuous infusion (12.18), it can be seen that they areequivalent:

Cpss = S · F · k0

Cl(12.18)

A general equation for chronic drug administration may be written as equation (12.17),where Ra may be either a continuous constant administration rate (as for an infusion)or an average rate of administration (D/� ), achieved by individual discrete doses duringmultiple-dose therapy :

Cpss = S · F · Ra

Cl(12.19)

Some important points were made from a consideration of this equation in Chapter11. Owing to the importance of these points, they will be reiterated here. When a drug isadministered over an extended period, either at a constant continuous rate or as individualdiscrete doses, the average steady-state plasma concentration is:

1. Directly proportional to the effective rate of drug administration, S · F · Ra (this is acharacteristic of linear pharmacokinetics)

2. Inversely proportional to clearance

3. Independent of the volume of distribution

Equation (12.19) also provides a handle to use to calculate dosing regimens to achievethe desired steady-state plasma concentrations. It is very frequently used clinically for thispurpose.

Example 12.5 Doses of digoxin are usually administered every 24 h. Calculate an ap-propriate dose of digoxin (to achieve a concentration of 1 �g/L in a patient whose digoxinclearance is estimated to be 123 L/day.

Page 262: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

242 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

Solution Substituting into equation (12.19), assuming intravenous doses (F = 1) and S= 1, we obtain

1 �g/L = Ra

123 L/day

Ra = 123 �g/day

A daily dose of 123�g, rounded to 125 �g, is recommended.

Example 12.6 An average steady plasma concentration of 50 �g/L is required from theadministration of multiple intravenous bolus injections of the fictitious drug lipoamide. Vd= 420 L and Cl = 62 L/h. What rate of drug administration should be used?

Solution Substituting into equation (12.19) and rearranging yields

Ra = 62 L/h × 50 �g/L

1 × 1= 3.1 mg/h

Any combination of dose and � that gives a rate of administration of 3.1 mg/h will achievean average steady-state plasma concentration of 50 �g/L. The following regimens could beused:

3.1 mg/h—infusion no fluctuation3.1 mg every hour little fluctuation24.8 mg every 8 h more fluctuation37.2 mg every 12 h even greater fluctuation74.4 mg every 24 h large amount of fluctuation

12.5.3 Fluctuation

Fluctuation refers to the difference between the peak and the trough plasma concentrationswithin a dosing interval. As such it reflects the amount of drug that is eliminated duringa dosing interval. As the amount eliminated increases, the amount of fluctuation willincrease. The amount of elimination during a dosing interval is dependent on a drug’s rateof elimination (t1/2 or k) and the length of the dosing interval.

Elimination, and by extension fluctuation, will obviously be greatest when the dosinginterval is long and the drug’s t1/2 short. This can be shown mathematically by consideringthe ratio of Cpmax,ss to Cpmin,ss [see equation (12.14)]:

Cpmax,ss

Cpmin,ss= 1

e−k�(12.20)

The value of the ratio is a measure of fluctuation, and equation (12.20) demonstrates that itis dependent only on k (or the half-life) and � . The value of e−k� will be smallest, and thefluctuation greatest, when � and k are large (t1/2 short). Thus, short half-lives and long dosingintervals promote large fluctuation. Conversely, long half-lives and short dosing intervalspromote little fluctuation. Since the half-life is a constant for a given drug (assuming normalconditions and health), the length of the dosing interval controls fluctuation.

Page 263: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

STEADY STATE 243

Time into Therapy

Cp

r = ratio of Cp at steady state to Cp atthe same time after the first dose

min,ss

min,1

Cpr

Cp=

max,ss

max,1

Cpr

Cp=

t=x,ss

t= x,1

Cpr

Cp=

11 kr

e− Τ=−

FIGURE 12.5 Assessment of accumulation. Accumulation can be assessed using the accumulationratio (r), which is the ratio of a plasma concentration at steady state and the equivalent concentrationafter the first dose.

12.5.4 Accumulation

Accumulation is the increase in drug concentration that occurs with each additional dose(Figure 12.1). It occurs whenever a dose is given when drug from a previous dose is still inthe body. With each successive dose, the accumulation between doses decreases more andmore and eventually stops altogether when steady state is achieved. The ultimate amountof accumulation that occurs at steady state can be quantified by comparing a steady-stateplasma concentration to the equivalent concentration after the first dose. For example, thesteady-state peak could be compared to the peak after the first dose, the troughs could becompared, the average plasma concentrations during the intervals could be compared, andso on (Figure 12.5). The ratios of these values is known as the accumulation ratio, r:

r = Cpss

Cp1at equivalent times in the dosing interval (12.21)

Comparing the peaks gives us

r = Cpmax,ss

Cpmax,1= (S · F · D/Vd) · 1/(1 − e−k� )

S · F · D/Vd(12.22)

r = 1

1 − e−k�(12.23)

Accumulation can also be expressed by comparing the average amount of drug in the bodyat steady state (Abav,ss) to the dose:

Abav,ss

S · F · D= Cpav,ss · Vd

S · F · D= (S · F · D/Cl · � ) · Vd

S · F · D(12.24)

Page 264: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

244 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

Given that t1/2 = 0.693Vd/Cl,

Abav,ss

S · F · D= 1.44

t1/2

�(12.25)

Equations (12.23) and (12.25) both demonstrate that like fluctuation, accumulation is con-trolled by the elimination half-life and the dosing interval. But accumulation and fluctuationrun counter to each other. If little drug is eliminated during a dosing interval (small � andlong t1/2), fluctuation will be small, but the buildup of drug from one dose to the next willbe large, and accumulation will be large.

In summary, the plasma concentration during a steady-state dosing interval is r timeshigher than the corresponding plasma concentration after a single dose [equations (12.21)and(12.23)]. Accumulation will be most apparent when the dosing interval is short and thedrug’s half-life long. If a drug is administered every half-life, there will be 1.44 times moredrug in the body at steady state than after a single dose [equation (12.25)].

Clinically, accumulation may become an important consideration in patients who haveimpaired clearance, which in turn will result in an increased half-life. If the normal dosinginterval is used in these patients, a greater degree of accumulation will occur. If the drug hasa narrow therapeutic range, this accumulation may be associated with toxicity. For example,the elimination half-life of methadone, which is being used increasingly to treat severe pain,displays wide variability in the population. It has been found that the accumulation of thedrug in patients with long half-lives can cause accumulation and potentially life-threateningtoxicity. Accumulation is also a problem with environmental pollutants that have long half-lives. Continuous exposure to only small amounts of these compounds can result in theirsignificant buildup in the body.

12.5.5 Time to Reach Steady State

Recall that n · � represents time into therapy. As n increases with each dose, 1 − e−nk� getscloser and closer to 1. When 1 − e−nk� reaches 1, steady state is achieved and the plasmaconcentration does not increase further with each dose. If an equation that applies beforesteady state is compared to the equivalent steady-state equation [e.g., compare Cpmax,n inequation (12.7) to Cpmax,ss in equation (12.12)], it can be observed that the equation beforesteady state can be expressed

Cpmax,n = Cpmax,ss · (1 − e−nt� ) (12.26)

Thus, 1 − e−nk� represents the fraction of steady state achieved at any time:

fraction of steady state = 1 − e−nk� (12.27)

In common with intravenous infusions, the time n · � to achieve steady state is dependentonly on k or t1/2. The number of half-lives necessary to achieve a certain fraction of steadystate are shown in Table 12.2. Clinically, it is usually considered to take 3 to 5 half-lives toachieve steady state.

Page 265: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

BASIC FORMULA REVISITED 245

TABLE 12.2 Fraction of Steady-Sate Achieved atVarious Times in Terms of Half-Life

Time into Therapy(t1/2) Fraction of Steady State (%)

1 503.3 904.4 956.6 99

12.5.6 Loading Dose

If it is necessary to achieve steady-state plasma concentrations immediately, a loadingdose may be administered. The purpose of the loading dose is to provide all the drug thatultimately accumulates at steady state in a single dose. At steady state there is r times moredrug in the body than after a single dose. Thus, the loading dose will need to be r timesgreater than the usual maintenance dose.

DL = r · DM

DL = DM

1 − e−k�(12.28)

Note that if a drug is administered every half-life, the loading dose is double the maintenancedose.

12.6 BASIC FORMULA REVISITED

The phenomenon of accumulation differentiates single and multiple doses. During thecourse of this chapter, expressions have been presented to describe:

� The ultimate amount of accumulation that occurs at steady state� The fraction of steady state achieved at any time during therapy

These expressions for accumulations (Figure 12.6) can be used to construct the basicequation for multiple doses. Figure 12.6 shows that the plasma concentration at any timeafter a dose during multiple doses is equal to:

The plasma concentration at the same time after a single dose, multiplied by

The fraction of steady state achieved at this point in the therapy, multiplied by

The ultimate accumulation that occurs at steady state

Breaking down the basic equation in this way should help demystify the equation andpromote better understanding.

Page 266: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

246 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

CpMultiple Doses

CpMultiple Doses

CpSingle Dose

Fraction of Steady State

Achievedat Any Time

UltimateAccumulation at

Steady State(r)

S•F•DCp •e

Vd−kt=

• •

• •

=

= 1 nke τ−−1

1 ke τ−−

FIGURE 12.6 Component parts of the multiple-dosing equation. The basic equation for multipledoses consists of three parts: the equation for a single dose, the fraction of steady state achieved atany time; and the ultimate accumulation that occurs at steady state.

12.7 PHARMACOKINETIC-GUIDED DOSING REGIMEN DESIGN

12.7.1 General Considerations for Selection of the Dosing Interval

Dosing regimens must be determined for new drugs as they progress through the devel-opment process. Clinically, dosing regimens may be determined for established drugs inindividual patients if it is thought that the patient may experience suboptimal concentra-tions from a standard dose. The dose is chosen to achieve therapeutic concentrations thatproduce minimal side effects or toxicity. The specific value of the dose is based on a drug’spharmacokinetic and pharmacodynamic properties. The value of the dosing interval is se-lected to ensure that the therapeutic response is maintained between doses. In many cases,a drug’s pharmacokinetic properties, specifically the elimination half-life, dictate the valueof the dosing interval. However, as will be demonstrated in subsequent chapters, a drug’spharmacodynamic properties can affect, and in some cases control, the optimum dosinginterval.

Once-daily dosing, which is associated with the greatest patient convenience and ad-herence, is generally regarded as the preferred dosing interval. However, if a drug has ashort elimination half-life, daily dosing may result in excessive fluctuation, which maybe associated with toxic and/or subtherapeutic plasma concentrations. As a result, dosingintervals of 12, 8, and 6 h may have to be used.

Many therapeutic drugs have half-lives within the range 24 to 8 h, and they are frequentlyadministered approximately every half-life. As a result, plasma concentrations will fall byabout 50% during a dosing interval, the trough will be about half the peak, and the loadingdose will be about twice the maintenance dose. For drugs that have short half-lives (6 hor less), from a perspective of patient convenience it is desirable to use a dosing intervalgreater than the half-life. But this will result in a large fluctuation, and the therapeutic rangewill dictate whether or not the fluctuation will be tolerated. If the range is wide, such as forthe penicllins, large fluctuations in the plasma concentrations will be tolerated, and dosingintervals of several half-lives can be used. If, however, a drug’s therapeutic range is narrow,it may not be possible to use a dosing interval greater than the half-life. For example,theophylline has a very narrow therapeutic range (5 to 15 mg/L), and it also has a short

Page 267: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

PHARMACOKINETIC-GUIDED DOSING REGIMEN DESIGN 247

half-life. As a result, short dosing intervals may be necessary. For example, the eliminationhalf-life in smokers and in children can be less than 6 h, and the normal regular releasepreparations of theophylline may have to be administered every 6 h.

The inconvenience of short dosing intervals usually results in the development ofprolonged-release preparations. Several of these are available for theophylline, and theycan be administered twice daily to patients with very short half-lives and once daily in otherpatients. A very short half-life may limit the marketability of a drug if it is dosed on phar-macokinetics principles. It is interesting to note that several therapeutic drugs that have veryshort half-lives have long dosing intervals because their pharmacodynamic properties, nottheir pharmacokinetics, control the dosing interval. For example, both aspirin (antiplateletaction) and proton pump inhibitors such as omeprazole have very short half-lives (�1 h),and both are eliminated from the body within a few hours of the dose. But both act bydestroying their target irreversibly. So their duration of action is dependent not on theircontinued presence in the body but on the new synthesis of their target. For both drugs thistakes over 24 h so both can be administered daily.

In theory, extended dosing intervals of greater than 24 h could be used for drugs that havevery long half-lives (greater than 24 h). However, for patient adherence, dosing intervalsare frequently limited to 24 h. For example, phenobarbital (t1/2 ∼ 5 days) and digoxin (t1/2

∼ 2 days) are both given daily. Weekly or monthly dosing may be used for a small numberof drugs that have very long half-lives. For example, the antimalarial drug chloroquine(t1/2 = 10 to 24 days) is administered weekly, and alendronate, which binds to bone, fromwhich it is released only very slowly (t1/2 ∼ 10 years), is also administered weekly. If adrug has a long half-life, it will take a long time to reach steady state and a loading dosemay be necessary. The long half-lives will produce a large accumulation, which will requireloading doses that are much greater than the maintenance doses.

12.7.2 Protocols for Pharmacokinetic-Guided Dosing Regimens

Clinically, it may be necessary to apply pharmacokinetic principles to determine dosingregimens for individual patients when one or more of the following criteria are met:

1. A drug has a narrow therapeutic range, and serious clinical consequences result whenthe plasma concentrations are outside the range.

2. A drug displays wide interpatient variability in its pharmacokinetic parameters.

3. A patient possesses a characteristic that is frequently associated with altered phar-macokinetics. This may include renal disease, hepatic disease, low activity of adrug-metabolizing enzyme due to genetic factors or concomitant medications, andlow transporter activity.

Drugs that are commonly subject to pharmacokinetic-based dosage individualizationinclude aminoglycosides, phenytoin, lithium, immunosuppressants, and digoxin. Warfarinis also a very important example of a drug whose dose must be individualized for eachpatient. However, since the response to warfarin is easily measured (clotting time or inter-national normalized ratio), dosage adjustments to warfarin are based on the response itselfrather than on plasma concentrations and pharmacokinetics.

Several approaches are available to determine dosing regimens based on pharmacokineticprinciples. The most appropriate approach will depend on the goal of the therapy. Two

Page 268: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

248 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

approaches are presented below. In protocol I the goal of the therapy is to produce adesired average steady-state plasma concentration. In protocol II the regimen is based onthe attainment of specific desired peaks and troughs.

12.7.2.1 Protocol I: Targeting the Average Steady-State Plasma ConcentrationStep 1.

Note the drug’s therapeutic range.

Step 2.

Note the patient’s estimated pharmacokinetic parameters (Cl, t1/2, Vd). In the absence ofany specific information about the patient’s pharmacokinetic parameters, populationaverage values must be assumed. Even then, any patient characteristics that are knownto affect certain pharmacokinetic parameters should be considered. For example, in theabsence of any other information, the clearance of theophylline in a smoker shouldbe considered to be 1.6 times the average value, and its clearance for someone onconcomitant fluvoxamine should be considered to be 30% of the average value (1).

Step 3.

Select a suitable Cpav,ss value from the middle of the therapeutic range.

Step 4.

Calculate the rate of drug administration necessary to achieve the average steady-stateplasma concentration desired (similar to determination of the infusion rate):

S · F · Ra = Cpav,ss · Cl (12.29)

Step 5.

Determine the dosing interval based on the drug’s therapeutic range, the drug’s half-life,and patient convenience. If the therapeutic range is narrow, a suitable interval can becalculated using equation (12.5):

� = −1

kln

Cpmin,ss

Cpmax,ss(12.30)

Step 6.

Calculate the specific dose:

D = Ra · �

S · F(12.31)

Step 7.

If the therapeutic range is narrow, the estimated steady-state peaks and troughs can becalculated to ensure that they are within the range.

Page 269: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

PHARMACOKINETIC-GUIDED DOSING REGIMEN DESIGN 249

Example 12.7 Following a severe blow to the head, a 80-kg man (L.K.) developedseizures. Recommend a suitable dosing regimen for phenobarbital sodium. Even thoughthe drug will be administered orally, use the equations associated with intravenous bolusinjections for your calculations. Would you recommend a loading dose? If so, calculateone. TR = 10 to 30 mg/L; F = 0.9; S = 0.9; Vd = 0.7 L/kg; Cl = 4.0 mL/h · kg.

Solution The half-life is a very important parameter for these calculations and it isimportant to calculate it as soon as possible:

t1/2 = 0.693Vd

Cl= 0.693 × 0.7 × 80 L

(4 × 80)/1000 L/h= 121.3 h = 5.05 days

Phenobarbital has a very long half-life of 5 days.A goal for the average plasma concentration is chosen in the middle of the therapeutic

range: 20 mg/L. The rate of administration necessary to achieve 20 mg/L is determinedfrom (12.29):

S · F · D

�= 20 mg/L ×(4 × 80)/1000 L/h = 6.4 mg/h

The dosing interval is selected. The t1/2 is over 5 days, which will permit the optimumdosing interval of 24 h to be used. This represents only 1

5 or 20% of the half-life. Soduring a 24-h period the plasma concentration will easily remain within the therapeuticrange.

� = 24 h. The dose is now calculated.

S · F · D

�= 6.4 mg/h

D = 6.4�

S · F= 6.4 × 24

0.9 × 0.9= 190 mg of phenobarbital sodium per day

It will take about 15to 25 days to reach steady state, so a loading dose may be advantageous.Using equation (12.28) yields

DL = 190

1 − e−1×0.693/5.05= 1482 mg of phenobarbital sodium

The estimated steady-state peaks [equation (12.12)] and troughs [equation (12.13)] may becalculated to ensure that they are within the therapeutic range.

Cpmax,ss = 21 mg/L

Cpmin,ss = 18.7 mg/L

A maintenance dose of 190 mg of phenobarbital sodium daily is recommended, with aloading dose of around 1500 mg. Generally, the loading dose is divided into three or foursmaller units that can be administered over a period of several hours.

Page 270: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

250 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

Example 12.8 It was shown previously that lipoamide (Cl = 62 L/h; Vd = 420 L) mustbe administered at a rate of 3.1 mg/h to achieve a desired plasma concentration of 50 �g/L.If peaks and troughs with the range 90 to 25 �g/L are desired, recommend a suitable dosingregimen.

Solution The rate of drug administration to achieve a desired steady-state plasma concen-tration of 50 �g/L was determined for this drug using the infusion formula (Problem 11.5).The same average rate of administration should be maintained to achieve 50 �g/L withmultiple doses. Lipoamide’s t1/2 = 0.693 × 420/62 = 4.68 h. From the perspective ofpatient convenience, a dosing interval greater than the t1/2 of this drug is preferred. Its ther-apeutic range will permit a fluctuation of greater than 50% [(Cpmax,ss − Cpmin,ss)/Cpmax,ss] ·100%, so it is possible that a dosing interval greater than the half-life could be used. In thisexample the formula to calculate the dosing interval [equation (12.30)] would be useful.Let the desired steady-state peak and trough be 90 and 25 mg/L, respectively:

� = − 4.68

0.693ln

25

90= 8.65 h

The dosing interval will be rounded off to 8 h and the dose is determined from the calculatedrate (3.1 mg/h) and the dosing interval (8 h) with S and F = 1:

D = 8 × 3.1 = 24.8 mg

A dosing regimen of 25 mg of lipoamide administered every 8 h is recommended.

12.7.2.2 Protocol II: Targeting Specific Steady-State Peaks and TroughsStep 1.

Note the therapeutic range of the drug.

Step 2.

Note the patient’s estimated pharmacokinetic parameters (Cl, t1/2, Vd). As discussed above,wherever possible the patient’s individual characteristics should be considered in esti-mation of the parameter values.

Step 3.

Note the desired steady-state trough and peak and calculate a dosing interval necessary toachieve the desired trough/peak ratio using equation (12.5):

T = −1

kln

Cpmin,ss

Cpmax,ss(12.32)

Step 4.

Calculate the specific dose by substituting into the equation for either Cpmax,ss or Cpmin,ss.

Example 12.9 Determine a suitable dose and dosing interval for gentamicin to achievesteady-state peaks and troughs of 8 and 0.5 mg/L in a patient who is estimated to have thefollowing pharmacokinetic parameters: Vd = 17.5 L and t1/2 = 2.0 h.

Page 271: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

SIMULATION EXERCISE 251

Solution Note that the aminoglycosides are administered as short intermittent infusions,but this calculation will be performed assuming administration by multiple bolus injections.The doses determined may differ from those used clinically.

The dosing interval to achieve the desired peak and trough is calculated from equation(12.32):

� = − 2.0

0.693 ln

0.5

8= 8 h

The dose can be determined using the equation for either the peak or trough at steady state.Using equation (12.12) for the peak at steady state,

8 = 1 × 1 × D

17.5(1 − e−8×0.693/2)

D = 131 mg

A dose of around 130 mg of gentamicin administered every 8 h is recommended.

Example 12.10 A drug company is developing a new anticancer drug. Phase I studieshave established the population average pharmacokinetic parameters (Cl = 15 L/h andVd = 2.3 L/kg). Animal studies indicated that optimum response is obtained with a troughand peak of 0.35 and 0.65 mg/L, respectively. Determine a suitable dosing regimen in anaverage 70-kg patient.

Solution The drug’s t1/2 = 0.693 × 70 × 2.3/15 h = 7.44 h. The dosing interval to achievethe desired peak and trough is calculated from equation (12.32):

� = − 7.44

0.693 ln

0.35

0.65= 6.64 h

The dosing interval would probably be rounded to 6 h and the dose could be calculatedfrom the equation for either the peak or trough at steady state. Using the equation for thepeak at steady state, equation (12.12), yields

0.65 = 1 × 1 × D

70 × 2.3 (1 − e−6×0.693/7.44)

D = 44.8 or 45.0 mg

The recommended dose is 45 mg every 6 h.

12.8 SIMULATION EXERCISE

Open the model “Multiple Intravenous Bolus Injections” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter12

Page 272: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

252 MULTIPLE INTRAVENOUS BOLUS INJECTIONS IN THE ONE-COMPARTMENT MODEL

Default settings for the model are dose = 120 mg, � = 6 h, Cl = 4 L/h, and Vd = 50 L.

1. Review the objectives and the “Model Summary” page.

2. Go to the “Cp–Time Profile” page. Do a simulation and note that the peaks occurwhen a dose is given (t = 0) and a trough occurs just before the next dose(t = � ). Also note that the plasma concentration at any time during a dosinginterval increases with dose. But note that this increase lessens with each dose.Eventually, the accumulation stops. At this time all the peaks and troughs areexactly the same; steady state has been achieved.

3. Go the “Fluctuation and Accumulation” page to observe how the dosing intervalinfluences fluctuation and accumulation. To maintain the same average plasmaconcentration, the same rate of drug administration (20 mg/h) must be usedthroughout these simulations. Thus, when a dosing interval is altered, propor-tional changes to the dose must be made.

(a) Assess fluctuation, [(Cpmax,ss − Cpmin,ss)/Cpmax,ss] · 100%.

(b) Assess accumulation, Cpmax,ss/Cpmax,1.

(c) Record in Table SE12.1 how dosing intervals of 2, 6, and 12 h affect fluctua-tion and accumulation.

TABLE SE12.1 Effect of the Dosing Interval on Fluctuation and Accumulation

DosingInterval(h)

Rate ofAdministration

(mg/h)Dose(mg)

Fluctuation[(Cpmax,ss − Cpmin,ss)/

Cpmax,ss] · 100%Accumulation

(Cpmax,ss/Cpmax,1)2 206 20

12 20

4. Go the “Effect of Clearance” page and conduct simulations with clearance equalto 2, 4, and 12 L/h. Record in Table SE12.2 how clearance affects the parameters.

TABLE SE12.2 Influence of Clearance on the Cp–Time Profile for Multiple IVBolus Injections

Cl(L/h)

Cpmax,1

(mg/L)Cpav,ss

(↓, ↑, or ↔a)

Time toSteady State(↓,↑, or ↔a)

Fluctuation[(Cpmax,ss − Cpmin,ss)/

Cpmax,ss] · 100%Accumulation

(Cpmax,ss/Cpmax,1)2 N.A. N.A.4

12aCompare to the first simulation. Use ↑ for increase, ↓ for decrease, and ↔ for no effect.

5. Go to the “Effect of Volume of Distribution” page and conduct simulations with avolume of distribution equal to 20, 40, and 80 L. Record in Table SE12.3 how thevolume of distribution affects the parameters.

Page 273: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc12 JWBS062-Rosenbaum February 16, 2011 7:59 Printer Name: Yet to Come

REFERENCE 253

TABLE SE12.3 Influence of Volume of Distribution on the Cp–Time Profile forMultiple IV Bolus Injections

Vd(L)

Cpmax,1

(mg/L)Cpav,ss

(↓, ↑, or ↔a)

Time toSteady State(↓, ↑, or ↔a)

Fluctuation[(Cpmax,ss − Cpmin,ss)/

Cpmax,ss] · 100%Accumulation

(Cpmax,ss/Cpmax,1)25 N.A. N.A.50

100aCompare to the first simulation. Use ↑ for increase, ↓ for decrease, and ↔ for no effect.

PROBLEMS

12.1 Recommend a pharmacokinetic-based multiple intravenous bolus dosing regimen forthe fictitious drug nosolatol. Single-dose studies indicate that plasma concentrationsin the range 1900 to 750 �g/L are therapeutic. The major pharmacokinetic parametersof nosolatol are Cl = 12.6 L/h, Vd = 210 L/70 kg, and S = 1. Design a dosing regimenthat will achieve an average steady-state plasma concentration of 1200 �g/L.

12.2 Recommend a pharmacokinetic-based multiple intravenous bolus dosing regimen forthe fictitious drug disolvprazole. Single-dose studies indicate that plasma concen-trations in the range 1000 to 100 �g/L are therapeutic. The major pharmacokineticparameters of disolvprazole are Cl = 12 L/h, Vd = 35 L/70 kg, and S = 1.

12.3 A synthetic analgesic has the following pharmacokinetic and pharmacodynamicparameters: Vd = 0.2 L/kg, Cl = 0.012 L/h/kg, and S = 1; the therapeutic range isbetween 0.5 and 1.5 mg/L.

(a) Determine an appropriate dosing regimen for a 75-kg male who has just under-gone major orthopedic surgery.

(b) Would you recommend a loading dose? If so, calculate one.

(c) After 10 days of treatment the patient develops toxicity, and therapy iswithdrawn:

(1) How long will it take to eliminate 99% of the drug in the body?

(2) What is the Cp value 24 h after the last dose?

12.4 A 70-kg man is to receive quinidine, an antiarrthymic drug used in the treatmentof atrial fibrillation and other cardiac arrhythmias. The pharmacokinetic parametersin this man are estimated to be Cl = 4 ml/min/kg, Vd = 3.0 L/kg, and measuredhalf-life = 6.9 h. Use the intravenous bolus formula to devise a dosing regimen forquinidine sulfate (S = 0.82; F = 0.73) that will maintain the plasma concentrationsin the range 3 to 1.5 mg/L.

REFERENCE

1. Winter, M. E. (2010) Basic Clinical Pharmacokinetics, 5th ed., Lippincott Williams & Wilkins,Baltimore.

Page 274: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

13MULTIPLE INTERMITTENT INFUSIONS

13.1 Introduction

13.2 Steady-State Equations for Multiple Intermittent Infusions

13.3 Monoexponential Decay During a Dosing Interval: Determination of Peaks, Troughs, andElimination Half-Life13.3.1 Determination of Half-Life13.3.2 Determination of Peaks and Troughs

13.4 Determination of the Volume of Distribution

13.5 Individualization of Dosing Regimens

13.6 Simulation Exercise

Problems

Objectives

The material presented in this chapter will enable the reader to:

1. Understand the characteristics of the plasma concentration profile after multipleintermittent infusions

2. Understand the derivation of an expression for the steady-state plasma concentrationsachieved by intermittent infusions

3. Use two steady-state plasma concentrations to determine a drug’s half-life, the steady-state peaks and troughs, and volume of distribution

4. Individualize doses for patients receiving multiple intermittent infusions

13.1 INTRODUCTION

When drugs are administered as bolus injections, typically they are injected over a periodof a minute or more to avoid very high initial concentrations. Even with this approach, theinitial plasma concentrations of some drugs may still be high enough to cause toxicity. This

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

254

Page 275: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

INTRODUCTION 255

can be especially problematic for drugs that display two-compartment pharmacokinetics,where the initial distribution volume is small. These drugs can be administered more safelyby extending the administration period and infusing the dose at a constant rate over a periodof anywhere from half an hour, up to 2 h or more. The administration is then repeated with thesame frequency that it would be for bolus injections. This type of drug administration thusconsists of multiple intermittent infusions or multiple short infusions. The aminoglycosideantibiotics are important examples of drugs administered in this way. These drugs areused to treat serious life-threatening gram-negative infections, and their use is complicatedby their potential to cause serious renal and hearing impairment. As a result of bothwide interindividual variability in their pharmacokinetics and the serious consequencesof either subtherapeutic or toxic concentrations, plasma concentrations of these drugsare usually monitored and doses individualized. A knowledge and understanding of thepharmacokinetics and associated equations of multiple intermittent infusions is necessaryto perform this process.

The characteristics and equations for multiple intermittent infusions are very similarto those for multiple bolus injections. Figure 13.1 shows the typical plasma concentrationprofile associated with this type of administration, which in common with bolus injections,demonstrates fluctuation and an accumulation of drug in the buildup to steady state. Againin common with multiple bolus injections, it takes 3 to 5 elimination half-lives to achievesteady state. When a specific dosing interval is studied, some important differences betweenthe two forms of drug administration become apparent (Figure 13.2). Although the troughconcentration occurs at the end of the dosing interval, the peak plasma concentrationsobserved with multiple infusions do not occur at the beginning of the dosing interval butwhen the infusion is stopped.

The symbols used are the same as those used for bolus injections: t is the time that haselapsed since the dose was administered (start of the infusion) and � is the dosing intervalor time between doses. Tau varies constantly from zero to � , to zero to � , and so on. Theequations for multiple infusions incorporate an additional time parameter, the duration ofthe infusion (tinf). The dosing interval can be broken down into two parts: the period from

0

1

2

0 8 16 24 32 40 48

Cp

(mg/

L)

Time into Therapy (h)tinf

FIGURE 13.1 Typical plasma concentration–time profile observed with multiple intermittentinfusions. The duration of the infusion is tinf and the interval between the start of consecutiveinfusions is � .

Page 276: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

256 MULTIPLE INTERMITTENT INFUSIONS

Cp

Cpmax,ss

max,ssssCp •e−k(t − tinf)= Cp

t: 0 τe.g.

MonoexponentialDecay

tinf

tinf

A B

t – tinf

8 (h)0 1

inf)min,ss max,ss

k(Cp •e τ− − t= Cp

FIGURE 13.2 Multiple intermittent infusions: plasma concentrations during a steady-state dosinginterval. Time t is the time after the start of the infusion. The dosing interval can be separated intotwo periods: A and B. During period A the infusion is running and plasma concentrations increase.Note that the peak occurs when the infusion stops (t = tinf). During period B (from t = tinf to t = � ),there is no ongoing drug input, and plasma concentrations decay monoexponentially.

time 0 to tinf when the infusion is running, during which plasma concentrations increase(A in Figure 13.2); and the period from the end of the infusion to the end of the dosinginterval (period tinf to � ) when the infusion is not running. During the latter period, plasmaconcentrations fall monoexponentially under the influence of first-order elimination (B inFigure 13.2). Note in Figure 13.2 that the peak occurs when t = tinf, and a trough is obtainedat the end of the dosing interval when t = � .

In a dosing interval, once the infusion stops, the plasma concentration falls monoexpo-nentially from the peak. As a result, the plasma concentration at any time during this period(B in Figure 13.2) can be expressed in terms of its decay from the peak and the time thathas elapsed from the peak (t − tinf). For example, at steady state:

Cpss = Cpmax,ss · e−k(t−tinf ) (13.1)

where Cpss is the concentration at any time, t, during the dosing interval and Cpmax,ss isthe steady-state peak. Cp at the trough during a steady-state dosing interval (Cpmin,ss) isexpressed as

Cpmin,ss = Cpmax,ss · e−k(�−tinf ) (13.2)

13.2 STEADY-STATE EQUATIONS FOR MULTIPLEINTERMITTENT INFUSIONS

In Chapter 12 we demonstrated that an equation for multiple dosing could be constructedas follows (Figure 12.6):

Cpmultiple doses = Cpsingle dose × fraction of steady state at anytime

× final accumulation at steady state

Page 277: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

STEADY-STATE EQUATIONS FOR MULTIPLE INTERMITTENT INFUSIONS 257

Expressions for the fraction of steady state achieved at any time and the final accumulationat steady state were derived in Chapter 12 [equations (12.27) and (12.23), respectively].Thus,

Cpmultiple doses = Cpsingle dose · (1 − e−nk� ) · 1

1 − e−k�(13.3)

If the equation is to be limited to steady state, the fraction of steady state (1 − e−nk� ) equals1 and can be removed from the expression:

Cpmultiple doses,ss = Cpsingle dose · 1

1 − e−k�(13.4)

The equation for the plasma concentration at any time after a single infusion was presentedin Chapter 11 [equation (11.16)]:

Cpsingle infusion = S · F · k0 · (1 − e−kT )

Cl· e−kt ′

(13.5)

where T is the time the infusion was terminated and t′ is the time elapsed since termination.Next we substitute the time symbols used for multiple infusions: T = tinf and t′ = t − tinf.Substituting these symbols into equation (13.5) and then substituting into equation (13.4)yields

Cpss = S · F · k0 · (1 − e−ktinf )

Cl · (1 − e−k� )· e−k(t−tinf ) (13.6)

where Cpss is the plasma concentration at any time during a steady-state dosing interval,k0 the infusion rate, S the salt factor of the drug, F the bioavailability, k the eliminationrate constant, tinf the duration of the infusion, t the time elapsed since the infusion wasstarted, Cl the clearance, and � the dosing interval or time between the start of consecutiveinfusions.

A peak plasma concentration occurs at the end of the infusion when t = tinf:

Cpmax,ss = S · F · k0 · (1 − e−ktinf )

Cl · (1 − e−k� )(13.7)

A trough plasma concentration occurs at the end of the dosing interval (t = � ), immediatelybefore the next infusion is started:

Cpmin,ss = S · F · k0 · (1 − e−ktinf )

Cl · (1 − e−k� )· e−k(�−tinf ) (13.8)

Example 13.1 A drug (Cl = 8.67 L/h; Vd = 100 L; and S = 1) was administered asmultiple intravenous infusions. The dose (80 mg) was administered over a 1-h period every8 h. Calculate the estimated plasma concentrations in the third day of therapy, at (a) 1 h,(b) 2 h, (c) 4 h, and (d) 8 h after the start of the first infusion of the day.

Page 278: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

258 MULTIPLE INTERMITTENT INFUSIONS

Cp

Cpmax,ss

0 8 t (h)Infusion

Startsk0 = 80 mg/h

InfusionStops

??

?

?

inf)Cpss •e−k(t − t= Cpmax,ss

inf)min,ss max,ss

k(Cp •e τ− − t= Cp

421

FIGURE 13.3 Example problem using multiple intermittent infusion equations. The drug is ad-ministered at a rate of 80 mg/h over a 1-h period. Plasma concentrations at 1, 2, 4, and 8 h must becalculated after the first dose on the third day.

Solution The drug has a t1/2 = 0.693 × 100/8.67 = 8 h. A dose of 80 mg is infused over1 h: k0 = 80 mg/h. S = 1, F = 1. The problem is summarized in Figure 13.3. It will takeabout 24 to 40 h to get to steady state. By the third day, steady state will have been achievedand the profiles from the three infusions of the day will be exactly the same.

(a) When t = 1 h, the infusion has just been terminated, Cp = Cpmax,ss. Substitutinginto equation (13.7) yields

Cpmax,ss = 80(1 − e−1×0.693/8)

8.67(1 − e−8×0.693/8)= 1.53 mg/L

(b) When t = 2 h, it is 1 h after the infusion stops.

Cpss,t=2 = 80(1 − e−1×0.693/8)

8.67(1 − e−8×0.693/8)· e−(2−1)0.693/8

or

Cpss,t=2 = Cpmax,ss · e−(2−1)0.693/8 = 1.53e−(2−1)0.693/8 = 1.40 mg/L

(c) When t = 4 h, it is halfway through the dosing interval and 3 h after the infusionhas stopped:

Cpss,t=4 = Cpmax,ss · e−(4−1)0.693/8 = 1.53e−(4−1)0.693/8 = 1.18 mg/L

(d) When t = 8 h, it is the end of the dosing interval and 7 h from the peak:

Cpss,t=8 = Cpmin,ss = Cpmax,ss · e−(8−1)0.693/8 = 1.53e−(8−1)0.693/8 = 0.83 mg/L

Page 279: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

MONOEXPONENTIAL DECAY DURING A DOSING INTERVAL 259

Example 13.2 A 0.5 h infusion is used to administer the dose (80 mg) of the drugdescribed in Example 13.1. Calculate the estimated plasma concentrations on the third dayof therapy, at (a) 0.5 h, (b) 1 h, (c) 2 h, and (d) 8 h after the start of the infusion.

Solution The drug has a t1/2 = 0.693 × 100/8.67 = 8 h. A dose of 80 mg is infused over0.5 h: k0 = 160 mg/h. S = 1, F = 1. It will take about 24 to 40 h to get to steady state. Bythe third day, steady state will have been achieved. A diagram of the steady-state dosinginterval is shown in Figure 13.3 but in this example tinf = 0.5 h.

(a) When t = 0.5, the infusion has just been terminated, Cp = Cpmax,ss. Substitutinginto equation (13.7), we have

Cpmax,ss = 160(1 − e−0.5×0.693/8)

8.67(1 − e−8×0.693/8)= 1.58 mg/L

(b) When t = 1, it is 0.5 h after the infusion stops.

Cpss,t=1 = Cpmax,ss · e−(1−0.5)0.693/8 = 1.58e−(1−0.5)0.693/8 = 1.51 mg/L

(c) When t = 2, it is 1.5 h after the infusion stops.

Cpss,t=2 = Cpmax,ss · e−(2−0.5)0.693/8 = 1.58e−(2−0.5)0.693/8 = 1.38 mg/L

(d) When t = 8, it is the end of the dosing interval and 7.5 h from the peak

Cpss,t=8 = Cpmin,ss = Cpmax,ss · e−(8−0.5)0.693/8 = 1.58e−(8−0.5)0.693/8 = 0.82 mg/L

13.3 MONOEXPONENTIAL DECAY DURING A DOSING INTERVAL:DETERMINATION OF PEAKS, TROUGHS, AND ELIMINATION HALF-LIFE

13.3.1 Determination of Half-Life

A drug’s elimination half-life can be determined from two plasma concentrations measuredduring the period when the infusion is not running (period tinf to � ), when plasma con-centrations are falling monoexponentially as a result of first-order elimination. During thisperiod,

Cpn = Cpmax,n · e−k(t−tinf ) (13.9)

where Cpmax,n and Cpn are the peak and the plasma concentration at time t, respectively,during the nth dosing interval. Taking the logarithmic of equation (13.9) yields

ln Cpn = ln Cpmax,n − k · (t − tinf) (13.10)

Thus, k can be determined:

k = ln(Cp1/Cp2)

t2 − t1(13.11)

Page 280: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

260 MULTIPLE INTERMITTENT INFUSIONS

and the half-life

t1/2 = 0.693

k(13.12)

Example 13.3 A patient is being treated with gentamicin. A 140-mg dose is administeredas a short infusion over a 1-h period every 8 h. Plasma concentrations of the drug weredetermined during the first dosing interval and are given in Table E13.3. Assume onecompartmental pharmacokinetics and calculate the drug’s half-life.

TABLE E13.3

Time After Start ofInfusion (h)

Gentamicin Concentration(mg/L)

1.5 6.16 2.2

Solution The problem is summarized in Figure E13.3. The plasma samples were takenduring a period when the infusion was not running. Thus, the only process affecting plasmaconcentrations is first-order elimination, and equation (13.11) can be used to determine therate constant and half-life.

k = ln(6.1/2.2)

6 − 1.5= 0.227 h−1

t1/2 = 0.693

0.227= 3.05 h

Cpmax,1

0 1 1.5 6 8 t (h)Infusion

Starts140 mg/h

?

?

6.1

2.2

Cp

max,nnCp Cp •e−k(t − tinf )=

min,n max,nCp Cp •e τ−k( − tinf)=

InfusionStops

FIGURE E13.3 Calculation of the half-life. If two plasma concentrations are known in the periodwhen the infusion is not running, the half-life can be calculated. Once the half-life is known, thepeaks and troughs associated with this dosing interval may also be calculated.

Page 281: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

DETERMINATION OF THE VOLUME OF DISTRIBUTION 261

13.3.2 Determination of Peaks and Troughs

If a drug’s half-life or elimination rate constant is known, and if at least one plasmaconcentration during the dosing interval is known, the values of the peak and troughassociated with the dosing interval can be calculated.

Example 13.4 Continuing with Example 13.3, determine the trough and peak of thedosing interval described above.

Solution The drug’s elimination rate constant is 0.227 h−1. Once the infusion is stopped,plasma concentrations decay monoexponentially (Figure 13.4):

Cpn = Cpmax,n · e−k(t−tinf ) (13.13)

Cpn = 6.1 mg/L when t = 1.5 h and t − tinf = 0.5 h. Substitute in equation (13.13) todetermine the peak:

6.1 = Cpmax,n · e−0.227×0.5

Cpmax,n = 6.83 mg/L

The trough occurs when t = � and t − tinf = 7 h:

Cpmin,n = Cpmax,n · e−0.227×7

= 6.83e−0.22×7 = 1.39 mg/L

13.4 DETERMINATION OF THE VOLUME OF DISTRIBUTION

The volume of distribution can be calculated if a peak plasma concentration and its previoustrough are known.

Theory The equation for the volume of distribution is derived by considering the amountof drug in the body at the time of a peak (Abmax,n) (Figure 13.4). The amount of drug inthe body at the time of a peak can be partitioned into two parts: drug from the most recentinfusion; and drug remaining from previous infusions. At the peak:

Abmax,n = drug from the most recent infusion

+ the amount remaining from previous infusions (13.14)

The amount of drug from the most recent infusion is determined by multiplying Vd by theequation for the plasma concentration from a single infusion [see equation (13.5)], withT = tinf and t′ = 0):

Abrecent = Vd · S · F · k0

Cl· (1 − e−ktinf ) (13.15)

As Cl = k · Vd,

Abrecent = Vd · S · F · k0

k · Vd· (1 − e−ktinf ) (13.16)

Page 282: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

262 MULTIPLE INTERMITTENT INFUSIONS

Ab

Abmax,n = amount from most recent infusion + amount remaining from Abmin,(n–1)

0 tinf τ tInfusion

Starts

Abmin,(n–1)

InfusionStops

FIGURE 13.4 Determination of the volume of distribution. The derivation of the equation for Vdis based on partitioning the amount of drug in the body at the time of a peak into two components: (1)the amount of drug that came from the most recent infusion, and (2) the amount of drug remainingfrom the previous trough.

The amount remaining from previous infusion(s) is equal to the amount at the time of theprevious trough (Abmin,(n −1)) minus the amount eliminated during the recent infusion:

Abprevious = Abprevious trough − elimination during infusion= Vd · Cpmin,(n−1) · e−ktinf

(13.17)

Substituting the expression of the amount from the most recent infusion (13.15) and theamount from previous infusion(s) [equation (13.17)] into equation (13.14) gives us

Abmax,n = Vd · S · F · k0

k · Vd· (1 − e−ktinf ) + Vd · Cpmin,(n−1) · e−ktinf (13.18)

Substituting Cpmax,n · Vd for Abmax,n results in

Vd · Cpmax,n = Vd · S · F · k0

k · Vd· (1 − e−ktinf ) + Vd · Cpmin,(n−1) · e−ktinf

Cpmax,n = S · F · k0

k · Vd· (1 − e−ktinf ) + Cpmin,(n−1) · e−ktinf

Vd = S · F · k0 · (1 − e−ktinf )

k(Cpmax,n − Cpmin,(n−1) · e−ktinf )

(13.19)

Practice From equation (13.19) it can be seen that the volume of distribution can becalculated if a peak concentration (Cpmax,n) and a previous trough (Cpmin,(n − 1)) are known.If the therapy is at steady state, any peak and trough may be used because the troughs andpeaks are exactly the same at steady state.

Page 283: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

DETERMINATION OF THE VOLUME OF DISTRIBUTION 263

Example 13.5 The suitability of a gentamicin regimen (110 mg every 8 h infused over a1-h period) is being assessed on the third day of treatment. A steady-state trough concen-tration is found to be 3.9 mg/L. The plasma concentration 1.5 h after the start of the mostrecent infusion is found to be 7.5 mg/L.

(a) Determine the elimination half-life.

(b) Determine the peak plasma concentration after the most recent infusion.

(c) Determine the volume of distribution.

Solution The infusion is at steady state, so all the peaks and troughs will be the same.The profile associated with this problem is shown in Figure 13.5.

The elimination rate constant may be calculated:

k = ln(7.5/3.9)

8 − 1.5= 0.101 h−1

(a) The elimination half-life is determined:

t1/2 = 0.693

k= 6.86 h

(b) The steady-state peak can be determined:

7.5 = Cpmax,n · e−0.101×0.5

Cpmax,n = 7.89 mg/L

Cp

Cpmax,ss

0 8 t (h)Infusion

Startsk0 = 110 mg/h

3.9 mg/L3.9 mg/L

7.5 mg/L?

1.51

StopsInfusion

FIGURE 13.5 Example calculation of the volume of distribution. This parameter is determinedfrom a peak plasma concentration and its previous trough. Initially, the half-life must be estimated inorder to calculate the peak plasma concentration.

Page 284: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

264 MULTIPLE INTERMITTENT INFUSIONS

(c) The volume of distribution may be calculated from equation (13.19):

Vd = 110(1 − e−0.101×1)

0.101(7.89 − 3.9 × e−0.101×1)= 23.9 L

13.5 INDIVIDUALIZATION OF DOSING REGIMENS

The methods used to design the dosing regimens discussed in Chapter 12 can be applied tomultiple intermittent infusions.

Example 13.6 Continuing with Example 13.5, the trough concentration found in the pa-tient was considered to be too high. Design another regimen that will provide a gentamicinsteady-state peak and trough of 8 and 0.5 mg/L, respectively. The pharmacokinetic parame-ters in this patient were found to be k = 0.101 h−1, Vd = 23.9 L, and Cl = k · Vd = 2.41 L/h.

Solution Here the focus of the regimen is on the achievement of specific peaks andtroughs. Recall the equation introduced in Chapter 12 to calculate a dosing interval toprovide specific peaks and troughs with multiple intravenous injections:

� = −1

kln

Cpmin,ss

Cpmax,ss(13.20)

For multiple infusions, the time between the peak and the trough is � − tinf (Figure 13.2)and not � as it is for multiple bolus injections. Thus, equation (13.20) will have to bemodified appropriately:

� − tinf = −1

kln

Cpmin,ss

Cpmax,ss(13.21)

Using equation (13.21) to calculate the necessary dosing interval for peaks and troughs of8 and 0.5 mg/L, respectively, yields

� − tinf = − 1

0.101ln

0.5

8= 27.5 h

� = 28.5 h, which would be rounded to 24 h

The appropriate dose could be calculated using the equation for either the peak (8 mg/L)or the trough (0.5 mg/L). Using the peak equation (13.7), we have

Cpmax,ss = 8 = k0 · (1 − e−1×0.101)

2.41(1 − e−8×0.101)

k0 = 111 mg

To obtain peaks and troughs of 8 and 0.5 mg/L, 111 mg of gentamicin should be infusedover a 1-h period every 24 h.

Page 285: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

PROBLEMS 265

13.6 SIMULATION EXERCISE

Open the model “Multiple Intermittent Infusions” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter13

Default settings for the model are dose = 80 mg, � = 8 h, t inf = 1 h, Cl = 8.67 L/h, andVd = 20 L.

1. Review the objectives and the “Model Summary” page

2. Go to the “Cp–Time Profile” page. Do a simulation and note that the peaks occurwhen the infusion stops (t = t inf) and a trough occurs just before the next dose(t = � ). Also note that accumulation occurs with successive doses but stops atsteady state.

3. Go the “Influence of Infusion Duration” page. The overall rate of drug adminis-tration or the dose administered will remain constant (80 mg every 8 h), but theduration of the infusion will be altered. Since the individual dose will remain con-stant, the infusion rate will change in inverse proportion to the infusion duration.This adjustment will be made automatically by the software. Observe how theprofile changes with infusion of duration 0.5, 2, and 4 h. (The corresponding infu-sion rates will be 160, 40, and 20 mg/h). As the duration of the infusion increases,the peak gets lower and the trough gets higher; that is, fluctuation decreases.

4. Go to the “Determine a Dosing Regimen” page. Use the drugs pharmacokineticparameters (Cl = 8.67 L/h and Vd = 20 L) and a 1 h infusion to calculate adose and a dosing interval that will provide a peak and trough of 10 and 1 mg/L,respectively. Check your answers using the model.

PROBLEMS

13.1 A drug (Cl = 1.73 L/h, Vd = 30 L, S = 1) was administered as multiple shortintravenous infusions. A dose of 40 mg was administered over a 1-h period every12 h.

(a) How long will it take to get to steady state?

(b) Calculate the peak plasma concentration during a steady-state dosing interval.

(c) Calculate the plasma concentration 6 h into a steady-state dosing interval.

(d) Calculate the trough plasma concentration during a steady-state dosing interval.

13.2 If the dose of the drug discussed above was infused over a 0.5-h period, calculatethe steady-state peak and trough concentrations.

13.3 If the dose of the drug above was infused over a 2-h period, calculate the steady-statepeak and trough concentrations.

13.4 A 38-year-old male patient (75 kg) is given gentamicin (120 mg every 8 h infusedover a 1-h period). Plasma samples taken after the second dose were analyzed forgentamicin and the results are shown in Table P13.4.

Page 286: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc13 JWBS062-Rosenbaum February 16, 2011 8:4 Printer Name: Yet to Come

266 MULTIPLE INTERMITTENT INFUSIONS

TABLE P13.4

Time After Startof Infusion (h)

GentamicinCp (mg/L)

2 3.847 1.10

(a) What is gentamicin’s half-life in this patient?

(b) What is the peak plasma concentration of this dosing interval?

(c) What is the trough plasma concentration of this dosing interval?

13.5 Tobramycin (120 mg) is being infused over a 1 h period every 8 h. Three days intotherapy, at steady state, tobramycin concentrations at 1 and 8 h after the start of aninfusion were found to be 7.12 and 0.75 mg/L, respectively. Calculate the drug’s half-life and it volume of distribution. Note that because steady state has been achieved,the trough concentration of the previous dosing interval can also be assumed to be0.75 mg/L.

13.6 Gentamicin (140 mg) is infused over a 1-h period every 8 h. A steady-state peak andtrough of 10 and 0.25 mg/L, respectively, are required. At steady state, gentamicinconcentrations at 2 and 8 h after the start of an infusion were found to be 5.5 and0.75 mg/L, respectively. Is this regimen satisfactory? If not, recommend a regimenthat would achieve the desired peaks and troughs of gentamicin concentrations.

Page 287: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

14MULTIPLE ORAL DOSES

14.1 Introduction

14.2 Steady-State Equations14.2.1 Time to Peak Steady-State Plasma Concentration14.2.2 Maximum Steady-State Plasma Concentration14.2.3 Minimum Steady-State Plasma Concentration14.2.4 Average Steady-State Plasma Concentration14.2.5 Overall Effect of Absorption Parameters on a Steady-State Dosing Interval

14.3 Equations Used Clinically to Individualize Oral Doses14.3.1 Protocol to Select an Appropriate Equation

14.4 Simulation Exercise

Objectives

The material presented in this chapter will enable the reader to:

1. Understand the characteristics of the plasma concentration profile after multiple oraldoses

2. Compare the profile to other modes of chronic drug administration

3. Understand how F and ka influence the plasma concentration profile

4. Identify an appropriate intravenous formula to use to individualize oral doses clini-cally

14.1 INTRODUCTION

The oral route is the most popular and common form of drug administration. Consequently,it is important for health professions and scientists involved in evaluation of the dose–plasmaconcentration relationship to have good knowledge and understanding of any special phar-macokinetic characteristics of this route. It has the same accumulation properties in thebuildup to steady state, and it takes the same time to reach steady state as do other forms ofmultiple doses (Figure 14.1). Fluctuation also occurs, and in common with the fluctuation

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

267

Page 288: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

268 MULTIPLE ORAL DOSES

0.0

2.0

4.0

0 1 2 3 4 5 6 7 8 9Time into Therapy (t1/2)

Cp

FIGURE 14.1 Typical plasma concentration–time profile observed with multiple oral doses for adrug administered every half-life.

observed with multiple short infusions, the peak is seen slightly later than the time at whichthe dose is administered (Figure 14.1).

The pharmacokinetics of oral drug administration was introduced in Chapter 9, whereit was seen that the pharmacokinetic model contains two absorption parameters: F, thefraction of the dose that reaches the systemic circulation and ka, the first-order rate constantfor absorption. It follows that a focus of the discussion on the pharmacokinetics of multipleoral doses should be on how these two special absorption parameters influence the plasmaconcentration–time profile.

The clearest way to demonstrate the influence of these parameters is, first, through aconsideration of the equations for multiple oral doses, and then through computer simu-lations. In this chapter we concentrate on steady-state concentrations. It is not uncommonfor bioavailability and bioequivalence studies to be carried out at steady state. Thus, anadditional and related topic discussed in this chapter is the justification and validation forconducting bioavailability and bioequivalence studies at steady state.

14.2 STEADY-STATE EQUATIONS

As discussed in Chapter 13, steady-state equations for multiple dosing therapies can beobtained as follows:

Cpmultiple doses, ss = Cpsingle doses, ss · accumulation ratio(r )

and

r = 1

1 − e−kn ·� (14.1)

where kn is a rate constant in an exponential expression in the single-dose equation.

Page 289: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

STEADY-STATE EQUATIONS 269

In the equation for single oral doses,

Cp = S · F · D · ka

Vd · (ka − k)· (

e−kt − e−ka t)

(14.2)

it can be seen that it contains two exponential functions. To derive the steady-state equationsfor multiple doses, each exponential function has to be multiplied by its own accumulationratio. The steady-state plasma concentration after multiple oral doses may be expressed

Cpss = S · F · D · ka

Vd · (ka − k)·(

e−kt

1 − e−k�− e−ka t

1 − e−ka �

)(14.3)

where t is the time since the last dose and � is the dosing interval or time between doses.The typical profile observed after multiple oral doses and the one predicted by equation

(14.3) is shown in Figure 14.1, where the typical accumulation of drug up to steady state ispresent, as is fluctuation. We also see that the peak plasma concentration (Cmax) does notoccur at time zero (i.e., when a dose is given) but later. In common with the symbols usedfor single oral doses, the time of the peak plasma concentration is Tmax. After single doses,these parameters are important measures of the rate and extent of drug absorption, and as aresult they are used to assess bioavailability. The determinants of Cmax and Tmax at steadystate are addressed next.

14.2.1 Time to Peak Steady-State Plasma Concentration

At the time of a peak during multiple oral doses, the rate of absorption is momentarily equalto the rate of elimination, and the rate of change of Cp with time is zero. The expression forthe time of the maximum steady-state plasma concentration can be obtained by differenti-ating equation (14.3) and setting dCp/dt = 0. Upon rearrangement, the following equationis obtained (1):

Tmax − Tmax,ss = 1

ka − k· ln

1 − e−ka �

1 − e−k�(14.4)

where Tmax is the time of the peak plasma concentration after a single dose and Tmax,ss isthe time of the peak during a steady-state dosing interval. Recall from Chapter 9 that

Tmax = ln(ka/k)

ka − k

Several important points can be made from a consideration of equation (14.4):

1. The time of the peak steady-state plasma concentration for a given drug (k is constant)is a function only of the absorption rate constant. As a result, in common with itsapplication in single-dose studies, it can be used to assess the rate of drug absorptionin multiple-dose bioavailability studies. If the absorption rate constant increases, thetime to the peak will decrease, and vice versa.

2. Because the right-hand side of the equation must always be positive, it can beconcluded that it takes less time to reach the peak at steady state than to reach the

Page 290: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

270 MULTIPLE ORAL DOSES

peak after a single dose. Recall that after a dose, plasma concentrations increasebecause the rate of absorption is greater than the rate of elimination. As absorptionproceeds after a dose, the rate of absorption decreases as drug is depleted from thegastrointestinal tract and the rate of elimination increases as more drug gets into thebody. The peak occurs when the rates of the two processes are momentarily equal.As a result of accumulation, the amount of drug in the body increases with each dose.As a result, with each successive dose, it takes less time for the rate of eliminationto equal the rate of absorption, and Tmax decreases with each dose. At steady state,accumulation stops and Tmax,ss remains constant.

The determinants of Tmax,ss are discussed further in Section 14.2.5.

14.2.2 Maximum Steady-State Plasma Concentration

The peak plasma concentration at steady state occurs when the time after the dose is equalto Tmax,ss. Thus, the expression for the peak plasma concentration at steady state is obtainedby substituting the expression for Tmax,ss [equation (14.4)] into the equation for the plasmaconcentration during a steady-state dosing interval [equation (14.3)] (2).

Cmax,ss = S · F · D

Vd· 1

1 − e−k�· e−kTmax,ss (14.5)

A consideration of equation (14.5) shows that in common with the peak after a singledose, the peak at steady state is directly proportional to bioavailability (F), and through itsdependence on Tmax,ss is also dependent on the rate of drug absorption. If F increases, thevalue of the peak plasma concentration will increase, and vice versa. If the absorption rateconstant increases, Tmax,ss will decrease and Cmax,ss will increase. These relationships arediscussed further in Section 14.2.5.

14.2.3 Minimum Steady-State Plasma Concentration

Assuming that ka � k at later times during a dosing interval, e−kat in equation (14.3) willtend to zero. Additionally, the trough concentration occurs when t = � . Thus, equation(14.3) will simplify to

Cpmin,ss = S · F · D · ka

Vd · (ka − k)· e−k�

1 − e−k�(14.6)

From equation (14.6) it can be seen that the trough at steady state is equal to the plasmaconcentration at the same time after a single dose multiplied by the accumulation ratio.

14.2.4 Average Steady-State Plasma Concentration

The average steady-state plasma concentration is equal to the area under the curve duringa steady-state dosing interval divided by � :

AUC�0 =

∫Cpss · dt = S · F · D

Cl(14.7)

Page 291: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

STEADY-STATE EQUATIONS 271

0 2 4 6 8 10Time into Therapy (t1/2)

Cp

FIGURE 14.2 Area under the curve (AUC) at steady state and after a single dose. The AUC fromzero to � during a steady-state dosing interval is the same as the AUC from zero to infinity after asingle dose: S·F·D/Cl.

Note that the area under the curve during a steady-state dosing interval is exactly the sameas the area under the plasma concentration time curve from zero to infinity after a singledose (Figure 14.2). Additionally, equation (14.7) shows that for a given drug (Cl is constant)the AUC during a steady-state dosing interval is dependent only on the fraction of the doseabsorbed (F) and demonstrates that this area can be used to assess bioavailability.

The expression for the average steady-state plasma concentration can now be obtained:

Cpav,ss = S · F · D

Cl · �(14.8)

The equation demonstrates the dependency of the average steady-state plasma concentra-tion on clearance and the effective rate of drug administration. It also demonstrates itsindependence of the volume of distribution and the rate of drug absorption. The equationcan be used to determine the rate of drug administration to achieve a desired average con-centration. Note that equation (14.8)is exactly the same as that for the average steady-stateplasma concentration achieved by multiple intravenous bolus injections. However, withintravenous injections, bioavailability (F) is always equal to 1. In contrast, bioavailabilityassociated with oral doses may be less than 1. This is an important factor to consider whenconverting an intravenous dosing regimen to an oral dosing regimen. For example, thebioavailability of morphine after oral administration is about 0.3. Thus, for equivalency,the rate of drug administration for oral doses must be about 3.3 times greater than theintravenous rate.

14.2.5 Overall Effect of Absorption Parameters on a Steady-State Dosing Interval

It has been shown that the first-order absorption rate constant influences the Tmax,ss andCmax,ss but does not influence either AUC�

0 or Cpav,ss. The fraction of the dose absorbed

Page 292: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

272 MULTIPLE ORAL DOSES

0 Tmax,ss Tmax,ss Time

Cp

A

B

C

A B and C

AUC ,A = AUC ,B > AUC ,C

0 0 0

FIGURE 14.3 Steady-state plasma concentrations of three different formulations of a drug. Eachformulation has the same dose and is administered with the same frequency. Note that FA = FB �FC, and kaB = kaC � kaA . As a result of the differences, AUCA = AUCB � AUCC; Tmax,ss,B = Tmax,ss,C

� Tmax,ss,A. The Cmax,ss values of all three are different because they are influenced by both F and ka:Cpav,ss,A = Cpav,ss,B � Cpav,ss,C. Where AUC is the area under the concentration–time curve during asteady-state dosing interval.

(F) influences AUC�0, Cpav,ss, and Cmax,ss but not Tmax,ss. Figure 14.3 demonstrates these

principles and shows the steady-state profile obtained from three different formulations ofthe same drug. Each formulation contained the same dose and was administered with thesame frequency. The formulations differed with respect to the rate and extent of absorptionof the drug: FA = FB � FC and kaB = kaC � kaA. It can be seen that AUC�

0, Cmax,ss, andCpav,ss are proportional to F, but F does not affect Tmax,ss. It can be seen that if ka increases,Tmax,ss decreases and Cmax,ss increases, but ka has no influence on AUC�

0 and Cpav,ss.

14.3 EQUATIONS USED CLINICALLY TO INDIVIDUALIZE ORAL DOSES

Doses of drugs that have both a narrow therapeutic range and a wide interindividualvariability in their pharmacokinetics parameters are frequently individualized for eachpatient. The individualization process is usually conducted by applying pharmacokineticprinciples to the values of one or more plasma concentrations measured in a patient atspecific times after a dose(s). Many of these drugs are given orally and include digoxin,theophylline, cyclosporine, lithium, and others. The equations for multiple oral doses arerarely used clinically to individualize doses, as they include the parameter ka, which isdifficult to measure under the best of circumstances and impossible to measure in clinicalpractice where only one or two plasma concentrations of the drug may be available froma patient. Additionally, the equations are long and cumbersome and mistakes can easilybe made if only a handheld calculator is used. It has been found that provided that certainprecautions are taken, the simpler equations associated with intravenous administrationprovide a satisfactory degree of accuracy when applied to orally administered drugs.

Three different types of intravenous drug administrations have been presented in thisbook: continuous constant infusion; multiple bolus injections, and multiple intermittentinfusions. The most appropriate approximation for orally administered drugs depends on

Page 293: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

EQUATIONS USED CLINICALLY TO INDIVIDUALIZE ORAL DOSES 273

1. How much fluctuation? If absorption is constant and continuous throughout the dosing interval

If τ < 1/3 t1/2: Little dose eliminated in a dosing interval

Minimal Fluctuation: Use Continuous Infusion Equations

If τ > 1/3 t1/2: Much drug is eliminated in a dosing interval

Significant Flucutation: Use Discrete Dose Equations

ContinuousInfusion

DiscreteDoses

0.5

23

IntermittentInfusions

BolusInjections

2. How much elimination occurs during absorption period? If absorption period < 1/6 t1/2: little elimination during absorption

Minimal Elimination: Use Bolus Equations

If absorption Period > 1/6 t1/2: much elimination during absoprtion

Significant Elimination Use Intermittent Infusion Equations

1. Fluctuation ?

2. Elimination During Drug Input ?

or

FIGURE 14.4 Contrasting steady-state plasma concentration profiles from continuous drug infu-sion, multiple bolus injections, and multiple intermittent infusions. The upper panel compares theprofiles of continuous drug infusion to multiple discrete doses. The lower panel compares the profilefrom multiple bolus injections to multiple intermittent infusions. To evaluate which equations wouldbe most accurate to substitute for oral equations, the amount of fluctuation that occurs during thedosing interval is assessed. If little fluctuation occurs, continuous infusion equations may be used.If a significant amount of fluctuation occurs, multiple discrete dose equations must be used. To dis-tinguish between bolus injections and intermittent infusions, the amount of elimination that occursduring the absorption period is evaluated. If little elimination occurs, the bolus equations may beused; otherwise, the intermittent infusions equation will have to be used.

a drug’s elimination characteristics in a given patient and on the rate of absorption of aspecific formulation. A commonly used protocol to select an appropriate equation (3) isprovided below.

14.3.1 Protocol to Select an Appropriate Equation

Continuous Input or Multiple Discrete Dose Equations? The degree of fluctuation thatoccurs during a dosing interval differentiates these two modes of drug administration. Nofluctuation is present with continuous infusions, and clear fluctuation is present with thepulse-like administration of discrete doses (Figure 14.4). Multiple oral doses would producelittle fluctuation if:

A. The dose is absorbed at a constant rate throughout the entire course of the dosinginterval. If this is the case, the equations for a continuous infusion may be used. Forexample, the continuous infusion model is frequently applied to the slow release andabsorption of drugs from sustained-release preparations such as theophylline.

or

B. If the drug’s half-life is much greater than the dosing interval, little of the drug iseliminated during the dosing interval, fluctuation will be minimal, and the continuous

Page 294: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

274 MULTIPLE ORAL DOSES

infusion equations can be used. Commonly, a cutoff of a 20% loss of drug is used(3). If less than 20% of the drug in the body is lost during the dosing interval, thecontinuous infusion model may be used. Recall that 20% of the drug in the bodyis eliminated in a period equal to 1

3 t1/2 (Appendix B). Thus, if the dosing intervalis less than 1/3 t1/2, the continuous infusion equations may be used. For example,the continuous infusion model is often applied to oral digoxin, which has a dosinginterval (1 day) of about 20% its half-life (5 days). Thus, if:� � � 1/3t1/2: the continuous infusion model may be used.� � � 1/3t1/2: much drug is eliminated in the dosing interval and multiple discrete

dose equations must be used.

Multiple Bolus Injection or Multiple Intermittent Infusion Equations? When a drug isadministered as a bolus injection, the administration process is very rapid. The entire doseis administered almost instantaneously and no drug is eliminated during the administrationperiod. In the case of intermittent infusions, the administration period is longer (0.5 to 2 h)and elimination occurs during this input. To decide whether to use the multiple bolus injec-tion model or the intermittent infusion model for orally administered drugs, it is necessaryto assess how much elimination occurs during drug absorption. Commonly, a cutoff of 10%is used (3). If drug absorption is rapid relative to elimination and less than 10% of the drugin the body is eliminated during absorption, the bolus equations may be used. If not, theintermittent infusion equations must be used. Recall that 10% of the drug in the body is lostin a period of 1

6 t1/2 (Appendix B). For example, bolus equations are usually used for regular-release theophylline and valproic acid, both of which are absorbed very rapidly. Thus, if:

� Duration of absorption phase � 16 t1/2, multiple bolus equations may be used

� Duration of absorption phase � 16 t1/2, multiple intermittent infusion equations should

be used

14.4 SIMULATION EXERCISE

Open the model “Multiple Oral Doses” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter14

Default settings for the model are: the unit of time is the elimination half-life (t1/2); dose =200 mg; Cl = 34.7 L/t1/2; Vd = 50 L; ka = 8 t1/2

−1; � = 1 t1/2; F = 1; S = 1.

1. Review the objectives and the “Model Summary” Page.

2. Go to the “Cp–Time Profile” page. Do a simulation and note:� The peaks occur later than the time at which the dose is administered, but the

trough occurs just before the next dose (t = � ).� Accumulation causes the plasma concentration at any time during a dosing

interval to increase with dose. Accumulation decreases more and more witheach dose and eventually stops at steady state.

� It takes about 5 t1/2 to reach steady state.

Page 295: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

SIMULATION EXERCISE 275

� Hold the mouse over the first peak and a steady-state peak and note that T max,1

and T max,ss.

3. Go to the “Influence of ka” page. The influence of ka on the bioavailability param-eters and on fluctuation during a dosing interval will be addressed.

Bioavailability parameters. For drugs that have long half-lives, and for drugsthat cannot be administered to healthy volunteers, such as anticancer drugs,bioavailability and bioequivalence studies may be conducted during a steady-state dosing interval after multiple doses. Simulate with different values of ka andnote the effects on T max,ss, Cmax,ss, and the AUC.� Note that as ka increases, T max,ss decreases, Cmax,ss increases, and AUC is

unaltered.

Fluctuation. The default value of ka is 8 1/t1/2. This compares to a value of k of0.693 1/t1/2. Thus, as is typically observed, ka > k (in this case ka is just over 10times larger than k). Sustained-release products typically have values of ka thatare substantially less than k.

a. Clear the graph and simulate the plasma concentration with a ka value of 0.31/t1/2, which is about half the value of k. A more prolonged absorption resultsand little fluctuation is observed during the dosing interval. This is a flip-flopmodel (see Chapter 9), where the drug’s absorption and not its eliminationcontrols the fall in the plasma concentration at later times. When absorptionis prolonged, the equations for continuous infusions can be used clinically tocalculate doses.

b. Now increase ka to 8 1/t1/2. Note that much greater fluctuation is observedand the absorption period which, as shown in the display is 0.43 t1/2 (5 ∗

0.693/8). Thus, about 25% of a dose will be eliminated during this period andthe equations for multiple intermittent infusions will need to be used clinically.

c. Increase ka to 24 1/t1/2. Note that much sharper peaks are obtained and theprofile is similar to multiple bolus doses. Note also that the absorption periodis equal to (5 ∗ 0.693

24 ) or 0.14 t1/2. Less than 10% of the dose will be eliminatedin this period. The equations for multiple bolus injections may be used underthese circumstances.

4. Go to the “Influence of F” page. Simulate with the different values of F and notethe effects on T max,ss, Cmax,ss, and AUC (shown in the display window) during asteady-state dosing interval.� As was the case with a single oral dose, reductions in F result in decreases in

Cmax,ss and AUC, but T max,ss is unaffected.

5. Go to the “Influence of � ” page. In these simulations, � will be altered, and thedose must also be altered proportionally to maintain the default rate of drugadministration (200 mg/t1/2). In this way the same average steady-state plasmaconcentration will be achieved.

a. Let � = 0.25 t1/2. During a dosing interval slightly less than about 20%of the dose will be eliminated. Note that this value of � results in minimalfluctuation. When � is < 1

3 t1/2, the equations for a continuous infusion can beapproximated for multiple oral doses.

b. Let tau = 2 t1/2’s. Note the much greater fluctuation. About 75% of the drugin the body will be lost during a dosing interval. In this situation, equations

Page 296: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc14 JWBS062-Rosenbaum February 16, 2011 8:5 Printer Name: Yet to Come

276 MULTIPLE ORAL DOSES

for multiple discrete doses must be used. As discussed in 14.4.3 above, theamount of elimination that occurs during the absorption period determineswhether multiple bolus or multiple infusion equations would be most appro-priate. If a significant portion (>10%) of the dose is lost during absorption,the intermittent infusion equations should be used. Otherwise the equationsfor multiple intravenous doses may be used.

REFERENCES

1. Gibaldi, M., and Perrier, D. (1982) Pharmacokinetics, 2nd ed., Marcel Dekker, New York.

2. Jambhekar, S. S., and Breen, P. J. (2009) Basic Pharmacokinetics, Pharmaceutical Press, London.

3. Winter, M. E. (2010) Basic Clinical Pharmacokinetics, 5th ed., Lippincott Williams & Wilkins,Baltimore.

Page 297: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

15NONLINEAR PHARMACOKINETICS

15.1 Linear Pharmacokinetics

15.2 Nonlinear Processes in Absorption, Distribution, Metabolism, and Elimination

15.3 Pharmacokinetics of Capacity-Limited Metabolism15.3.1 Kinetics of Enzymatic Processes15.3.2 Plasma Concentration–Time Profile

15.4 Phenytoin15.4.1 Basic Equation for Steady State15.4.2 Estimation of Doses and Plasma Concentrations

15.4.2.1 Simulation Exercise15.4.3 Influence of Km and Vmax and Factors That Affect These Parameters

15.4.3.1 Simulation Exercise15.4.4 Time to Eliminate the Drug15.4.5 Time to Reach Steady State15.4.6 Individualization of Doses of Phenytoin

15.4.6.1 Modification of Dose Based on One Data Pair15.4.6.2 Modification of Dose Based on Two Data Pairs

Problems

Objectives

The material in this chapter will enable the reader to:

1. Differentiate the characteristics of linear and nonlinear pharmacokinetics

2. Understand how nonlinearity can arise in pharmacokinetics

3. Understand thoroughly the pharmacokinetics of single capacity-limited elimination

4. Understand how nonlinear pharmacokinetics is handled clinically

5. Gain experience individualizing doses based on estimates of patients’ Km and Vmax

values obtained from one and two plasma concentrations

15.1 LINEAR PHARMACOKINETICS

The vast majority of drugs used clinically follow linear pharmacokinetics. The pharmacoki-netics discussed up to this point in the book are all examples of linear pharmacokinetics.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

277

Page 298: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

278 NONLINEAR PHARMACOKINETICS

The term linear clearly cannot refer to the relationship between plasma concentration andtime, which is linear only when the plasma concentration is under the sole influence offirst-order elimination, and even then, only when the plasma concentration is converted tothe logarithm domain. The term linear refers to the relationship between the dose and theplasma concentration at any time after drug administration. In linear pharmacokinetics,the plasma concentration at any time after a dose is proportional to the dose. Linearity is aconsequence of the fact that all processes in drug disposition are first order and/or that thepharmacokinetic parameters are constant and do not change with dose.

Figure 15.1 shows the outcome of linear pharmacokinetics for single doses. Plasma con-centrations at any time after an extravascular or intravenous dose are directly proportionalto dose (Figure 15.1a). If plasma concentrations are normalized by dose, the plots fromdifferent doses are superimposed (Figure 15.1b). A distinctive characteristic that is oftenused as a check for linearity is that the area under the plasma concentration–time curvefrom time zero to infinity (AUC) is directly proportional to dose (Figure 15.1c).

Figure 15.2 shows some important consequences of linear pharmacokinetics with ex-tended drug administration. In Figure 15.2a it can be seen that plasma concentration doubleswith every doubling of the rate of drug administration. Figure 15.2b shows an extremelyimportant characteristic of linear pharmacokinetics: The average steady-state plasma con-centration is directly proportional to the rate of drug administration. This relationship is usedextensively when measured plasma concentrations are used to make dosage adjustments.For example, if a steady-state plasma concentration is half of its goal value, a doubling of therate of drug administration (double the dose or halve the dosing frequency) should achievethe goal. If a patient is found to have only one-third of the normal clearance, the usual rate ofdrug administration would produce three times the usual plasma concentration (S · F · Ra =Cl · Cpss). Thus, for this patient the normal rate of administration should be reduced byone-third (reduce the dose by one-third or increase the dosing interval threefold).

Linear pharmacokinetics provides the basis of bioavailability and bioequivalence studies.As seen in Chapter 9,

AUC = F · D

Cl(15.1)

In linear pharmacokinetics, clearance is a constant for a given drug:

F ∝ AUC

D(15.2)

Thus, the bioavailability (F) of a drug can be assessed from the dose-normalized AUC.Absolute bioavailability of an extravascular dose (FPO) is determined by comparing itsdose-normalized AUC to that after intravenous administration (FIV = 1).

FPO = AUCPO/DPO

AUCIV/DIV(15.3)

In bioequivalence studies, where the bioavailability of two different formulations of a drugare compared,

FT

FS= AUCT · DS

AUCS · DT(15.4)

where the subscripts T and S stand for test and standard, respectively.

Page 299: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

LINEAR PHARMACOKINETICS 279

Cp

Time

Cp

Time

D2D

3D

D

2D

3D

Time

IntravenousAdministration

ExtrvascularAdministration

Cp/

DC

p/D

Time

(a) (b)

AU

C

Dose(c)

FIGURE 15.1 Characteristics of the plasma concentration–time profile for drugs that display linearpharmacokinetics. Part (a) demonstrates that the plasma concentration is proportional to the dose atany time; (b) shows that dose-normalized plasma concentrations for a drug are superimposed overeach other; and (c) demonstrates that the area under the plasma concentration time curve (AUC) isproportional to the dose.

If DT = DS, relative bioavailability can be assessed simply by comparing the AUC valuesof the two products:

FT

FS= AUCT

AUCS(15.5)

Nonlinearity in the pharmacokinetics of a drug greatly complicates drug development,therapeutic drug use, and assessment of bioavailability. Nonlinearity can have a number of

Page 300: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

280 NONLINEAR PHARMACOKINETICS

0

10

20

30

40

0 1 2 3 4 5 6 7 8

0

10

20

30

40

0 1 2 3 4 5

Cp

Time

Cp S

S

Rate of Administration

(a)

(b)

Successive doubling of rate of administration

FIGURE 15.2 Relationship between plasma concentration and the rate of drug administration fora drug that displays linear pharmacokinetics. A successive doubling of the rate of administrationdoubles the plasma concentrations at any time (a). The steady-state plasma concentration (Cpss) isdirectly proportional to the rate of drug administration (b).

different origins but arises when a process in absorption, distribution, metabolism, or ex-cretion (ADME) deviates from a first-order process, and/or when a drug’s pharmacokineticparameters change with dose.

15.2 NONLINEAR PROCESSES IN ABSORPTION, DISTRIBUTION,METABOLISM, AND ELIMINATION

Most commonly, nonlinearity in pharmacokinetics arises when therapeutic drug concen-trations are high enough to saturate an enzyme or another protein involved in ADME.Consequently, nonlinear pharmacokinetics has the potential to arise whenever a protein isinvolved in ADME. Table 15.1 lists processes in ADME that involve proteins and which asa result have the potential to become saturated. The outcome expected for the saturation isalso shown.

Some clinical examples of the saturation of the processes listed in Table 15.1 includereduced absorption of riboflavin at higher doses as a result of a saturation of its uptaketransporter in the gastrointestinal membrane; reduced presystemic extraction of propranolol

Page 301: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHARMACOKINETICS OF CAPACITY-LIMITED METABOLISM 281

TABLE 15.1 Examples of Processes in ADME That Can Become Saturated

Process Typical Outcome of Saturation

AbsorptionUptake transporters Less absorption at higher dosesEfflux transporters More absorption at higher dosesEnzymes in enterocytes More absorption at higher dosesHepatic first-pass enzymes More absorption at higher

DistributionPlasma proteins Higher free fractions at higher dosesUptake transporters Lower tissue concentrations at higher dosesEfflux transporters Higher tissues concentrations at higher doses

MetabolismHepatic enzymes Lower clearance, slower elimination at higher doses

ExcretionUptake transporters Lower clearance, slower elimination at higher dosesEfflux transporters Lower clearance, slower elimination at higher doses

with higher doses, resulting in higher bioavailbility; dose-dependent protein binding ofvalproic acid; saturable tissue uptake of methotrexate; and saturation of the metabolism ofphenytoin and ethanol at higher doses.

Nonlinearity can also arise through some other mechanisms. For example, drugs suchas carbamazepine that induce their own metabolism will display nonlinear pharmacoki-netics until the induction process stabilizes, which generally takes about 10 to 14 days.The bioavailability of drugs that are poorly soluble in gastrointestinal fluid may decreasewith dose or changes in pH. For example, the dissolution and bioavailability of ketocona-zole change with changes in gastric pH. Nonlinear pharmacokinetics can also arise frompharmacological or toxicological actions of a drug. For example, theophylline inducesconcentration-dependent diuresis, which results in increased renal excretion with dose. In-terestingly, theophylline’s overall pharmacokinetics tend to be linear because it also displayssaturable metabolism, and the increase in renal clearance and decrease in hepatic clearancethat occur with higher plasma concentrations tend to offset each other. The clearance ofaminoglycosides can decrease with dose as a result of dose-dependent renal toxicity.

The outcome of nonlinearity will depend on the specific process involved. For example,saturable absorption resulting from saturable uptake transporters or poor dissolution willresult in larger doses, producing lower plasma concentrations than those predicted by linearpharmacokinetics. Conversely, with increasing doses, saturation of the other absorptionprocesses listed in Table 15.1 will result in plasma concentrations that are higher thanthose expected from linear processes. Nonlinear or capacity-limited metabolism is the mostcommon example of nonlinearity observed clinically, and the remainder of the chapter isdevoted to this topic.

15.3 PHARMACOKINETICS OF CAPACITY-LIMITED METABOLISM

The most clinically important type of nonlinear pharmacokinetics is saturable metabolism,which is also referred to as capacity-limited metabolism, biotransformation, or elimination.It arises when therapeutic concentrations of a drug partially or fully saturate an enzyme(s)that plays an important role in elimination of the drug. To understand the factors that

Page 302: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

282 NONLINEAR PHARMACOKINETICS

control nonlinearity and explain why only a fraction of the drugs that undergo metabolismare subject to nonlinearity, it is necessary to review the kinetics of enzymatic metabolism.This material, which was covered in Chapter 5, is reviewed here.

15.3.1 Kinetics of Enzymatic Processes

The kinetics of an enzymatic process such as metabolism often follow Michaelis–Mentonkinetics, which provides the following relationship between the rate of the process (V) andthe plasma concentration (Cp):

V = Vmax · Cp

Km + Cp(15.6)

where Vmax is the maximum rate of metabolism that is observed when all the enzyme issaturated with the substrate drug, and Km is the Michaelis–Menton constant, which is adissociation constant (as affinity for the enzyme increases, Km decreases). The units of Vare amount per unit time, and the units of Km are concentration.

Accordingly, the typical hyperbolic relationship of a capacity-limited system is observedbetween the rate of metabolism and the plasma concentration (Figure 15.3). Also, as wasshown in Chapter 5, the concentration that is associated with half the maximum rate isequal to the drug’s Km value. Two limiting situations exist for the rate of metabolism:

1. At low drug concentrations (Cp � Km), there is an excess amount of enzyme present.Under these circumstances, as the concentration of the drug increases, the rate canincrease proportionally: The rate is proportional to the plasma concentration, andmetabolism is an apparent first-order process (Figure 15.3). Mathematically, Cp �Km, Km + Cp ≈ Km, and

V = Vmax · Cp

Km + Cp= Vmax · Cp

Km= constant · Cp

Rat

e of

Met

abol

ism

(V

)

Cp

Vmax

Vmax/2

Km

Therapeutic concentrationsof most drugs are less than Km.Thus, metabolism is first order.

Some drugs have therapeuticconcentrations that are greater thantheir Km. As their concentrations increase,these drugs display first order, mixed,and zero-order metabolism

The enzymes are saturated;the rate of metabolism is itsmaximum value.

FIGURE 15.3 Michaelis–Menton kinetics. At low drug concentrations, excess enzyme is availableto metabolize the drug, and the rate can increase in direct proportion to increases in concentration (firstorder). As the drug concentration increases, some saturation is seen and the rate of metabolism canno longer keep up with increases in the drug concentration. At high drug concentrations, the enzymeis completely saturated and the maximum rate of metabolism is observed. At this point, increases indrug concentration are not associated with any further increases in the rate of metabolism.

Page 303: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHARMACOKINETICS OF CAPACITY-LIMITED METABOLISM 283

2. When the drug concentration becomes very high, the enzyme is fully saturated andthe rate is constant at its maximum value, Vmax. The rate of metabolism is a zero-orderprocess (Figure 15.3). Mathematically, Cp � Km, Km + Cp ≈ Cp, and

V = Vmax · Cp

Km + Cp= Vmax · Cp

Cp= Vmax

Between these two extremes, metabolism is mixed order and nonlinear.

It is very important to note that the therapeutic concentrations of most drugs are wellbelow their Km values. As a result, the enzymatic metabolism of the majority of drugsused in clinical practice follow apparent first-order kinetics, and their pharmacokineticsare linear. A small number of drugs, however, have therapeutic plasma concentrations thatapproach or exceed their Km value. For example, the average Km value for phenytoin is4 mg/L, which compares to a therapeutic range of 10 to 20 mg/L.

15.3.2 Plasma Concentration–Time Profile

Figure 15.4 shows the fall in the plasma concentration with time on a semilogarithmicscale after a series of intravenous doses of a drug that displays saturable metabolism(Figure 15.4a). The different plots represent a successive doubling of the dose. It can be seenthat the profile from the smallest dose resembles that observed with linear pharmacokinetics:a typical straight-line relationship between ln Cp and time. But notice that with the largerdoses, the initial fall in the plasma concentration is much less steep than that found withlinear pharmacokinetics. At higher concentrations the initial fall takes on a concave profile.This is the result of a saturation of the enzymes at higher concentrations and the inabilityof the rate of metabolism to increase in proportion to the increase in concentrations. Therate is less than it would be for a first-order process. As a result, the AUC increasesdisproportionately with dose (Figure 15.4b). The initial plasma concentration (Cp0) for

ln C

p

Time

AU

C

Dose

(a) (b)

Successive doubling of dose

FIGURE 15.4 Impact of nonlinear elimination on the plasma concentration–time profile after sin-gle doses. Graph (a) is a semilogarithmic plot of plasma concentration against time after a successivedoubling of an intravenous dose. After large doses, the initial fall in the plasma concentration withtime is less steep than with linear pharmacokinetics. Graph (b) shows that the area under the plasmaconcentration time curve (AUC) increases with dose for drugs that display saturable elimination.

Page 304: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

284 NONLINEAR PHARMACOKINETICS

0

10

20

30

40

50

0 10 20 30

Cp

Time

Cp s

s

Rate of Administration, Ra

Successive doublingof rate of administration

(a) (b)

Ra

2Ra

4Ra

FIGURE 15.5 Impact of nonlinear elimination on the pharmacokinetics of chromic drug admin-istration. Graph (a) shows the plasma concentration–time profile after a successive doubling of therate of administration from an infusion. After the first doubling, the plasma concentration at any timeincreases by slightly more than double. After the second doubling, the plasma concentration at anytime into therapy increases severalfold. Steady state has not been achieved over the observation periodbut the steady-state plasma concentration will be over eightfold greater than that from the previousdose. Graph (b) shows that the steady-state plasma concentration (Cpss) increases disproportionallywith the rate of drug administration. At a high rate of administration, the Cpss tends toward infinity.

drugs that display saturable metabolism is proportional to dose, as it is dependent on thedrug’s volume of distribution and is independent of its elimination characteristics.

The typical plasma concentration profile seen with the extended administration of drugsthat display nonlinear metabolism is shown in Figure 15.5. The profile shows the effect ofa successive doubling of the rate of drug administration. Notice that the first doubling pro-duces about a three- to fourfold increase in the steady-state plasma concentration. A furtherdoubling results in an over eightfold increase in the plasma concentration. Figure 15.5bshows the relationship between the steady-state plasma concentration and a wide range ofrates of drug administration. It can be seen that increases in the rate result in dispropor-tionate increases in the steady-state plasma concentrations. At high rates the concentrationtends toward infinity.

15.4 PHENYTOIN

Phenytoin is an important example of a drug that displays nonlinear pharmacokinetics.Although it has a number of different indications, it is used primarily as an anticonvulsantin the treatment and prophylaxis of seizure disorders. It is very commonly prescribed, andmany patients take oral phenytoin chronically, over periods of many years. Its widespreaduse has provided extensive experience with nonlinear pharmacokinetics and the effect ithas on therapeutic drug use. Also, since phenytoin has a narrow therapeutic range, it hasforced clinical pharmacists and pharmacologists to closely evaluate the dose–responserelationship and to develop procedures for the application of nonlinear pharmacokineticsto dosage individualization. As a result, phenytoin provides an interesting example to usein a discussion of the therapeutic implications of nonlinearity.

There are many interesting aspects to the pharmacokinetics and pharmacodynam-ics of phenytoin. It has a very narrow therapeutic range (10 to 20 mg/L), and plasma

Page 305: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHENYTOIN 285

concentrations outside these values are associated with very serious clinical consequences:Subtherapeutic concentrations place patients at risk for dangerous breakthrough seizures,and concentrations above the maximum tolerated concentration are associated with a num-ber of concentration-dependent conditions. With increasing concentrations of phenytoin,these include nystagmus, ataxia, slurred speech, confusion, and coma. Thus, it is importantto maintain plasma concentrations of phenytoin in the therapeutic range.

Phenytoin is eliminated primarily by saturable metabolism, which is described by theMichaelis–Menton equation and parameterized using Km and Vmax. The population averagevalue of phenytoin’s Km is around 4 mg/L, which is lower than the therapeutic range andillustrates why nonlinear pharmacokinetics are observed. The population average Vmax

value of phenytoin is about 7 mg/kg/day or about 500 mg/day for the standard 70-kg male.Phenytoin displays wide interindividual variability in the values of both its Km and Vmax.

As a result, a standard dose will be subtherpaeutic in some patients, therapeutic in others,and toxic in some. A study demonstrated that a standard dose of 300 mg/day of phenytoinachieved a therapeutic plasma concentration in only about 30% of patients (1). The remain-der had either subtherapeutic or toxic concentrations. The study illustrated the importanceof individualizing the dose of phenytoin for each patient. Typically, this is accomplished bymeasuring a steady-state plasma concentration about two weeks after the start of therapy.If the plasma concentration is too low, the dose is increased. Conversely, if the plasmaconcentration is too high, the dose is decreased. However, nonlinear pharmacokineticscomplicate the adjustment process, and proportional adjustments in the dose must not bemade. A doubling of the dose in response to a plasma concentration of 6 mg/L could quitepossibly produce toxic concentrations and place the patient at risk for the dangerous sideeffects of this drug.

The extensive and variable plasma protein binding of phenytoin adds another interestingdimension to the evaluation of phenytoin’s pharmacokinetics and pharmacodynamics, butthis topic is not addressed in this chapter.

15.4.1 Basic Equation for Steady State

The pharmacokinetic model developed for phenytoin must reflect its primary clinical useas an oral preparation that is used over an extended period of time. The key features of themodel are as follows:

� A one-compartmental model can be used to describe the pharmacokinetics of pheny-toin after oral doses (Figure 15.6).

E

Initial Conditions: t = 0, Ab = Ab0 = 0

VdCpAb

S•F•D/τ

Ra

Vmax•Cp

Km+Cp

FIGURE 15.6 Pharmacokinetic model for phenytoin consistent with a one-compartment model.The input is the rate of drug administration (Ra) is S·F·D/� . Elimination (E) is given by theMichaelis–Menton equation.

Page 306: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

286 NONLINEAR PHARMACOKINETICS

� Chronic drug administration is incorporated as the input into the compartment usingthe rate of drug administration, or dose/dosing interval.

� Phenytoin is eliminated primarily by saturable metabolism (fe = 0.01 to 0.05), mainlyby CYP2C9, but CYP2C19 is also involved. Since renal excretion is minor, it isignored, and the various metabolic pathways are merged into a single capacity-limitedprocess. Thus, elimination is modeled using the Michaelis–Menton equation:

rate of elimination = Vmax · Cp

Km + Cp

−dAb

dt= Vmax · Cp

Km + Cp

(15.7)

� Drug absorption from phenytoin, particularly from the innovator product Dilantin isvery slow, and it can be assumed to occur at a constant rate throughout the entire dosinginterval. This enables the infusion model (constant, zero-order drug administration)to be used to derive equations for phenytoin (see Chapter 14). The constant rate ofadministration of phenytoin is the effective dose divided by the dosing interval, whichis typically 0.5 or 1 day. Overall, the rate of change of the amount of drug in the bodywith time is

dAb

dt= rate of inputs − rate of outputs

= S · F · D

�− Vmax · Cp

Km + Cp

(15.8)

where S is the salt factor, F the bioavailability, D the administed dose, and � the dosinginterval.

� Phenytoin’s plasma concentrations are monitored primarily at steady state when theequations can be simplified quite considerably. At steady state, the plasma concentra-tion (Cpss) is constant and the rate of input is equal to the rate of output (Figure 15.7):

rate of administration = rate of elimination

S · F · D

�= Vmax · Cpss

Km + Cpss

(15.9)

Cp

Time into Therapy

Cpss:Rate of Administration= Rate of Elimination

FIGURE 15.7 Predicted plasma concentrations of phenytoin that result from its constant, contin-uous administration. At steady state, the plasma concentration (Cpss) remains constant. Thus, the rateof elimination must be equal to the rate of drug administration.

Page 307: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHENYTOIN 287

This single equation is used extensively for phenytoin and other drugs that displaysaturable elimination. Population average values or literature values of the pharma-cokinetic parameters can be used in equation (15.9) to estimate plasma concentrationsof phenytoin from different doses. It can also be used to estimate doses to achieve de-sired plasma concentrations. If an individual patient’s Vmax and Km values are known,patient-specific doses can be determined. Also, as demonstrated in a later section,this equation is used clinically to estimate a patient’s Km and/or Vmax values frommeasured plasma concentrations of phenytoin.

In addition to the arrangement shown above, equation (15.9) has two other usefularrangements. These are provided below.

Cpss = Km · S · F · Ra

Vmax − S · F · Ra(15.10)

where Ra is the rate of drug administration or D/� . The denominator in equation (15.10)demonstrates the importance of ensuring that the rate of drug administration does notexceed the drug’s Vmax value. If this happens, the steady-state plasma concentration willtend toward infinity. For example, suppose that a patient who has a Vmax of phenytoin of350 mg/day received a dose of 400 mg of phenytoin per day. For every day that the patienttook this dose, he or she would accumulate 50 mg phenytoin. Unlike linear pharmacoki-netics, the accumulation would not abate but would continue for as long as the patient took400 mg of phenytoin daily.

Vmax = S · F · Ra · (Km + Cpss)

Cpss(15.11)

Some example applications of these formulas are presented below.

15.4.2 Estimation of Doses and Plasma Concentrations

Next we look at the use of the equations to estimate doses and plasma concentrations.

Example 15.1 A patient is to begin taking phenytoin. A steady plasma concentration of15 mg/L is desired. Assuming a Vmax of 400 mg/day and a Km value of 4 mg/L, what rateof administration of phenytoin sodium (S = 0.92, F = 1) should be used?

Solution Using equation (15.9)

0.92 × 1 × D

�= 400 × 15

4 + 15D

�= 343 mg per day

Thus, a dose of 350 mg of phenytoin sodium per day would be administered.

Page 308: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

288 NONLINEAR PHARMACOKINETICS

Example 15.2 A patient’s individual Vmax and Km values are found to be 450 mg/dayand 5 mg/L, respectively. What are her estimated steady-state plasma concentrations fromdoses of (a) 350 mg/day, (b) 400 mg/day, and (c) 450 mg/day?

Solution(a) For a rate of administration of 350 mg/day, using equation (15.10), we obtain

Cpss =5 × 0.92 × 350

450 − 0.92 × 350= 12.6 mg/L

(b) For a rate of administration of 400 mg/day,

Cpss =5 × 0.92 × 400

450 − 0.92 × 400= 22.4 mg/L

(c) For a rate of administration of 450 mg/day,

Cpss =5 × 0.92 × 450

450 − 0.92 × 450= 57.5 mg/L

Example 15.2 demonstrates how the steady-state plasma concentration increases dispro-portionately to dose. A 15% increase in the rate of administration from 350 mg/day to400 mg/day increased the steady-state plasma concentration by about 80%. A similar in-crease from 400 to 450 mg per day increased the steady-state plasma concentration about150%.

15.4.2.1 Simulation ExerciseOpen the model “Nonlinear Pharmacokinetics” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter15

Default settings for the model are dose = 350 mg, S = 0.92, F = 1; � = 1 day; V max =450 mg/day; Km = 6 mg/L; and Vd = 45.5 L.

1. Review the objectives, explore the model, and review the model summary.

2. Go to the “Cp–Time Profile” page. Observe the effects of increasing the dose from350, to 400, then to 450 mg. Note that, in contrast to linear pharmacokinetics:� Increases in dose produce disproportionate increases in the steady-state

plasma concentration.� As the dose increases, it takes longer to achieve steady state.

3. Observe the effect of doubling the original dose of 350 mg, which achieved asteady-state plasma concentration of about 15 mg/L in about two to three weeks.Doubling the dose produces plasma concentrations that exceed the therapeu-tic range (>20 mg/L) during the third day of treatment and which appear to beheading toward infinity. Note that this rate of administration (700 mg of pheny-toin sodium per day or about 650 mg of pure phenytoin per day) exceeds V max

(450 mg/day). As therapy continues, the patient will accumulate 200 mg of pheny-toin daily.

Page 309: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHENYTOIN 289

TABLE 15.2 Factors That Can Alter the Value of Vmax and Km

Parameter Factors Affecting Parameter

Direction ofChange inParameter Outcome

Vmax Enzyme induction (e.g., carbamazepine) ↑ ↓CpLiver disease (e.g., cirrhosis) ↓ ↑Cp

Km Competitive inhibitors (e.g., cimetidine, valproicacid, fluoxetine)

↑ ↑Cp

Displacement from plasma proteins, valproic acid,hypoalbinemia

↓ ↓Cp

15.4.3 Influence of Km and Vmax and Factors That Affect These Parameters

For drugs that display nonlinear pharmacokinetics, the relationship between dose and thesteady state plasma concentration is controlled by Vmax and Km. Example 15.2 demonstratedthe very sensitive nature of this relationship. It is important to understand the role that eachparameter plays in the dose–plasma concentration relationship. Additionally, it is importantto be aware of any situations where either of the parameters may differ from normal and toknow how to plan therapy in the light of any known or suspected changes.

Vmax, the maximum rate of metabolism, is a function of the amount of enzyme present.Enzyme induction will increase Vmax, and hepatic diseases such as cirrhosis, which reducethe number of functioning hepatocytes, will decrease Vmax. Km is a dissociation constantand is a reciprocal expression of the affinity of the drug for the enzyme. As Km increases,affinity decreases. Concomitant medications that competitively inhibit enzymes responsiblefor the elimination of the subject drug will increase Km. The displacement of the subjectdrug from its plasma protein binding sites will produce an apparent decrease in Km. This isbecause displacement increases the unbound fraction, which enables more drug to interactwith the enzyme at a given concentration. Thus, based on total plasma concentration, it willappear that affinity has increased (that Km has decreased).

Table 15.2 shows some factors that can affect phenytoin’s Vmax and Km. It also showshow the changes affect the plasma concentration. However, the clearest way to appreciatethese effects is through simulation.

15.4.3.1 Simulation ExerciseOpen the model “Nonlinear Pharmacokinetics” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter15

The default settings for the model are dose = 350 mg, � = 1 day, V max = 450 mg/day,Km = 6 mg/L, and Vd = 45.5 L.

Go to the “V max and Km” page.

1. While maintaining the default values of Km (6 mg/L), increase V max from 300 to400 to 600 mg/day. V max represents the maximum rate of metabolism and isa function of the amount of enzyme present in a system. Note that as V max

increases, the plasma concentration at any time decreases. Note also that asV max increases, it takes less time to reach steady state.

Page 310: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

290 NONLINEAR PHARMACOKINETICS

2. Now maintain the default value of V max (450 mg/day) and increase Km from 2 to4 to 8 mg/L. Km is a dissociation constant, so it is a reciprocal measure of affinity.As Km increases, the affinity of the drug for the enzyme decreases. Note that asKm increases (affinity decreases), the Cp at any time increases and it takes moretime to get to steady state.

The simulations have demonstrated that the dose of phenytoin may have to be re-duced (increased) in patients with decreased (increased) V max values and/or increased(decreased) Km values.

15.4.4 Time to Eliminate the Drug

When elimination is not a first-order process, the concept of a half-life does not hold andthere is no simple way to estimate the time it takes the plasma concentration to fall by50% or any other amount. The rate of elimination of phenytoin was presented previously[equation (15.7)]. If there is no ongoing drug input, the drug in the body is only under theinfluence of elimination:

−dAb

dt= Vmax · Cp

Km + Cp

Substituting for Ab where Ab = Cp · Vd, we obtain

−dCp

dt= Vmax · Cp

Vd · (Km + Cp)(15.12)

This equation can be integrated (2) to yield

t = Vd

Vmax

(Cp0 − Cpt + Km ln

Cp0

Cpt

)(15.13)

where Cp0 is the plasma concentration at t = 0 and Cpt is the plasma concentration at time t.Equation (15.13) cannot be solved explicitly and used to calculate the plasma concentration,but it can be used to calculate the time for the plasma concentration to fall from one valueto another.

Example 15.3 A patient (Vd = 45 L, Vmax = 400 mg/day, and Km = 4 mg/L) presentswith a phenytoin concentration of 40 mg/L.

(a) How long will it take for the plasma concentration to reach the therapeutic range(20 mg/L)?

(b) How long will it take for plasma concentrations to fall another 50%, to 10 mg/L?

Solution(a) Substituting the values into equation (15.13), we have

t = 45

400

(40 − 20 + 4 · ln

40

20

)= 2.56 days

Page 311: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHENYTOIN 291

(b) Substituting into equation (15.13) gives us

t = 45

400

(20 − 10 + 4 × ln

20

10

)= 1.43 days

Note that the time for the plasma concentration to fall by 50% is not constantbut decreases as the plasma concentration decreases. This is because at higherconcentrations there is a greater degree of saturation of the enzymes and a greaterfraction of the drug present cannot be metabolized.

In certain circumstances it may be necessary to estimate how long it takes a certainamount of drug in the body to fall to a specific amount. Equation (15.13) can be adaptedfor this:

t = 1

Vmax

(Ab0 − Abt + Km · ln

Ab0

Abt

)(15.14)

where Ab0 is the initial amount in the body at time t = 0, Abt is the amount at time = t, andKm is in units of amount [i.e., Km (mg/L) · Vd (L)].

15.4.5 Time to Reach Steady State

Once again, because elimination is not a first-order process, there is no simple way toestimate the time it takes to reach steady state. Information on the time to takes to get tosteady state can be obtained by deriving an equation for the plasma concentration at anytime. This is accomplished using the fundamental equation for the model (15.8), which isreproduced here:

dAb

dt= S · F · D

�− Vmax · Cp

Km + Cp

This equation is integrated, solved for plasma concentration, and then rearranged to providean expression for the time it takes to get to 90% steady state (t90%) (2,3):

t90% = Km · Vd

(Vmax − S · F · Ra)2(2.3Vmax − 0.9 · S · F · Ra) (15.15)

Equation (15.15) demonstrates that the time to steady state, or 90% steady state, isdependent on the pharmacokinetic parameters Km and Vmax, and in contrast to linearpharmacokinetics, it is also dependent on the rate of administration. As demonstrated inthe simulations, it takes longer to get to steady state with higher rates of administration.More specifically, it is the difference between Vmax and the rate of drug administration thatdetermines the time to reach steady state. As the rate of administration approaches Vmax

(as complete saturation of the enzyme is approached), the denominator in equation (15.15)gets smaller and it takes longer to get to steady state. As S · F · Ra approaches Vmax, t90%

tends toward infinity.

Page 312: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

292 NONLINEAR PHARMACOKINETICS

Example 15.4 The population average parameters for phenytoin in a standard 70-kg maleare: Vd = 45.5 L, Vmax = 400 mg/day, and Km = 6 mg/L. How long will it take to get to90% steady state when the dose is administered at a rate of 300 mg of phenytoin sodiumper day?

Solution Substituting the parameter values into equation (15.15) gives

t90% = 6 × 45.5

(400 − 0.92 × 300)2(2.3 × 400 − 0.9 × 0.92 × 300) = 11.9 days

Note that if the dose is only 250 mg, 90% steady state is reached in about 7 days, butwhen the dose is increased to 350 mg and approaches Vmax, the time to reach steady stateincreases to around 28 days.

15.4.6 Individualization of Doses of Phenytoin

When patients are started on phenytoin therapy, the wide interindividual variability inVmax and Km, combined with phenytoin’s narrow therapeutic range, makes it essential tomonitor the plasma concentrations and individualize the dose if necessary. When patientsstart therapy, they are evaluated to determine if any obvious factors are present that wouldwarrant a dose that is different from the standard. Factors to be considered include:

� Concomitant medications that could alter intrinsic clearance or alter plasma proteinbinding

� The presence of liver disease, which could reduce intrinsic clearance and/or alterprotein binding

� The presence of renal disease, which could alter protein binding

Assuming that none of the foregoing factors are present, the patient may be startedon a standard dose of about 7 mg/kg/day of phenytoin sodium. Once steady state hasbeen achieved, after about two weeks, the phenytoin concentration can be evaluated. If theplasma concentration is in the therapeutic range, the dose will be maintained. If not,the dose has to be modified, and the process should be based on the patient’s history: that is,the steady-state plasma concentration achieved by the initial rate of drug administration (oneS · F · Ra–Cpss data pair).

15.4.6.1 Modification of Dose Based on One Data PairThe process of individualization involves estimating the patient’s individual pharmacoki-netic parameters for phenytoin. Recall the basic formula that relates the rate of drugadministration to the steady-state plasma concentration:

Cpss = Km · S · F · R

Vmax − S · F · R

In contrast to linear pharmacokinetics (Cpss = S · F · Ra/Cl), the relationship between Cpss

and S · F · Ra is controlled by two parameters (Vmax, Km) rather than one (Cl). With only oneCpss–S · F · R data pair, it is possible to estimate only one of phenytoin’s pharmacokineticparameters. Since it is critical that the drug not be administered at a rate that exceeds Vmax,

Page 313: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PHENYTOIN 293

this parameter is usually estimated first. The population average value of Km (4 mg/L) isassumed for the patient.

Recall the arrangement of the basic equation that solves for Vmax:

Vmax = S · F · Ra · (Km + Cpss)

Cpss(15.16)

The initial rate of drug administration (S · F · Ra) and the steady-state concentration itachieved are substituted in the equation along with the population average Km of 4 mg/L,and Vmax is estimated. The Vmax calculated, the population average Km (4 mg/L), and thesteady-state plasma concentration desired (usually 15 mg/L) are then substituted into thearrangement of the basic equation that solves for S · F · Ra:

S · F · Ra = Vmax · Cpss

Km + Cpss(15.17)

A new rate of drug administration is then determined based on the patient’s previous historywith the drug.

Example 15.5 A patient has been taking dilantin 300 mg daily for two weeks. At thistime a plasma sample reveals a phenytoin concentration of 8 mg/L. Recommend a newdose to acheive a steady-state concentration of 15 mg/L.

Solution The population average Km (4 mg/L) will be assumed. Substituting in equation(15.16) to estimate the patient’s Vmax leads to

Vmax = 0.92 × 300 × (4 + 8)

8= 414 mg/day

This estimated Vmax value and the population average Km are now used in equation (15.17)to estimate a rate of administration to provide the desired Cpss of 15 mg/L:

Ra = 414 × 15

(4 + 15) × 0.92= 355 mg/day

The new rate of drug administration may also be assessed about two weeks later whenthe new steady state would be expected. If this steady-state plasma concentration is notsatisfactory, a new rate of drug administration must be determined. If this is the case, twoS · F · Ra–Cpss data pairs are now available. These can be used to estimate both the Vmax

and Km values of the patient.

15.4.6.2 Modification of Dose Based on Two Data PairsThe method of Ludden is a common mathematical approach used to determine Km and Vmax

from two or more data pairs. Taking the basic equation that is expressed in terms of Cpss

Cpss = Km · S · F · Ra

Vmax − S · F · Ra(15.18)

Page 314: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

294 NONLINEAR PHARMACOKINETICS

S•F

•Ra

S•F•Ra/Cpss

Vmax

slope = –Km

FIGURE 15.8 Method of Ludden to determine Km and Vmax. Plot of the effective rate of adminis-tration (S · F · Ra) against S · F · Ra divided by the steady-state plasma concentration (Cpss).

Rearranging gives us

Vmax − S · F · Ra = Km · S · F · Ra

Cpss(15.19)

Rearranging again, we obtain

S · F · Ra = Vmax − Km · S · F · Ra

Cpss(15.20)

A plot of S · F · Ra against S · F · Ra/Cpss yields a straight line of slope −Km and interceptVmax (Figure 15.8). If more than two data pairs are available, a full plot can be constructed.If only two data pairs are available, they are simply used to calculate the slope of the lineS · F · Ra against S · F · Ra/Cpss. This provides a value for Km. The estimate of Vmax canbe obtained using equation (15.16).

Assuming the same F and S values the slope may be calculated as follows

slope = Ra1 − Ra2

Ra1/Cpss1− Ra2/Cpss2

Km = −slope

= Ra2 − Ra1

Ra1/Cpss1− Ra2/Cpss2

(15.21)

Example 15.6 Continuing with Example 15.5, recall that an initial dose of 300 mg/dayphenytoin sodium provided a steady-state plasma concentration of 8 mg/L. The dose wasincreased to 350 mg/day. Two weeks later the second dose was found to provide a steady-state plasma concentration of 11 mg/L. Recommend a dose to provide a steady-state plasmaconcentration of 15 mg/L.

Page 315: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

PROBLEMS 295

Solution The existing history on this patient can be summarized as follows: The first rateof drug administration is

Ra1 (300 mg/day) gave a Cpss1value of 8 mg/L

and the second rate of drug administration is

Ra2 (350 mg/day) gave a Cpss2value of 11 mg/L

The patient’s Km value is estimated first using equation (15.21):

Km = 350 − 300

300/8 − 350/11= 8.8 mg/L (15.22)

Vmax is calculated, using equation (15.16) and either data pair:

Vmax = 0.92 × 300 × (8.8 + 8)

8= 579.6 mg/day

The new dose is calculated using equation (15.17):

S · F · Ra = 579.6 × 15

8.8 × 15= 365

Ra = 397 mg

A new dose of 400 mg of phenytoin sodium daily is recommended.

Several other methods are available for estimating phenytoin’s Vmax and Km values andcan be found in clinical pharmacokinetics textbooks (3–5).

PROBLEMS

15.1 C.R. is a 45-year-old 85-kg male (height 180 cm), who experiences simple partialseizures. He has normal liver and renal function and is not taking any other medica-tions. He is started taking 400 mg of extended phenytoin sodium daily. Two weekslater a plasma concentration, which is assumed to be steady state, is found to be6.6 mg/L. The patient says that he has taken all the doses as directed. Suggest adosage regimen to achieve a steady-state phenytoin concentration of 15 mg/L.

15.2 L.M. is a 29-year-old female who has been taking carbamazepine for seven monthsto control her epilepsy. She still experiences several seizures each month. It isdecided to discontinue carbamazepine and try phenytoin. She is prescribed dilantin(300 mg b.i.d.). Three weeks later she says that she has not experienced a seizureand feels great. However, about a month later she returns to the physician andcomplains of feeling unsteady, “out of it,” and having difficulty keeping focused onactivities. A blood sample is taken and the phenytoin plasma concentration is found

Page 316: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc15 JWBS062-Rosenbaum February 16, 2011 8:7 Printer Name: Yet to Come

296 NONLINEAR PHARMACOKINETICS

to be 28 mg/L. Suggest possible explanations for these observations and recommenda more appropriate dose of phenytoin.

15.3 A patient who is suspected to have reduced intrinsic clearance of phenytoin is startedon a dose of 250 mg of phenytoin sodium daily.

(a) Two weeks after the start of therapy a steady-state plasma concentration is foundto be 4 mg/L. Estimate her Vmax and recommend a new dose.

(b) Her dose is increased to 400 mg of phenytoin sodium daily. Two weeks latera steady-state plasma concentration is found to be 10 mg/L. Determine her Km

and Vmax values and recommend a new dose.

15.4 A 65-kg 19-year-old male took an overdose of phenytoin. He is admitted to a hospital24 h later. A blood sample reveals a phenytoin concentration of 55 mg/L. How longwill it take for the plasma concentration to reach the upper limit of the therapeuticrange (20 mg/L)? Assume a population average Vmax of 7 mg/kg/day, a Km of 4mg/L, and a Vd of 0.65 L/kg.

15.5 A drug is eliminated by a single metabolic pathway which has a Km value of 10 �g/mL and a Vmax value of 500 mg/day (range 250 to 1500 mg/day). The therapeuticrange of this drug is 10 to 25 �g/mL.

(a) Based on the information provided, comment on any special considerationsneeded when selecting a dosage regimen for this drug.

(b) Calculate the steady-state plasma concentration achieved when 300 mg of thefree drug is administered daily to a patient who has the population averageparameter values.

(c) Calculate the steady-state plasma concentration if the dose is increased slightlyto 350 mg.

(d) Calculate the steady-state plasma concentration if while on the latter dosage thepatient develops hepatotoxicity, which causes a decrease in Vmax to 375 mg.

REFERENCES

1. Koch-Weser, J. (1975) The serum level approach to individualization of drug dosage, Eur J ClinPharmacol 9, 1–8.

2. Gibaldi, M., and Perrier, D. (1982) Pharmacokinetics, 2nd ed., Marcel Dekker, New York.

3. Winter, M. E., and Tozer, T. N. (2006) Phenytoin, In Applied Pharmacokinetics and Pharma-codyamics (Burton, M. E., Shaw, L. M., Schentag, J. J., and Evans, W. E., Eds.) 4th ed., LippincottWilliams & Wilkins, Baltimore.

4. Bauer, L. A. (2008) Applied Clinical Pharmacokinetics, 2nd ed., McGraw-Hill, New York.

5. Winter, M. E. (2010) Basic Clinical Pharmacokinetics, 5th ed., Lippincott Williams & Wilkins,Baltimore.

Page 317: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

16INTRODUCTION TOPHARMACODYNAMIC MODELS ANDINTEGRATED PHARMACOKINETIC–PHARMACODYNAMIC MODELS

16.1 Introduction

16.2 Classic Pharmacodynamic Models Based on Traditional Receptor Theory16.2.1 Receptor Binding16.2.2 Response–Concentration Models

16.2.2.1 Intrinsic Activity Model16.2.2.2 Efficacy Model

16.3 Empirical Pharmacodynamic Models Used Clinically16.3.1 Sigmoidal Emax and Emax Models16.3.2 Linear Adaptations of the Emax Model

16.4 Integrated PK–PD Models: Emax Model Combined with a PK Model for Intravenous BolusInjection in a One-Compartment Model16.4.1 Simulation Exercise

16.5 Hysteresis and the Effect Compartment16.5.1 Simulation Exercise

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand classic receptor theory of drug action

2. Understand the basis and application of the sigmoidal Emax and related models

3. Understand the time course of drug response based on the sigmoidal Emax model

4. Understand how distributional delays cause counterclockwise hysteresis

5. Understand the use of an effect compartment to model distributional delays in drugresponse

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

297

Page 318: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

298 INTRODUCTION TO PHARMACODYNAMIC MODELS

16.1 INTRODUCTION

Mathematical models are important tools for the study of drug actions in vivo. Tobe complete, such models should include a pharmacodynamic component for theconcentration–response relationship and a pharmacokinetic component for the dose–plasmaconcentration relationship. In theory, such integrated pharmacokinetic–pharmacodynamic(PK–PD) models could predict responses at any time, after any dose, administered by anyroute. These models would be of enormous value for estimating doses and dosing intervalsto achieve specific desired responses. They could also be used to gain insight into thebehavior of a drug in situations not yet studied.

For example, if an appropriate PK–PD model is identified and the model parametersdetermined after a single dose of a drug, simulations can be performed to predict steady-state responses and optimum dosing regimens to achieve desired responses. IntegratedPK–PD models are being applied increasingly to optimize the design of clinical trials,increase the efficiency of these studies, and decrease the cost of drug development (1,2).Integrated PK–PD models are also being used increasingly in translational research inwhich parameters identified in vitro and small animals are used to predict drug response inhumans (2,3). These studies are proving to be valuable during drug discovery to help identifythe specific molecules most likely to progress successfully throughout the developmentprocess and succeed in humans. Recently, an integrated PK–PD model has been developedspecifically for clinical practice to estimate optimum doses of several anticancer drugs inindividual patients (4).

As discussed in Chapter 1, the development of mathematical models forconcentration–response relationships in humans has been hampered by several factors.Among these are the inability to measure the drug concentration at the site of action, thedifficulty in measuring response, and the complex and varied nature of the chain of eventsbetween receptor activation and the emergence of response. The processes associated withthe pharmacodynamic phase of drug response are much more varied and complex in naturethan those involved in the pharmacokinetic phase. A drug’s pharmacokinetics are drivenprimarily by passive diffusion and the common pathways of renal excretion and hepaticmetabolism, which are usually simple first-order processes. By contrast, the interaction ofa drug with its receptors can lead to a vast array of disparate effects. The ultimate responsemay occur almost immediately or may take several days or even weeks to become apparent.The response may initiate homeostatic processes that can further complicate its evaluation.Thus, in comparison to pharmacokinetics, the design of models of drug response is muchmore challenging, and a greater diversity of models is required to accommodate differenttypes of processes. Over the last 10 to 15 years a diverse group of pharmacodynamic modelshave been developed and applied successfully to the modeling of response data in vivo.These models require accurate objective data, ideally measured on a continuous scale. Thisrequirement has been another hurdle for pharmacodynamic studies since responses to mostdrugs in human beings cannot be measured in this way. This problem has been overcometo a large extent by the identification of biomarkers of drug response. A biomarker may bedefined as a concrete biological characteristic that can be measured objectively as a parallelindicator of a drug response (5). Biomarkers used for the development of pharmacodynamicmodels should possess the following qualities:

� Valid for the response being assessed.� Measured objectively rather than relying on subjective evaluations of a patient or

health care provider.

Page 319: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

CLASSIC PHARMACODYNAMIC MODELS BASED ON TRADITIONAL RECEPTOR THEORY 299

� Continuous rather than an all-or-none response.� Sensitive for the response; that is, changes in the response should lead consistently to

changes in the biomarkers.� Specific for the response. Ideally, changes in the biomarker should be associated with

the drug response, not with other events.� Reproducible from one occasion to another and from one clinical center to another.� Occurs in the same concentration range as the therapeutic response.� Repeatable in the same patient at later times to enable the response to be evaluated over

an extended period. Evaluations that are invasive or those that may involve learningwould not meet this criterion.

Examples of biomarkers include measurement of the amplitude of certain frequencybands on an electroencephalograph to evaluate the effects of centrally acting drugs, and themeasurement of relevant endogenous compounds such as proteins in biological fluids. Forexample, reductions in amyloid beta peptide levels in the cerebrospinal fluid and increasedconcentrations of total � protein have been used as biomarkers in Alzheimer’s disease.Advances in analytical technology have enabled a wide variety of endogenous compoundsto be used as biomarkers.

In this chapter we discuss the receptor theory of drug action, the classic pharmacody-namic models, and the parameters that are used to evaluate a drug’s pharmacodynamiccharacteristics. We also discuss the development and application of the pharmacodynamicmodels most commonly used to model drug response in vivo.

16.2 CLASSIC PHARMACODYNAMIC MODELS BASED ON TRADITIONALRECEPTOR THEORY

The classic receptor theory of drug action is an important starting point for understandingdrug action in vivo and the development of pharmacodynamic models in humans. Theclassic receptor-based models have been developed primarily through a study of drugaction in the more controlled and isolated in vitro environment, where response and thedrug concentration at the site of action can easily be measured. Additionally, the action ofa drug in vitro can be studied in isolation without the interference of homeostatic and othercompeting processes.

As discussed in Chapter 1, most drugs exert their effects by interacting with theirreceptors in a reversible manner. The interaction of an agonist with its receptor leads toa conformational change in the receptor that results in a signal or stimulus. The stimulusthen initiates other actions that ultimately result in a biological response (Figure 16.1).The events between generation of the stimulus and the final response constitute what iscalled the response chain. This can be a simple direct process such as the opening orclosing of an ion channel, or it may involve a long transduction process that includes acascade of several events and the action of second messengers. The overall intensity ofdrug response is a function of two types of properties: drug-specific and tissue- or system-specific properties. The drug-specific properties are the ability of the drug to interact withthe receptors (affinity) and the ability of the drug to produce a stimulus per unit receptor(intrinsic efficacy). The tissue-specific properties are the number or density of the receptorsand the process that converts the initial signal into a response. Ideally, a pharmacodynamicmodel should separate these two types of properties.

Page 320: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

300 INTRODUCTION TO PHARMACODYNAMIC MODELS

STIMULUS

Cell Membrane

Receptor

RESPONSE

Drug

Transduction

FIGURE 16.1 Diagramatic representation of the drug–receptor interaction. The drug interactswith the receptor to produce a conformational change in the receptor. This results in a stimulus. Thestimulus initiates the events that culminate in the biological response.

The first mathematical model for the relationship between agonist drug response andconcentration was developed in 1937 by Clark. Over the years this model was modified—byAriens (in 1954), Stephenson (in 1956), and Furchgott (in 1966) [see (6)]—but key elementsof the original model remain. Based on their work, two models that have relevance to drugevaluation in humans are presented. The first is the simpler model of Ariens, the intrinsicactivity model, which incorporates the term intrinsic activity. The second model, theefficacy model, a more sophisticated extension of the first model, incorporates a parameterfor a drug’s efficacy to accommodate the concept of spare receptors. Both models arebased on the assumption that drug action is a function of a drug’s quality of binding to itsreceptors. Thus, the characteristics of receptor binding are fundamental to both models.

16.2.1 Receptor Binding

The response to a drug is assumed to be a function of the number of receptors occupied.Receptor occupancy is another example of a capacity-limited process that is described bythe law of mass action:

C + Rkon−→←−koff

RC (16.1)

where C is the molar concentration of drug, R the molar concentration of unoccupiedreceptors, RC the molar concentration of the drug’s receptor complex, kon the rate constantfor the forward process, and koff the rate constant for the backward process.

Once equilibrium is established, the rates of the forward and backward processes becomeequal:

(RT − RC) · C · kon = RC · koff (16.2)

Page 321: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

CLASSIC PHARMACODYNAMIC MODELS BASED ON TRADITIONAL RECEPTOR THEORY 301

where RT is the total molar concentration of receptors. After rearrangement, we have

RC = RT · C

Kd + C(16.3)

or

RC

RT= C

Kd + C(16.4)

where Kd is the drug’s dissociation constant (koff/kon) and is a reciprocal measure of thedrug’s affinity for the receptors. As Kd decreases, affinity increases and there is greaterbinding at a given drug concentration. Kd is equal to the drug concentration at 50% receptoroccupancy.

Because the system has only a finite number of receptors, the concentration of occu-pied receptors (RC) in equation (16.3) is limited by the total concentration (or capacity)of receptors in a system. As a result, the relationship between occupancy and drug con-centration has the hyperbolic shape typical of capacity-limited processes (Figure 16.2).At low concentrations there are plenty of free receptors, and the concentration of thecomplex can increase in direct proportion to increases in concentration. But as the con-centration increases further, some saturation begins to occur and the concentration of thecomplex cannot increase in proportion to the drug concentration. Eventually, all the recep-tors are occupied, the concentration of the drug–receptor complex achieves its maximumvalue, and increases in drug concentration have no further effect on the concentration ofthe complex.

1

0

RC

/RT

Drug Concentration

0.5

Kd

FIGURE 16.2 Relationship between fraction of receptors occupied and drug concentration. Thefraction of the receptors occupied (RC/RT) is shown as a function of the drug concentration (C).Binding of a drug to its receptors is an example of a capacity-limited process. At high drug concen-trations, binding is limited by the number or capacity of the receptors. RC is the concentration of thedrug–receptor complex and RT is the total concentration of receptors. When RC

RT= 0.5, C = Kd, the

dissociation constant.

Page 322: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

302 INTRODUCTION TO PHARMACODYNAMIC MODELS

Res

pons

e (%

of

Sys

tem

’s M

axim

um)

Drug Concentration

100

0

Em

FIGURE 16.3 Relationship between the response and drug concentration. The response increaseswith drug concentration, but the increase gets proportionally less as the drug concentration increases(law of diminishing returns). Eventually, at higher drug concentrations, a maximum response isachieved. In this figure the drug is able to achieve the system’s maximum response (Em).

16.2.2 Response–Concentration Models

Response to a drug is assumed to be a function of the number or fraction of the total recep-tors that are occupied. The relationship between response and drug concentration is usuallyeither hyperbolic (Figure 16.3) or sigmoidal in nature; that is, high drug concentrationsproduce a maximum response, and once this has been achieved, further increases in con-centration produce no additional increases in response. This suggests that response is alsocontrolled by a capacity-limited process. The two models presented below differ in their as-sumptions regarding the origin of the capacity-limited nature of the response–concentrationrelationship.

16.2.2.1 Intrinsic Activity ModelThe simplest pharmacodynamic model the intrinsic activity model, assumes that thecapacity-limited nature of the response–concentration relationship is a direct consequenceof the capacity-limited nature of receptor occupancy. In this model, developed by Ariens,response is assumed to be directly proportional to receptor occupancy. Specifically, thefraction of maximum response (E/Em) is assumed to be proportional to the fraction of thetotal receptors occupied (RC/RT), and the constant of proportionality is the intrinsic activity:

E

Em= � · RC

RT(16.5)

where E is the response, Em the system’s maximum possible response, and � an efficacyterm that Ariens called intrinsic activity.

The maximum response that a drug can achieve (Emax) may be equal to or less thanthe maximum response that the system can produce (Em) (Figure 16.4). Intrinsic activityis the constant of proportionality between the system’s maximum response and the drug’smaximum response. Full agonists will have an intrinsic activity of 1; a partial agonist willhave an intrinsic activity greater than 0 and less than 1. For example, a drug that canproduce only 60% of the system’s maximum effect (Emax = 0.6Em) will have an � value of

Page 323: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

CLASSIC PHARMACODYNAMIC MODELS BASED ON TRADITIONAL RECEPTOR THEORY 303

Res

pons

e (%

of

Sys

tem

’s M

axim

um)

Drug Concentration

100

0

Emax = Em, α = 1

Emax = 0.6•Em, α = 0.6

FIGURE 16.4 Response–concentration relationship based on the intrinsic activity model. Thefractional response is directly proportional to the fraction of the total receptors occupied. The constantof proportionality is intrinsic activity (�). Drugs that have intrinsic activities of 1 (full agonist) and0.6 (partial agonist) have maximum responses (Emax) of 100% and 60% of the system’s maximumresponse (Em), respectively.

0.6 (Figure 16.4). An antagonist will have an intrinsic activity of zero. According to thismodel, the same degree of receptor occupancy for a series of full agonists (� = 1) willresult in the same response.

Substituting for RC/RT in equation (16.4) yields

E

Em= � · C

Kd + C(16.6)

Note that the drug concentration that produces 50% of its maximum response, which isknown as in EC50, in this model is equal to the Kd value, the drug concentration at 50%receptor occupancy.

During the course of this discussion, several pharmacodynamic parameters will beintroduced. To help readers keep track of them, and to distinguish them from each other,they will be summarized after they have been introduced. In this section we introduced:

1. Em, the maximum response of a system.

2. Emax, the maximum response of a drug; for a full agonist, Emax = Em.

3. EC50, the drug concentration that produces half the drug’s maximum response.

4. Kd, the drug concentration that results in 50% receptor occupancy.

5. �, the intrinsic activity or fraction of the system’s maximum response that an agonistcan illicit.

16.2.2.2 Efficacy ModelThe efficacy model superseded the simple model described above because the simple modelcould not account for the observation that some drugs are able to produce a maximumresponse at less than maximum receptor occupancy. To accommodate this observation:

1. The concept of spare receptors, receptors that are not occupied when some drugsproduce a maximal response, was introduced.

Page 324: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

304 INTRODUCTION TO PHARMACODYNAMIC MODELS

2. The model separated the two stages of drug response (Figure 16.1). First, the druginteracts with its receptors to produce a conformational change in the receptor andgenerate a stimulus. Second, the tissue translates the stimulus into a biological re-sponse, a process that may involve signal transduction and second messengers.

3. It is assumed that the intensity of the stimulus that results from a given level ofreceptor occupancy is not the same among drugs. Drugs that possess high efficacyare able to produce a larger stimulus per occupied receptor than drugs with lowerefficacy. Further, drugs with high efficacy are able to produce the system’s maximumresponse at less than full receptor occupancy. Drugs with lower efficacy may need tooccupy all the receptors to produce a full response, while others are unable to producethe system’s maximum response (partial agonists) even when all the receptors areoccupied.

4. It is assumed that the capacity-limited nature of the overall response concentrationrelationship can arise from either saturable receptor occupancy (weak agonists andpartial agonists) or an unknown process farther down the response chain: for example,a simple explanation could be the depletion of a second messenger.

The value of the stimulus depends on two factors: the fraction of the receptors occupied(drug concentration and affinity), and the efficacy (e), the efficiency with which the drugtranslates its binding to the receptors into the stimulus. The value of the stimulus can beexpressed as

S = e · RC

RT(16.7)

where S is the value of the stimulus and e is the efficacy.A drug’s efficacy is a constant of proportionality that links receptor occupancy to the

stimulus. Because the stimulus does not represent the fraction of maximal response (E/Em),it can achieve values greater than 1 (E/Em cannot be greater than 1). As a result, unlikeintrinsic activity, efficacy can achieve a value greater than 1. In contrast to the simpleintrinsic activity model, where all full agonists had an intrinsic activity of 1, the value ofefficacy can vary among full agonists, which allows the model to distinguish the differentefficacies of full agonists.

The system then converts the stimulus into a biological response. This may be a fast,simple process or may involve a cascade of several steps and may require a significantamount of time. The magnitude of response is a function of the value of the initial stimulusand may be expressed

E

Em= f (S) (16.8)

To accommodate the capacity-limited characteristics of the relationship between re-sponse and drug concentration (as discussed above, E/Em has a maximum value of 1), theright-hand side of the equation must resolve to a value between zero (the stimulus producesno response) and 1 (the stimulus produces the maximum response), so a hyperbolic function[E/Em = S/(1 + S)] is frequently used for the relationship between the value of the stimulus

Page 325: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

CLASSIC PHARMACODYNAMIC MODELS BASED ON TRADITIONAL RECEPTOR THEORY 305

and the magnitude of the biological effect. Combining equations (16.7) and (16.8) yields

E

Em= f

(e · RC

RT

)(16.9)

Substituting for RC/RT from equation (16.4) gives us

E

Em= f

(e · C

Kd + C

)(16.10)

A drug’s efficacy (the efficiency with which the receptor binding is converted into theinitial stimulus) is a function not only of the drug but also of the number of receptors. The ef-ficacy of a drug can be expressed per unit receptor. This is known as the intrinsic efficacy (ε):

ε = e

RT(16.11)

Thus, equation (16.10) may be written

E

Em= f

(ε · RT · C

Kd + C

)(16.12)

From equation (16.12), it can be seen that the effect produced by a given drug concentrationis dependent on:

1. The drug property intrinsic efficacy (ε) is the efficiency with which the drug is ableconvert its binding to a receptor into a stimulus. Compared to drugs with low efficacy,drugs with high intrinsic efficacy need to occupy fewer receptors to produce a givenresponse.

2. The drug property affinity of the drug for the receptor, which is expressed using thedissociation constant (Kd), which is a reciprocal measure of affinity. It is equal to thedrug concentration when 50% of the receptors are occupied. Low values of Kd areassociated with high affinity, and vice versa.

3. The tissue property total concentration of receptors, RT.

4. The tissue property that translates the initial stimulus into the biological response.

The parameters of this model thus separate the drug-specific parameters of intrinsicefficacy (ε) and affinity (Kd) from tissue-specific properties of the total concentration ofreceptors (RT) and the tissue property that converts the stimulus into the biological response.

Figure 16.5 shows the typical concentration–response profile associated with this model.In the figure, drugs with efficacies of 1 and 10 are partial agonists because they cannotproduce the maximum response produced by other drugs (the full agonists). Note thatat low concentrations, drugs with higher efficacies produce larger responses at equivalentconcentrations (i.e., they are more potent). At very high concentrations, there is no differenceamong full agonists in the magnitude of the response, but partial agonists produce lowermaximum responses. Note that although the two curves on the left both represent responsesto full agonists, they are not superimposed because the agonists producing them differin efficacy.

Page 326: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

306 INTRODUCTION TO PHARMACODYNAMIC MODELS

0

20

40

60

80

100

% M

axim

um R

espo

nse

Log Drug Concentration

e = 1000

e = 100

e = 10

e = 1

FIGURE 16.5 Semilogarithmic plots of response against drug concentration for a series of agonistswith different efficacies (e). The affinity of all four drugs is the same. In the figure the drugs thathave efficacies of 1 and 10 are partial agonists because they cannot produce the maximum responseproduced by other drugs (full agonists).

Figure 16.6 shows how the value of a drug’s affinity influences the response–concentration relationship for a series of full agonists with the same efficacy. As affinityincreases (Kd decreases), response at a given concentration increases. Figures 16.5 and 16.6demonstrate that a drug’s potency is a function of both its affinity and its efficacy. A sum-mary of some of the pharmacodynamic terms introduced in this section are provided below.

1. Affinity. The affinity of a drug is a measure of the strength with which the drug bindsto the receptor and is the reciprocal of the dissociation constant (i.e., 1/Kd). Kd is equal tothe drug concentration when half the receptors are occupied. Drugs that have high affinities(Kd small) require a lower concentration to occupy half the receptors compared to drugswith relatively low affinities.

0

20

40

60

80

100

% M

axim

um R

espo

nse

Log Drug Concentration

Kd = 0.1

Kd = 1

Kd = 10

FIGURE 16.6 Semilogarithmic plot of response against drug concentration for a series of agonistswith different values of affinity. The efficacy of all three drugs is the same. A drug’s Kd value is areciprocal form of affinity; as Kd increases, affinity decreases.

Page 327: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

EMPIRICAL PHARMACODYNAMIC MODELS USED CLINICALLY 307

C R RC⎯⎯⎯→+ ←⎯⎯ ⎯ STIMULUS

RESPONSE

Transduction

AffinityEfficacyIntrinsic

kon

koff

FIGURE 16.7 Affinity and intrinsic efficacy in the drug response chain.

2. Efficacy and intrinsic efficacy. Efficacy is an expression of the efficiency with whicha drug converts its interaction with the receptors of a system into a stimulus. Full agonistshave large values of efficacy that vary among drugs. Partial agonists will have low efficacies,and antagonists will have zero efficacy. Intrinsic efficacy is a measure of a drug’s efficacyper unit receptor. As such, in contrast to efficacy, it is dependent only on the drug andis a pure measure of the drug’s ability to produce a stimulus from its interaction with areceptor. Affinity and intrinsic efficacy constitute the drug-specific parameters that controlthe response–concentration relationship. Their individual roles in the response chain areshown in Figure 16.7.

3. Intrinsic activity. Intrinsic activity is the fraction of the full system response that adrug can produce. A full agonist that can produce the maximal system response has anintrinsic activity of 1, a partial agonist has an intrinsic activity between 0 and 1, and anantagonist has an intrinsic activity of 0. In contrast to pure efficacy, intrinsic activity doesnot make any distinction among full agonists—they all have intrinsic activities of 1.

4. Potency. Potency reflects the concentration of a drug that is required to produce agiven effect. A drug with high potency will produce a given effect at a lower concentrationthan will one with low potency. Potency, a function of tissue and drug factors, is controlledby the drug factors of affinity and intrinsic efficacy. Affinity controls the number of receptorsoccupied at a certain drug concentration, and intrinsic efficacy determines the magnitude ofthe effect that results from the occupancy. Potency is also controlled by two tissue-specificfactors: the number of receptors present in a system and how the receptor stimulus isconverted to a response. Within a biological system, the relative potency of two drugs isdependent on affinity and intrinsic efficacy.

5. EC50. EC50 is the drug concentration that produces half the drug’s maximum response.In the simpler intrinsic efficacy model, EC50 = Kd (the drug concentration that results in50% receptor occupancy). Note that in the efficacy model, if a drug has high efficacy, itsEC50 will be much less than the Kd value.

16.3 EMPIRICAL PHARMACODYNAMIC MODELS USED CLINICALLY

As discussed previously, the evaluation of drug response in humans is complicated byseveral factors. These include the difficulty in measuring drug concentrations at the siteof action, the identification of the events that constitute the response chain, the poten-tial involvement of homeostatic mechanisms, and the difficulty associated with measur-ing drug response. Recognizing the capacity-limited nature of the response–concentration

Page 328: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

308 INTRODUCTION TO PHARMACODYNAMIC MODELS

relationship in vivo, Wagner (7) proposed using an empirical, capacity-limited model, basedon the Hill equation, to describe the relationship between response and concentration. Thisequation expresses the binding of a ligand to a macromolecule and was developed initiallyto describe the binding of oxygen to hemoglobin (8). In pharmacodynamics it is known asthe Emax or sigmoidal Emax model.

16.3.1 Sigmoidal Emax and Emax Models

The sigmoidal Emax model and its simpler sister, the Emax model, are among the most fre-quently used pharmacodynamic models in clinical studies. The Emax model is summarizedas

E = Emax + C

EC50 + C(16.13)

where E is the drug effect or response, Emax the maximum effect of the drug, C the drugconcentration, and EC50 the concentration that produces 50% maximum response.

This model is empirical and makes no assumptions about the factors that are responsiblefor the capacity-limited or saturation characteristics of the response–concentration relation-ship. However, the Emax model bears a close resemblance to the simple intrinsic activitymodel, which assumed that response was directly proportional to receptor occupancy, didnot incorporate the concept of spare receptors, and combined the processes of stimulusgeneration and transduction. The similarities of the models are clear from a comparison ofequation (16.13) to the equation for the simple intrinsic activity model [equation (16.6)]:

E = � · Em · C

Kd + C(16.14)

The drug’s maximum response (Emax) is equivalent to the intrinsic activity (�) multipliedby the system’s maximum response (Em). Recall that the intrinsic activity represents thatfraction of the system’s total response that a drug can illicit: � is equal to 1 for a drug (fullagonist) that can produce the system’s maximum response, and � � 1 and � 0 for partialagonists. The drug concentration that gives half the drug’s maximum response (EC50) isequal to the drug concentration when half the receptors are occupied (Kd). However, it isimportant to recognize that despite its similarity to the intrinsic efficacy model, the Emax

model is empirical. It does not consider the events that constitute the response chain andhas, for example, been used to model the response to antagonists, such as �-blockers.

In considering the relationship between response and concentration, it is helpful torecall the very similar Michaelis–Menton model for the rate of an enzymatic reaction (see5.4.6). The hyperbolic relationship between the drug effect and concentration is shown inFigure 16.8a. At low concentrations the system is well below saturation, and the effectincreases almost linearly with concentration. As the concentration increases further, somesaturation of the system is observed and the effect can no longer increase proportionatelywith concentration. Eventually, when the system is fully saturated, the maximum effectis observed. In common with Michaelis–Menton kinetics, it is the relationship betweenthe concentration and the EC50 (Km in the Michaelis–Menton equation) that determinesthe nature of the response–concentration relationship. When drug concentrations are much

Page 329: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

EMPIRICAL PHARMACODYNAMIC MODELS USED CLINICALLY 309

100%

0

Res

pons

e

Res

pons

e

Drug Concentration Drug Concentration

100%

50%50%

EC50 EC50

(a) (b)

FIGURE 16.8 Relationship between response and drug concentration at the site of action for theEmax model (a) and the sigmoidal Emax model (b). The EC50 is the concentration that produces halfthe drug’s maximum response.

lower than EC50 (the response is less than 20% maximum), a linear relationship is observedbetween the response and the concentration. As the concentration increases, the increasein response with concentration becomes nonlinear, and eventually, at high concentrations,the response approaches maximum. At this point, increases in concentrations produce littleincrease in effect. Additionally, after the administration of a dose that approaches maximumeffect, the dissipation of the response will be slow because there is little change in responsewith concentration at high drug concentrations.

Equation (16.13) can be adapted further to take account of the fact that more than onedrug molecule may bind to each receptor:

E = Emax + Cn

ECn50 + Cn

(16.15)

where n is the number of drug molecules bound per receptor or the slope factor. The modelexpressed by equation (16.15) is known as the sigmoidal Emax model because it results in asigmoidal relationship between effect and concentration (Figure 16.8b). The parameter n isknown as the slope factor because it influences the slope of the effect–concentration relation-ship. It can be seen in Figure 16.9 that as n increases, the slope of the response–concentrationrelationship becomes steeper. In theory, n represents the number of drug molecules bindingper receptor, but it is often found to achieve noninteger values. Owing to the empirical natureof the model, n should be considered as the parameter necessary to describe the steepnessof the concentration–response curve. A summary of the parameters of the model follows.

1. EC50, the concentration of a drug that produces 50% of its maximum response. It isa measure of a drug’s potency. It is dependent on a drug’s affinity and its intrinsicefficacy. It will also depend on how the biological system relays the stimulus thatresults from activation of the receptor to generate the response.

2. Emax, an efficacy parameter, is dependent on a drug’s efficacy but also on the biologicalsystem. Specifically, it will be dependent on the number of receptors and how thestimulus resulting from receptor activation is relayed to generate the response. If the

Page 330: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

310 INTRODUCTION TO PHARMACODYNAMIC MODELS

0

20

40

60

80

100

0 0.5 1 1.5 2 2.5

Res

pons

e (%

)

Drug Concentration

n = 6

n = 3

n = 1 EC50

FIGURE 16.9 Effect of the slope factor n on the response–concentration relationship in the sig-moidal Emax model. It can be seen that as n increases, the slope of the response–concentration curvegets steeper.

Emax of a series of drugs were measured in the same system, the values could becompared to provide values of their relative intrinsic activity and thus distinguishbetween full and partial agonists.

3. n, a factor that expresses the slope of the response–concentration relationship.

Baseline Effects and Drug-Induced Decreases in Response The basic equation for thesigmoidal Emax model (16.15) predicts that the response to an agonist will be zero whenthe drug concentration is zero and that the response will increase with concentration. Inmany cases, this does not apply. First, a positive (nonzero) baseline response may exist. Forexample, antihypertensive agents modify a resting blood pressure, hypoglycemic agentsaffect existing blood glucose levels, and H2- blockers modify an existing gastric pH. Second,many drugs reduce rather than increase an existing response. In the examples above, boththe antihypertensive agents and the hypoglycemic agents reduce a baseline response. Thesigmoidal Emax model can easily be adapted to account for both of these situations:

E = E0 ± Emax + Cn

ECn50 + Cn

(16.16)

where, E0 is the baseline state of the system, and drugs will either increase or decrease theresponse relative to this baseline value.

16.3.2 Linear Adaptations of the Emax Model

Prior to the availability of computer software to perform nonlinear regression analysis,linear forms of the Emax model were useful. These models may still be of value in situationswhere it may not be possible to approach the maximum effect of a drug, possibly becauseof toxicity. Two linear versions exist: the linear model and the logarithmic model.

Page 331: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

EMPIRICAL PHARMACODYNAMIC MODELS USED CLINICALLY 311

Linear Model Notice in equation (16.13) that when C � EC50, the denominator resolvesto EC50, which leads to the linear model, where there is a linear relationship between effectand concentration:

E = Emax · C

EC50(16.17)

or

E = a · C (16.18)

where a is a proportionality constant equal to Emax/EC50. This can be seen at low concen-trations in Figure 16.10. The linear model holds in the range 0 to 20% maximum response.The disadvantage of this model is that it predicts that the response will continue to increaseindefinitely in a proportional manner as concentration increases. It does not reflect clinicallyobserved responses at higher concentrations.

Logarithmic Model In the logarithmic model, response is expressed as a function of thelogarithm of concentration. There is no mechanistic reason for this; it simply allows agreater range of concentrations to be plotted. However, in the response range 20 to 80%a linear relationship exists between response and the logarithm of concentration (Figure16.11). The logarithmic model is

E = a + b log C (16.19)

The logarithmic model suffers from several disadvantages. In common with the simplelinear model, it predicts that the effect will rise indefinitely with increases in concentration.The logarithmic model cannot predict effects when the concentration is zero, and if themaximum response is not known, it is difficult to identify the lower (20%) and upper (80%)bounds of the model.

Res

pons

e

Drug Concentration

100

0

50 EC50

Res

pons

e

Drug Concentration

20

0

FIGURE 16.10 Linear model at concentrations producing less than 20% response.

Page 332: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

312 INTRODUCTION TO PHARMACODYNAMIC MODELS

0

20

40

60

80

100

0.1 1 10

Res

pons

e

Drug Concentration

FIGURE 16.11 Log-linear model. A linear relationship between response and the logarithm ofdrug concentration exists in the region of 20 to 80% maximum response.

16.4 INTEGRATED PK–PD MODELS: Emax MODEL COMBINED WITH APK MODEL FOR INTRAVENOUS BOLUS INJECTION IN AONE-COMPARTMENT MODEL

To create a complete model of drug response, a pharmacokinetic model that describes theplasma concentration at any time after the administration of a dose of a drug must belinked to a pharmacodynamic model that describes the response produced by any givenconcentration at the site of action (Figure 16.12). A drug’s pharmacokinetic parameters,such as clearance and volume of distribution, determine the plasma concentration at anytime after the dose. The pharmacodynamic parameters, such as Emax and EC50, determinethe response to any concentration at the site of action. When they are linked, the responseat any time after a dose can be estimated. The simplest approach to linking the two modelsis to assume that the concentration at the site of action is always in equilibrium with the

PHARMACOKINETICMODEL

PHARMACODYNAMICMODEL

Cl Vd

1-, 2-, or 3-Compartments

Sigmoidal, Linear

EC50n

LINKMODEL

Ce RESPONSEDOSE Cp

DrugSystemic

Circulation

Drug atSite ofAction

DrugInteraction

with ReceptorRESPONSE

FIGURE 16.12 Modeling the complete dose–response relationship can be accomplished by linkinga pharmacokinetic model to a pharmacodynamic model.

Page 333: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

INTEGRATED PK–PD MODELS 313

PHARMACOKINETICMODEL

PHARMACODYNAMICMODEL

Cl Vd

1-, 2-, or 3-Compartments

Ce RESPONSEDOSE Cp α

Sigmoidal, Linear

EC50n

FIGURE 16.13 Direct link between the pharmacokinetic and pharmacodynamic models. The drugconcentration at the site of action is assumed to be in equilibrium with the plasma at all times. Thisallows response in the pharmacodynamic model to be driven by the plasma concentration.

plasma concentration (Figure 16.13). As a result, the plasma concentration can be used asthe concentration that drives response:

E = Emax · Cp

EC50 + Cp(16.20)

Because plasma concentrations are used to drive the response in both the Emax and sigmoidalEmax models, EC50 will be in units of equivalent plasma concentrations.

Figure 16.14 shows the plasma concentration and response simulated when a model foran intravenous bolus injection is linked to an Emax model. Note that the units of time inFigure 16.14 are elimination half-lives and the units of concentration are EC50 equivalents.It can be seen that the initial plasma concentration falls by 50% in one unit of time, and fallsan additional 50% over each subsequent unit of time. In contrast, the initial fall in responseis much less steep. In one elimination half-life (one unit of time), the initial response

0

20

40

60

80

100

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8

Cp

(EC

50)

Time (elimination half-lives)

Res

pons

e (%

Max

imum

)

Response

Cp

FIGURE 16.14 Graph of plasma concentration and response against time. Note that time has unitsof the drug’s elimination half-life, and the units of concentration are the drug’s EC50.

Page 334: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

314 INTRODUCTION TO PHARMACODYNAMIC MODELS

0

10

20

30

40

50

60

0

0.5

1

0 1 2 3 4 5 6 7 80

20

40

60

80

100

0

20

40

60

80

100

0 1 2 3 4 5 6 7 8

Cp

(EC

50)

Cp

(EC

50)

Time ( t1/2) Time ( t1/2)

(a) (b)

Res

pons

e (%

max

)

Res

pons

e (%

max

)

FIGURE 16.15 Graph of plasma concentration and response against time after a small (a) and alarge (b) dose. Note that time has units of the drug’s elimination half-life, and the units of concentrationare the drug’s EC50. After a small dose (a), the plasma concentrations (dashed line) are around, orless than, the drug’s EC50, and response (solid line) falls almost in parallel with plasma concentration.After large doses (b), plasma concentrations (dashed line) are much larger than the EC50, and the fallin response (solid line) is much slower than the fall in the plasma concentration.

falls by only about 10%. The differing slopes of the fall in response and concentrationare illustrated further in Figure 16.15, which shows plasma concentration and responseover time after a low dose (10 units) (Figure 16.15a) and a high dose (1000 units) (Figure16.15b). It can be seen that the initial plasma concentration from the high dose is 100concentration units, or 100 times greater than the drug’s EC50. Under these conditions amaximum response of 100% is obtained. The system is saturated and there is excess drugavailable to produce maximum response. As a result, even as drug is eliminated from thebody, sufficient drug is still available to sustain the maximum response. When the drugconcentration is much greater than the drug’s EC50 (or when the response is around maximalresponse), the fall is response is much less steep than the fall in the plasma concentration,and response lingers. Under these conditions, it takes several elimination half-lives for theinitial response to fall by 50% (Figure 16.15b). In contrast, a small dose that achieves aninitial concentration around EC50 produces around 50% maximal response, and the fallin response approximately parallels the fall in plasma concentration (Figure 16.15a). Thisphenomenon has several implications, among which is its impact on dosing regimen design.Drugs that are administered in doses that achieve plasma concentrations much larger thantheir EC50 values and which, as a result, produce responses approaching maximal canbe administered using dosing frequencies much longer than their half-lives. For example,atenolol has a half-life of about 6 h but can be administered daily because it is used in dosesthat produce close to maximum response (9).

16.4.1 Simulation Exercise

Open the model “Sigmoidal Emax Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter16a

The model consists of a one-compartment model with intravenous input linked to asigmoidal Emax model. The model is set up so that time is in units of elimination

Page 335: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

HYSTERESIS AND THE EFFECT COMPARTMENT 315

half-lives and concentration is in units of EC50. The parameters have the followingdefault values: Vd = 10 L, Cl = 6.93 L/t1/2, Emax = 100, and EC50 = 1 EC50; dose unitsare EC50 · L.

1. Explore the model and note the model summary.

2. Go to the “Cp and Response” page. Give a dose and observe that when Cp =EC50, response is 50% maximum. Note that the fall in response does not parallelthe fall in Cp. It takes one unit of time (one elimination t1/2) for the initial Cp tofall by 50%. In the same period, response falls by only about 10%. Note that at 6units of time, the response is about 20%, and it takes about one unit of time (oneelimination half-life) for it to fall 50%.

3. Go to the “Effect of Cp:EC50 Ratio” page. Give a dose of 10 units. Note that Cptakes one unit of time to fall 50%, and during this time the response also fallsby about 50%. When Cp is small compared to the EC50 (Cp � EC50), the fall inresponse parallels the fall in Cp. Give a dose of 1000 units. Note that the initialCp is 100 times greater than the EC50 and the initial response is maximal. Inaddition, during the first elimination half-life, Cp falls by 50% but response doesnot change. Under these circumstances, it takes about 6 elimination half-livesfor the response to fall by 50%. When Cp >> EC50, the response is maximal orclose to maximal. The system is saturated and there is an excess amount of drugat the site of action. Even after drug has been eliminated, there is still enoughdrug to elicit a large response. Under these circumstances, the fall in responseis much slower than the fall in Cp. Drugs that are administered in doses thatproduce a close to maximal effect (Cp >> EC50) do not have to be administeredevery half-life to avoid wide changes in response. The dosing interval can bemuch greater than the half-life.

4. Observe the maximum response (Rmax) from doses of 10, 100 and 1000 units.Note that Rmax is not proportional to dose. The response concentration relation-ship is nonlinear.

5. Go to the “Effect of n” page. Without clearing the graph between doses, givedoses using n values of 0.5, 1, 2, and 5. Note that as n increases the steepnessof the concentration–response curve increases. When n is large, small increasesin the concentration can lead to large increases in response. Note that changesin n do not alter the EC50.

16.5 HYSTERESIS AND THE EFFECT COMPARTMENT

The drug concentration in the plasma is generally used to drive response in pharmaco-dynamic models because it is the only concentration that can be measured easily. Thisapproach is justified as long as the drug concentration at the site of action is in equi-librium with the plasma. When these two concentrations are not in equilibrium, strangeresponse–concentration profiles can be observed. These unusual effects must be understoodand addressed before pharmacodynamic modeling can be conducted.

Frequently in clinical pharmacodynamic studies, a plot of drug response against plasmaconcentration appears as a counterclockwise circular path such as the profile shown in

Page 336: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

316 INTRODUCTION TO PHARMACODYNAMIC MODELS

Res

pons

e

CpC

once

ntra

tion

1 2 31

23

Plasma

Site of Action

(a) (b)

Time

FIGURE 16.16 Hysteresis due to a distributional delay. Panel (a) shows response plotted as a func-tion of plasma concentration (solid line). Also shown is the expected response–plasma concentrationrelationship (dashed line). Panel (b) shows the drug concentrations in the plasma (solid line) and atthe site of action (dashed line) as a function of time after a dose. As a result of slow distribution, thepeak concentration at the site of action occurs later than the peak plasma concentration. It is importantto appreciate that the response is driven by the drug concentration at the site of action, not that in theplasma. Note in area 1 of part (b) that the plasma concentration and the concentration at the site ofaction both increase. Because of the latter, the effect increases. In area 2 the concentration at the sitecontinues to increase; thus the effect increases, but the plasma concentration is decreasing. It at thispoint that the curve of response against Cp begins to move in a counterclockwise direction [panel (a)].Eventually, the concentration at the site of action decreases. As a result, response decreases (area 3).

Figure 16.16. For comparison the figure also shows the relationship that would be expectedbased on the Emax model. This phenomenon is called hysteresis, a word derived from aGreek word meaning to be behind or to be late. It occurs when there is a time lag betweenthe rise and fall of plasma concentration and the rise and fall in response.

Hysteresis can have several different causes (discussed in Chapter 17), including indirectdrug effects and a long transduction process. But a simple and common cause is a slow dis-tribution of a drug to its site of action. The phenomenon may be understood by recognizing,first, that it is the drug at the site of action, not that in the plasma, that drives the response.Second, it is important to appreciate that distribution of the drug from the plasma to thesite of action may be slow and that the peak concentration at the site may occur later thanthat in the plasma. Figure 16.15b shows the drug concentration–time profile for the plasmaand the drug’s site of action when distribution is slow. The overall curve has been split intothree phases. It can be seen that the peak plasma concentration (solid line) occurs earlierthan the peak at the site of action. In the initial period after drug administration (phase 1)there is a slow distribution of the drug to the site: The concentration at the site of actionincreases and the response increases. During this phase the plasma concentration is alsoincreasing. Eventually, the plasma reaches its peak concentration, and in phase 2 the plasma

Page 337: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

HYSTERESIS AND THE EFFECT COMPARTMENT 317

DrugSystemic

Circulation

Drug at Site ofAction

DrugInteraction

with ReceptorRESPONSE

PHARMACOKINETICMODEL

PHARMACODYNAMICMODEL

Ce RESPONSEDOSE Cp

Effect Compartment

Ce

FIGURE 16.17 An effect compartment is used to link the pharmacokinetic and pharmacodynamicmodels when there is a delay in drug reaching the site of action.

concentration falls. During this phase the concentration at the site continues to increase, andas a result so does the response. Thus, it is in this area (plasma concentration falling, responseincreasing) that the plot of response against the plasma concentration begins to move in acounterclockwise pattern. This pattern continues until the concentration at the site of actionbegins to fall (Figure 16.16, area 3). In phase 3 the concentration at the site, the concentra-tion in the plasma, and the effect all decrease. Note that hysteresis would be eliminated if theresponse were plotted as a function of the drug concentration at the site of action rather thanin the plasma. An interactive video is available that demonstrates and explains hysteresis(see Section 16.5.1).

The distributional delay to the site of action can be accommodated using an effect com-partment to link the pharmacokinetic and pharmacodynamic models (10) (Figure 16.17).The effect compartment represents the site of action or biophase, and it is attached tothe central compartment of multicompartment models (Figure 16.18). Drug distribution toand redistribution from the compartment are modeled as first-order processes. The rate ofchange of the drug concentration in the effect compartment may be expressed as

dCe

dt= Cp · k1e − Ce · ke0 (16.21)

where Ce is the concentration of drug in the effect compartment or the concentration at thesite of action, Cp the plasma concentration, and k1e and ke0 the first-order rate constants fordistribution into and out of the effect compartment, respectively.

The amount of drug that distributes to the site of action is assumed to be very small. Asa result, drug redistributing from the effect compartment is not returned to the system butis assumed to be lost (Figure 16.18). This simplifies the pharmacokinetics of the drug.

The prominence of hysteresis and the amount of delay in the response is determined bythe time it takes for the effect site to equilibrate with the plasma. Just as a drug’s eliminationrate constant or elimination half-life controls the time to reach steady state during aninfusion or any other type of chronic drug administration, ke0 controls the time it takes the

Page 338: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

318 INTRODUCTION TO PHARMACODYNAMIC MODELS

ke0•Ce

AeCeVe

k1e•Cp

A1CpV1

Central Compartmentof PK Model

Effect Compartment

Ce drives response

FIGURE 16.18 The effect compartment is linked to the central compartment of the pharmacoki-netic model. The distribution (D) and redistribution (R) of the drug to and from the effect compartmentare first-order processes with rate constants of k1e and ke0, respectively. The amount of drug that dis-tributes to the effect compartment is assumed to be so small that rather than returning it to the system,it can be assumed to be lost from the system. A, C, and V represent the amount, concentration, andvolume, respectively, and 1 and e signify the central and effect compartments, respectively.

effect compartment to equilibrate with the plasma. Thus, minimal hysteresis is associatedwith large ke0 values, and significant hysteresis is associated with small ke0 values. Sinceredistribution or loss of drug from the site is a first-order process, the ke0 half-life (0.693/ke0)can be used to estimate the time it takes for drug distribution to be complete (3 to 5 t1/2, ke0 ).Once distribution to the site is complete, the ratio of the concentration of drug in the plasmaand the site of action will be constant and the concentrations will parallel each other. Atthis time, a normal relationship between response and the plasma concentration will beobserved. Thus, hysteresis is not observed when plasma concentrations and concentrationsat the site of action are at steady state but is only observed in non-steady-state conditions,such as the period after drug administration.

The value of the ke0 can be estimated from the hysteresis curve and the time courseof drug response. It is not possible to estimate k1e, and as a result it is not possible toestimate drug concentrations at the site of action. The value of k1e is usually assumed toequal ke0, and as a result, after distribution has gone to completion, Ce will equal Cp. As aresult when the effect compartment is linked to an Emax model, the value of the EC50 will beexpressed in terms of the plasma concentration. Examples of drugs whose effects have beenmodeled using the effect compartment include d-tubocurarine (muscle paralysis), digoxin(LVET shortening), disopyramide (QT prolongation), and fentanyl (respiratory depression)[see (11)].

16.5.1 Simulation Exercise

Open the model “Sigmoidal Emax Model with an Effect Compartment” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter16b

The model is a one-compartment model with first-order absorption linked through aneffect compartment to an Emax model. The default model parameters are Vd = 20 L,

Page 339: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

PROBLEMS 319

Cl = 4 L/h (this results in k and t1/2 of 0.2 h−1 and 3.5 h, respectively), F = 1, ka = 1h−1, Emax = 100, EC50 = 1 mg/L, ke0 = 0.5 h−1, and dose = 20 mg.

1. Explore the model and review the model summary.

2. Review the interactive video.

3. Go to the “Effect of keo” page. The graph on the left shows the values of Cp and Ceafter each simulation. The graph on the right shows the response after each simu-lation. The graph on the right will not be cleared after a simulation, so the responsefrom different simulation runs can be compared. Give doses using values of ke0

of 0.5, 1, 2, and 4 h−1. Note that as ke0 increases, the time lag between the riseand fall of Cp and that of Ce decreases. Note that the delay in response also de-creases. It can also be seen that changes in the value of ke0 influence the durationof action. If the response must be greater than 20% to be therapeutic, the onset ofaction would increase and the duration of action would decrease as ke0 increased.Comparing ke0 values of 0.5 and 4 h−1, the onset of action would decrease fromaround 1.5 h to 0.5 h, and the duration of action would decrease from around 8.8 hto 7.6 h, respectively.

4. Go to the “Effect of Dose” page. Give doses of 20, 50, 100, and 200 mg andobserve how Rmax (the maximum response) and T max (the time of maximumresponse) are influenced by dose. Note that the relationship between responseand dose is not linear. Increases in dose result in less than proportional in-creases in response. Eventually, when a peak plasma concentration producesthe maximum response (Emax), further increases in dose will not produce largerresponses. Note that T max is the same for each dose. This is an important featureof hysteresis produced by distributional delays that distinguishes it from othercauses of hysteresis and delays in response.

PROBLEMS

16.1 The dosing regimen for nosolatol (Cl = 12.6 L/h, Vd = 210 L/70 kg, S = 1)was calculated in Chapter 12 using pharmacokinetic principles. A dosing regimenof 200 mg every 12 h was selected based on a target average steady-state plasmaconcentration of 1.2 mg/L, a t1/2 value of about 12 h, and the desire to have theplasma concentrations fall no more than about 50% in a dosing interval and staybetween 750 to 1900 mg/L.

The pharmacodynamics of nosolatol have recently been studied. The response(reduction in exercise heart rate) and the plasma concentration were measured afterseveral doses and the data fit to a one-compartment pharmacokinetic model linkedto an Emax model. The values of nosolatol’s pharmacokinetic parameters agreedwell with previous estimates, and the estimated pharmacodynamic parameters ofnosolatol were (±S.D.) Emax = 30 ± 3.2 beats per minute (bpm) and EC50 = 75± 15�g/L. Ideally, the drug’s effect should always be in the range of 28 to 22 bpmresponse (reduction in exercise heart rate). Assume that the drug is administered asa multiple IV bolus injections and that there is no hysteresis or delay in its effect.

Page 340: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

320 INTRODUCTION TO PHARMACODYNAMIC MODELS

Use equation (16.20) to identify the desired peak and through concentrations. De-termine a dosing regimen to achieve the desired response profile.

16.2 The pharmacokinetics and pharmacodynamics of a new antihistamine are beingevaluated in human volunteers. The response is assessed by measuring the inhibitionof a wheal generated in response to a subcutaneous injection of histamine in theforearm. Volunteers received a 10-mg oral dose of the drug. Plasma concentrationand response were measured at various times after the dose, and the data are providedin Table P16.2A.

TABLE P16.2A

Time Cp Response(h) (�g/L) (% inhibition)

0 0 0.00.4 1.69 5.11 3.05 23.12 3.76 44.02.4 3.78 48.73 3.69 53.53.8 3.45 57.14 3.39 57.65 3.04 59.15.4 2.9 59.26 2.7 59.07 2.39 58.18 2.11 56.5

10 1.65 52.212 1.29 46.916 0.78 35.818 0.61 30.4

The data are to be used to assess the concentration–response relationship and identifya multiple dosing schedule.

(a) Plot the response as a function of Cp. If, for example, a response of 60%inhibition is desired, is it possible to identify therapeutic plasma concentrations?

(b) Previous studies have shown that the drug has a bioavailability of 0.5. Theworksheet used for the analysis of oral data can be used to perform pharma-cokinetic analysis on the data. It will be found that the drug has the followingpharmacokinetic parameters: Cl = 123 L/h, Vd = 1000 L, and ka = 1 h−1.

Nonlinear regression analysis was used to model the data to the Emax modelwith an effect compartment. The following parameter values were obtained:Emax = 100%, EC50 = 2 �g/L, ke0 = 0.4 h−1. As a result, the concentration ofdrug at the site of action could be estimated in units of plasma concentration.These data are shown in Table P16.2B. Plot the data and comment on theconcentration–effect relationship.

Develop a multiple intravenous dosing regimen that will provide a steady-state peak effect of about 80% and a trough effect of about 40% inhibition of

Page 341: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

REFERENCES 321

TABLE P16.2B

CeTime (Cp equivalents) Response(h) �g/L (% inhibition)

0 0 0.00.4 0.11 5.11 0.6 23.12 1.57 44.02.4 1.9 48.73 2.3 53.53.8 2.66 57.14 2.72 57.65 2.89 59.15.4 2.9 59.26 2.88 59.07 2.77 58.18 2.6 56.5

10 2.18 52.212 1.77 46.916 1.11 35.818 0.87 30.4

the wheal. Assume that at steady state the effect compartment is in equilibriumwith the plasma.

REFERENCES

1. Peck, C. C., Barr, W. H., Benet, L. Z., Collins, J., Desjardins, R. E., Furst, D. E., Harter, J. G., Levy,G., Ludden, T., Rodman, J. H., et al. (1994) Opportunities for integration of pharmacokinetics,pharmacodynamics, and toxicokinetics in rational drug development, J Clin Pharmacol 34,111–119.

2. Danhof, M., de Jongh, J., De Lange, E. C., Della Pasqua, O., Ploeger, B. A., and Voskuyl, R.A. (2007) Mechanism-based pharmacokinetic–pharmacodynamic modeling: biophase distribu-tion, receptor theory, and dynamical systems analysis, Annu Rev Pharmacol Toxicol 47, 357–400.

3. Mager, D. E., Woo, S., and Jusko, W. J. (2009) Scaling pharmacodynamics from in vitro andpreclinical animal studies to humans, Drug Metab Pharmacokinet 24, 16–24.

4. Wallin, J. E., Friberg, L. E., and Karlsson, M. O. (2009) A tool for neutrophil guided doseadaptation in chemotherapy, Comput Methods Programs Biomed 93, 283–291.

5. Wagner, J. A. (2009) Biomarkers: principles, policies, and practice, Clin Pharmacol Ther 86,3–7.

6. Clarke, W. P., and Bond, R. A. (1998) The elusive nature of intrinsic efficacy, Trends PharmacolSci 19, 270–276.

7. Wagner, J. G. (1968) Kinetics of pharmacologic response: I. Proposed relationships betweenresponse and drug concentration in the intact animal and man, J Theor Biol 20, 173–201.

Page 342: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc16 JWBS062-Rosenbaum February 16, 2011 8:13 Printer Name: Yet to Come

322 INTRODUCTION TO PHARMACODYNAMIC MODELS

8. Hill, A. (1910) The possible effects of the aggregation of the molecules of haemoglobin on itsdissociation curves, J Physiol 40, IV–VII.

9. Tozer, T. N., and Rowland, M. R. (2006) Introduction to Pharmacokinetics and Pharmacody-namics, Lippincott Williams & Wilkins, Baltimore.

10. Sheiner, L. B., Stanski, D. R., Vozeh, S., Miller, R. D., and Ham, J. (1979) Simultaneous modelingof pharmacokinetics and pharmacodynamics: application to d-tubocurarine, Clin Pharmacol Ther25, 358–371.

11. Lalonde, R. L. (1992) Pharmacodynamics, In Applied Pharmacokinetics (Evans, W. E., Schentag,J. J., and Jusko, W. J., Eds.) 2nd ed., Applied Therapeutics, Vancouver, WA.

Page 343: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

17MECHANISM-BASED INTEGRATEDPHARMACOKINETIC–PHARMACODYNAMIC MODELS

17.1 Introduction

17.2 Models Incorporating Receptor Theory: Operational Model of Agonism17.2.1 Simulation Exercise

17.3 Physiological Turnover Model and Its Characteristics17.3.1 Points of Drug Action17.3.2 System Recovery After Change in Baseline Value

17.4 Indirect Effect Models17.4.1 Characteristics of Indirect Effect Drug Responses17.4.2 Characteristics of Indirect Effect Models Illustrated Using Model I

17.4.2.1 Time Course of Response17.4.2.2 Effect of Dose17.4.2.3 Maximum Response and Maximum Achievable Response of a Drug17.4.2.4 Influence of Physiological System Parameters17.4.2.5 Influence of Pharmacokinetics: Elimination Rate Constant17.4.2.6 Pharmacodynamic Parameters of the Drug: Effect of IC50 and Imax

17.4.2.7 Time to Steady State During an Intravenous Infusion17.4.3 Other Indirect Models

17.5 Transduction and Transit Compartment Models17.5.1 Simulation Exercise

17.6 Tolerance Models17.6.1 Counter-regulatory Force Model

17.6.1.1 Simulation Exercise17.6.2 Precursor Pool Model of Tolerance

17.6.2.1 Simulation Exercise

17.7 Irreversible Drug Effects17.7.1 Application of the Turnover Model to Irreversible Drug Action

17.7.1.1 Simulation Exercise17.7.2 Model for Hematological Toxicity of Anticancer Drugs

17.7.2.1 Simulation Exercise

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

323

Page 344: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

324 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

17.8 Disease Progression Models17.8.1 Generation of Drug Response17.8.2 Drug Interaction with a Disease17.8.3 Disease Progression Models

17.8.3.1 Linear Disease Progression Model17.8.3.2 Exponential Decay or Zero Asymptotic Model17.8.3.3 Exponential Model with a Nonzero Maximum Disease Status

Problems

Objectives

The material in this chapter will enable the reader to:

1. Understand the limitations of the simple sigmoidal Emax model to characterize a drug’spharmacodynamic properties and to summarize the time course of drug response

2. Understand the advantages offered by the operational model of agonism

3. Appreciate the value of several examples of mechanism-based pharmacodynamicmodels

4. Identify the structure and characteristics of the mechanism-based models for indirecteffects, transduction, tolerance, irreversible drug effects, and disease progression

17.1 INTRODUCTION

The sigmoidal Emax model has been used extensively in clinical pharmacology and hasproven to be valuable in modeling response data from a wide variety of drugs, includingboth agonists and antagonists. Models developed for individual drugs have provided a bet-ter understanding of the dose–response relationship, assisted in the determination of doserequirements, and helped identify situations where pharmacodynamics and dose require-ments may be altered. However, the model is essentially empirical and, as such, suffersfrom several disadvantages.

The sigmoidal Emax model takes the final response and simply condeness it into asingle capacity limited process driven by drug concentrations. Consideration is not givento any of the events along the response chain (Figure 17.1). Although the assumptions aredifferent, it can be considered to be equivalent to the intrinsic activity model (16.2.2.1). Itdoes not accommodate the concept of spare receptors, and as a result it cannot distinguishdifferent efficacies among full agonists. It does not accommodate time delays betweenreceptor activation and the emergence of the response. An effect compartment can beadded to explain distributional delays. However, in some situations, the mechanism ofaction of the drug may suggest that the delay in response has a pharmacodynamic, nota pharmacokinetic basis. For example, the peak effect in response to a dose of warfarinoccurs at a time when most of a dose has already been eliminated from the body. Thiscannot be explained by a delay in the drug reaching its site of action. Additionally, thesigmoidal Emax model cannot support the possibility of exposure-dependent changes in theconcentration–response relationship, such as the development of tolerance.

Finally, the model does not separate drug-specific (efficacy and affinity) and tissue-specific (receptor density) parameters. The drug-specific pharmacodynamic parametershave been found to be remarkably consistent among different species and from in vitro

Page 345: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

MODELS INCORPORATING RECEPTOR THEORY 325

PKMODEL

PDMODEL

LINKMODEL

Ce RESPONSEDOSE Cp

RESPONSE

SigmoidalEmax

1. Signal (S) Generation

S

2. Physiological Phase

- Sigmoidal Emax - Operational Model of Agonism

- Indirect Effect- Transduction- Tolerance- Irreversible Effects- Disease Progression

PhysiologicalPhase

1. Simple PK–PD Models

2. Mechanism-Based PK–PD Models

( )m TRCE fE R=

E

E

DOSE Cp Ce

Cp DRIVESEFFECT

LINKMODEL

Cp DRIVESEFFECT

or

or

FIGURE 17.1 Diagrammatic representation of the simple sigmoidal Emax and mechanism-basedpharmacodynamic models. S is the initial stimulus resulting from the drug receptor interaction, andE is the direct biological response.

studies (1). Thus, if an integrated PK–PD model for drug response in vivo separated thedrug- and tissue-specific parameters, it would be possible to estimate drug response inhumans based on parameters found in laboratory animals or in vitro. This translationalresearch could have a major impact on drug development. It could allow drug candidateswith the greatest chance of success to be identified early during development and couldhelp to optimize the design of clinical trials, which could decrease both the cost and timeof drug development. The sigmoidal Emax parameters are, however, dependent on both thedrug and the system in which they were determined. As a result, this model has limitedapplications in translational research.

The disadvantages of the sigmoidal Emax model have been addressed through the de-velopment of a range of mechanism-based pharmacodynamic models that incorporateadditional steps in the chain of events between receptor activation and the emergence ofthe response [see (1–3)]. A description of some of these models is presented below.

17.2 MODELS INCORPORATING RECEPTOR THEORY: OPERATIONALMODEL OF AGONISM

As discussed above, the parameters of the sigmoidal Emax model are combined drug- andtissue-specific parameters. Black and Leff’s operational model of agonism (4) incorporatesreceptor theory into in vivo models of drug response. As a result, the model providesestimates of the drug-specific parameters of efficacy and affinity.

Page 346: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

326 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

As presented in Chapter 16, according to classical receptor theory, the interaction ofa drug with its receptors is controlled by the law of mass action. The concentration ofreceptors occupied is expressed as follows:

RC = RT · C

Kd + C(17.1)

where RC is the concentration of the drug–receptor complex, C the concentration of drug,RT the total concentration of receptors, and Kd the drug’s dissociation constant, a reciprocalmeasure of a drug’s affinity. Furthermore, the response that results from the drug–receptorinteraction is believed to be a function of the number of receptors occupied:

E

Em= f

(RC

RT

)(17.2)

where E is the effect and Em is the maximum possible response of the system.The Emax model is mathematically equivalent to the intrinsic activity model, which

assumes a linear relationship between response and receptor occupancy (Figure 17.2). Asshown previously [equations (16.15) and (16.16)], Emax is equivalent to the product ofthe maximum response of the system or the maximum response of a full agonist, and theintrinsic activity: Emax= Em · � (Note: For full agonists, � = 1; for partial agonists, 0 � � �1). As a result, Emax is both a drug-specific (�) and a system-specific (Em) parameter. This

RC

/RT

C

RC

/RT

C

100

0

E

RC/RT

E

RC/RT

Kd

Kd = EC50

Emax = Em for a full agonist

KdKe or RCE50

Em

1

50

100

0

50

100

0

50

100

0

50

0.5

Upper Graphs the Emax Model

Lower Graphs the Operational Model of Agonism

FIGURE 17.2 Relationship between fractional receptor occupancy and response for the Emax

model (upper panel) and the operation model of agonism (lower panel). Kd is an inverse measureof affinity and corresponds to the drug concentration that results in 50% receptor occupancy. TheEmax model is equivalent to a model that assumes that the response (E) is directly proportional tothe fraction of receptors occupied (RC/RT). The OMA assumes a hyperbolic relationship betweenresponse and receptor occupancy. Ke or RCE50 is the drug–receptor concentration that results in 50%of the system’s maximum response.

Page 347: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

MODELS INCORPORATING RECEPTOR THEORY 327

model also assumes that full agonists will all produce the same quality of drug–receptorinteraction and that a given fractional occupancy will produce the same response amongdifferent drugs. But this is not necessarily the case, as many drugs are able to produce amaximum response at less than 100% receptor occupancy.

The Black and Leff operational model of agonism (4) is an alternative way of modelingdrug action. The model accommodates the concept of spare receptors and incorporates aterm to represent a drug’s efficacy, which enables it to distinguish among full agonists thathave different efficacies. Thus, it addresses the fact that the response to a given level ofreceptor occupancy varies among drugs, and that a maximum response can be observed atless than 100% receptor occupancy.

The normal hyperbolic, capacity-limited relationship between drug concentration andreceptor occupancy is assumed [equation (17.1)]. A second hyperbolic capacity-limited pro-cess is then used to model the relationship between the concentration of the drug–receptorcomplex and the response (Figure 17.2):

E

Em= RC

Ke + RC(17.3)

where Em is the maximum possible response of the system (as distinct from Emax, whichis the maximum possible response of a given drug) and Ke is the concentration of thedrug–receptor complex that produces 50% Em.

This model allows the response elicited by a given drug–receptor concentration to varyamong drugs. Drugs with higher efficacy will be able to elicit a given effect at a lowerdrug–receptor concentration than will drugs with low efficacy. The Ke value (Figure 17.2,lower right) is the concentration of the drug–receptor complex that produces half themaximum response of the system. A more meaningful symbol for Ke is RCE50. Thus, RCE50

is a measure of efficacy. Drugs with low RCE50 values will be able to produce 50% of thesystem’s maximal response at lower drug–receptor complex concentrations than will drugswith high RCE50 values.

Substituting the expression for RC shown in equation (17.1) into equation (17.3), wehave

E = Em · RT · C

RCE50 · Kd + C · (RCE50 + RT )(17.4)

Equation (17.4) is simplified by introducing an efficacy expression known as the trans-duction ratio (� ), which is defined as the ratio of the total concentration of receptors tothe concentration of drug–receptor complex that produces 50% of the maximum response(RT/RCE50). The larger the value of the transduction ratio, the greater is a drug’s efficacy. If,for example, a relatively low concentration of the drug–receptor complex produces a 50%maximum response (RCE50 is small), the transduction ratio (� ) is large.

Substituting for the transduction ratio in equation (17.4), the response may be ex-pressed as

E = Em · � · C

Kd + C · (1 + � )(17.5)

The parameters of the Emax model (Emax and EC50) can be related to those of the operationalmodel of agonism (OMA). When a drug’s response is maximum (E = Emax), C is high,

Page 348: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

328 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

C � Kd, and the expression in the denominator of equation (17.5) may be modified: Kd +C + C · � = C + C · � :

Emax = Em · �

1 + �(17.6)

A drug’s EC50 is the concentration that produces half of the drug’s maximum response.Substituting for Emax/2 for E and EC50 for C in equation (17.5) yields

Emax

2= Em · � · EC50

Kd + EC50 · (1 + � )(17.7)

Substituting for Emax from equation (17.6), and rearranging, we have

EC50 = Kd

1 + �(17.8)

Full agonists with high intrinsic efficacy can produce a large response at relatively lowconcentrations (Figure 16.3). They will have low RCE50 values and, as a result, hightransduction ratios. The denominator in equation (17.6) will then simplify to 1 + � ≈� , and the drug’s Emax will be the system maximum. For example, if � = 99, EC50 =Kd/100, and Emax = (99% Em). The fact that the EC50 is much less than the Kd (the drugconcentration when 50% of the receptors are occupied) illustrates that the drug has highefficacy. This drug, which is able to produce the full system response, can produce half themaximum response when only about 1% (1/99 × 100%) of the receptors are occupied. Ifa drug has a low � (e.g., 0.01), EC50 is approximately equal to Kd and the maximum effectis about 1% of Em (4). When � = 1, the agonist produces half of the system’s maximumresponse at 100% receptor occupancy, and this constitutes the drug’s Emax [Emax = Em · � /(1 + � ) = Em/2]. This would occur with a partial agonist or in a tissue that either had a lowconcentration of receptors or one in which the receptors had become desensitized.

This model can also incorporate a slope factor (n) similar to the slope factor of thesigmoidal Emax model. The model equations that incorporate this factor are

E = Em · � n · Cn

(Kd + C)n + Cn · � n(17.9)

Emax = Em · � n

1 + � n(17.10)

EC50 = Kd

(2 + � n)1/n − 1(17.11)

In clinical studies, the values of Em and n can be determined using a full agonist and thesigmoidal Emax model. The response–concentration data can then be modeled to determine� and Kd. The OMA has been used extensively in studies on the �-opioid (MOP) agonists.It was used to successfully simulate the effects of a series of MOP agonists in rats usingvalues of the affinity and efficacy determined in vitro, in conjunction with system-specificparameters (Em and n), (5). Determination of the transduction ratio in vivo has enabled therelative efficacies of some MOP agonists to be determined in mice [see (6)]. These studiesfound an efficacy of fentanyl � etorphine � methadone = morphine � hydromorphone

Page 349: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

PHYSIOLOGICAL TURNOVER MODEL AND ITS CHARACTERISTICS 329

� oxycodone � hydrocodone. In another study conducted in rats, the development oftolerance to alfentanil was associated with large increases in the drug’s EC50. Theoretically,this could be explained by either a decrease in affinity or a decrease in efficacy. Simulationsperformed using the OMA demonstrated that the observations could be explained by a 40%loss of functional MOP receptors (7), which indicated that normally the system has a verylarge receptor reserve (5,7). Since drug-specific parameters have been found to be consistentbetween species, the OMA is expected to be of great value in translational research wherestudies conducted in vitro or in animal models generate drug-specific parameters, whichcan then be applied to humans (1,8).

17.2.1 Simulation Exercise

Open the model “Operational Model of Agonism” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17a

The model has the following default parameters: RCE50 = 5 mg/L, RT = 200 units, Kd =10 mg/L, and Em = 100%. A problem based on the OMA (Problem 17.1) reviews theproperties and behavior of the model.

17.3 PHYSIOLOGICAL TURNOVER MODEL AND ITS CHARACTERISTICS

The direct action of a drug can be modeled using the sigmoidal Emax model, any of its derivedmodels (Emax and linear models), or the operational model of agonism. Mechanism-basedPK–PD models address the events that occur along the response chain subsequent to thereceptor activation and the generation of the initial stimulus (Figure 17.1). For example,models have been developed to address long transduction processes, the developmentof tolerance, indirect drug effects, irreversible drug effects, and the impact of drugs onan underlying disease that changes over time (disease progression models). A variety ofdifferent models have been developed to address these issues, and many incorporate aphysiological turnover model for the synthesis and degradation of a biological quantity orfactor (Figure 17.3). These turnover models were first introduced into pharmacodynamics

R

Zero-OrderFormation

Rate In = kin

First-OrderLoss

Rate Out = R•kout

At equilibrium, R = R0, and Rate In = Rate Outkin = R0•koutR0 = kin/kout

FIGURE 17.3 Turnover model for the formation and loss of a biological factor or response variable(R). Its formation is assumed to be a zero-order process (kin), and loss is assumed to be first orderwith a rate constant of kout.

Page 350: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

330 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

with the indirect effect models (9–12) and have subsequently been incorporated into othermodels in diverse and creative ways. In pharmacodynamic models, the biological factor (R)that is being produced in the turnover model is an entity that is usually directly involved inmediating the biological response. As a result, it is often referred to as the response variable.In models, R has been a variety of different things, including the amount or concentrationof an endogenous compound, an enzyme, white blood cells, red blood cells, and gastricacid secretion. Most commonly, formation of the biological quantity is assumed to be azero-order process with a rate constant of kin. Loss or degradation of R is assumed to be afirst-order process with a rate constant of kout.

The physiological system for the production and degradation of the biological factor isshown in Figure 17.3. The assumptions and characteristics of the system are as follows:

� It is assumed that the precursor pool for the biological factor is very large and is neverdepleted. As a result, it is assumed to be formed by a constant continuous (zero-order)process: rate = kin.

� The factor is assumed to be degraded or removed by a first-order process, the rate ofwhich is dependent on the value of the factor and a first-order rate constant kout.

� The rate of change of R with time is given by

dR

dt= kin − kout · R (17.12)

where R is a biological quantity, kin is a zero-order rate constant for the formation ofR, and kout is a first-order rate constant for the loss of R.

� Under normal circumstances, the system is stationary and at equilibrium. The biolog-ical factor (R) will be constant and have a baseline level of R0. At baseline,

rate of production = rate of degradation

kin = R0 · kout (17.13)

Rearranging yields

R0 = kin

kout(17.14)

17.3.1 Points of Drug Action

External factors such as drugs and/or disease can interfere with the physiological systemthrough actions on kin and/or kout, or by a direct effect on R. These actions would destroythe equilibrium and produce changes in R. An inhibition of kin, a stimulation of kout, ordirect destruction of the biological factor would all result in a decrease its value. In contrast,stimulation of kin or inhibition of kout would increase the value of the biological factor.

17.3.2 System Recovery After Change in Baseline Value

Assuming that the action of the external influence is only temporary, once the influ-ence is removed, the system would return to equilibrium and its normal baseline level.

Page 351: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

INDIRECT EFFECT MODELS 331

0.00 12.00 24.00 36.00 48.00 0.00 12.00 24.00 36.00 48.00–0.5

–0.3

0.0

–0.5

–0.3

0.0

R0

– R

R0

R0

– R

R0

Time (h) Time (h)

kout decreasing from 0.2, 0.1, and 0.05 h–1

Values of kin of 5,10, and 20 units/h

FIGURE 17.4 Influence of the values of the rate constants on the time it takes a system to restoreequilibrium when the baseline is reduced by 50%. Values of kout of 0.05, 0.1, and 0.2 h−1 were used,and kin was set to 10 units/h. Values of kin of 5, 10, and 20 units/h were used and kout was fixed at0.1 h−1. Response is fractional change from baseline.

Figure 17.4 shows how the values of kin and kout influence the return to baseline after anexternal influence reduced the response variable to 50% of its normal baseline value. Itcan be seen that the time for the system to restore equilibrium is dependent on kout andindependent of kin. Since the loss of the biological factor is a first-order process, it takesabout 4 degradation (kout) half-lives for equilibrium to be restored (Appendix B). The sys-tem is analogous to the kinetics of an intravenous infusion, where the time to steady stateis dependent on a drug’s elimination half-life and independent of the infusion rate and thevalue of the steady-state plasma concentration. In common with the infusion model, thetime it takes for the system to restore equilibrium is also independent of the magnitude ofthe initial change. Thus, if the baseline is disrupted by 20, 50, or 70%, it will still take 4degradation (kout) half-lives for equilibrium to be restored.

In summary:

� The time it takes the system to restore the usual baseline level is dependent on thevalue of kout. As a first-order process, it will take 3 to 5 kout half-lives for the systemto return to baseline.

� The time it takes the system to restore the usual baseline level is independent of thevalue of kin.

� The time it takes the system to restore the usual baseline level is independent of themagnitude of the change from baseline.

17.4 INDIRECT EFFECT MODELS

The product of a physiological system such as the one discussed above may be a naturalligand (R) that is directly responsible for a physiological response. For example, R couldbe gastric acid production, a substance that clots blood, or a low-density lipoprotein. Adrug could exert its action by affecting the concentration of R. It could reduce R either byinhibiting its formation or by stimulating its degradation. Alternatively, it could increaseR by either stimulating its production or inhibiting its degradation. Such drugs are saidto act indirectly. Warfarin is an example of a drug that acts indirectly. Warfarin is usedtherapeutically to inhibit the clotting of blood, but it does not have any direct effect on thisprocess. Instead, it inhibits the synthesis of clotting factors that play an integral part in the

Page 352: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

332 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

RR0 = k in /kout

Zero-OrderFormation

kin

First-OrderLoss

R• kout

Stimulate (S)Model III

Inhibit (I)Model I

Stimulate (S)Model IV

Inhibit (I)Model II

FIGURE 17.5 Four indirect effect models. Drugs could inhibit kin or kout (models I and II, respec-tively) or stimulate kin or kout (models III and IV, respectively).

clotting of blood, and it is the concentration of these clotting factors (response variables)that is directly related to the action of warfarin.

Pharmacodynamic models for drugs that act indirectly need to incorporate, in addition tothe model for the drug’s direct action (e.g., Emax model), the physiological turnover systemdescribed above. The four potential ways that drugs can interfere with the physiologicalturnover models [inhibition or stimulation of either kin or kout (Figure 17.5)] give rise tofour basic indirect models (9):

� Inhibition of kin: model I� Inhibition of kout: model II� Stimulation of kin: model III� Stimulation of kout: model IV

The direct action of a drug on the physiological system is usually modeled using theEmax or sigmoidal Emax model. To differentiate the four models, the “effect” is categorizedas either stimulation (S) or inhibition (I), and it is measured as the fractional change in therate constant affected. Thus, the stimulatory (S) or inhibitory (I) effect of 0.3 would mean a30% increase or a 30% decrease, respectively, in the rate constant affected. The equationsfor the direct effect of the drug are

I = Imax · Cp

IC50 + Cp(17.15)

S = Smax · Cp

SC50 + Cp(17.16)

where I and S represent the fractional change in the rate constant, Imax is the maximalpossible fraction inhibition that a drug can produce [(Imax can vary between zero (noinhibition) and 1 (complete inhibition)], IC50 the drug concentration that produces 50%of Imax, Smax the maximal fraction stimulation that a drug can produce (Smax can be anynumber greater than zero), and SC50 the drug concentration that produces 50% of Smax. Cpis the plasma concentration, which is assumed to drive the direct effect.

The action of the drug on kin or kout is then incorporated into the differential equationfor the physiological model [equation (17.12)]. For example, for model I, the inhibition of

Page 353: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

INDIRECT EFFECT MODELS 333

Zero-OrderFormation

kin

First-OrderLoss

I: Model I S: Model III I: Model III S: Model IV

max •

50

I CpI

IC Cp=

+

max •

50

S CpS

SC Cp=

+

•in outdR

k k Rdt

= −

I. II.

III. IV.

(1 ) •in outdR

k I k Rdt

= − −

(1 ) •in outdR

k S k Rdt

= + −

(1 )•in outdR

k k I Rdt

= − −

(1 )•in outdR

k k S Rdt

= − +

RR0 = k in /kout

R• kout

FIGURE 17.6 Equations for the four indirect models. The rate of change of the response variable(R) with time is equal to the rate in minus the rate out. The direct effect of the drug to either inhibit(I) or stimulate (S) kin or kout is modeled using the Emax model. The direct effect is expressed in termsof the fraction decrease (inhibition) or increase (stimulation) in the rate constant affected.

kin, the inhibitory action of the drug, would be incorporated as follows:

dR

dt= kin(1 − I ) − kout · R (17.17)

Substituting I from equation (17.15) into equation (17.17), we have

dR

dt= kin

(1 − Imax · Cp

IC50 + Cp

)− kout · R (17.18)

The equations for all four indirect effect models are shown in Figure 17.6. It is not possibleto solve the differential equations to obtain explicit solutions. As a result, simulations areextremely useful in understanding the characteristics of indirect effect models.

17.4.1 Characteristics of Indirect Effect Drug Responses

Indirect effect models have been used extensively to model the response of a variety ofdrugs, including the action of warfarin on the synthesis of clotting factors; the inhibitoryaction of H2-blockers on acid secretion; the inhibition of water reabsorption by furosimide,bronchodilation produced by �2-agonists, and the action of terbutaline in reducing potas-sium concentrations (13).

In two of the four models the drug reduces the response variable (inhibition of kin andstimulation of kout), while in the other two models the drug increases the response variableabove its baseline (stimulation of kin and inhibition of kout). Figure 17.7 shows the typicalresponse profiles from the four indirect effect models at three dose levels. The data weresimulated by connecting a one-compartment model with intravenous bolus input to eachof the four indirect effect models. Two special characteristics of these models are clearly

Page 354: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

334 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

–80

–60

–40

–20

0

0 12 24 36

(R –

R0/

R0)

•100

0

20

40

60

80

100

120

140

0 12 24 36

0

50

100

150

200

250

300

0 12 24 36–80

–60

–40

–20

0

0 12 24 36

(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

Time (h) Time (h)

Time (h)Time (h)

Model IInhibition of kin

Increasing Dose

Increasing Dose

Increasing Dose

Increasing Dose

Model IIIStimulation of kin

Model IVStimulation of kout

Model IIInhibition of kout

FIGURE 17.7 Response (percent change from baseline) profiles of the four indirect effect modelsafter a series of intravenous bolus injections. Simulations were performed using doses of 10, 100,and 1000 mg in each of the four models. The models had the following parameters: Vd = 20 L, k =0.7 h−1, kin = 10 units/h, kout = 0.2 h−1, Imax (models I and II) = 1, Smax (models III and IV) = 5,IC50 (models I and II) = 0.1 mg/L, SC50 (models III and IV) = 1 mg/L.

visible in Figure 17.7. First, the response is delayed relative to the plasma concentration(with an intravenous injection, the peak plasma concentration occurs at time zero). Second,although, as expected, the maximum response (Rm) increases with dose, unusually, the timeof the maximum response (TRm ) also increases with dose.

The four models share many characteristics and some have their own unique features. Adetailed discussion of the properties of the models, the selection of an appropriate modelfor a specific drug effect, and the estimation of model parameters are beyond the scopeof this discussion but can be found in the literature (10–12). The discussion below willprovide a general description of the models, with particular emphasis on how the modelparameters affect the time course of drug response (the onset, magnitude, and duration) andhow this affects the design of dosing regimens. The discussion is presented using model I,the inhibition of kin, as the example. Interactive simulation models of all four indirectmodels are provided to allow readers to evaluate the characteristics of all the indirect effectmodels.

17.4.2 Characteristics of Indirect Effect Models Illustrated Using Model I

In the indirect effect model I, the drug inhibits the zero-order rate constant (kin) for theproduction of the response variable. This model has been applied to the action of warfarin,which is used widely to increase the clotting time of blood in patients who are susceptibleto blood clots and at risk for strokes. Warfarin exerts its action by inhibiting the synthesisof clotting factors that are involved in the clotting process. Indirect effect model I results

Page 355: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

INDIRECT EFFECT MODELS 335

in a decrease in the value of the response variable according to the equation

dR

dt= kin

(1 − Imax · Cp

IC50 + Cp

)− kout · R (17.19)

If high concentrations of the drug are able to inhibit kin completely, Imax = 1 and theparameter can be removed from the numerator. Initially, when no drug is present, theresponse is at its equilibrium baseline value (kin/kout) (see Figure 17.3). After an intravenousdose, assuming no distributional delays, the drug will immediately inhibit kin, and thesynthesis of the response variable will fall. Translation of the drug action into a decreasein the response variable itself will depend on the time it takes for the existing responsevariable to be removed, which will depend on the value of kout.

The characteristics of the model will be demonstrated using simulations. A one-compartment model with intravenous bolus input was combined with the indirect effectmodel. The simulations were carried out using the following default parameter values:dose = 100 mg, k = 0.7 h−1, Vd = 20 L, IC50 = 0.1 mg/L, Imax = 1, kin = 10 units/h, andkout = 0.2 h−1. Note that for these simulations, k � kout; the loss of the response variable israte limiting (the drug’s elimination half-life is shorter than the half-life for the turnover ofthe response variable). The model used to simulate these figures may be found at

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17b

17.4.2.1 Time Course of ResponseNote that for clarity the elimination half-life of the drug has been set to 1 h. The timecourse of the response can be observed in Figure 17.8; the plasma concentration (dashedline) is at its peak at time zero, but the maximum response does not occur until about 5 h(5 elimination half-lives). At this time, the drug has been almost completely removed fromthe body. For drugs with indirect action, the time course of the drug action extends beyondthe drug’s presence in the body. In this example, it takes about 16 h (16 drug elimination

0

10

20

30

40

50

60

0

1

2

3

4

5

6

0 10 20 30 40

Cp

(mg/

L)

Res

pons

e

Time (h)

FIGURE 17.8 Plasma concentration (dashed line) and response (solid line) after an intravenousbolus injection (100 mg). Simulations were carried out using parameter values for model I given inthe legend of Figure 17.7.

Page 356: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

336 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

half-lives) for the baseline to return to about 90% of its original value. Recall from Section17.3.2 that once the external force is removed, it takes about 4 kout half-lives for the systemto be restored to equilibrium. In this example the t1/2 of kout is 3.5 h.

17.4.2.2 Effect of DoseIt was shown previously (Figure 17.7) that as the value of the dose increases, the responseincreases. Note, however, that the dose–response relationship is nonlinear. In model I, adose of 10 produces a maximum response (Rm) of about a 25% change from baseline, adose of 100 produces an Rm of about a 50% change from baseline, and a dose of 1000 mgproduces an Rm of about a 70% change from baseline. Also note that the time for themaximum response (TRm ) increases with dose. This is characteristic of these models andcan be used to distinguish delays in response caused by indirect effects from delays causedby a slow distribution of the drug to its site of action. Recall that in the effect compartmentmodel, the time for the maximum response remains constant with dose (see 16.5.1). Thetime of the maximum effect in the indirect models increases with dose because larger dosesproduce greater saturation of the system (e.g., receptors) and, as a result, the duration of thedirect action of the drug is longer (see Figure 16.12b). Because kin is inhibited for a longerperiod at higher doses, the peak or maximum response occurs later. Thus, the duration ofaction of action increases with dose.

17.4.2.3 Maximum Response and Maximum Achievable Response of a DrugAt the peak response (Rm), the rate of production of the response variable equals the rateof removal, and the rate of change of R with time is momentarily zero:

dR

dt= 0 (17.20)

Substituting into (17.19) and rearranging gives us

Rm = kin

kout

(1 − Imax · Cp

IC50 + Cp

)(17.21)

Substituting for kin/kout according to equation (17.14) yields

Rm = R0

(1 − Imax · Cp

IC50 + Cp

)(17.22)

As the dose increases the drug’s effect (I) approaches Imax and Rm approaches the maximumeffect the drug can achieve (Rmax), since Cp � IC50, IC50 + Cp ≈ Cp and:

Rmax = R0(1 − Imax) (17.23)

If a drug has the maximum possible value of Imax (1), Rmax tends to zero and the responseexpressed as the percentage change from baseline tends to 100%. Thus, the maximumresponse possible in model I is zero.

Page 357: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

INDIRECT EFFECT MODELS 337

17.4.2.4 Influence of Physiological System ParametersWhen the physiological turnover system was studied in isolation earlier in the chapter, itwas shown that the time for complete recovery of the system after its equilibrium has beendisturbed, is dependent on kout and independent of kin. The independence of recovery onkin for the indirect effect model is shown in Figure 17.9a, which shows the time course ofthe therapeutic response (expressed as a percentage change from baseline) at three valuesof kin. The influence of kout on the system is shown in Figure 17.9b. Note that kout controlsboth the onset of action and the recovery of the system. The larger the value of kout, thefaster the onset, the shorter TRm and the faster the recovery. This has important implicationsfor dosing regimen design. Small values of kout will result in a long duration of action andwill enable the drug to be administered less frequently. When the kout is 0.05 h−1 (kout

t1/2 = 14 h), the action of the drug persists for over 36 h. Thus, this drug could easily beadministered daily, even though it has an elimination half-life of 1 h.

Figure 17.9c shows the influence of kout in a system where kout � k (elimination is ratecontrolling). Note that the time scale of this plot has been expanded to 72 h. Under thesecircumstances (k � kout), kout has little influence on the duration of action. Elimination is arate-limiting process and it controls the duration of action. The dosing interval of the drugwould then be based on the pharmacokinetic not the pharmacodynamic characteristics ofthe drug.

–100

–80

–60

–40

–20

0

0 12 24 36 48 60 72–80

–60

–40

–20

0

0 12 24 36

–60

–40

–20

0

0 12 24 36–80

–60

–40

–20

0

0 12 24 36

Time (h) Time (h)

Time (h)Time (h)

Varying kin

Varying kout: k < kout

Varying k: k > kout

Varying kout: k > kout

kout (h–1)

0.05

0.1

0.2

kout (h–1)

0.20.4

0.8

k (h–1)1.4

0.7

0.35

(a) (b)

(c) (d)

(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

FIGURE 17.9 Effect of variability in the three rate constants on a response profile in indirect effectmodel I after IV bolus administration. Response is the percent change from baseline. In part (a), kin

was varied (5, 10, and 20 units/h); in part (b), kout was varied (0.05, 0.1, and 0.3 h−1), with k at itsdefault value (0.7 h−1); in part (c), kout was varied (0.2, 0.4, and 0.8 h−1) with k set to 0.1 h−1); and inpart (d), k was varied (0.35, 0.7, and 1.4 h−1). The dose was set to 100 mg. Unless otherwise stated,all other parameters were set to the default values given in the legend of Figure 17.7.

Page 358: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

338 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

15

35

55

0 12 24 36

R

10

70

0 12 24 36

decreasing kin from 7.5,10, and 12.5 units/h

increasing kout from 0.15,0.2, and 0.3 1/h

R

Time Time(a) (b)

50

30

FIGURE 17.10 Effect of changes in kin and kout on the absolute value of the response variable (R).The values of the rate constants that were varied are given in the figure. All other parameter valueswere set to their default values given in the legend of Figure 17.7.

The initial values of kin and kout determine the baseline value of the biological variable.As a result, when the initial value of either parameter is altered, the baseline and the absolutechange in R will be affected (Figure 17.10). It can be seen that smaller values of kin areassociated with lower baseline values of R. The response curve is shifted downward andthe maximum response is greater, but recall from Figure 17.9a that the relative change in Ris the same for all values of kin. It can be seen that as the initial value of kout increases, thebaseline decreases and the response curve is shifted downward. A given dose produces alarger maximum response, which occurs earlier (recall from Figure 17.9b that larger initialvalues of kout also produce larger relative responses). The smaller the value of kout, thelonger it takes to return to baseline.

17.4.2.5 Influence of Pharmacokinetics: Elimination Rate ConstantThe elimination rate constant is a measure of the rate of drug elimination. It can be seen(Figure 17.9d) that slower elimination (smaller values of the elimination rate constant)results in increased maximum responses, an increase in TRm , and a more prolonged durationof action of the drug. Slower elimination results in a more prolonged inhibition of kin,which will result in a more profound, longer-lasting therapeutic effect. The value of k doesnot affect the onset of response (the initial slope).

17.4.2.6 Pharmacodynamic Parameters of the Drug: Effect of IC50 and Imax

Variability in IC50 is essentially the same as variability in the dose. A decrease in thevalue of IC50 (the potency of the drug increases) is equivalent to an increase in the dose.Thus, when IC50 decreases, the response and the time for maximum response increase(Figure 17.11a). In contrast, as the Imax value increases, the response increases but thetime for maximum response remains the same (Figure 17.11b). Note that variability in Imax

produces proportional changes in Rm.

17.4.2.7 Time to Steady State During an Intravenous InfusionThe time to steady-state response during a constant, continuous intravenous infusion isshown in Figure 17.12. Figure 17.12a shows the response over time for model I and, forcomparison, Figure 17.12b shows the response for model IV (stimulation of kout), which

Page 359: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

INDIRECT EFFECT MODELS 339

–80

–60

–40

–20

0

0 12 24 36–60

–40

–20

0

0 12 24 36Time (h) Time (h)

(a) (b)

IC50 (mg/ L)

1.0

0.1

0.01

Imax0.25

0.5

1.0(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

FIGURE 17.11 Effect of variability in IC50 (a) and Imax (b) on the response profile of indirecteffect model I after IV bolus administration. Response (percent change from baseline) was simulatedusing values of IC50 of 0.01, 0.1, and 1 mg/L (a) and values of Imax of 0.25, 0.5, and 1 (b). A doseof 100 mg was used, and unless otherwise stated, all other parameters were set to the default valuesgiven in the legend of Figure 17.7.

also produces a reduction in the response variable below baseline. Note that for model I(Figure 17.12a) the time to steady-state response is about the same for the various infusionrates, and it is approximately 24 h. In contrast, the time to steady-state response in model IVdecreases as the infusion rate increases (Figure 17.12b). This is because the value of kout

controls the time to steady-state response. In model I, kout does not vary with dose; it isconstant (0.2 h−1, kout t1/2 = 3.5 h), and by about 7 kout half-lives (24.5 h), the response is atsteady state. In contrast, kout is the subject of the drug’s action in model IV. Larger infusionrates will produce larger stimulations of kout, and as a result (kout t1/2 decreases), it will takeless time to reach steady state. If these data are available, the time to steady-state responsefrom different infusion rates can be used to distinguish models I and IV. Simulations usingthe other indirect effect models will demonstrate that the time to steady state is constantfor model III (stimulation of kin) but varies with the infusion rate for model II. Specifically,for model II (inhibition of kout), as the infusion rate increases, a greater inhibition of kout isobserved and the time to steady-state response increases.

–100

–80

–60

–40

–20

0

0 12 24 36 48–80

–60

–40

–20

0

0 12 24 36Time (h) Time (h)

(a) (b)

1 mg/h

4 mg/h

20 mg/h

1 mg/h

4 mg/h

20 mg/h(R –

R0/

R0)

•100

(R –

R0/

R0)

•100

FIGURE 17.12 Effect of the infusion rate on the time to steady state response in indirect responsemodels I (a) and IV (b). Parameter values for the models are those given in the legend of Figure 17.7.

Page 360: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

340 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

TABLE 17.1 Default Parameters Used in the Simulation Models for the Four IndirectEffect Models

Parameter Model I Model II Model III Model IV

Dose (mg) 100 500 500 500k (h−1) 0.7 0.7 0.7 0.7Vd (L) 20 20 20 20kin (units/h) 10 10 10 10kout (h−1) 0.2 0.2 0.2 0.2Imax or Smax 1 1 5 5IC50 or SC50 (mg/L) 0.1 0.1 1 1

17.4.3 Other Indirect Models

The characteristics of the three other models can be investigated by computer simulations.The default parameter values of the simulation models are shown in Table 17.1, and theindividual models can be found at the following sites:

� Model I: http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17b� Model II: http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17c� Model III: http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17d� Model IV: http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17e

A summary of the characteristics of the four models is shown in Table 17.2, which alsoshows the limiting value of the maximum achievable response (Rmax) in the four models.These values were identified using the same approach for model I as presented in equations(17.20) to (17.23).

17.5 TRANSDUCTION AND TRANSIT COMPARTMENT MODELS

Transduction refers to the process and steps involved in the conversion of drug–receptorinteraction into a measured biological response. During transduction the stimulus that isgenerated from the drug–receptor interaction is relayed along a sequence of cascadingevents that may involve G-protein activation, second messengers, ion-channel activation,and/or gene transcription. If the process is slow, it may become rate limiting and theremay be a significant time lag between the changes in response and changes in the plasmaconcentrations of the drug. The delayed response–time profile that occurs as a result oftransduction can be captured using a series of transit compartments to relay or transferthe initial stimulus (14). Generally, it is not possible to determine either the number oftransduction steps or the duration of each step. Thus, the number of transit compartmentsand the time for the response to move between compartments is modeled empirically to fitthe response data. A model with three transit compartments is shown in Figure 17.13.

Generation of the initial stimulus or biosignal (E) that results from interaction of a drugwith its receptors can be modeled using any of the models presented previously, such asthe Emax model (Figure 17.13). If it is assumed that there is rapid distribution of the drugto its site of action, plasma concentrations can be used as the driving force in the equation.

Page 361: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

TRANSDUCTION AND TRANSIT COMPARTMENT MODELS 341

TABLE 17.2 Summary of the Characteristics of the Four Indirect Response Models

Model I:Inhibition: kin

Model IIInhibition: kout

Model IIIStimulation: kin

Model IVStimulation: kout

As dose ↑: Response ↑TRm ↑

Response ↑TRm ↑

Response ↑TRm ↑

Response ↑TRm ↑

As kin ↑ R0 ↑Relative R ↔

R0 ↑Relative R ↔

R0 ↑Relative R ↔

R0 ↑Relative R ↔

As kout ↑ R0 ↓Relative R ↑Time of onset ↓Duration ↓

R0 ↓Relative R ↑Time ofonset ↓Duration ↓

R0 ↓Response ↑Time of onset ↓Duration ↓

R0 ↓Response ↑Time of onset ↓Duration ↓

As k ↑ Response ↓TRm ↓Duration ↓

Response ↓TRm ↓Duration ↓

Response ↓TRm ↓Duration ↓

Response ↓TRm ↓Duration ↓

As IC50/SC50 ↑ Response ↓TRm ↓

Response ↓TRm ↓

Response ↓TRm ↓

Response ↓TRm ↓

As Imax/Smax ↑ Response ↑TRm ↔

Response ↑TRm ↑

Response ↑TRm ↔

Response ↑TRm ↓

Time to steadystate

↔ Dose ↑ Dose ↔ Dose ↓ Dose

Rmax = R0 · (1 − Imax);tends to zero

= R0/(1 − Imax);tends to ∞

= R0 · (1 + Smax) = R0/(1 + Smax)

C + R RCInitial

Stimulus

τττ

E3 E2 E1

max •CpEE

EC50 + Cp =

τResponse

FIGURE 17.13 Transit compartment model with three transit compartments. In this model, themagnitude of the initial signal (E) is estimated from the Emax model. The signal is then transferredthroughout a series of transit compartments, each of which has an intercompartmental transit timeof � .

The stimulus is then transmitted throughout the transit compartments. These models areusually parameterized for the mean transit time (MTT) for the entire process and for thenumber of compartments (n). The individual intercompartmental transit times (� ) for thevarious compartments are usually fixed to the same value, MTT/(n + 1). A transit timeis equivalent to the reciprocal of a first-order rate constant, and the transfer of the effectbetween compartments can be modeled using first-order kinetics. Generally the followingequation can be used for the effect (E) in each compartment:

dEn

dt= 1

�· En−1 − 1

�· En = 1

�· (En−1 − En) (17.24)

Page 362: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

342 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

The equations for each compartment are:

dE1

dt= 1

�·(

Emax · Cp

EC50 + Cp− E1

)(17.25)

dE2

dt= 1

�· (E1 − E2) (17.26)

dE3

dt= 1

�· (E2 − E3) (17.27)

where En is the effect in the nth transit compartment and � =MTT/(n + 1) is the intercom-partmental transit time. A power function can be added to the value of the effect in anycompartment to amplify or dampen the effect during transduction (14).

Response data simulated using a four transit compartment model is shown in Fig-ure 17.14. It can be seen that as the response is transferred from one compartment to thenext, the response is delayed, and it is dampened. As it moves through successive com-partments, the peak of the signal is lower and the overall profile becomes wider and moresymmetrical.

Figure 17.15 shows how the number of compartments influences the profile. Thedata were simulated by fixing the total mean transit time (6 h) and using between oneand four transit compartments. The number of compartments has the clearest impact onthe time of the final response. Even though to total mean transit time is the same inthe various simulations (a to d), as the number of compartments increases, the delay in thefinal response increases. Also, as the initial stimulus is divided into an increasing numberof compartments, the profiles in equivalent compartments (e.g., compartment 2 of twocompared to compartment 2 of four) become sharper, larger, and the peak occurs sooner.As a result, when a large number of compartments are used, the final response can be largerthan that obtained in a model with fewer compartments. For example, in Figure 17.15, the

0

20

40

60

0 2 4 6 8 10 12 14 16 18

% M

axim

um R

espo

nse

Time

1st

2nd

3rd

4th

FIGURE 17.14 Value of the signal as it is transferred through four transit compartments. The datawere simulated using an IV bolus injection (dose = 100 mg, Vd = 20 L, k = 0.7 h−1) with a direct linkto an Emax model (EC50 = 1 mg/L, Emax = 100). The initial signal was then transferred throughoutfour transit compartments with a total transit time of 6 h and an intercompartmental transit time of6/5 = 1.2 h.

Page 363: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

TRANSDUCTION AND TRANSIT COMPARTMENT MODELS 343

0

20

40

60

0 2 4 6 8 10 12 14 16 18

0

20

40

60

0 2 4 6 8 10 12 14 16 18

0

20

40

60

0 2 4 6 8 10 12 14 16 18

1st

3rd

1st

2nd

1st

4th

2nd

2nd

3rd

% M

axim

um R

espo

nse

% M

axim

um R

espo

nse

% M

axim

um R

espo

nse

% M

axim

um R

espo

nse

TimeTime

TimeTime(a) (b)

(c) (d)

0

20

40

60

0 2 4 6 8 10 12 14 16 18

FIGURE 17.15 Effect of the number of transit compartments on the shape of the final responseafter an IV bolus injection. Values of the dose, pharmacokinetic and pharmacodynamic are the sameas those in the legend of Figure 17.14. The total mean transit time was maintained constant at 6 h,and response was generated using one (a), two (b), three (c), and four (d) transit compartments, withintercompartmental transit times of 3, 2, 1.5, and 1.2 h, respectively. The solid line represents thefinal response.

peak response in the four-compartment model is 37%, whereas that in the two-compartmentmodel is 35%. The model for the hematological toxicity of anticancer drugs that is presentedin Section 17.7.2 is an example of a transit compartment model.

17.5.1 Simulation Exercise

Open the model “Transit Compartment Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17f

The model consists of a pharmacokinetic model of a one-compartment model (Vd =20 L, k = 0.7 h−1) with intravenous bolus input. The generation of the stimulus is mod-eled using an Emax model (Emax = 100, EC50 = 1 mg/L) linked to Cp. The stimulus istransferred through a series of transit compartments. For each model the intercompart-mental transit time, and the transit time for the dissipation of the final response, are allthe same and equal to the total mean transit time/(n + 1), where n is the number oftransit compartments. The default value of n and the total mean transit time are 4 and6 h, respectively.

1. Explore the model and note its structure.

2. Go to the “Response Profiles” page. Choose a dose and note the time profilesof the plasma concentration, the initial stimulus, and the response in the differenttransit compartments.

Page 364: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

344 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

Observe:� Cp falls monoexponentially from its maximum value at time zero.� There is no delay in the generation of the initial stimulus; its maximum value

corresponds to the maximum value of the plasma concentration at time zero.The stimulus decreases as plasma concentrations decay.

� Transit compartments produce a delay in response. The response in eachcompartment starts at zero and gradually increases to reach a peak and thendecreases.

� The time of the peak response increases with each successive compartment.Note that the peak of the final response in compartment 4 occurs at around6 h, by which time all the drug will have been eliminated (t1/2 = 1 h).

� The compartments have a dampening effect on the response. This dampeningeffect increases as the response moves through the compartments. Thus,the maximum response decreases and the profile widens and becomes moresymmetrical in each successive compartment.

3. Go to the “Effect of Dose” page. Perform simulations with the default model withfour compartments and a total mean transit time of 6 h. Choose doses of 10, 100,and 1000 mg. Notice that the maximum response is not proportional to dose. Thetime of the maximum response increases with dose.

4. Go to the “Number of Compartments (No. Comp.) and MTT” page.Observe in turn how:� The total mean transit time affects the profile when the number of compartment

are maintained constant.� The number of compartments affects the profile when the total mean transit

time is held constant.� Probe different ways to obtain a peak response at 4 h.

17.6 TOLERANCE MODELS

Tolerance may be defined as a process that results in a reduction in the response to a specificdrug concentration following repeated drug exposure. This definition of tolerance excludespharmacokinetics tolerance, which can, for example, arise with a drug that induces its ownmetabolism. Only pharmacodynamic-based tolerance is addressed in this section. A modelfor the time course (onset and duration) and the magnitude of tolerance would be useful touse to probe potential modifications of dosing regimens to minimize its impact as well asto estimate the type of dose adjustments that may be necessary to overcome its effect.

Pharmacodynamic-based tolerance can develop through several mechanisms, one ofwhich is the production of a compensatory or counter-regulatory response called homeo-static tolerance. Tolerance can also occur due to changes in the chain of events betweenreceptor activation and the production of a response. These include desensitization of thereceptors, down-regulation of the receptors, and the depletion of endogenous compounds,such as second messengers, that play an important role in mediating the response.

Owing to the complex underlying biological processes associated with the develop-ment of tolerance, most approaches to modeling tolerance use empirical models that at-tempt to explain the observations. Some very simple models for receptor down-regulationand desensitization have been created using decay and growth functions. Thus, receptor

Page 365: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

TOLERANCE MODELS 345

down-regulation has been modeled by adding a decay function (e−kt) to the Emax param-eter of the sigmoidal Emax model. Desensitization has been modeled by adding a growthfunction to the drug’s EC50.

17.6.1 Counter-regulatory Force Model

A common approach that has been used to model tolerance is to assume that the drug or itsresponse stimulates the production of a counter-regulatory compound or force. The lattercan be modeled to either produce the opposite effect of the drug or to act as an antagonist ora partial agonist to the drug. This approach has been used to model tolerance from nicotine,nitroglycerin, caffeine, and morphine (15–18). The specific details of the models used fordifferent drugs vary somewhat, but the model used for the development of the tolerancefor nicotine represents a clear example. Tolerance of the chronotropic effect of nicotinewas assumed to be the result of the formation of a hypothetical nicotine antagonist (15).This antagonist or tolerance mediator is assumed to be formed from the drug in the centralcompartment, but since it is only hypothetical, its production does not deplete or in anyway influence the amount of drug in the central compartment. The rate of production ofthe mediator is assumed to be first-order, driven by the plasma concentration of the drug(Figure 17.16). The mediator is assumed to be lost by a first-order process:

dCm

dt= Cp · k1m − Cm · km0 (17.28)

where Cm is the concentration of mediator or counterregulatory compound, k1m is a first-order rate constant for the formation of the mediator, and km0 is the first-order rate constantfor its destruction.

Often, k1m and km0 are set equal to each other, and as result, at steady state, the concen-tration of the hypothetical mediator is the same as the plasma concentration of the drug.This model is very similar to the effect compartment model presented in Chapter 16. Incommon with the effect compartment model, the rate constant for the loss of the mediatorcontrols the onset and dissipation of tolerance. Thus, it will take 3 to 5 t1/2,km0 for toleranceto reach its maximum value at a given drug concentration, and it will take the same time todissipate once the drug has been withdrawn.

Any of the pharmacodynamic models discussed previously, such as the a sigmoidal Emax

model, can be used for the direct action of the drug. The plasma concentration could beused as the driving force for the response, or an effect compartment could be incorporated.The model for tolerance to the chronotropic action of nicotine did not incorporate an effectcompartment and used a linear pharmacodynamic model on the assumption that the nicotineconcentration is always much less than the EC50:

E = E0 + S · Cp (17.29)

where E is the heart rate, E0 the resting heart rate, Cp the plasma concentration of nicotine,and S the slope factor.

The pharmacodynamic model is then modified to account for the action of the modifier(Figure 17.16). In the nicotine example the modifier was modeled as a noncompetitiveantagonist. Thus, the overall effect of the drug was expressed as

E = E0 + S · Cp

1 + Cm/Cm50(17.30)

Page 366: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

346 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

Antagonistor

Mediator Cm

Cp•k1m Cm•km0

0501+ Cm/Cm

S•CpE = E +

dCm

dt= Cp•k1m−Cm•km0

FIGURE 17.16 Pharmacodynamic model for tolerance. Tolerance is assumed to be produced bythe action of a hypothetical metabolite of the parent drug. The metabolite is assumed to be producedand removed by first-order processes driven by the concentration of the parent drug (Cp) and thehypothetical metabolite (Cm), respectively. The rate constant for formation (k1m) and loss (km0) areusually set equal to each other. The effect of the drug is modeled as the change from baseline (E0)using linear model with a slope of S. The metabolite is assumed to act as an antagonist, and Cm50 isthe steady-state concentration of the metabolite that produces 50% maximum tolerance. This is themodel used for the development of tolerance to nicotine. [From Ref. (15).]

where E is the net effect, Cm the concentration of the counter-regulatory mediator, and Cm50

the steady-state concentration of the mediator that produces 50% maximum tolerance.The pharmacokinetic parameters are estimated from the plasma concentration data. The

pharmacodynamic parameters, S, km0, Cm50, and E0 are estimated from the time course ofresponse. Figure 17.17 shows the plasma concentration and response (heart rate) simulatedfrom the model parameters reported for nicotine (15). Data were simulated after two shortinfusions of 5 mg of nicotine over 30 min. It can be seen in Figure 17.17a that as a resultof accumulation, the nicotine concentration after the second dose is higher than that afterthe first. In contrast, and as a result of tolerance, the response after the second dose is lower

50

70

90

0 10 20 30 40 50 C(µg/L)

50

70

90

0

10

20

30

40

50

0 100 200 300 400 500

Hea

rt R

ate

(bpm

)

C (

µg/

L)

Time (min)(a) (b)

Heart Rate

BloodConcentration

FIGURE 17.17 Tolerance to nicotine. The nicotine blood concentration (�g/L) (dotted line) andresponse (heart rate) (solid line) against time are shown after two successive infusions of nicotine(a). Figure 17.17(b) shows response (heart rate) plotted against nicotine blood concentration after thetwo infusions. [Based on data from Ref. (15).]

Page 367: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

TOLERANCE MODELS 347

than that after the first. When response is plotted as a function of nicotine concentration,a circular or clockwise relationship between response and concentration is observed. Thisphenomenon is known as proteresis, which means “comes earlier.” In Figure 17.17b it canbe seen that proteresis becomes apparent on the downward fall in the plasma concentration,when the response at a given concentration is less than it was during the initial upwardmovement in the concentration.

A similar model was used for the development of tolerance to nitroglycerin, but in thiscase the hypothetical mediator was assumed to produce an action (Ec, vasoconstrictor) inopposition to that of the drug (Ed, vasodilatorary) (17). The net effect of nitroglycerin wasthe sum of the vasodilatory and vasoconstrictor effects (Ed + Ec). A comparison of thedifferent approaches to tolerance has been reviewed (19). As discussed in Section 17.2, theOMA has proved to be useful for studying tolerance. For example, it has been used to studythe mechanism behind the development of tolerance to the MOP (7) and to investigate therelationship between efficacy and tolerance (6).

17.6.1.1 Simulation Exercise

Open the model “Tolerance: Counter-Regulatory Force” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17g

The model is based on the published model of tolerance to the cardio-accelerating ef-fects of nicotine (15). The pharmacokinetic model consists of a two-compartment phar-macokinetic model with parameters k10, k12, and k21 of 0.0112, 0.03, and 0.0325 min−1,respectively, and Vc = 114 L. Note that these parameters give an elimination half-lifeof nicotine of about 150 min. The pharmacodynamic model consists of a linear effectmodel [equation (17.29)] with tolerance produced by a hypothetical noncompetitiveantagonist [equations (17.28) and (17.30)]. The pharmacodynamic parameters are asfollows: k1m = km0 = 0.020 min−1, S = 1.31 bpm per �g/L, Cm50 = 7.72 �g/L, and E0 =61.2 bpm. Drug administration is modeled as two short (30-min) intravenous infusionsof 5 mg.

1. Explore the model and review the “Model Summary” page.

2. Simulate plasma concentration and the response. Notice that although the plasmaconcentration increases with the second dose, the effect decreases.

3. Try to increase the value of the second dose to achieve the same response asthat obtained after the first dose.

4. Go to the “km0 and Cm50” page. Probe the influence of km0. Note that as thevalue of km0 increases, tolerance occurs more quickly, is more prominent (theeffect of nicotine is less) and its effect wears off faster, so the fall in response ismore gentle because the effects of nicotine are greater.

5. Conduct simulation changing the value of Cm50. This is the concentration ofthe “antagonist” that produces a 50% effect. Since it is nicotine that drives thetolerance and since the concentration of nicotine at steady state is the same asthat of the antagonist, Cm50 can be considered the steady-state nicotine con-centration that produces 50% tolerance. As Cm50 decreases, tolerance becomesmore prominent at a given drug concentration; the nicotine-induced increase inthe heart rate is smaller.

Page 368: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

348 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

6. Go to the “Change � ” page, which allows the time between the multiple doses tobe altered. Notice that as the dosing interval is increased, the tolerance effect onthe next dose becomes less. The loss of tolerance is controlled by km0, which isabout 1.2 h, and has a half-life of 0.58 h. Thus, tolerance should dissipate in about7 × 0.58 h = 4 h. The elimination half-life of nicotine (about 2 h) also controls theduration of tolerance. Reduce the elimination half-life by increasing the value ofk10. Note that when k10 is increased to large values (rapid elimination), tolerancedissipates about 4 h after a dose.

17.6.2 Precursor Pool Model of Tolerance

Tolerance would occur if a drug’s action is dependent on an endogenous compound (pre-cursor) that gets depleted during the response to the drug. The full action of the drug wouldonly be restored once the amount of the endogenous compound returned to its normalresting value. An indirect response model has been applied to model this phenomenon (20).In previous discussions of the indirect model, it was assumed that the precursor pool forthe production of the response variable was large and could never be depleted. Thus, theproduction of the response variable was modeled as a constant (zero-order) process. In theprecursor pool depletion model for tolerance this assumption is not made. The responsevariable (R) is assumed to be produced from its precursor (P) by a first-order process with arate constant (kp) (Figure 17.18). The loss of the response variable is modeled in the usualway as a first-order process with a rate constant kout.

dR

dt= kp P − kout R (17.31)

The drug is assumed to increase the amount of the response variable by stimulating its rateconstant for production (kp). The direct action (E) is the fractional stimulation of (kp).

dR

dt= kp(1 + E)P − kout R (17.32)

The direct effect of the drug can be modeled using the Emax model:

E = Emax ∗ Cp

EC50 + Cp(17.33)

The usual turnover model is used for the precursor pool (Figure 17.18). The precursor isassumed to be produced by a zero-order process and lost by a first-order process throughits conversion to the response variable:

dP

dt= k0 − Pkp (17.34)

Drug Effect on P: In the absence of the drug, the precursor pool is in equilibrium andP0 = k0/kp. When the drug stimulates kp to increase the response variable, P decreases(Figure 17.19). Once the drug’s action stops, the return of P to equilibrium value (P0) isdetermined by the value of (kp). If a second dose of the drug is administered before theprecursor pool is restored, the second dose will not be able to produce the same magnitudeof effect as the first dose.

Page 369: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

TOLERANCE MODELS 349

RR = P•k /k0 p out

First-OrderLoss

R• kout

PP = k /k0 0 p

Zero-OrderFormation

k0

First Order P• kp

Indirect EffectModel

Turnover Modelfor Precursor

FIGURE 17.18 Precursor pool depletion model of tolerance. The response variable (R) is producedby a first-order process (rate constant kp) from a precursor (P) that gets depleted. The loss of theresponse variable is a first-order process with a rate constant kout. The precursor is formed by azero-order process (k0). The drug stimulates the formation of the response variable by increasing kp

300

350

400

450

500

550

40

60

80

0 12 24 36 48 60

Response

Time (h)

Precursor Pool

Dose Dose Dose

FIGURE 17.19 Precursor model predicted plot of response (solid line) and precursor pool status(dashed line) against time. The simulation was conducted with multiple IV bolus injections (100 mgevery 24 h) and the following parameters: Vd = 20 L, k = 0.4 h−1, k0 = 50 units/h, kp = 0.1 h−1,kout = 1 h−1, EC50 = 1 mg/L, Emax = 1.

Drug Effect on R: In the absence of the drug, R is at its baseline value (R0 = kp ∗ P0/kout).The drug stimulates kp and increases R. When the drug action ceases, R decreases towardsR0 (Figure 17.19). However, bacause of the time it takes P to increase back to P0, R canachieve a value that is less than R0 (Figure 17.19). This is known as rebound. If P has notbeen able to return to its baseline value by the time a second dose is given, the effect fromthis second dose will be less than that after the first (Figure 17.19).

If a drug decreases the response variable, an inhibitory effect on kp can be used in themodel.

17.6.2.1 Simulation Exercise

Open the model “Tolerance: Precursor Pool Depletion Model” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17h

Page 370: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

350 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

The model has the following parameter values: k = 0.4h−1; Vd = 20L; kp = 0.1h−1;k0 = 50 units per h; P0 = k0/kp = 500 units; kout = I h−1; R0 = kp ∗ P0/kout = 50/1 =50 units; EC50 = 1unit/L; Emax = 1; dose = 100 mg; and dosing interval = 24 h.

1. Explore the model. Perform a simulation on the “Response Profile” page. Thedrug stimulates the production of the response variable (R) (response), whichdepletes the precursor pool (P). As the drug effect dissipates, R decreasestowards its baseline value, but it overshoots the original value. This is known asrebound and occurs because P is still depleted. Notice that even by the time asecond dose is given, P is not yet fully restored to its baseline value (500 units).As a result the response to the second dose is less than that to the first.

2. Go to the “Dosing Regimen” page. Note that as the dose increases, the reboundeffect and tolerance are more prominent. Restore the default setting and probe theinfluence of the dosing interval. Note that tolerance and rebound become moreprominent with smaller dosing intervals. When the dosing interval is increased to48 h P is able to return to its original baseline value and tolerance is not observed,but the rebound effect is still present.

17.7 IRREVERSIBLE DRUG EFFECTS

Some drugs bind covalently to their receptors. As a result, the target is destroyed and itsfunction returns only when it has been replaced by newly synthesized product. The targetmay be a protein, DNA, an enzyme, or a cell at any stage of development. The proton pumpinhibitors are examples of drugs that act irreversibly. They bind to and destroy the H+,K+-ATPase pumps in the parietal cells of the gastric mucosa, and normal proton secretion isrestored only when the pumps are replaced by newly synthesized functioning pumps (i.e.,the usual turnover time of the system). For proton pumps the turnover time is over 24 h, andas a result, proton pump inhibitors can be administered daily, even though most of them havevery short elimination half-lives. For example, the duration of action of omeprazole (t1/2 �1 h) is sufficiently long to allow it to be administered daily. Aspirin is another example ofa drug that acts irreversibly. Aspirin reacts covalently with prostaglandin cyclo-oxygenase,an enzyme that is responsible for the formation of thromboxane B2, which in turn promotesplatelet aggregation when platelets become fractured. Aspirin destroys the activity of theenzyme, and as a result it can no longer synthesize thromboxane B2. Aspirin’s action persistsuntil new platelets are synthesized with functional cyclo-oxygenase that can resume thesynthesis of thromboxane. Once again, although aspirin has a very short elimination half-life (around 15 min), the drug can be given daily because of prolonged inhibition ofcyclo-oxygenase.

Cancer chemotherapy also provides examples of irreversibly acting drugs, including thealkylating agents, such as cylcophosphamide and chlorambucil, which bind covalently toDNA, and drugs such as etoposide and topotecan, which destroy developing white bloodcells in the bone marrow. Additionally, some inhibitors of drug metabolism act by bindingirreversibly to, and destroying the activity of, enzymes. Again, the action of the inhibitorcan last long after it has been eliminated from the body, and will persist until the damagedenzyme is turned over and replaced with new, functioning enzyme.

Page 371: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

IRREVERSIBLE DRUG EFFECTS 351

R

Zero-Order Formation kin

First-Order Loss R•kout

C + R Inactive ProductDrug

•Rin out irr

dR

dt= k − R•k −C•k

FIGURE 17.20 Model for an irreversible pharmacological effect. The model consists of a physi-ological turnover model with zero-order formation (kin) and first-order loss (rate constant kout) of theresponse variable (R). The irreversible interaction of the drug with the response variable is driven bythe drug concentration (C) and expressed using the second-order rate constant kirr.

17.7.1 Application of the Turnover Model to Irreversible Drug Action

The physiological turnover model presented in Section 17.3 has been used to model theirreversible effects of several drugs, including proton pump inhibitors. In this applicationthe target of the drug is the biological factor, or response variable, itself (Figure 17.20). Asusual, the response variable is assumed to be synthesized by a zero-order process, with arate constant equal to kin and degraded by a first-order process with a rate constant kout.The irreversible drug effect is modeled by incorporating drug concentration-dependentdestruction of the response variable, R, according to the equation

C + Rkirr−→ product

where C is the drug concentration, R the response variable and target of the drug action,and kirr a second-order rate constant for the concentration-dependent effect of the drug onthe factor.

Based on the model shown in Figure 17.20, an expression can be written for the actionof the drug on the response variable:

dR

dt= kin − R · kout − C · kirr · R (17.35)

This model has been used to model the action of proton pump inhibitor pantoprazoleusing the rate of acid output as the response variable (21). Thus, R was the rate of acidsecretion, kin a zero-order rate constant for the rate of acid production, kout a first-order rateconstant for the natural endogenous degradation of the rate of acid secretion, and C theplasma concentration of the drug. A simulation model has been built for the ficticious drugdisolvprazole based on the published model for pantoprazole.

17.7.1.1 Simulation Exercise

The model developed for pantoprazole (21) was used as the basis of an integratedPK–PD model for the fictitious drug disolvprazole. The characteristics of this model willbe demonstrated through simulation. The model “Irreversible Effects: Disolvprazole”may be found at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17i

Page 372: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

352 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

The pharmacokinetic model consists of a one-compartment model with first-order ab-sorption with the parameters presented previously for disolvprazole: Cl = 12 L/h,Vd = 35 L, F = 0.5, and ka = 3 h−1. The elimination half-life is 2 h. The pharma-codynamic model consists of a physiological turnover model that incorporates drugconcentration–dependent irreversible destruction of the response variable (acid pro-duction). The pharmacodynamic parameters are based on those of pantoprazole, butwere modified slightly when appropriate to match the characteristics of disolvprazolediscussed previously. They are as follows: k in = 0.8 m mol/h/h and kout = 0.03 h−1

k irr = 1 L/mg/h. These compare to the following pharmacodynamic parameters of pan-toprozole: k in = 0.416 mmol/h/h, kout = 0.031 h−1 k irr = 0.751 L/mg/h (21).

1. Review the model.

2. Notice the time course of the plasma concentration and the response. Note that:

a. The maximum response is delayed with respect to the plasma concentration.

b. The effect of the drug persists long after the drug is eliminated.

3. Go to the “Dose and � ” page. For doses of 25, 50, 100, and 200 mg, observethat:

a. Response increases with dose but not in a proportional manner.

b. As the dose increases, the peak effect occurs earlier.

4. Increase � to observe how long the effect from a single dose persists. Note thateven though the drug’s elimination half-life is 2 h, the effect of a single dose lastsover 3 days.

This simulation model is required to answer Problem 17.3.

17.7.2 Model for Hematological Toxicity of Anticancer Drugs

Doses and the duration of therapy for many anticancer drugs, including docetaxel, paclitaxel,etoposide, and topotecan, are limited by drug-induced neutropenia. This is a serious toxicitythat can put patients at risk for serious life-threatening infections. An integrated PK–PDmodel has been developed for the time course of this effect (22) (Figure 17.21). The modelcontains some unique elements as well as components of the models discussed previously.The model is outlined below.

1. A unique part of the model is the formation of neutrophils from the stem cell pool inthe bone marrow. Their formation is assumed to be a first-order process driven by thenumber of precursor cells and a first-order rate constant (kprol). A feedback processis incorporated into the rate of proliferation of the neutrophils based on the baselineneutrophils count (N0) and the number of neutrophils circulating at any time (Nt):

feedback =[

N0

Nt

]n

(17.36)

where n is a power factor. The feedback process becomes operative whenever thecirculating neutrophil count deviates from the normal circulating baseline value.

Page 373: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

IRREVERSIBLE DRUG EFFECTS 353

ττ

Circulation32StemCells

kprol= 1/τ

1

ττ

Drug Effect:Cp•k

N0

Nt

n

kcirc = 1/τ

ktr = 1/τ

( )

Nt

FIGURE 17.21 Pharmacodynamic component of the integrated PK–PD model for the neutropeniabrought about by anticancer drugs. The neutrophils are assumed to be produced in the stem cells.Their prolonged maturation prior to release in the circulation is modeled using three transit com-partments, each of which has the same intercompartmental transit time (� ). The proliferation rate ofthe neutrophils is dependent on the proliferation rate constant (kprol), the number of precursor cellsand a feedback mechanism that is dependent on the baseline neutrophil count (N0), and the numbercirculating at any time (Nt). Feeback is equal to (N0/Nt)n, where n is a power function. Drug toxicity ismodeled as an action on kprol, where ktr and kcirc are first-order rate constants for progression betweencompartments and the loss of neutrophils respectively.

Overall, the rate of proliferation of the neutrophils is given by

rate of proliferation = number in pool · kprol · feedback (17.37)

2. Once new cell proliferation begins, it takes about 90 to 135 h for the cells to developfully and to be released into the circulation (22). This extended development processis modeled using the transit compartment transduction model. A series of threetransit compartments are incorporated between the stem cells and the circulation(Figure 17.21). The intercompartmental transit time between each compartment isset to the same value. For example, if the total time for cell development (total meantransit time) is 135 h, the transit time between compartments is 135/4 = 33.75 h,which is equivalent to a first-order rate constant of 0.0296 h−1.

3. The target of the drug action is assumed to be the first-order rate constant for neutrophilproliferation (kprol). Thus, this component of the model is similar to that of an indirectresponse model (model I, inhibition of kin), and similarly, the effect of the drug isassessed in terms of the fractional inhibition of kprol:

kprol = kprol0 (1 − I ) (17.38)

where kprol is the first-order rate constant for proliferation, kprol0 is the rate constantin the absence of the drug, and I is the inhibitory effect of the drug.

4. The inhibitory action of the drug on kprol is modeled as a linear function of the plasmaconcentration and is expressed

I = Cp · KF (17.39)

where I is the effect (fractional inhibition of kprol) and KF is the kill factor, whichhas units of reciprocal concentration and is often called the slope. It is the constantof proportionality that determines the potency of the drug in inhibiting proliferation.

Page 374: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

354 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

0

1

2

3

4

5

6

0

5

10

15

20

0 5 10 15 20 25 30

Cp

(µg/

L)

Neu

trop

hil C

ount

× 1

09 /L

Time After Start of Cycle (days)

FIGURE 17.22 Typical plasma concentration (solid line) and neutrophil count (dashed line) afterfive daily infusions of an anticancer drug that causes neutropenia. The data were simulated usingparameters of the fictitious drug mirotecan. The parameter values are given in the text.

Owing to the difficulty assessing the rate constants for proliferation (kprol) and the loss ofneutrophils from the circulation (kcirc), as well as to keep the number of model parametersto a minimum, these rate constants are both set equal to the rate constant for progressionbetween the transit compartments (Figure 17.21).

Pharmacodynamic models of hematological toxicity have been developed for severalanticancer drugs, including docetaxel, paclitaxel, etoposide (22), and topotecan (23). Thesystem-specific parameters (mean transit time and the power factor n for the feedback con-trol of circulating neutrophils) of the models were found to be consistent among the variousdrugs (22). A typical model-predicted profile of the plasma concentration and neutrophilcount after a cycle of five daily doses of a myelotoxic drug is shown in Figure 17.22.These plots were generated for a fictitions myelotoxic drug, mirotecan, using parametervalues similar to those reported in the literature. The parameters used for the simulationare provided in Section 7.2.1. For comparison, the published values of topotecan are alsoprovided. From Figure 17.22 it can be seen that the nadir neutrophil count occurs about 12days after the start of therapy, at which time drug from all five doses has been eliminated.Notice that by 21 days, the time when the next cycle of doses is typically administered,the neutrophil count has recovered. In fact, it can be seen that at around 22 to 24 days, theneutrophil count overshoots its original baseline value. This phenomenon occurs as a resultof the feedback mechanism, which stimulates proliferation when the circulating neutrophilcount is low.

The application of these models in cancer chemotherapy offers the potential of providinga way to optimize doses for individual patients. From a therapeutic perspective, doses ofthese drugs should be as large as possible to maximize their effects on cancer cells, but theirhematological toxicity limits the dose. The importance of the dose in determining patientoutcome has been illustrated in studies which indicate that patients who experience midlevelneutropenia (grade 2 to 3) (Table 17.3) have a higher survival rate than do those who expe-rience milder (grade 1) or more severe neutropenia (grade 4) [see (24)]. Doses are generallybased on body surface area or weight and do not address the possibility that a patient’s phar-macokinetic or pharmacodynamic parameters may be significantly different from average.

Page 375: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

IRREVERSIBLE DRUG EFFECTS 355

TABLE 17.3 Neutropenia Classification System

Grade of neutropenia 1 2 3 4

Neuotrophil count (109/L) 2.0–1.5 1.5–1.0 1.0–0.5 �0.5

Some patients may require lower-than-average doses, due, for example, to slow elimina-tion (pharmacokinetic), a high kill factor (pharmacodynamic), or a low baseline neutrophilcount. This group of patients will be at risk for developing severe neutropenia. Usually,these patients are identified as a result of routine monitoring of the neutrophils, which isusually performed during treatment. If patients are found to have severe neutropenia, astandard protocol is used to reduce their dose and/or to delay the next drug cycle.

The pharmacokinetic and/or pharmacodynamic properties in other patients may resultin higher-than-normal dose requirements, and since there are no measurable indicators ofthis situation, it may go unnoticed and can result in patients being chronically underdosed.The group of scientists who first developed these models have now developed softwareto estimate optimum doses of etoposide in individual patients (24,25). The software usesthe model for hematological toxicity in conjunction with a patient’s pretreatment baselineneutrophil count and a measured neutrophil count during therapy.

17.7.2.1 Simulation ExerciseA simulation model based on the published models (22) and parameters has beencreated for a fictitious drug (mirotecan) and may be found at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17i

Mirotecan is assumed to follow two-compartmental pharmacokinetics with the followingparameters: Cl = 15 L/h, Cld = 18 L/h, V 1 = 26 L, and V 2 = 45 L. The parametersof the pharmacodynamic model are baseline neutrophil count = 5 × 109 L−1, totalmean transit time = 135 h, kill factor = 0.25 L/�g, and power function for feedback loop(n) = 0.18. For comparison, the published pharmacokinetic and pharmacodynamicparameters for topotecan are Cl = 25 L/h (normal renal function), Cld = 49.9 L/h,V 1 = 39.9 L/70 kg, and V 2 = 44.5 L. The baseline neutrophil count = 4.89 × 109/L−1,total mean transit time = 116 h, kill factor = 0.183 L/�g, and power function for feed-back loop (n) = 0.130 (23). The dosing schedule for mirotecan is five daily doses of1.0 mg administered intravenously as short infusions over a 30-min period. Explore thepharmacokinetic and pharmacodynamic models.

1. Go to the “Plasma Concentration and Cell Profile” page. Give the doses andobserve the time course of plasma concentrations and neutrophil count. Notethat the nadir of the neutrophil level occurs at around 12 days. The typical patientwill experience a nadir of about 1.3 × 109 neutrophils/L, which is a grade 2neutropenia.

2. Observe the effects of pharmacokinetic and pharmacodynamic variability onresponse.

(a) Select parameter values that would represent the worst-case scenario of apatient who would be most resistant to the effects of the drug. Observe thenadir of the neutrophil count and recovery time in this patient.

Page 376: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

356 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

(b) Select parameter values that would represent the worst-case scenario of apatient who would be most sensitive to the effects of the drug. Observe thenadir of the neutrophil count and recovery time in this patient.

17.8 DISEASE PROGRESSION MODELS

Many diseases that are treated with drugs are not stationary. Chronic progressive diseasessuch as Parkinson’s disease, Alzheimer’s disease, and osteoporosis deteriorate over time.Other conditions, such as postoperative pain or recovery from an injury, are self-limitingand gradually resolve even without drug treatment. In many cases, drugs used to treat theseconditions have no effect on healthy, disease-free individuals. Thus, models for the responseof these diseases to drugs must incorporate a model for the underlying disease, and thismodel must address the continuously changing status of the disease. These models, calleddisease progression models, consist of three parts: a model for the response resulting fromthe direct action of the drug, a model for how the drug interacts with the disease, and amodel for the underlying disease.

17.8.1 Generation of Drug Response

The direct drug action that results from drug–receptor interaction can be generated using apharmacokinetic model linked to any of the previously discussed pharmacodynamic models,such as the sigmoidal Emax model or linear model. If necessary, an effect compartment ortransit compartments can be added to accommodate delays in the response of the diseaseto the drug. In this discussion, the simple Emax model without delays will be used to modelthe direct effect of the drug. Additionally, for simplicity, drug input will be modeled usinga constant continuous intravenous infusion.

17.8.2 Drug Interaction with a Disease

Drugs can theoretically interact with diseases in two ways:

1. Drugs can relieve the symptoms without altering the underlying progression of adisease. This is called symptomatic action. When a symptomatic drug is withdrawn,the disease is at the same place that it would have been at without the treatment.

2. Drugs can halt or slow the progression of a disease. This is known as a protectiveaction. In this case, the improvement in disease status remains even when the drug iswithdrawn.

17.8.3 Disease Progression Models

A disease progression model represents how a disease changes over time in the absenceof drug therapy. The disease status may be evaluated by measuring a symptom of thedisease, an outcome of the disease (e.g., blood pressure, bone density), or a biomarker.Three models of disease progression are presented: the linear model, the exponential decayor zero asymptotic model, and the exponential model with nonzero maximum diseasestatus. These models have been used successfully to model a variety of conditions, such

Page 377: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

DISEASE PROGRESSION MODELS 357

as Parkinson’s disease, Alzheimer’s disease, muscular dystrophy, and the treatment of HIVdisease (26–29).

17.8.3.1 Linear Disease Progression ModelThe linear model is frequently used to model the progression and treatment of chronicprogressive diseases. It simply assumes that the disease deteriorates at a constant rate andcan carry on doing so indefinitely. The disease status (St) at a given time is given by

St = S0 + � · t (17.40)

where St is the status of the disease at time t, S0 the status of the disease at time zero, and� the slope representing the rate at which the status of the disease changes over time.

Depending on how disease status is measured, it may increase or decrease during thenatural progression of a disease. For example, the loss of bone density due to osteoporosisdecreases as the disease progresses, and if renal function is assessed using creatinineclearance, the disease status will increase over time. Thus, � may be positive or negative. Forthis discussion, a positive slope will be used. Because many chronic conditions deterioratevery slowly, the period over which the diseases and their treatment are studied can beextremely long and may span several years.

Symptomatic Drugs The action of symptomatic drugs, which reduce the disease statuswithout altering the underlying progression, is modeled by incorporating an additive term,Et, to represent the action of the drug at time t. Thus, the linear disease progression modelwill be modified as follows:

St = S0 + � · t − Et (17.41)

where Et is the effect of the drug.The effect of a symptomatic drug in a linear model is shown in Figure 17.23, which

shows that as the symptomatic action is characterized by an immediate change in the diseasestatus, the curve is shifted downward but the slope is unchanged. In practice, frequentlythere is a lag time before the effects of the drug become apparent and a more gradual onsetof action is observed. This can be accommodated in a model by incorporating a link model,such as a series of transit compartments. As soon as the drug is withdrawn, the status revertsback to the value that it would have had without drug therapy. This feature distinguishesthe symptomatic effect from the protective effect (see below). However, in practice, asymptomatic drug effect may wear off more slowly than is shown in Figure 17.23, and thedifference between the symptomatic and protective effects become less noticeable.

Protective Drugs Protective drugs slow the progression of a disease. Their action ismodeled by an effect on the progression slope:

St = S0 + (� − Et ) · t (17.42)

Thus, the drug effectively reduces the slope (Figure 17.23). It can be seen that the effectof protective drugs is more gradual than those of symptomatic drugs. When the drug iswithdrawn, the slope will return to its original value, but the patient is left with a lower(better) disease status than that of a patient who had no treatment.

Page 378: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

358 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

60

80

100

120

140

160

0 50 100 1500

20

40

60

80

100

0 50 100

S S

Time (days) Time (days)

natural progression

protectiveaction

symptomaticaction

natural progression

protectiveactionsymptomatic

action

Drug Therapy Drug Therapy

(a) (b)

FIGURE 17.23 Linear (a) and exponential (b) disease progression models. Disease status (S) isshown as a function of time. Data were simulated using an IV infusion of 100 mg/h in a one-compartment model (Vd = 20 L, Cl = 4 L/h). The linear progression model has an initial diseasestatus of 100 units and a slope of 0.4 unit/day. The symptomatic and protective effects were modeledusing the Emax model with an EC50 of 10 mg/L and Emax values of 50 units and 0.35 day−1, respectively.The infusion ran from day 10 to day 100. The exponential model has an initial disease status of 100units and a kprog of 0.02 day−1. The EC50 of the drug was 10 mg/L, and Emax values of 25 units and0.03 day−1 were used for the symptomatic and protective effects, respectively. The infusion ran fromday 10 to day 50.

17.8.3.2 Exponential Decay or Zero Asymptotic ModelDiseases that are only transient in nature and resolve themselves over time can be viewedas conditions that eventually decay to zero. These diseases are modeled by adding a decayfunction to the disease status:

St = S0 · e−kprog · t (17.43)

where kprog is a first-order rate constant for disease progression. In this model, S decays toa minimum (zero), and as a result this model is called an asymptotic model.

Symptomatic Drugs Once again, the action of symptomatic drugs is modeled using anadditive term:

St = S0 · e−kprog · t − Et (17.44)

The effect of a symptomatic drug in an exponential model is shown in Figure 17.24. Inthe zero asymptotic model, the action of the drug is in the same direction as the diseaseprogression. In common with the linear model, the symptomatic action is rapid, the curveis shifted downward, the slope is unchanged, and when the drug is withdrawn, the statusreverts back to the value that it would have had without drug therapy. Once again a moregradual onset of action can be accommodated using a link model.

Protective Drugs In the exponential model, the action of protective drugs is incorporatedinto kprog. In the treatment of a self-resolving condition, the drug increases the value of kprog

to expedite the decay to zero. The action of protective drugs is modeled as follows:

St = S0 · e−(kprog+Et ) · t (17.45)

Page 379: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

DISEASE PROGRESSION MODELS 359

0

50

100

150

200

0 50 100

S0S

time0

0 • (1−e )t ssS = S • e−kprog • t −kprog • t+ S

Drug Therapy

natural progression

symptomaticaction

protectiveaction

S

Time (days)

(a) (b)

FIGURE 17.24 Maximum asymptotic disease progression model. Disease status (S) is shownover time. Time is measured from the first measured disease status value (S0) (a). Sss represents themaximum terminal value for the disease status, and kprog is the rate constant for disease progression.Part (b) shows the profile for symptomatic and protective drug effects on kprog. The data were simulatedusing an intravenous infusion of 100 mg/h in a one-compartment model (Vd = 20 L, Cl = 4 L/h).The infusion ran from day 10 to day 20. The progression model had kprog and Sss of 0.02 day−1 and200 units, respectively. The symptomatic and protective effects were modeled using the Emax modelwith an EC50 of 10 mg/L and Emax values of 50 units and 0.5, respectively.

It can be seen in Figure 17.23 that as was the case for the linear model, the protective effecthas a more gradual onset of action, and once again when the treatment is withdrawn, theslope reverts to its original value but the patient is in a better place because of the treatment.These types of conditions that resolve naturally often are much shorter in duration thanchronic progressive conditions. This leads to several important points. First, because of theirfaster onset of action, drugs that act symptomatically may be superior to disease-modifyingdrugs. Also, because these conditions resolve naturally, it is not as important that the naturalprogression of the disease be affected. Second, when evaluating the action of drugs, it isimportant to assess their effect early in the treatment process. If their effect is assessed toolate, the disease may have almost resolved itself, and the drug may appear to be no differentfrom a placebo.

17.8.3.3 Exponential Model with a Nonzero Maximum Disease StatusSome degenerative diseases deteriorate to a final concluding maximal state. These diseasescan be modeled using a model for exponential growth (Figure 17.24a). The equation forthis profile is

St = Sss · (1 − e−kprog · t ) (17.46)

where kprog is the first-order rate constant for disease progression, Sss the terminal status ofthe disease, and t the time since the beginning of the disease.

The equation is modified because the time when the disease started is usually unknownand progression is assessed relative to when it is first measured (baseline) (Figure 17.24a).Thus, the equation for progression takes the form

St = S0 · e−kprog · t + Sss · (1 − e−kprog · t ) (17.47)

where S0 is the measured baseline and t is the time from the baseline measurement.

Page 380: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

360 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

Symptomatic drug treatment is modeled by an additive term:

St = S0 · e−kprog · t + Sss · (1 − e−kprog · t ) − Et (17.48)

Protective action can be modeled through an effect on kprog:

St = S0 · e−(kprog−Et ) · t + Sss · (1 − e−(kprog−Et ) · t ) (17.49)

Figure 17.24b shows the typical profile of symptomatic and protective effects from thismodel.

Alternatively, the protective effect can be modeled through an effect on the terminalstate of the disease:

St = S0 · e−kprog · t + (Sss − Et ) · (1 − e−kprog · t ) (17.50)

These models have been further adapted to incorporate placebo effects, which can bevery important for some drugs, such as antidepressants. Other models have combined bothsymptomatic and protective effects. An alternative approach to modeling the effects of drugson disease progression has been proposed based on the physiological turnover system (30)discussed at the beginning of this chapter. In this application the biological quantity orresponse variable is the disease status (St).Under normal conditions of health, the diseasestatus, which is a disease symptom or a biomarker used to assess the disease, is constant,as are kin and kout (S = kin/kout). Disease is assumed to disrupt homeostasis by decreasingeither kin or kout. In effect, these disease progression models are essentially models I andII of the indirect effect models with ongoing modification of kin and kout. This approachwas used to model the action of several hypoglycemic drugs in the treatment of type 2diabetes (31).

PROBLEMS

17.1 Open the model “Operational Model of Agonism” at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17a

The model has the following default parameters: RCE50 = 5 mg/L, RT = 200 units,Kd = 10 mg/L, and Em = 100%.

A. Effect of changes in efficacy. Perform simulations with the total number ofreceptors set to 1000 units and the default values of all the other parameters exceptthe drug’s RCE50. Do runs with RCE50 set to 10, 5, and 1 mg/L. The RCE50 isthe concentration of occupied receptors that produce half the system’s maximumresponse. In the various simulation runs, record the values of the transduction ratio,Emax, and EC50 in Table P17.1A.

(a) Are the drugs used for these simulations full or partial agonists?

(b) How are their different efficacies reflected?

(c) How do changes in efficacy influence the relative values of the EC50 and Kd fora given simulation?

Page 381: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

PROBLEMS 361

TABLE P17.1A Influence of Efficacy

RT = 1000 mg/L

RCE50 (mg/L) 10 5 1

Emax (%Em)

EC50 (mg/L)

TABLE P17.1B Influence of the Number of System Receptors

RC50 = 5 mg/L

RT (mg/L) 1000 100 10

Emax (%Em)

EC50 (mg/L)

TABLE P17.1C Influence of the Number of System Receptors When the Efficacy IsVery High

RCE50 = 5 mg/L, RT = 200 mg/L

RT (mg/L) 200 120 (60%) 60 (30%) 40 (20%) 20 (10%) 10 (5%)

Emax (%Em)

EC50 (mg/L)

B. Influence of the concentration of receptors. Perform simulations with the num-ber of receptors set to 1000, 100, and 10 units and the remaining parameters set totheir default values. Record in Table P17.1B the values of the transduction ratio,Emax, and EC50 for the three different simulations.

(a) The drug properties do not change for this simulation, so it can be considered tobe the same drug. Is the drug used for these simulations a full or partial agonist?

(b) How are Emax and the transduction ratio (efficacy) affected by the number ofreceptors?

C. Influence of the concentration of receptors when the efficacy is very high. Pressthe restore button to reset the parameters to their default values. To represent anagonist of high efficacy, the default value of RCE50 (5 mg/L) will be used. Per-form a simulation, then reduce the receptor concentration sequentially according toTable P17.1C. Record the parameter values for each simulation in Table P17.1C.

(a) What fraction of the original receptors has to be lost before the drug’s maximumresponse is affected?

Page 382: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

362 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

(b) How are the parameters in Table P17.1C affected? Which parameter(s) consis-tently reflects the change in efficacy?

(c) Can a change in efficacy be concluded from the effects on EC50?

D. Influence of affinity on the drug parameters. Perform simulations with thedefault parameter values and change only the drug affinity. Recall that as Kd increases,the affinity decreases. Use Kd values of 1, 10, and 20 mg/L and record the valuesof the transduction ratio, Emax, and EC50 in Table P17.1D. Which parameters areaffected by changes in the affinity and which are not?

TABLE P17.1D Influence of Affinity on the Measurable DrugParameters

RT = 200 mg/L; RCE50 = 5 mg/L

Kd (mg/L) 1 10 20

Emax (%Em)

EC50 (mg/L)

17.2 The development of tolerance to a synthetic opioid is under investigation. Its phar-macokinetics and pharmacodynamics are being evaluated after two consecutive in-fusions. No change is observed in the values of the pharmacokinetic parametersbetween infusions. However, after the second infusion, the drug’s EC50 is aboutdouble that after the first infusion. Provide possible explanations for the observation.Explain how the transduction ratio could be helpful to understand the mechanismbehind the change in the EC50.

17.3 An intravenous dosing regimen of 25 mg of lipoamide every 8 h was developed inSection 17.7.2.1. The regimen was designed on pharmacokinetic principles, basedon the belief that peaks and troughs of around 90 and 25 �g/L, respectively, aredesirable. Based on lipoamide’s reported bioavailability of 0.21, the equivalent oraldose would be about 120 mg. Prior to starting a clinical trial with lipoamide, amore thorough understanding of the dose–response relationship is desired so thatoptimum dosing regimens can be used in the trial. A small integrated PK/PD studywas conducted in which response of lipoamide was measured by observing thereduction in body temperature. The data from a single patient are shown in TableP17.3.

(a) Plot concentration and response against time. Plot response against concentra-tion. Comment on these profiles.

Is it possible to determine the optimum concentration to produce a particularresponse? Suggest models that could be applied to the response data?

(b) Several mechanism-based models were evaluated to see if they would fit thedata. The best fit of the data was obtained with indirect effect model I (inhibitionof kin), which has been used previously to model antipyretic activity (32). Feveris assumed to be the result of the action of a response variable. Lipoamide’santipyretic activity is assumed to be the result of its inhibition of the synthesisof this substance.

Page 383: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

PROBLEMS 363

TABLE P17.3 Plasma Concentration and Response After aSingle Oral Dose (120 mg) of Lipoamide

Time (h) Cp (�g/L) Reduction in Temp ◦F

0 0.0 00.5 49.9 1.151 53.2 2.251.5 50.2 2.92 46.7 3.272.5 43.3 3.463 40.2 3.554 34.7 3.566 25.8 3.328 19.2 2.98

12 10.6 2.2514 7.9 1.9

The model used to fit the lipoamide data may be found at

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17k

Use the model to probe optimum dosing regimens for this drug. Ideally, a sustainedantipyretic action is desired that will maintain the temperature at or below 100◦Fduring the dosing interval. If possible, a dosing interval of either 12 or 24 h wouldbe preferred to 8 h. More recent studies indicate that the incidence of side effectsis minimal if the plasma concentration is below 300 �g/L. Can the dosing regimenbased on pharmacokinetics (120 mg q 8 h) be improved upon?

17.4 A model to examine the action of a fictitious proton pump inhibitor, disolvprazole,is provided at the link

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html#chapter17i

Open the model and go to the “Problem” page. Flip the switch to observe the effectsof single doses. Observe the effects of changing the rate constants individually whilemaintaining all the others at their default value. Use the following values for each ofthe rate constants when they are altered individually:

kin: 0.4, 0.8, and 1.6 mmol/h/h (Note that every time the graphs are cleared, thesingle-dose switch will have to be turned on again.)

kout: 0.015, 0.03, and 0.06 h−1

kirr: 15, 30, and 60 L/mg/h

k: 0.17, 0.34, and 0.68 h−1

Answer the following questions:

(a) When all the parameters have their default values, what is the maximum effect?What is the duration of action of the drug? How does it compare to the drug’selimination half-life?

(b) Summarize how each individual parameter affects the intensity and duration ofthe effect.

Page 384: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

364 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

(c) How would you predict that inhibitors of the metabolism of the proton pumpinhibitors would affect their therapeutic effect?

17.5 A drug is under development for the treatment of an autoimmune disease that appearsto be mediated by a newly identified protein that has been called �-in. Studies inanimals show that the drug reduces the concentrations of the protein and that thesymptoms of the disease (response), which is assessed using a biomarker, improve.Trials are being conducted in humans. Three separate oral doses of the drug (100,50, and 25 mg) were administered to several patients with the autoimmune disease,and plasma concentrations of the drug and the fall in the level of the biomarker weremeasured at different times. The data from one subject are given in Table P17.5.

(a) Plot response against concentration for one of the doses. Comment on the profileand suggest possible explanations and ways in which it could be addressed inthe modeling process.

(b) Plot response against time for the three doses on the same chart. Based on theprofile, do you think that an effect compartment model or an indirect effectmodel, would be more appropriate for this data.

(c) The drug’s pharmacokinetic parameters are k = 0.02 h−1 and Vd = 20 L. Basedon pharmacokinetics alone, how frequently do you think the drug should bedosed? Revise your estimate of the dosing frequency based on the response–timeprofiles. How do you explain the difference between the two estimates?

17.6 A double-blind clinical trial was conducted on a new corticosteroid for the treatmentof muscular dystrophy. Trial participants were split randomly into two groups. Thetest group took a 40-mg daily dose of the drug under study. The control group tookplacebo daily. Response was assessed from tests of muscle strength. After 8 months,the trial had to be discontinued due to the concern of toxicity. The average data forthe test (drug) and placebo group are given in Table P17.6. Additionally, data areprovided on the natural progression of the disease in the absence of intervention. Plot

TABLE P17.5 Plasma Drug Concentration (Cp) and Change in Concentration of aBiomarker (R) at Three-Dose Levels

Dose

100 mg 50 mg 25 mg

Time (h) Cp (mg/L) � R Cp (mg/L) � R Cp (mg/L) � R

0 0.000 0.0 0.000 0.0 0.000 0.03 2.919 −23.9 1.460 −22.8 0.730 −20.96 2.049 −42.7 1.024 −40.9 0.512 −37.9

10 0.979 −59.4 0.490 −56.3 0.245 −51.014 0.442 −68.3 0.221 −62.9 0.111 −54.515 0.362 −69.5 0.181 −63.2 0.090 −54.116 0.296 −70.1 0.148 −63.1 0.074 −53.317 0.242 −70.4 0.121 −62.6 0.060 −52.018 0.197 −70.2 0.099 −61.6 0.049 −50.424 0.059 −61.3 0.029 −49.2 0.015 −37.140 0.002 −20.0 0.001 −14.1 0.001 −9.6

Page 385: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

PROBLEMS 365

TABLE P17.6 Response (Change in AverageMuscle Strength) at Various Times

Time(months)

NaturalProgression Placebo

NewDrug

0 10.0 10.0 10.01 9.2 11.6 9.82 8.4 11.5 9.03 7.6 10.7 7.84 6.8 9.9 6.85 6.0 9.1 6.06 5.2 6.0 5.27 4.4 4.4 4.4

the data and comment on the effects of placebo and the drug relative to the naturalprogression. Do you think the drug has a protective or a symptomatic effect?

17.7 A totally new class of drug was evaluated for the treatment of Alzheimer’s disease.The clinical status of each patient was evaluated using a dementia score based on the

TABLE P17.7

Time Natural New Existing(weeks) Progression Drug Therapy

0 1.0 1.0 1.02 1.8 1.8 1.86 3.4 3.4 3.4

10 5.0 5.0 5.015 7.0 6.9 −2.220 9.0 8.1 −14.524 10.6 8.8 −18.028 12.2 9.4 −18.332 13.8 9.9 −17.236 15.4 10.4 −15.842 17.8 11.2 −13.448 20.2 11.9 −11.156 23.4 12.9 −7.864 26.6 14.0 −4.772 29.8 15.0 −1.580 33.0 16.0 1.888 36.2 17.0 5.096 39.4 18.0 8.2

100 41.0 18.5 9.8102 41.8 18.8 11.3104 42.6 19.1 16.8106 43.4 19.5 24.3108 44.2 20.0 31.4112 45.8 21.3 41.0120 49.0 24.3 48.6

Page 386: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

366 MECHANISM-BASED INTEGRATED PHARMACOKINETIC– PHARMACODYNAMIC MODELS

Alzheimer’s Disease Assessment Scale, which tests memory, language, orientation,reason, praxis, and concentration. The total score could vary from 0 (no impairment)to 100 (maximum impairment). Response was evaluated as the mean change frombaseline. The patients were enrolled in the trial and monitored over a 10-weekperiod. At 10 weeks, half of the participants in the trial took an existing medicationfor Alzheimer’s disease, and the remainder received the new treatment. The drugswere discontinued after 100 weeks. The average data for the test (drug) and control(existing medication) are given in Table P17.7. Additionally, data are provided onthe natural progression of the disease in the absence of any interventions. Plot thedata and comment on the effect of the new drug.

REFERENCES

1. Danhof, M., de Jongh, J., De Lange, E. C., Della Pasqua, O., Ploeger, B. A., and Voskuyl,R. A. (2007) Mechanism-based pharmacokinetic-pharmacodynamic modeling: biophase dis-tribution, receptor theory, and dynamical systems analysis, Annu Rev Pharmacol Toxicol 47,357–400.

2. Mager, D. E., Wyska, E., and Jusko, W. J. (2003) Diversity of mechanism-based pharmacody-namic models, Drug Metab Dispos 31, 510–518.

3. Mager, D. E., Woo, S., and Jusko, W. J. (2009) Scaling pharmacodynamics from in vitro andpreclinical animal studies to humans, Drug Metab Pharmacokinet 24, 16–24.

4. Black, J. W., and Leff, P. (1983) Operational models of pharmacological agonism, Proc R SocLond B Biol Sci 220, 141–162.

5. Cox, E. H., Kerbusch, T., van der Graaf, P. H., and Danhof, M. (1998) Pharmacokinetic-pharmacodynamic modeling of the electroencephalogram effect of synthetic opioids in therat: correlation with the interaction at the mu-opioid receptor, J Pharmacol Exp Ther 284,1095–1103.

6. Madia, P. A., Dighe, S. V., Sirohi, S., Walker, E. A., and Yoburn, B. C. (2009) Dosing protocoland analgesic efficacy determine opioid tolerance in the mouse, Psychopharmacology (Berl)207, 413–422.

7. Cox, E. H., Kuipers, J. A., and Danhof, M. (1998) Pharmacokinetic-pharmacodynamic modellingof the EEG effect of alfentanil in rats: assessment of rapid functional adaptation, Br J Pharmacol124, 1534–1540.

8. Ploeger, B. A., van der Graaf, P. H., and Danhof, M. (2009) Incorporating receptor theory inmechanism-based pharmacokinetic–pharmacodynamic (PK–PD) modeling, Drug Metab Phar-macokinet 24, 3–15.

9. Dayneka, N. L., Garg, V., and Jusko, W. J. (1993) Comparison of four basic models of indirectpharmacodynamic responses, J Pharmacokinet Biopharm 21, 457–478.

10. Sharma, A., and Jusko, W. J. (1996) Characterization of four basic models of indirect pharma-codynamic responses, J Pharmacokinet Biopharm 24, 611–635.

11. Sharma, A., and Jusko, W. J. (1998) Characteristics of indirect pharmacodynamic models andapplications to clinical drug responses, Br J Clin Pharmacol 45, 229–239.

12. Krzyzanski, W., and Jusko, W. J. (1998) Mathematical formalism and characteristics of four basicmodels of indirect pharmacodynamic responses for drug infusions, J Pharmacokinet Biopharm26, 385–408.

13. Jusko, W. J., and Ko, H. C. (1994) Physiologic indirect response models characterize diversetypes of pharmacodynamic effects, Clin Pharmacol Ther 56, 406–419.

14. Mager, D. E., and Jusko, W. J. (2001) Pharmacodynamic modeling of time-dependent transduc-tion systems, Clin Pharmacol Ther 70, 210–216.

Page 387: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCc17 JWBS062-Rosenbaum February 16, 2011 9:7 Printer Name: Yet to Come

REFERENCES 367

15. Porchet, H. C., Benowitz, N. L., and Sheiner, L. B. (1988) Pharmacodynamic model of tolerance:application to nicotine, J Pharmacol Exp Ther 244, 231–236.

16. Shi, J., Benowitz, N. L., Denaro, C. P., and Sheiner, L. B. (1993) Pharmacokinetic–pharmacodynamic modeling of caffeine: tolerance to pressor effects, Clin Pharmacol Ther53, 6–14.

17. Bauer, J. A., and Fung, H. L. (1994) Pharmacodynamic models of nitroglycerin-induced hemo-dynamic tolerance in experimental heart failure, Pharm Res 11, 816–823.

18. Ouellet, D. M., and Pollack, G. M. (1995) A pharmacokinetic-pharmacodynamic model oftolerance to morphine analgesia during infusion in rats, J Pharmacokinet Biopharm 23, 531–549.

19. Gardmark, M., Brynne, L., Hammarlund-Udenaes, M., and Karlsson, M. O. (1999) Interchange-ability and predictive performance of empirical tolerance models, Clin Pharmacokinet 36,145–167.

20. Sharma, A., Ebling, W. F., and Jusko, W. J. (1998) Precursor-dependent indirect pharmacody-namic response model for tolerance and rebound phenomena, J Pharm Sci 87, 1577–1584.

21. Ferron, G. M., McKeand, W., and Mayer, P. R. (2001) Pharmacodynamic modeling of panto-prazole’s irreversible effect on gastric acid secretion in humans and rats, J Clin Pharmacol 41,149–156.

22. Friberg, L. E., Henningsson, A., Maas, H., Nguyen, L., and Karlsson, M. O. (2002) Model ofchemotherapy-induced myelosuppression with parameter consistency across drugs, J Clin Oncol20, 4713–4721.

23. Leger, F., Loos, W. J., Bugat, R., Mathijssen, R. H., Goffinet, M., Verweij, J., Sparreboom,A., and Chatelut, E. (2004) Mechanism-based models for topotecan-induced neutropenia, ClinPharmacol Ther 76, 567–578.

24. Wallin, J. E., Friberg, L. E., and Karlsson, M. O. (2009) Model-based neutrophil-guided doseadaptation in chemotherapy: evaluation of predicted outcome with different types and amountsof information, Basic Clin Pharmacol Toxicol 106, 234–242.

25. Wallin, J. E., Friberg, L. E., and Karlsson, M. O. (2009) A tool for neutrophil guided doseadaptation in chemotherapy, Comput Methods Programs Biomed 93, 283–291.

26. Chan, P. L., and Holford, N. H. (2001) Drug treatment effects on disease progression, Annu RevPharmacol Toxicol 41, 625–659.

27. Holford, N. H., Chan, P. L., Nutt, J. G., Kieburtz, K., and Shoulson, I. (2006) Disease progressionand pharmacodynamics in Parkinson disease: evidence for functional protection with levodopaand other treatments, J Pharmacokinet Pharmacodyn 33, 281–311.

28. Griggs, R. C., Moxley, R. T., 3rd, Mendell, J. R., Fenichel, G. M., Brooke, M. H., Pestronk,A., and Miller, J. P. (1991) Prednisone in Duchenne dystrophy: a randomized, controlled trialdefining the time course and dose response. Clinical Investigation of Duchenne DystrophyGroup, Arch Neurol 48, 383–388.

29. Mould, D. R. (2007) Developing models of disease progression, In Pharmacometrics: TheScience of Quantitative Pharmacology (Ette, E. I., and Williams, P. J., Eds.), pp. 547–581,Wiley, Hoboken, NJ.

30. Post, T. M., Freijer, J. I., DeJongh, J., and Danhof, M. (2005) Disease system analysis: basicdisease progression models in degenerative disease, Pharm Res 22, 1038–1049.

31. de Winter, W., DeJongh, J., Post, T., Ploeger, B., Urquhart, R., Moules, I., Eckland, D., andDanhof, M. (2006) A mechanism-based disease progression model for comparison of long-termeffects of pioglitazone, metformin and gliclazide on disease processes underlying type 2 diabetesmellitus, J Pharmacokinet Pharmacodyn 33, 313–343.

32. Garg, V., and Jusko, W. J. (1994) Pharmacodynamic modeling of nonsteroidal anti-inflammatorydrugs: antipyretic effect of ibuprofen, Clin Pharmacol Ther 55, 87–88.

Page 388: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

APPENDIX A

REVIEW OF EXPONENTSAND LOGARITHMS

A.1 Exponents

A.2 Logarithms: log and ln

A.3 Performing Calculations in the Logarithmic DomainA.3.1 MultiplicationA.3.2 DivisionA.3.3 ReciprocalsA.3.4 Exponents

A.4 Calculations Using Exponential Expressions and Logarithms

A.5 Decay Function: e−kt

A.6 Growth Function: 1 − e−kt

A.7 Decay Function in Pharmacokinetics

Problems

A.1 EXPONENTS

When a number is expressed in exponential notation it takes the following form:

number = baseexponent

The following are examples of exponential expressions using different bases:

23, 63, 106, 158, e3, 202

Exponents to the base 10 and base e are the most common, and the base 10 form is usedmost frequently in everyday life. Examples include

106 = 1,000,000

103 = 1000

101 = 10

100 = 1

10−1 = 0.1

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

368

Page 389: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

LOGARITHMS: log AND ln 369

The base e form is used most frequently in the mathematical sciences, including phar-macokinetics. This exponent form is introduced into a formula when calculus is used tointegrate equations.

e = 2.718

Examples of exponents to the base e:

e2 = 7.389

e1 = 2.718

e0.639 = 2

e0 = 1 important to know

e−1 = 0.368 (1/e or 1/2.718)

e−∞ = 0 important to know

In pharmacokinetics it is necessary to know how to use a calculator to solve exponentexpressions such as e6.8, e−0.43, and e−0.3×2. (answers: 897.8, 0.651, and 0.549)

A.2 LOGARITHMS: log AND ln

The logarithm of a number is the power or exponent when that number is converted toexponential form. The power or exponent will clearly depend on the base. If the base 10 isused, the logarithm is known as the Log. Examples are provided in Table A.1.

When the base e is used, the logarithm is known as the natural logarithm or ln. Examplesare provided in Table A.2.

TABLE A.1

Number Exponential Form Log

1000,000 106 6100 102 240 101.6 1.62 100.301 0.301

10 101 11 100 00.1 10−1 −1

TABLE A.2

Number Exponential Form Ln

7.389 e2 22 e0.693 0.6932.718 e1 11 e0 00.368 e−1 −1

Page 390: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

370 REVIEW OF EXPONENTS AND LOGARITHMS

When the base 10 logarithm scale is used for expressions containing e, it may benecessary to convert a log to an ln. This is done using the factor 2.303:

ln X = log X × 2.303

Thus if log 5 = 0.6989, then

ln 5 = 0.6989 × 2.303 = 1.609

If log 2 = 0.3010, then

ln 2 = 0.3010 × 2.303 = 0.693

Generally, there is no longer any reason to use the base 10 logarithms in pharmacoki-netics. They were used in the past because it was the most convenient scale to use onsemilogarithmic graph paper to plot pharmacokinetic data. Now data can be plotted easilyusing spreadsheets on computers, where the natural logarithm scale can be used as easilyas the base 10 scale.

A.3 PERFORMING CALCULATIONS IN THE LOGARITHMIC DOMAIN

A.3.1 Multiplication

To multiply two numbers, they can be converted to logarithm form and added. Consider102 × 103:

Nonlog domain Log domain

100 × 1000 = 100,000 2 + 3 = 5

Thus,

102 × 103 = 102+3 = 105

B · C · D:

ln(B · C · D) = lnB + lnC + lnD

3 × 5:

ln(3 × 5) = ln 3 + ln 5 = 1.10 + 1.61 = 2.71 ∗anti-ln (2.71) = 15

∗The conversion from the logarithm domain back to the nonlogarithm domain is referredto as taking an anti-logarithm.

B · e3:

ln(B × e3) = lnB + lne3 = lnB + 3

Page 391: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

CALCULATIONS USING EXPONENTIAL EXPRESSIONS AND LOGARITHMS 371

A.3.2 Division

To divide two numbers, they can be converted to the logarithm domain and subtracted.

B/A:

ln(B/A) = lnB − lnA

3/5:

ln(3/5) = ln 3 − ln 5 = 1.10 − 1.61 = −0.51 anti-ln(−0.51) = 0.6

A.3.3 Reciprocals

The reciprocal of a number is the negative ln of the number.

ln(1/A) = −ln(A)

ln(B/A) = −ln(A/B)

Proof: − ln(A/B) = −(lnA − lnB) = lnB − lnA = ln(B/A)

ln(1/10) = −ln10 = −2.303 anti-ln(−2.303) = 0.1

A.3.4 Exponents

To determine the solution of an exponential expression, in the logarithmic domain, expo-nents are multiplied.

Ab:

lnAb = b · lnA

82:

ln 82 = 2 · ln 8 = 2 × 2.08 = 4.16 anti-ln(4.16) = 64

A.4 CALCULATIONS USING EXPONENTIAL EXPRESSIONSAND LOGARITHMS

Example A.1 Solve for x in the following expressions: (a) ex = 3; (b) ex = 23; (c) ex =106; (d) ex = 563.

Solution(a) ex = 3

x = ln 3 = 1.10

(b) 3.14

(c) 4.66

(d) 6.33

Page 392: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

372 REVIEW OF EXPONENTS AND LOGARITHMS

Example A.2 Solve for x in the following expressions: (a) ln x = 4.3; (b) ln x = −1.4;(c) ln x = −0.3.

Solution(a) ln x = 4.3

x = e4.3

x =73.7

(b) 0.247

(c) 0.741

Example A.3 Evaluate e−1.3.

Solution

e−1.3 = 0.273

Example A.4 Find the value of k in the following expression: e−1.3k = 2

Solution Take the logarithms

k · ln e−1.3 = ln 2

k · (−1.3) = 0.693

−1.3k = 0.693

k = −0.693

1.3= −0.533

Example A.5 A common expression in pharmacokinetics is Cp = Cp0 · e−kt. EvaluateCp when Cp0 = 35, k = 1.5, and t = 2.

Solution

Cp = 35e−1.5×2 = 35e−3 = 35 × 0.0498 = 1.74

Example A.6 Given Y = Y0 · e−kt, convert the equation to the ln domain.

Solution

ln Y = ln Y0 − kt (A.1)

Expression (A.1) is the equation for a straight line (y = a − bx) with a slope of −k and anintercept of ln Y0 (Figure A.1).

Example A.7 Given Y = Y0 · e−kt, calculate k given Y1 = 66.3 mg, t1 = 2 h; Y2 = 29.2mg, t2 = 6 h.

Page 393: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

DECAY FUNCTION: e−kt 373

ln Y

Time

Intercept = ln Y0

Slope = –k

FIGURE A.1 Plot of lnY against time.

Solution k = − slope of the plot of ln Y against time:

slope = ln Y1 − ln Y2

t1 − t2or

ln(Y1/Y2)

t1 − t2

= ln(29.2/66.3)

6 − 2

= −0.205

k = −slope = 0.205 h−1

Example A.8 Given Y = Y0 · e−kt, calculate the intercept, Y0.

Solution Use either data pairs to substitute in the basic equation (1.1):

ln(66.3) = lnY0 − 0.205 × 2 or ln(29.2) = lnY0 − 0.205 × 6

4.19 = lnY0 − 0.41 or 3.37 = lnY0 − 1.23

lnY0 = 4.19 + 0.41 = 4.6 lnY0 = 3.37 + 1.23 = 4.6

Y0 = 100 Y0 = 100

A.5 DECAY FUNCTION: e−kt

This is an extremely important expression in pharmacokinetics and is utilized in almost allpharmacokinetic equations. It is important to understand this expression and not be intimi-dated by it. It is actually very simple, and a little time spent considering and understandingthis function will demystify all pharmacokinetic equations are help prevent calculationerrors.

e−kt is a decay function

Page 394: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

374 REVIEW OF EXPONENTS AND LOGARITHMS

where k is a constant and t is time, which starts at zero and increases to ∞.

At t = 0 : e−kt = 1

At t = ∞ : e−kt = 0

That is, as time increases, e−kt decays from 1 to zero. The rate constant k controls the speedwith which e−kt decays: The larger the value of k, the more rapidly the expression decaystoward zero.

A.6 GROWTH FUNCTION: 1 − e−kt

The growth function is also frequently encountered in pharmacokinetics and runs counterto the decay function (e−kt):

1 − e−kt is a growth function

where k is a constant and t is time, which starts at zero and increases to ∞.

At t = 0 : (1 − e−kt ) = 0

At t = ∞ : (1 − e−kt ) = 1

That is, as time increases, 1 − e−kt grows from zero to 1. The rate constant k controls thespeed with which the expression grows toward 1. The larger the value of k, the more rapidlythe expression grows toward 1.

A.7 DECAY FUNCTION IN PHARMACOKINETICS

Cp = A · e−kt

This is the most important mathematical equation in pharmacokinetics. In basic pharma-cokinetics it appears in the following forms:

A. Cp = A · e−kt .

B. Cp = A · e−k1t + B · e−k2t , where k1 � k2.

C. Cp = A · (1 − e−kt ).

D. Cp = A · (e−k1t − e−k2t ), where k2 � k1.

These equations represent the fundamental form of the pharmacokinetic equations asso-ciated with an intravenous bolus injection in a one-compartment model (A); an intravenousbolus injection in a two-compartment model (B); constant continuous drug input in aone-compartment model (C); first-order drug absorption in a one-compartment model (D).

If the expressions e−kt and 1 − e−kt are understood, equations A to D all become verysimple.

Page 395: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

PROBLEMS 375

Example A.9 Evaluate equations A through D above for Cp at time zero and infinity.

SolutionA. t = 0, Cp = A; t = ∞, Cp = 0

B. t = 0, Cp = A + B; t = ∞, Cp = 0

C. t = 0, Cp = 0; t = ∞, Cp = A

D. t = 0, Cp = 0; t = ∞, Cp = 0

Example A.10 In equations B and D, which exponent term gets to zero first?

Solution The one with the larger value of k: Equation B. k1; Equation D. k2.

Example A.11 Write out the shortened forms of equations B and D when the first exponentterm has reached zero.

Solution B. Cp = B · e−k2t ; D. Cp = A · e−k1t .

Both are simple expressions of monoexponential decay.

PROBLEMS

A.1 The following equation, Cp = 100e−0.347 × t, represents the monoexponential decayof the plasma concentration after an intravenous dose of a drug, where the plasmaconcentration (Cp) is mg/L, t is time (h), and 0.347 h−1 is the first-order eliminationrate constant. Evaluate for Cp when

(a) t = 0 h

(b) t = 2 h

(c) t = 6 h

(d) t = 8 h

(e) t = 10 h

(f) t = 14 h

(g) t = 20 h

What are the ranges of Cp from time zero to infinity?

A.2 The equation Cp = 100e−1.386 × t represents the monoexponential decay of the plasmaconcentration after an intravenous dose of a drug, where the plasma concentration(Cp) is mg/L, t is time (h), and 1.386 h−1 is the first-order elimination rate constant.Evaluate for Cp when:

(a) t = 0 h

(b) t = 0.5 h

(c) t = 1.5 h

(d) t = 2 h

(e) t = 2.5 h

(f) t = 3.5 h

(g) t = 5 h

Page 396: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappA JWBS062-Rosenbaum February 16, 2011 9:12 Printer Name: Yet to Come

376 REVIEW OF EXPONENTS AND LOGARITHMS

What are the ranges of Cp from zero to infinity? How do these answers compare tothe answers in Problem A.1, and what accounts for the difference?

A.3 The equation Cp = 100(1 − e−0.347 × t) represents the growth in the plasma con-centration when a drug is administered at a constant continuous rate (e.g. during anintravenous infusion), where the plasma concentration (Cp) is mg/L, t is time (h) and0.347 h−1 is the first-order elimination rate constant. Evaluate for Cp when:

(a) t = 0 h

(b) t = 2 h

(c) t = 6 h

(d) t = 8 h

(e) t = 10 h

(f) t = 14 h

(g) t = 20 h

What are the ranges of Cp from zero to infinity?

A.4 The equation Cp = 100 (1 − e−1.386 × t) represents the growth in the plasma con-centration when a drug is administered at a constant continuous rate (e.g., during anintravenous infusion), where the plasma concentration (Cp) is mg/L, t is time (h), and1.386 h−1 is the first-order elimination rate constant. Evaluate for Cp when:

(a) t = 0 h

(b) t = 0.5 h

(c) t = 1.5 h

(d) t = 2 h

(e) t = 2.5 h

(f) t = 3.5 h

(g) t = 5 h

What are the ranges of Cp from zero to infinity? How do these answers compare tothe answers in Problem A.3, and what accounts for the difference?

A.5 Given Cp = 23e−0.3t:

(a) Convert to the equation for the logarithm domain.

(b) Draw a plot of the shape you would expect from a plot of ln Cp versus t.

A.6 Calculate the slope given that [ln 2 – ln (0.5)]/(1 − 15) = slope.

A.7 C = C0 · e−kt, where C is a variable that changes with time, C0 is C at time = 0, k isa constant, and t is time. Given that C = 22 mg/L at t = 2 h and C = 5 mg/L at t =10h:

(a) Determine k.

(b) Determine C0.

Page 397: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

APPENDIX B

RATES OF PROCESSES

B.1 Introduction

B.2 Order of a Rate Process

B.3 Zero-Order ProcessesB.3.1 Equation for Zero-Order FillingB.3.2 Equation for Zero-Order EmptyingB.3.3 Time for Zero-Order Emptying to Go to 50% Completion

B.4 First-Order ProcessesB.4.1 Equation for a First-Order ProcessB.4.2 Time for 50% Completion: The Half-Life

B.5 Comparison of Zero- and First-Order Processes

B.6 Detailed Example of First-Order Decay in PharmacokineticsB.6.1 Equations and Semilogarithmic PlotsB.6.2 Half-LifeB.6.3 Fraction or Percent Completion of a First-Order Process Using First-Order

Elimination as an Example

B.7 Examples of the Application of First-Order Kinetics to Pharmacokinetics

B.1 INTRODUCTION

Pharmacokinetics is the study of the manner in which drug concentrations in the bodychange over time after the administration of a dose. The plasma concentration (Cp) ofthe drug is usually the focus in pharmacokinetic studies because it is fairly easy to obtainsamples of plasma and analyze them to determine the drug concentration. A goal in phar-macokinetics is to develop fairly simple equations to describe how the plasma concentrationchanges with time. Clearly, the plasma concentration (dependent variable) at any time willdepend on the:

� Dose administered (a constant in a specific situation)� Time the sample was taken (this is the independent variable)

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

377

Page 398: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

378 RATES OF PROCESSES

Thus, the pharmacokinetic equation will express the dependent variable as a function ofdose and time and generally can be written

Cp = dose (function) time

These equations will have to include expressions for how the underlying processes of drugabsorption, distribution, and elimination (metabolism and excretion) (ADME) influencethe plasma concentration–time relationship. As discussed in this book, almost all processesin pharmacokinetics are either first or zero order. Thus, pharmacokinetic equations mustincorporate mathematical expressions for the rates of zero- or first-order processes inADME when appropriate. Next we discuss the equations and characteristics of zero- andfirst-order processes.

B.2 ORDER OF A RATE PROCESS

The rate of many chemical and physical processes, such as chemical reactions, radioactivedecay, and the draining of liquid from a tank, is proportional to the concentration or amountof one or more of the participants or dependent variables. In pharmacokinetics, whichinvolves primarily simple zero- and first-order processes, the order of the process can beconsidered to be the number of participants involved in this relationship.

B.3 ZERO-ORDER PROCESSES

The rate of a zero-order process is constant and independent of the dependent variable(none of the reactants control the rate). Examples of zero-order processes include filling acar with gas, removing water from a boat or basement using a pump, and the administrationof a drug by an intravenous infusion.

Consider a tank being filled with water by an electric pump:

rate of filling = k0(L/h) (B.1)

where k0 represents the rate of filling. It is a constant controlled by the pump setting. The vol-ume of water (Y) is the dependent variable. As time increases, the filling process proceeds,the volume of water in the tank changes (increases), but the rate of filling does not change.

B.3.1 Equation for Zero-Order Filling

� Let the volume of water in the tank be Y .� Let the initial volume be Y0 (e. g., 5 mL).� Let the rate of filling be k0 (e.g., 10 mL/min).

What volume of water (Y) is in the vessel after 15 min of filling?Fifteen minutes of filling at a rate of 10 mL/min will add 15 × 10 = 150 mL of water.

The tank already contained 5 mL, so the total volume now is 150 + 5 = 155 mL of water.More generally, for a zero-order process,

Y = Y0 + k0 · t (B.2)

Page 399: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

ZERO-ORDER PROCESSES 379

0

20

40

60

80

100

0 10 20 300

20

40

60

80

100

0 10 20 30

Y

Time Time

Slope = k0Slope = –k0

Y0

Y0

Y

(a) (b)

FIGURE B.1 Plot of volume (Y) against time for a zero-order fill (a) and a zero-order emptying(b) process.

Thus, a plot of Y as a function of time will yield a straight line with a slope equal to therate of filling, k0 and an intercept equal to the initial volume, Y0 (Figure B.1a).

B.3.2 Equation for Zero-Order Emptying

Similarly, it can be shown that for zero-order emptying:

Y = Y0 − k0 · t (B.3)

Equation (B.3) is also the equation of a straight line. In this case the slope of the line (therate) is negative (Figure B.1b).

Example B.1 A tank containing 100 L of a liquid is being emptied by a pump which isset at 5 L/h. How many liters would remain after 10 h?

Solution

Y = Y0 − k0 · t

= 100 − 5 × 10 = 50 L

Thus, after 10 h, 50 L of liquid would remain.

B.3.3 Time for Zero-Order Emptying to Go to 50% Completion

At 50% completion of a zero-order emptying process, the amount (volume) is half theoriginal amount: Y = Y0/2. Substituting in equation (B.3) yields

Y0

2= Y0 − k0 · t

Y0

2= k0 · t

t = Y0

k0 · 2

(B.4)

Page 400: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

380 RATES OF PROCESSES

Thus, the time for a zero-order process to go to 50% completion is dependent on the zeroorder rate constant k0 and the initial amount of the dependent variable. The larger the initialquantity, the longer it takes for it to fall by 50%.

B.4 FIRST-ORDER PROCESSES

There are many examples of first-order processes in both nature and medicine. Theseinclude radioactive decay, natural degradation of substances, drug absorption from thegastrointestinal tract, and the elimination of drugs from the body. The growth of money ina savings account is type of a first-order process. A first-order process is characterized by arate that is proportional to the quantity either growing or shrinking under its influence (thedependent variable).

For example, consider a tank full of water being drained through a hole in the bottom.The emptying process is driven by the volume of water in the tank (more specifically, theheight of the water). The volume or height of the water is also the quantity being affected bythe process—it is the dependent variable. As the process continues, the volume decreasesand the height decreases, so the rate of draining also decreases. The rate of a first-orderprocess changes in direct proportion to changes in the dependent variable. Mathematically,

rate of draining ∝ Y (volume of water)

−dY

dt∝ Y

−dY

dt= k · Y

(B.5)

where k is a constant of proportionality or rate constant, with the units t−1 (equivalentto the interest rate in the financial example). The rate of this process is dependent on theamount of a single dependent variable and as a result, it is a first-order process.

B.4.1 Equation for a First-Order Process

The expression for the rate of emptying is given by equation (B.5). To get an expressionfor the dependent variable Y , it is necessary to integrate this equation. Rearranging andintegrating the equation yields

dY

Y= −k · dt (B.6)

∫ ∞

0

dY

Y= −k

∫ ∞

0dt (B.7)

ln Y = −kt + c (B.8)

when t = 0, Y = Y0; thus,

ln Y = ln Y0 − kt

Y = Y0 · e−kt(B.9)

Page 401: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

FIRST-ORDER PROCESSES 381

0

20

40

60

80

100

0 10 20 30

Y

Time

FIGURE B.2 Plot of volume (Y) against time for a first-order emptying process.

Thus, Y decays monoexponentially from Y0 at time = 0 to zero at t = ∞. The relationshipbetween the dependent variable (Y) and time is shown in Figure B.2.

Example B.2 Assume that a tank with an initial volume of 100 mL is being emptied bya first-order process with a rate constant of 0.1 hr−1. Determine the volume at 2, 3.5,7,and 10 h.

Solution Let Y0 = 100 mL and k = 0.1 h−1.

Starting conditions: At t = 0, Y = Y0 = 100

Finishing conditions: At t = ∞, Y = 0

When t = 2 h, Y = 100−0.1×2 and Y = 81.9 mL

When t = 3.5 h, Y = 70.5 mL

When t = 7 h, Y = 50 mL

When t = 10 h, Y = 36.8 mL

B.4.2 Time for 50% Completion: The Half-Life

The half-life (t1/2) of a process is defined as the time it takes for the process to go to 50%completion: When t = t1/2, Y = Y0/2. Substituting into equation (1.15) yields

Y0

2= Y0 · e−kt1/2 (B.10)

1

2= e−kt1/2 (B.11)

Taking the reciprocal of each side gives us

2 = ekt1/2 (B.12)

Page 402: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

382 RATES OF PROCESSES

TABLE B.1 Comparison of Key Characteristics of Zero- and First-Order Processes

Zero-Order Decay First-Order DecayProcess Process

Equation Y = Y0 – kt Y = Y0 · e−kt

Rate: dY/dt k0 Y · kShape of relationship between Y and time Straight line Exponential curveTime for original quantity to fall by 50% Y0/(2 · k0) 0.693/kTime for process to go to completion Y0/k0 3–5 t1/2

a

aCovered in the next section.

and taking natural logarithms gives us

ln 2 = kt1/2 (B.13)

Rearranging, we have

t1/2 = 0.693

k(B.14)

Note: The time it takes a first-order process to go to 50% completion is independent of theinitial value of the dependent variable.

The t1/2 is a reciprocal form of k. Thus, large values of k are associated with small valuesof t1/2, and vice versa. They are constants for a process, and both reflect the rate at whichthe process proceeds. Note that in the previous example, the half-life of the process is 7 h(0.693/0.1), and after 7 h the initial volume had fallen by half.

B.5 COMPARISON OF ZERO- AND FIRST-ORDER PROCESSES

Table B.1 compares key characteristics for zero- and first-order processes.

B.6 DETAILED EXAMPLE OF FIRST-ORDER DECAY INPHARMACOKINETICS

B.6.1 Equations and Semilogarithmic Plots

There are many examples of first-order processes in pharmacokinetics, including mostdrug absorption, distribution, and elimination. This example concentrates on first-orderelimination. Consider the situation where a dose of a drug is injected as an intravenousbolus. Thus, the entire dose is injected at once. In this example it is assumed that the onlyprocess that affects the amount of drug in the body is elimination, a first-order process. Attime zero the entire dose is put into the body. Thereafter the amount of drug in the body(Ab) is solely under the influence of first-order elimination.

When the amount in the body is large at time zero, the rate of elimination is relativelyhigh. But as the amount decreases due to elimination, the rate of the process will also

Page 403: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

DETAILED EXAMPLE OF FIRST-ORDER DECAY IN PHARMACOKINETICS 383

Time Time

Slope = –kAb0 = DoseAb0 = Dose

Ln

Ab

(a) (b)

Ab

FIGURE B.3 Plot of amount of drug in the body (Ab) against time on a linear scale (a) and asemilogarithmic scale (b).

decrease. As a result, the relationship between the amount in the body and time is not linear(Figure B.3a).

The amount of drug in the body decays according to the equation for first-order decay:

Ab = Ab0 · e−kt or Ab = dose · e−kt (B.15)

Where Ab0 is the initial amount of drug in the body, which is equal to the dose and k isfirst-order rate constant for elimination.

Semilogarithmic Plot of Dependent Variable (Ab) Versus Time Taking natural logarithmsof equation (1.15), we have

ln Ab = ln(Ab0) − kt or ln Ab = ln(dose) − kt (B.16)

These are the equations of a straight line. Thus, a plot of ln(Ab) against time yields a straightline of slope (−k), and at the intercept, Ab = Ab0 or dose (Figure B.3b). The slope may becalculated as follows:

slope = ln(Ab1/Ab2)

t1 − t2(B.17)

The elimination rate constant can be determined:

k = −slope = ln(Ab1/Ab2)

t2 − t1(B.18)

B.6.2 Half-Life

As shown previously,

t1/2 = 0.693

k

The t1/2 is a reciprocal form of k. They are constants for a particular drug, and both reflectthe rate at which a drug is eliminated from the body. Obviously, if k is high (i.e., t1/2 is

Page 404: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

384 RATES OF PROCESSES

short), elimination (decay) will proceed rapidly. If k is low (i.e., t1/2 is long), the drugwill be eliminated slowly (slow decay). Because the half-life has much more conceptualmeaning than the rate constant, it is more commonly used clinically when discussing adrug’s pharmacokinetics.

B.6.3 Fraction or Percent Completion of a First-Order Process Using First-OrderElimination as an Example

It is often important in pharmacokinetics to get an approximate idea of how long it takesfor first-order processes such as drug absorption, drug distribution, or drug elimination togo to completion. For example, it may be important to get an idea of how long it will taketo eliminate a drug from the body after a dose.

How long does it take a first-order process to go to completion? The answer is: ∞.Although the answer is accurate, it is of little practical value. An alternative approach tothe problem is to determine the time for a first-order process to almost go to completion(e.g., 90% completion). Recall that

Ab = Ab0 · e−kt (B.19)

where Ab is the amount of drug in the body at time t and Ab0 is the amount at time zerobefore decay starts.

The decay function (e−kt), which starts at 1 and decays to 0, represents the fraction of theoriginal quantity remaining at any time. The fraction of completion is the ratio of amountdecayed to original amount:

fraction completion = Ab0 − Ab

Ab0= 1 − Ab

Ab0= 1 − Ab0 · e−kt

Ab0= 1 − e−kt (B.20)

If e−kt = 0.7, 70% of the original quantity remains (Ab = 0.7Ab0); and 1 − e−kt = 0.3, 30%has decayed or been eliminated. The process is 30% complete.

The number of half-lives for a first-order process to go to any fraction of completion canbe calculated using equation (B.20). For example, the number of half-lives to go to 90%completion may be calculated as follows. At 90% completion, 1 − e−kt = 0.90:

0.9 = 1 − e−kt

But k = 0.693/t1/2:

0.9 = 1 − e−0.693/t1/2t

ln(0.1) = −0.693

t1/2· t (B.21)

t = 3.32t1/2

Thus, it takes 3.32t1/2 for a first-order process to be 90% complete.Table B.2 shows the number of half-lives needed to get to various fractions of completion.

These values hold for any first-order process. Depending on a given situation, it is usuallyconsidered to take anywhere from 3 to 7 half-lives for a first-order process to go to

Page 405: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

EXAMPLES OF THE APPLICATION OF FIRST-ORDER KINETICS TO PHARMACOKINETICS 385

TABLE B.2 Number of Half-Lives for a First-OrderProcess to Go to Certain Fractions of Completion

PercentCompletion 1 − e−kt Time, t1/2

10 0.1 ∼ 16

a

20 0.2 ∼ 13

a

50 0.5 190 0.90 3.395 0.95 4.499 0.99 6.6

aApproximate values.

completion. From a practical standpoint, on average, it can be assumed to take 4 half-livesfor completion.

B.7 EXAMPLES OF THE APPLICATION OF FIRST-ORDER KINETICSTO PHARMACOKINETICS

To answer these questions, assume that all processes in drug absorption, distribution,metabolism, and excretion are first-order processes.

Example B.3 Antacids impair the absorption of many drugs. How long after the ad-ministration of Neoral (cyclosporine) should a patient wait before taking an antacid? Thefirst-order absorption rate constant for cyclosporine in the Neoral formulation is around1.35 h−1.

Solution The half-life for absorption is 0.693/1.35 = 0.51 h. It will take about 4 × 0.5 =2 h to absorb cyclosporine. Thus, the patient should wait at least 2 h after a dose of Neoralbefore taking an antacid.

Example B.4 A physician wishes to give warfarin to a patient who has just stopped takingamiodarone, which has an elimination half-life of around 25 days. Amiodarone is knownto inhibit the metabolism of warfarin. How long could the interaction between amiodaroneand warfarin last?

Solution Amiodarone is an inhibitor of several drug-metabolizing enzymes, includingone of the enzymes that metabolizes warfarin. The interaction can last until amiodarone iseliminated, which will take about 4 months (4 elimination t1/2’s).

Example B.5 A child accidentally takes his grandmother’s digoxin. At the emergencyroom a blood sample reveals a serum concentration of 6 �g/L. The serum concentrationshould be at least � 2, and ideally, � 1.2 �g/L to avoid toxicity. How long will it take forblood levels to become safe? Assume that the plasma concentration is influenced only byfirst-order elimination and that digoxin’s elimination t1/2 is 2 days.

Page 406: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappB JWBS062-Rosenbaum February 16, 2011 9:17 Printer Name: Yet to Come

386 RATES OF PROCESSES

Solution The blood level will fall by 50% each elimination half-life. Thus, it will take2 half-lives, or 4 days, to get to 1.5 �g/L and 3 half-lives, or 6 days, to get to 0.75 �g/L.The exact time to get to 1.2 �g/L can be estimated using the equation for first-order decay[equation (B.9)], the serum concentration is the dependent variable, and the rate constantis 0.693/2 = 0.347 day−1. This provides an answer of 4.6 days.

Example B.6 The distribution of a drug from the plasma to the tissues is usually a first-order process. The disappearance of the drug from the plasma due to tissue uptake can bedescribed by the equation

Cp = A · e−�t

where Cp is the plasma concentration, A is a constant, � is a first-order rate constant fordistribution, and t is time.

If gentamicin and digoxin have first-order distribution rate constants of about 0.14 min−1

and 0.52 h−1, respectively. Approximately how long will it take for distribution to go toabout 95% completion for each drug?

Solution It will take 4(� · t1/2) to get to 95% completion. For gentamicin this will be4(0.693/0.14), or 19.8 min, and for digoxin it will take 4(0.693/0.52), or 5.4 h.

Page 407: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

APPENDIX C

CREATION OF EXCEL WORKSHEETSFOR PHARMACOKINETIC ANALYSIS

C.1 Measurement of AUC and ClearanceC.1.1 Trapezoidal RuleC.1.2 Excel Spreadsheet to Determine AUC0→∞ and Clearance

C.2 Analysis of Data from an Intravenous Bolus Injection in a One-Compartment Model

C.3 Analysis of Data from an Intravenous Bolus Injection in a Two-Compartment Model

C.4 Analysis of Oral Data in a One-Compartment Model

C.5 Noncompartmental Analysis of Oral Data

Conventions

Worksheet Instructions Quantities that actually have to be entered in the cells of theworksheets are presented within quotation marks and are italicized.

Worksheet Layout Data or values that must be entered into the worksheet are coloredred and model parameters are colored green. Various other colors are used to distinguishdifferent manipulations of the given data.

C.1 MEASUREMENT OF AUC AND CLEARANCE

The measurement of the area under the plasma concentration–time curve (AUC) is oftenan important part of a pharmacokinetic analysis. It is used to assess drug exposure and tocalculate clearance (Cl). Recall that

Cl = F · D

AUC∞0

where F is the fraction of the dose (D) absorbed. The AUC is most commonly measuredusing the trapezoidal rule. Descriptions of the trapezoidal rule, the measurement of theAUC, and the estimation of clearance are given below.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

387

Page 408: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

388 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

C.1.1 Trapezoidal Rule

When a dose of a drug is administered, plasma concentrations can be determined at varioustimes and then plotted against time. Figure C.1 shows the typical profile observed after oraladministration.

1. The first step in using the trapezoidal rule to estimate the AUC is to use the measureddata points to split the curve into a series of segments, each of which (except thefirst, which is a triangle) is a trapezoid (a four-sided shape with two parallel sides).In Figure C.1 the 10 measured data points produce 10 segments.

2. The area of each segment must now be determined. The area of a trapezoid (FigureC.1) is given by

area of a trapezoid = average height · base (C.1)

Note that this is also the formula for the area of a triangle (the first segment). Thus,the area of each trapezoid produced from the plasma concentration–time data can becalculated as follows:

area = Cpn + Cpn+1

2· (tn+1 − tn) (C.2)

3. The areas of each of the 10 segments are combined to obtain the AUC from zero tothe last time point.

4. The AUC from the last time point to infinity must now be calculated. At the time ofthe last time point it is assumed that the fall in the plasma concentration is influencedonly by first-order elimination (no ongoing absorption or distribution are assumed tooccur by this time). Under these conditions, the AUC from the last time point (Cplast)

1 2 3 4 5 6 7 8 9 10

Cp

Time

Cpn

Cpn+1

tn tn+1

Area = Average height • Base = (Cpn+ Cpn+1)/2 • (tn+1 – tn)Trapezoid

FIGURE C.1 Typical plasma concentration–time profile after oral administration; The curve issegmented into trapezoids using the data points.

Page 409: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

MEASUREMENT OF AUC AND CLEARANCE 389

to infinity is given by

AUCCplast→∞ = Cplast

k

where k is the overall first-order elimination rate constant. When Cp is affected onlyby first-order elimination, k is the negative slope for the fall in ln Cp with time.

5. All the individual areas are then summed to obtain AUC0→∞.

6. The drug’s clearance may be determined from the AUC if the bioavailability factor(F) is known:

Cl = F · D

AUC∞0

If F is not known, the parameter Cl/F, which is known as oral clearance, is determined:

Cl

F= D

AUC∞0

C.1.2 Excel Spreadsheet to Determine AUC0→∞ and Clearance

The empty shell of a worksheet to determine the AUC and clearance is shown in Figure C.2.This will be used as a guide to set up an operational worksheet to determine the AUC andclearance. Open an empty worksheet in Excel and copy the column headings and layoutshown in Figure C.2. Save the worksheet as “Worksheet for AUC and Cl” or by a similarappropriate name.

The worksheet will be set up using the plasma concentration–time data in Table C.1.These data were obtained after the oral administration of a 100-mg dose of a drug.

1. Enter the time and Cp data in the upper Given Data section of the worksheet only.

2. Enter the drug dose (100 mg).

3. Plot the data to visualize it and ensure that there are no outliers. Highlight the data(not the column headings), click on the Insert tab on the tool bar, choose Scatter, thenselect the Scatter with Straight Lines and Markers. Click on the Layout tab and entera chart heading and label each axis.

4. All the calculations and data manipulation procedures will be done in the lowersection of the worksheet. The given data must be placed in the time and concentrationcolumns in this lower section. Do not copy and paste the data into these columns.Instead, the given data cells in the upper section will be referenced. In this way, whennew data are entered in the Given Data section, the lower section will automaticallyupdate with the new data. The cells containing the data are referenced as follows:In the lower section, go the first cell in the time column and enter “=” (without thequotation marks), then click on the first time cell in the given data column in theupper area. Then hit Enter. Copy this formula throughout the time column. To do this,go back to the first cell containing the reference and point to the lower right-handcorner of the cell. When a cross appears, hold down the mouse and drag it throughthe rest of the columns. Repeat the entire procedure for the concentration data.

Page 410: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

390 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

WORKSHEET FOR AUC AND CL

Given DataDos 100e (mg)

Time (hr) Cp (mg/L)CONDITION

0 00.4 0.380.8 0.61 0.56

1.6 0.662 0.623 0.534 0.548 0.49

10 0.3412 0.3224 0.11

AUC and Clearance Determination

Dose (mg) 0

Time (hr) Cp (mg/L) ln Cp AvCp ∆ Time AUC of segmentCONDITION

AUC0-24

Slope (hr-1) AUC

k (hr-1) AUC (mg.hr/ L)

Cl/F (L/hr)

FIGURE C.2 Empty Excel worksheet for the determination of AUC and clearance.

TABLE C.1 Plasma Concentration Time Data After a 100 mgOral Dose

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0 0 3 0.5300.4 0.380 4 0.5400.8 0.600 8 0.4901 0.560 10 0.3401.6 0.660 12 0.3202 0.620 24 0.110

Page 411: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

MEASUREMENT OF AUC AND CLEARANCE 391

5. Enter the dose in the lower section of the worksheet by referencing the cell containingits value in the upper area.

6. The values for ln Cp for the last three data points will be entered using the ExcelLn function. Thus, in the first of these three ln Cp cells, enter “=Ln(” (without thequotation marks) and then click on the corresponding value of Cp and close theparentheses. Copy this formula throughout the two additional ln Cp cells.

7. The slope of the ln Cp versus time plot must be determined to calculate k. Enter thecell where for the value of the slope is to appear. The slope is determined using theslope function in Excel. This is one of the function keys that are accessed from theAutosum (

∑) symbol on Excel’s tool bar. Click on the arrow next to the Autosum

key and locate the slope function. It will probably be found in Other Functions,in the Statistical category. Once the slope function has been found, it is necessaryto enter the range of y values (ln Cp) and x values (time) that are to be used forthe calculation. To do this, click on the known y’s box in the pop-up window andthen click throughout the relevant ln Cp cells in the worksheet. Do the same for theknown x’s. Make sure that you go in the same direction when you highlight the y’sand x’s. Hit Enter.

8. Go to the cell where the value of k (equal to the negative slope) is to appear and enter“= -” and then click on the cell containing the value of the slope

9. The AUC of each trapezoidal segment will be calculated in stages:

a. Average Cp. The formula (Cpn + Cpn+1)/2 must be entered in the cells of theaverage Cp column. In the top cell enter “=(”, then reference the cell containingCpn, enter “+” and click on the cell containing Cpn+1, and enter “)/2”. Copy theformula throughout the column to the cell corresponding to the next-to-last Cp(there isn’t another Cp with which to average the last Cp).

b. Time difference. The formula to calculate delta t, tn+1 − tn, now has to be enteredin the top cell of the delta t column. Enter “=(”, then click on the cell containingtn+1, then enter “−” and click on the cell containing tn. Close the parenthesis.Copy the formula down through the next-too last cell in the column.

c. Area of each segment. The formula to calculate the area of each segment, (Cpn

+ Cpn+1)/2 ∗ (tn+1 − tn), must now be entered in the top cell of this column.Enter “=”, then reference on the average Cp value, then enter “∗” and referencethe corresponding delta t value. Copy the formula throughout the column.

d. AUC from zero to last data point. Add up all the individual areas (AUC from 0to Cplast) using the AutoSum function on the tool bar.

e. AUC from last data point to infinity. The formula Cplast/k to calculate the areafrom the last Cp value to infinity must be entered the cell where this value is toappear. Enter “=”, reference the cell containing the value of the last Cp, thenenter “/”, reference the value of k, and hit Enter.

f. AUC from zero to infinity. Use the AutoSum function to calculate AUC0→∞.

g. Calculate Cl/F. (Note: Cl cannot be determined because F is unknown.) Clickon the cell where the value of Cl/F is to appear, enter “=”, reference the cellcontaining the dose, then enter “/”, reference the value of AUC0→∞, and then hitEnter.

Save the completed worksheet as “Worksheet for AUC and Cl.” The completed worksheetis shown in Figure C.3.

Page 412: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

392 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

WORKSHEET FOR AUC AND CL

Given DataDose (mg) 100

Time (hr) Cp (mg/L)CONDITION

0 00.4 0.380.8 0.61 0.56

1.6 0.662 0.623 0.534 0.548 0.49

10 0.3412 0.3224 0.11

AUC and Clearance Determination

Dose (mg) 100

Time (hr) Cp (mg/L) ln Cp AvCp Δ Time AUC of segmentCONDITION

0 0 0.19 0.4 0.0760.4 0.38 0.49 0.4 0.1960.8 0.6 0.58 0.2 0.116

1 0.56 0.61 0.6 0.3661.6 0.66 0.64 0.4 0.256

2 0.62 0.575 1 0.5753 0.53 0.535 1 0.5354 0.54 0.515 4 2.068 0.49 0.415 2 0.83

10 0.34 -1.08 0.33 2 0.6612 0.32 -1.14 0.215 12 2.5824 0.11 -2.21

AUC0-24 8.25

Slope (hr-1) -0.084 AUC 1.32

k (hr -1) 0.084 AUC (mg.hr/ L) 9.567

Cl/F (L/hr) 10.453

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0 5 10 15 20 25

Cp

(m

g/L

)

Time (hr)

Cp Versus Time

FIGURE C.3 Completed Excel worksheet for the determination of AUC and clearance.

Note: When this worksheet is used for other data, remember to:

� Reenter the appropriate dose.� Make sure that all the units are consistent throughout the worksheet and that they are

all labeled correctly. For example, if the units of Cp are �g/L, make sure that the doseis entered in �g.

Page 413: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

ANALYSIS OF DATA FROM AN INTRAVENOUS BOLUS INJECTION 393

C.2 ANALYSIS OF DATA FROM AN INTRAVENOUS BOLUS INJECTION INA ONE-COMPARTMENT MODEL

This worksheet will be created by adapting the previous worksheet for the determinationof AUC and clearance.

1. Open the completed worksheet for the determination of the AUC and clearance.

2. Change the title of the worksheet to “Worksheet for the Analysis of Data from anIV Bolus Injection in a One-Compartment Model.”

3. Save the worksheet as “PK Analysis: One-Compartment Model IV.”

4. Delete the diagram and/or the chart or graph.

5. Enter the data obtained after the IV administration of 50-mg dose of a drugfrom Table C.2 into the upper area of the worksheet. Make sure that the dose isupdated.

6. The data in the lower calculation area should update automatically.

7. In the upper Given Data area, plot the data on a linear and a semilogarithmicscale to make sure that it appears that the one-compartment model holds for thisdata set, and that there are no odd data points that are very different from theothers.

8. Select the two columns of data (not the headings).

9. Go to Insert; in the Chart area, choose Scatter, Smooth lines, and markers.

10. This first chart will be the linear plot of Cp versus time. From the Layout tab add atitle to the graph and label each axis.

11. Copy and paste this graph to create a duplicate alongside it. On the copy, reformatthe y-axis to the logarithmic scale: right click on the y-axis, select Format the Axis,and choose Logarithmic Scale. At this point it may be appropriate to change thepoint where the horizontal axis crosses. Modify the graph title and label for they-axis.

12. Rename the lower calculation area “Parameter Determination”.

13. Continue the list of model parameters below the cell labeled “k (1/h).” Thus, addthe following names:

a. t1/2(h)

b. Intercept (I) (note that this will be the ln scale)

c. Cp0 (mg/L) (note this is the anti-ln of the intercept)

TABLE C.2

Time (h) Cp (mg/L) Time (h) Cp (mg/L)

0.1 2.45 3 1.370.2 2.40 5 0.920.5 2.26 7 0.621 2.05 10 0.341.5 1.85 12 0.232 1.68 15 0.12

Page 414: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

394 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

d. Vd (L)

e. Cl (k ∗ Vd) (L/h) (note that this clearance will be determined from kand Vd)

f. Cl (Cp0/k) (L/h) (note that this clearance will be determined from AUC calcu-lated from Cp0 and k)

g. Cl (trapezoidal) (L/h) (note that this clearance will be determined from AUCmeasured using the trapezoidal rule) Note that in this problem the area fromtime zero to the first data point (0–1) must be added.

14. Extend the calculation of the ln Cp values from the third-to-last data point all theway up to the top of the column. Hold the mouse over the bottom right corner of afilled ln Cp cell, and drag it up the column.

15. Go to the cell that contains the calculation of the slope. Extend the data points usedto calculate the slope to include all the data points.

16. Enter the formula for the listed parameters in the cells adjacent to the label (bolditalic type below indicates that the cell containing the value of that quantity shouldbe referenced):

a. t1/2(h): Enter “= 0.693/k” (but not the quotation marks).

b. Intercept (I). Note that this is from the ln Cp versus time plot. It will be in theln scale. Find the intercept function from the list accessed from AutoSum onthe upper tool bar. It will be found in the statistical functions. Enter the rangeof y (lnCp) and x values to be used for its calculation (all of them).

c. Cp0 (mg/L): Enter “= EXP(I)”.

d. Vd (L): Enter “= Dose/Cp0”.

e. Cl (k ∗ Vd)): Enter “= k ∗ Vd”.

f. Cl (Cp0/k) (L/h)): Enter “Dose/(Cp0/k)”.

h. Cl (trapezoidal): Enter : “Dose/AUC∞0 ”.

Note: When this worksheet is used for other data, it is very important to check the units.Make sure that all the units are consistent throughout the worksheet and that they are alllabeled correctly. For example, if the units of Cp are �g/L, make sure that the dose isentered in �g.

The completed worksheet is shown in Figure C.4.

C.3 ANALYSIS OF DATA FROM AN INTRAVENOUS BOLUS INJECTION INA TWO-COMPARTMENT MODEL

1. Set up empty worksheet. Figure C.5 shows the empty worksheet for the determinationof the parameters of the two-compartment model. Use this layout and cell labels tocreate your own worksheet.

2. Enter data. Enter the data from Table E8.1 (page 176), obtained after a 100 mg dose,in the top Given Data section. Plot the data on a semilogarithmic scale to ensure thatvisually the data appear to fit a two-compartment model.

Page 415: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

ANALYSIS OF DATA FROM AN INTRAVENOUS BOLUS INJECTION 395

WORKSHEET FOR ANALYSIS OF DATA FROM AN IV BOLUS INJECTION IN A 1-COMPARTMENT MODELGiven Data

Dose (mg) 50

Time (hr) Cp (mg/L)CONDITION

0.1 2.450.2 2.400.5 2.261.0 2.051.5 1.852.0 1.683.0 1.375.0 0.927.0 0.62

10.0 0.3412.0 0.2315.0 0.12

Parameter Determination

Dose (mg) 50

Time (hr) Cp (mg/L) ln Cp AvCp Δ Time AUC of segmentCONDITION

0.1 2.45 0.896 2.426 0.1 0.2430.2 2.40 0.876 2.332 0.3 0.7000.5 2.26 0.816 2.155 0.5 1.0771.0 2.05 0.716 1.950 0.5 0.9751.5 1.85 0.616 1.764 0.5 0.8822.0 1.68 0.516 1.524 1.0 1.5243.0 1.37 0.316 1.146 2.0 2.2925.0 0.92 -0.083 0.768 2.0 1.5367.0 0.62 -0.485 0.477 3.0 1.431

10.0 0.34 -1.085 0.283 2.0 0.56512.0 0.23 -1.483 0.176 3.0 0.52715.0 0.12 -2.087

Slope (1/hr) -0.201 AUC1-last 11.751

k (1/hr) 0.201 AUC0-1 0.248t1/2 (hr) 3.454 AUC last−∞ 0.618Intercept (I) 0.917 AUC0−∞mg.hr/L 12.616Cp0 (mg/L) 2.50Vd (L) 20.0 Cl/F (L/hr) 3.96Cl -kVd (L/hr) 4.01Cl -Cp0 /k (L/hr) 4.01Cl Trapezoidal (L/hr) 3.96

0.000.501.001.502.002.503.00

0.0 5.0 10.0 15.0 20.0

Cp(m

g/L)

Time (hr)

Plot of Cp Versus Time

-3-2-1012

0 5 10 15 20

LnCp

Time (hr)

Plot of LnCp Verus Time

FIGURE C.4 Completed Excel worksheet for pharmacokinetic analysis of an intravenous bolusinjection in a one-compartment model.

3. Reference data in lower calculation area. Reference the given time and concentrationdata in the lower Calculation Area of the worksheet. Use these data in the lower areafor all future calculations. Create time and ln Cp columns. Reference the given datato set up time and ln Cp columns.

4. Determine B and �. Determine B and � from the last three data points, whichare described by the equation Cp′ = B ∗ e−�t, where Cp′ = Cp for the last threedata points. Use the built-in function to determine the slope and intercept forthe ln Cp′ versus time relationship for the last three data points. Determine �and B.

Page 416: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

396 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

Worksheet for the Analysis of data for an IV Injection in a 2-compartment ModelGIVEN DATADose mg 100

Time(hr) Cp (mg/L)0.1 7.950.2 6.380.4 4.250.7 2.57

1 1.791.5 1.27

2 1.073 0.894 0.766 0.568 0.42

12 0.23

Calculation AreaGIVEN DATA LN SCALE Cp'=B*exp(-β*t) Isolating DistributionExponentTime(hr) Cp (mg/L) Time LnCp Time Cp' Time Cp-Cp' LnCp-Cp' Time LnCp' LnCp-Cp'

0.1 7.950.2 6.380.4 4.250.7 2.57

1 1.791.5 1.27

2 1.073 0.894 0.766 0.568 0.42

12 0.23DOSE(mg) 100

ParametersβSlope αSlope T1/2

βhr-1 α hr-1 k21 hr-1LnB LnA k10 hr-1B mg/L A mg/L k12 hr-1

AUCmg.hr/L

CL L/hrV1 LVβ LVdss LCld L/hr

FIGURE C.5 Empty Excel worksheet for determination of the parameters of the two-compartmentmodel after an intravenous bolus injection.

5. Determine A and �:

a. Calculate Cp′ at early times. Consider the equation Cp′ = B ∗ e−�t; at latertimes, Cp = Cp, but at earlier times, when distribution is occurring, Cp′ � Cp.The values of Cp′ at these earlier times must be calculated. Use B and � andthe equation Cp′ = B ∗ e−�t to determine the values of Cp′ that correspond tothe times of the given data (Cp). When doing this it is important to use theabsolute address of the value of B and � since they must remain constant whenthe formula for Cp′ is copied throughout the column. In the example worksheet,B and � are in cells E41 and E39, respectively. The absolute address of a cellis given by including a $ sign before the column letter and row number. Thus,the absolute addresses of B and � are $E$41 and $E$39, respectively. Enterthe formula to calculate Cp′ in the first Cp′ cell: “=$E$41∗EXP(-$E$39∗G24)”.Copy the formula throughout the rest of the Cp′ column (time will change foreach cell, but B and � will remain constant).

Page 417: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

ANALYSIS OF DATA FROM AN INTRAVENOUS BOLUS INJECTION 397

b. Calculate Cp − Cp′ to isolate the line described by A and �: Cp − Cp′ = A ∗ e−�t.Calculate C − Cp′ for those values of Cp′ that are less than Cp. Enter the valuesof ln(Cp − Cp′). Use the built-in functions to determine the slope and interceptof the ln(Cp − Cp′)–time relationship. Determine A and �.

6. Plot lines corresponding to the two exponential functions. On the farthermost rightarea, create adjacent columns of time, ln Cp′ and ln(Cp − Cp′). Plot a scatter plot ofln Cp′ and ln(Cp − Cp′) against time.

7. Determine the micro rate constants and the pharmacokinetic parameters. Enter theformula for each parameter (see Sections 8.7.2 and 8.7.3). Reference any parameterfrom the worksheet that is needed for the calculation.

The complete worksheet is shown in Figure C.6.

Worksheet for the Analysis of data for an IV Injection in a 2-compartment ModelGIVEN DATADose mg 100

Time(hr) Cp (mg/L)0.1 7.950.2 6.380.4 4.250.7 2.57

1 1.791.5 1.27

2 1.073 0.894 0.766 0.568 0.42

12 0.23

Calculation AreaGIVEN DATA LN SCALE Cp'=B*exp(-β*t) Isolating DistributionExponentTime(hr) Cp (mg/L) Time LnCp Time Cp' Time Cp-Cp' LnCp-Cp' Time LnCp' LnCp-Cp'

0.1 7.95 0.1 2.07 0.10 1.36 0.10 6.59 1.89 0.10 0.31 1.890.2 6.38 0.2 1.85 0.20 1.34 0.20 5.04 1.62 0.20 0.29 1.620.4 4.25 0.4 1.45 0.40 1.30 0.40 2.95 1.08 0.40 0.26 1.080.7 2.57 0.7 0.94 0.70 1.24 0.70 1.33 0.28 0.70 0.22 0.28

1 1.79 1 0.58 1.00 1.19 1.00 0.60 -0.51 1.00 0.17 -0.511.5 1.27 1.5 0.24 1.50 1.10 1.50 0.17 -1.78 1.50 0.10 -1.78

2 1.07 2 0.07 2.00 1.02 2.00 2.00 0.023 0.89 3 -0.12 3.00 0.88 3.00 3.00 -0.134 0.76 4 -0.27 4.00 0.76 4.00 4.00 -0.286 0.56 6 -0.58 6.00 0.56 6.00 6.00 -0.578 0.42 8 -0.87 8.00 0.42 8.00 8.00 -0.87

12 0.23 12 -1.47 12.00 0.23 12.00 12.00 -1.47DOSE(mg) 100

ParametersβSlope -0.15 αSlope -2.62 T1/2 4.65

βhr-1 0.15 α hr-1 2.62 k21 hr-1 0.50LnB 0.32 LnA 2.14 k10 hr-1 0.79B mg/L 1.38 A mg/L 8.47 k12 hr-1 1.49

AUCmg.hr/L 12.47CL L/hr 8.02V1 L 10.16Vβ L 53.81Vdss L 40.67CLd L/hr 15.11

-2.00

0.00

2.00

4.00

0.00 2.00 4.00 6.00 8.00 10.00 12.00

LnCp' LnCp-Cp'

0

20

0 5 10 15

Cpm

g/L

Time (hr)

Cp Against Time

0.1

1

10

0 5 10 15

C p

Time (hr)

LnCp Against Time

FIGURE C.6 Completed Excel worksheet for determination of the parameters of the two-compartment model after an intravenous bolus injection.

Page 418: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

398 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

C.4 ANALYSIS OF ORAL DATA IN A ONE-COMPARTMENT MODEL

1. Set up empty worksheet. Figure C.7 shows the empty worksheet for the determinationof the parameters from data obtained after oral administration in a one-compartmentmodel (see Table E9.1A, page 190). Use this layout and cell labels to create yourown worksheet.

2. Enter data. Enter the data in the top Given Data section. Plot the data on the regularlinear and semilogarithmic scales to ensure that visually the data appears to fit aone-compartment model and make sure that there are no outliers.

3. Reference data in lower calculation area. Reference the given time and concentrationdata in the lower Calculation Area of the worksheet. Use these data in the lower areafor all future calculations.

4. Create time and ln Cp columns. Reference the given data to set up adjacent time andln Cp columns.

5. Determine I and k. Determine the intercept (I) and slope (k) of the line created fromthe last three data points of ln Cp against time. These points are described by theequation Cp′ = I ∗ e−kt, where Cp′ = Cp for the last three data points. Use the built-in

Worksheet for the Analysis of Oral Data in a 1-Compartment ModelGIVEN DATADose mg 100

Time(hr) Cp (mg/L)0 0

0.6 2.740.8 3.131 3.37

1.4 3.551.8 3.52 3.43

2.6 3.123 2.894 2.337 1.1712 0.37

Calculation Area

GIVEN DATA LN SCALE Cp`=I (e-k*t) Isolating Absorption ExponentTime(hr) Cp (mg/L) Time LnCp Time Cp' Time Cp`-Cp LnCp`-Cp Time LnCp' LnCp`-Cp

DOSE(mg)

ParametersElinSlope AbspSlope T1/2 hr

khr-1 ka hr-1 Vd/F LLnI LnI AUC

I mg/L I mg/L Cl/F L/hrAUC

Cl/F L/hr(k*Vd)

FIGURE C.7 Empty Excel worksheet for determination of the parameters of the one-compartmentmodel with first-order absorption.

Page 419: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

NONCOMPARTMENTAL ANALYSIS OF ORAL DATA 399

slope and intercept function to determine ln I and the slope from the ln Cp′ versustime relationship. Determine I [I = exp(ln I)], and k = (−slope).

6. Determine ka and I from the absorption line:

a. Calculate Cp′ at early times. Consider the equation Cp′ = I ∗ e−kt; at latertimes Cp′ = Cp, but at earlier times, when absorption is underway, Cp′ � Cp.The values of Cp′ at these earlier times must be calculated. Use I and k and theequation Cp′ = I ∗ e−kt to determine the values of Cp′ that correspond to the timesof the given data (Cp). When doing this it is important to use the absolute addressof the value of I and k since they must remain constant when the formula for Cp′

is copied throughout the column. In the example worksheet I and k are in cellsE41 and E39, respectively. The absolute address of a cell is given by includinga $ sign before the column letter and row number. Thus, the absolute addressesof I and k are $E$41 and $E$39, respectively. Enter the formula to calculate Cp′

in the first Cp′ cell: “=$E$41∗EXP(-$E$39∗G24)” Copy the formula throughoutthe rest of the Cp′ column (time will change for each cell but I and k will remainconstant).

b. Calculate Cp′−Cp and isolate the line described by ka: Cp′ − Cp = I ∗ e−kat.Calculate Cp′ − Cp for those values of Cp′ that are greater than Cp. Enter thevalues of ln(Cp′ − Cp). Use the built-in functions to determine the slope andintercept of the ln(Cp′ − Cp)–time relationship. Determine I and ka.

7. Plot the lines corresponding to the two exponential functions. On the farthermostright area create adjacent columns of time, ln Cp′ and ln(Cp′ − Cp). Plot a scatterplot of ln Cp′ and ln(Cp′ − Cp) against time.

8. Determine half-life, clearance, and volume of distribution. Enter the formula foreach parameter. Vd/F = dose ∗ ka/I ∗ (ka − k) and Cl/F = dose/AUC, where AUC =I ∗ (1/k − 1/ka). Reference any parameter from the worksheet that is needed for thecalculation.

The complete worksheet is shown in Figure C.8.

C.5 NONCOMPARTMENTAL ANALYSIS OF ORAL DATA

This worksheet will be created to enable AUC, Cl/F, t1/2, Cmax, and Tmax to be estimatedafter the administration of a dose by any route. In this example data obtained after oraladministration will be analyzed. The worksheet will be set up to analyze data presentedin Chapter 10 from a hypothetical drug–drug interaction study to evaluate if fluconazolealters the pharmacokinetics of fictitious drug lipoamide. The data obtained after a 120-mgoral dose of lipoamide in the absence (control) and in the presence (test) of fluconazole asdescribed in example 10.3 (page 208) are provided in Table C.3.

The new worksheet will be created from the worksheet for the determination of the AUCand clearance.

1. Open the completed worksheet for the determination of AUC and Cl. Delete anygraphs. Save it as “Worksheet for NCA After Oral Administration”.

2. Change the name of the lower calculation area of the worksheet from “AUC andClearance Determination” to “Parameter Determination Using NCA”.

Page 420: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

400 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

Worksheet for the Analysis of Oral Data in a 1-Compartment ModelGIVEN DATADose mg 100

Time(hr) Cp (mg/L)0 0

0.6 2.740.8 3.131 3.37

1.4 3.551.8 3.52 3.43

2.6 3.123 2.894 2.337 1.1712 0.37

Calculation Area

GIVEN DATA LN SCALE Cp`=I (e-k*t) Isolating Absorption ExponentTime(hr) Cp (mg/L) Time LnCp Time Cp' Time Cp`-Cp LnCp`-Cp Time LnCp' LnCp`-Cp

0 0 0 0.00 5.85 0.00 5.85 1.77 0.00 1.77 1.770.6 2.74 0.6 1.01 0.60 5.10 0.60 2.36 0.86 0.60 1.63 0.860.8 3.13 0.8 1.14 0.80 4.87 0.80 1.74 0.55 0.80 1.58 0.551 3.37 1 1.21 1.00 4.65 1.00 1.28 0.25 1.00 1.54 0.25

1.4 3.55 1.4 1.27 1.40 4.24 1.40 0.69 -0.37 1.40 1.44 -0.371.8 3.5 1.8 1.25 1.80 3.87 1.80 0.37 -1.00 1.80 1.35 -1.002 3.43 2 1.23 2.00 3.69 2.00 0.26 -1.34 2.00 1.31 -1.34

2.6 3.12 2.6 1.14 2.60 3.22 2.60 0.10 -2.33 2.60 1.17 -2.333 2.89 3 1.06 3.00 2.93 3.00 3.00 1.084 2.33 4 0.85 4.00 2.33 4.00 4.00 0.857 1.17 7 0.16 7.00 1.17 7.00 7.00 0.1612 0.37 12 -0.99 12.00 0.37 12.00 12.00 -0.99

DOSE(mg) 100

ParametersElinSlop -0.23 AbspSlo -1.57 T1/2 hr 3.01

khr-1 0.23 ka hr-1 1.57 Vd/F L 20.02LnI 1.77 LnI 1.80 AUC 21.71

I mg/L 5.85 I mg/L 6.07 Cl/F L/hrAUC 4.61

Cl/F L/hr(k*Vd 4.61

-3.00

-2.50

-2.00

-1.50

-1.00

-0.50

0.00

0.50

1.00

1.50

2.00

0.00 2.00 4.00 6.00 8.00 10.00 12.00 14.00

LnCp `(blue) and Ln(Cp`-Cp) (red)Versus Time

0

1

2

3

4

0 5 10 15

Cp

(m

g/L

)

Time (hr)

Cp Against Time

0.1

1

10

0 5 10 15

Ln

Cp

Time (hr)

LnCp Against Time

FIGURE C.8 Completed Excel worksheet for determination of the parameters of the one-compartment model with first-order absorption.

TABLE C.3

Cp (�g/L) Cp (�g/L)

Time (h) Control Inhibitor Time (h) Control Inhibitor

0.0 0.0 0.0 1.2 52.0 160.90.2 38.1 117.8 2.0 46.3 143.20.4 50.6 156.7 4.0 34.4 106.40.6 54.1 167.4 10.0 14.1 43.60.8 54.3 168.0 15.0 6.7 20.71.0 53.3 165.0 24.0 1.8 5.4

Page 421: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

NONCOMPARTMENTAL ANALYSIS OF ORAL DATA 401

TABLE C.4

Parameter Control (Placebo) Test (Fluconazole)

Cl/F (L/h)

t1/2 (h)

Cmax (�g/L)

Tmax (h)

3. Enter (or copy and paste) the control and test data in the upper Given Data area toexactly replace the existing data in the first two columns. There will be one more Cpcolumn in the new worksheet. Insert a scatter chart to view the data. Add a title andlabel the axis.

4. Enter the value of the dose (make sure that the units are labeled correctly and thatthat they are uniform throughout the worksheet).

5. Note that the control data should automatically have been placed in the lower calcu-lation area.

WORKSHEET FOR NCAGiven Data

Dose (mcg) 120000.00

Time (hr) Cp (mcg/L) Cp (mcg/L)PHASE Control Test

0 0.0 0.00.2 38.1 117.80.4 50.6 156.70.6 54.1 167.40.8 54.3 168.0

1 53.3 165.01.2 52.0 160.9

2 46.3 143.24 34.4 106.4

10 14.1 43.615 6.7 20.724 1.8 5.4

Parameter Determination Using NCA

Dose (mcg) 120000.00 Dose (mcg) 120000.00

Time (hr) Cp (mcg/L) ln Cp AvCp Δ Time AUC of segment Time (hr) Cp (mcg/L) ln Cp AvCp Δ Time AUC segmentPHASE Control (mcg/L) (hr) (mcg.hr/L) PHASE Test (mcg/L) (hr) (mcg.hr/L)

0 0.0 17.48 0.20 3.50 0 0.0 58.88 0.20 11.780.2 35.0 41.61 0.20 8.32 0.2 117.8 137.23 0.20 27.450.4 48.3 50.49 0.20 10.10 0.4 156.7 162.04 0.20 32.410.6 52.7 53.15 0.20 10.63 0.6 167.4 167.67 0.20 33.530.8 53.6 53.29 0.20 10.66 0.8 168.0 166.49 0.20 33.301 53.0 52.43 0.20 10.49 1 165.0 162.96 0.20 32.59

1.2 51.9 49.09 0.80 39.27 1.2 160.9 152.05 0.80 121.642 46.3 40.38 2.00 80.75 2 143.2 124.79 2.00 249.584 34.4 24.30 6.00 145.80 4 106.4 74.98 6.00 449.8510 14.2 2.65 10.46 5.00 52.28 10 43.6 3.77 32.15 5.00 160.7515 6.8 1.91 4.27 9.00 38.39 15 20.7 3.03 13.08 9.00 117.6824 1.8 0.58 24 5.4 1.69

AUC0-Cplast 410.17 AUC0-24 1270.55

Slope (hr-1) -0.15 AUC Cp last- 12.02 Slope (hr-1) -0.15 AUC 24-∞

36.50

λ (hr-1) 0.15 AUC0- mcg.hr/L 422.18 λ (hr-1) 0.15 AUC0- mcg.hr/L 1307.05

Cl/F (L/hr) 284.24 Cl/F (L/hr) 91.81t1/2 (hr) 4.68 t1/2 (hr) 4.66Cmax(mcg/L) 53.58 Cmax(mcg/L) 167.96Tmax(hr) 0.80 Tmax(hr) 0.80

Results Summary

CONTROL TESTAUC mcg.hr/L 422.18 1307.05Cl/F L/hr 284.24 91.81t1/2 (hr) 4.68 4.66Cmax (mcg/L) 53.58 167.96Tmax (hr) 0.80 0.80

0.0

30.0

60.0

90.0

120.0

150.0

180.0

0 5 10 15 20 25 30

Cp(mcg/L)

Tim e(hr)

Plasma Concentration of LipoamideWith (solid line) and Without (dashed

line) Fluconazole

FIGURE C.9 Completed Excel worksheet for noncompartmental analysis.

Page 422: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappC JWBS062-Rosenbaum February 16, 2011 9:26 Printer Name: Yet to Come

402 CREATION OF EXCEL WORKSHEETS FOR PHARMACOKINETIC ANALYSIS

6. In the lower calculation area, change the label k in the cell underneath slope to �.

7. Underneath the cell labeled Cl/F, add the other parameters to be estimated: t1/2, Cmax,and Tmax. Color Cmax and Tmax red as a reminder that all these values must be enteredby hand. An entry for Vd/F may be added if desired.

8. Enter the values of Cmax and Tmax for the control data. Enter the formula for the newparameters: t1/2 = 0.693/� and Vd/F = CL/(F ∗ �). (Note this will be V� if the drugfollows two-compartmental characteristics.)

9. Copy and paste the entire lower calculation area and make a duplicate to the right.Do not worry that the data change to “0” and that #NUM appears in some cells. Inthe “Phase” row, label the left calculation area “Control” and the right area “Test”.

10. Enter the test dose by referencing the dose in the upper area of the worksheet.

11. Enter the test data by referencing the given test data in the upper area of the worksheet.Make sure to manually add the values of Cmax and Tmax for the test data.

12. Create a results summary as shown by referencing all the parameters. Enter theseparameters into Table C.4.

The completed worksheet is shown in Figure C.9.

Page 423: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappD JWBS062-Rosenbaum February 4, 2011 13:58 Printer Name: Yet to Come

APPENDIX D

DERIVATION OF EQUATIONSFOR MULTIPLE INTRAVENOUSBOLUS INJECTIONS

D.1 Assumptions

D.2 Basic Equation for Plasma Concentration After Multiple Intravenous Bolus Injections

D.3 Steady-State Equations

D.1 ASSUMPTIONS

1. All doses are the same.

2. The dosing interval (� ) remains constant.

3. Pharmacokinetic parameters remain constant over entire course of therapy.

For simplicity, F and S are not included as potential modifiers of a dose during the initialderivation of the formula.

D.2 BASIC EQUATION FOR PLASMA CONCENTRATION AFTERMULTIPLE INTRAVENOUS BOLUS INJECTIONS

It is assumed that after a dose, the total amount of drug in the body is equal to the dose plusany drug remaining from previous dose(s). This is known as the principle of superpositionand is illustrated in Figure D.1.

The maximum and minimum amounts of drug in the body after sequential doses willnow be calculated, dose by dose. The maximum amount of drug in the body after the firstdose is the dose (D). During the first dosing interval, this amount decays monoexponentially

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

403

Page 424: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappD JWBS062-Rosenbaum February 4, 2011 13:58 Printer Name: Yet to Come

404 DERIVATION OF EQUATIONS FOR MULTIPLE INTRAVENOUS BOLUS INJECTIONS

Ab

Abmin,1

Abmax,2 = D + Abmin,1

D

Abmin,2

Abmax,3 = D + Abmin,2

Abmin,3

Abmin,1 = D•e

D1 D2 D3 D4

Time into Therapy

–k

FIGURE D.1 Principle of superposition. The amount of drug in the body after a dose is equal tothe dose (D) plus the amount remaining from a previous dose or doses.

and at the end of the dosing interval;

Abmin,1 = Abmax,1 · e−k� = D · e−k� (D.1)

The second dose is given and the maximum amount in the body is equal to the dose pluswhat remains from the first dose:

Abmax,2 = D + D · e−k� = D · (1 + e−k� ) (D.2)

During the second dosing interval the amount of drug in the body decays mono-exponentially and the minimum amount in the body after the second dose is

Abmin,2 = Abmax,2 · e−k� = D · (1 + e−k� ) · e−k� = D · (e−k� + e−2k� ) (D.3)

The results of the calculations are summarized in Table D.1, which also shows the resultsof the calculations continued through the third dose. A pattern can be observed, and it ispossible to use this to predict Abmax,5:

Abmax,5 = D · (1 + e−k� + e−2k� + e−3k� + e−4k� ) (D.4)

TABLE D.1 Maximum (Abmax) and Minimum (Abmin) Amountsof Drug in the Body After Sequential Doses

Dose Abmax Abmin

1 D D · e−k�

2 D + D · e−k�

D · (1 + e−k� )D (1 + e−k� ) · e−k�

D · (e−kt + e−2kt)3 D + D · (e−k� + e−2k� )

D · (1 + e−k� + e−2k� )D · (1 + e−k� + e−2k� ) · e−k�

D · (e−k� + e−2k� + e−3k� )

Page 425: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappD JWBS062-Rosenbaum February 4, 2011 13:58 Printer Name: Yet to Come

MULTIPLE DOSE EQUATIONS 405

More generally,

Abmax,n = D · (1 + e−k� + e−2k� + e−3k� + · · · + e−(n−1)k� ) (D.5)

The expressions contain a geometric series. Let R represent the geometric series:

R = 1 + e−k� + e−2k� + e−3k� + · · · + e−(n−1)k� (D.6)

Thus,

Abmax,n = D · R (D.7)

Multiply R by e−k� :

R · e−k� = e−k� + e−2k� + e−3k� + · · · + e−nk� (D.8)

Subtracting equation (D.8) from equation (D.6) gives us

R − R · e−k� = 1 − e−nk�

R = 1 − e−nk�

1 − e−k�

(D.9)

Recall from equation (D.7) that Abmax,n = D · R:

Abmax,n = D · 1 − e−nk�

1 − e−k�(D.10)

As Cp = Ab/Vd,

Cpmax,n = D · (1 − e−nk� )

Vd · (1 − e−k� )(D.11)

A trough concentration (Cpmin,n) occurs when the time after the dose, t = � and is given by(Cpmin,n = Cpmax,n · e−k� ):

Cpmin,n = D · (1 − e−nk� )

Vd · (1 − e−k� )· e−k� (D.12)

The plasma concentration at anytime, t after a dose (Cpn) during a dosing interval is givenby (Cp,n = Cpmax,n · e−kt)

Cpn = D · (1 − e−nk� )

Vd · (1 − e−k� )· e−kt (D.13)

Including the salt factor and bioavailability as potential modifiers of the dose, we obtain

Cpn = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−kt (D.14)

Page 426: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappD JWBS062-Rosenbaum February 4, 2011 13:58 Printer Name: Yet to Come

406 DERIVATION OF EQUATIONS FOR MULTIPLE INTRAVENOUS BOLUS INJECTIONS

In summary:

1. The basic equation for the plasma concentration at any time during therapy withmultiple intravenous bolus injections is given by equation (D.14):

Cpss = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−kt

2. Peak plasma concentrations occur at the time a dose is given (t = 0):

Cpmax,n = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )(D.15)

3. Trough concentrations occur just before the next dose is given, when t = T:

Cpmin.n = S · F · D · (1 − e−nk� )

Vd · (1 − e−k� )· e−k� (D.16)

D.3 STEADY-STATE EQUATIONS

The equations for steady-state plasma concentrations simplify somewhat. These equationsare derived in Chapter 12 but are summarized here. During therapy n increases with eachsuccessive dose. As steady state is approached:

1. nkt gets large.

2. e−nkt decreases and tends to zero.

3. (1 − e−nk� ) becomes equal to 1 and disappears from equation (D.14).

Cpss = S · F · D

Vd · (1 − e−k� )· e−kt (D.17)

The peak plasma concentration at steady state:

Cpmax,ss = S · F · D

Vd · (1 − e−k� )(D.18)

The trough plasma concentration at steady state:

Cpmin,ss = S · F · D

Vd · (1 − e−k� )· e−k� (D.19)

Page 427: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappE JWBS062-Rosenbaum February 16, 2011 9:28 Printer Name: Yet to Come

APPENDIX E

SUMMARY OF THE PROPERTIESOF THE FICTITIOUS DRUGSUSED IN THE TEXT

Lipoamide Lipoamide is a novel antipyretic drug that is believed to work by reducing thesynthesis of cytokines. It appears to be safe in both children and adults. At concentrationsabove 300 �g/L there is an increased incidence of fairly minor concentration-related sideeffects, including nausea and diarrhea. Lipoamide is available in both an IV formulationand an oral tablet.

Nosolatol Nosolatol is a �1-adrenergic antagonist selective for receptors located in theheart and vascular smooth muscle. However, like all selective �1 agents, at high doses(�300 mg/day for nosolatol), it loses its selectivity and can block �2-adrenergic receptors,with undesirable results. Nosolatol shows no inherent �-activity. Like many other agentsin this class, nosolatol has been found to be beneficial in treating hypertension, chronicstable angina, heart failure, and myocardial prophylaxis. Nosolatol is available in both anIV formulation and an oral tablet.

Disolvprazole A member of the proton pump inhibitor family of drugs, disolvprazole isan irreversible inhibitor of H+, K+-pumps in the parietal cells of the gastric mucosa andit shows many of the same characteristics as other drugs in this class. Disolvprazole hasshown similar efficacy in ulcer-healing rates as other drugs in this class and is effective inthe elimination of Helicobacter pylori when used at the doses recommended. Disolvprazoleis available in both an IV formulation and an oral tablet.

Detailed information on the physicochemical and pharmacokinetic properties of thesedrugs is provided in Table E.1.

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

407

Page 428: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappE JWBS062-Rosenbaum February 16, 2011 9:28 Printer Name: Yet to Come

408 SUMMARY OF THE PROPERTIES OF THE FICTITIOUS DRUGS USED IN THE TEXT

TABLE E.1

Lipoamide Nosolatol Disolvprazole

Physicochemicalproperties

Acid/base Base Acid BaseMW 396 365 221Log P 3.2 2.1 0.2Log D6.0 3 1.8 −2.8Highest dose strength (mg) 150 250 50

Solubility: pH 1–7.5 High: 1g/l low: 0.5g/L High: 5g/LFraction of oral dose

recovered as metabolitesin humans

99.00% 99.00% 0.05–0.03

PharmacokineticsCl (L/h) 62 12.6 12Vd (L/kg) 6 3 0.5k (h−1) 0.15 0.06 0.34t1/2 4.70 11.6 2.02ka (h−1) (fasting) 4 2 2fe �0.01 �0.01 ∼0.98fu 0.05 0.6 �0.95Main enzyme involved in

its metabolismCYP2C9 CYP3A4 none

Substrate for intestinaluptake transporter

none known none known OATP1A2

Substrate for intestinalefflux transporter

none known P-gp none known

Fa 1 1 0.5Fg 1 0.7 1Fh 0.21 1 1F 0.21 0.7 0.5Substrate for renal

transporterno no OAT

Cp/Cb 1 1 1

PharmacodynamicsTarget Cpss 50 �g/L 1.2 mg/L 400 �g/LOther PD information Indirect effect model I Emax = 30 ± 3.2 bpm See Section 17.7.1.1

EC50 75 ± 15 �g/L

Page 429: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCappF JWBS062-Rosenbaum February 4, 2011 20:56 Printer Name: Yet to Come

APPENDIX F

COMPUTER SIMULATION MODELS

All the computer simulation models were created using Stella 9.1.3 (isee Systems, Lebanon,NH). The models were published on the Web as isee NetSim files using Simulate (Forio,San Francisco, CA). All the underlying models can be downloaded as Stella or Runtimefiles. The model interfaces are not downloadable.

A complete list of the models can be found at the following link:

http://www.uri.edu/pharmacy/faculty/rosenbaum/basicmodels.html

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

409

Page 430: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCgloss JWBS062-Rosenbaum February 16, 2011 9:30 Printer Name: Yet to Come

GLOSSARY OF ABBREVIATIONSAND SYMBOLS

A, B Intercepts of exponential terms in a two compartment model� Intrinsic activity; slope representing how a disease status changes over time�,� Rate constants of exponential terms of a two compartment modelA1 Amount of drug in the central compartmentAb Amount of drug in the bodyAbmax,n Maximum amount of drug in the body after the nth doseAbmin.n Minimum amount of drug in the body after the nth doseAGI Amount of drug in the gastrointestinal tractADME Absorption distribution metabolism and excretionAp Amount of drug in the plasmaAe Amount of drug elimintaedAt Amount of drug in the tissuesAu Amount of drug in the urineAu∞ Amount of drug excreted in urine by infinityAUC Area under the plasma concentration–time curveAUMC Area under the first moment-time curveCa Drug concentration in arterial bloodCb Drug concentration in bloodCl ClearanceClh Hepatic clearance (based on plasma)Clint Intrinsic hepatic clearanceClr Renal clearanceClbh Hepatic blood clearanceCm Concentration of metabolite mediatorCmax Maximum Observed Plasma ConcentrationCp Plasma concentration

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

410

Page 431: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCgloss JWBS062-Rosenbaum February 16, 2011 9:30 Printer Name: Yet to Come

GLOSSARY OF ABBREVIATIONS AND SYMBOLS 411

Cpav,ss Average steady state plasma concentrationCpmax Peak plasma concentrationCpmin Trough plasma concentrationCpn Plasma concentration during the nth dosing intervalCpss Steady state plasma concentrationCpu Concentration of unbound drug concentration in the plasmaCt Tissue concentrationCv Drug concentration in venous bloodD Diffusion coefficient (Noyes–Whitney); Distribution coefficient (lipophilicity)D Dose administeredDL Loading doseDM Maintenance dosee Efficacyε Intrinsic efficacy or efficacy per unit receptorE Drug effectE Extraction RatioE0 Baseline effectEm System’s maximum responseEmax Drug’s maximum responseEC50 Concentration of drug that produces half its maximum responseEt Effect of a drug at time tF Bioavailability factor (fraction of dose absorbed)Fa Fraction of a dose that is absorbed into GI membraneFg Intestinal bioavailabilityFh Hepatic bioavailabilityfe Fraction of a dose excreted unchangedfu Fraction of a drug in the plasma that is unboundfut Fraction of a drug in the tissues that is unboundGFR Glomerular filtration rateGI GastrointestinalI Inhibitory effectImax A drug’s maximum inhibitory effectIC50 Drug concentration that produces 50% inhibitionICTT Intercompartmental transit timek Overall elimination rate constantk0 Zero order rate constant; rate of an intravenous infusionk10 Elimination rate constant in a two compartment modelk12 Rate constant for distribution between the first and second compartmentsk1m Rate constant for formation of tolerance mediatork21 Rate constant for redistribution between the second and first compartmentska First-order rate constant for absorptionkcirc First-order rate constant for the loss of neutrophilskd First-order rate constant for distribution to a tissueKd Dissociation constant; reciprocal measure of affinity; drug concentration at 50%

receptor occupancyKe Concentration of a drug–receptor complex that produces 50% of a system’s

maximum responseke0 Rate constant for removal of drug from the effect compartment

Page 432: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCgloss JWBS062-Rosenbaum February 16, 2011 9:30 Printer Name: Yet to Come

412 GLOSSARY OF ABBREVIATIONS AND SYMBOLS

kin Zero-order rate constant for the formation of a physiological response variablekirr Second-order rate constant for the irreversible destruction of a response variablekm First-order rate constant for the elimination of a drug by metabolismKm Michaelis–Menton constantkm0 Rate constant for the destruction of a tolerance mediatorkoff Rate constant for the disassociation of a drug with its receptorkon Rate constant for the interaction of a drug with its receptorkout First-order rate constant for the degradation of a physiological response

variable, RKp Tissue to blood partition coefficientkprog First-order rate constant for disease progressionkprol Rate constant for the proliferation of neutrophils from a stem cell poolkr First-order rate constant for the elimination of a drug by renal excretionlog D Log distribution coefficientlog P Log partition coefficientMEC Minimum effective concentrationMRT Mean residence timeMTC Maximum tolerated concentrationMTT Mean transit timen Power functionN0 Baseline neutrophil countNt Number of neutrophils at time tP Partition coefficientQ Tissue blood flowR Concentration of a physiological response variableR0 Baseline concentration of a physiological response variableRa Rate of drug administrationRm Maximum response achieved at a given dose in an indirect effect modelRmax Maximum achievable response of a drug in an indirect effect modelRT Total concentration of receptorsRC Concentration of a drug–receptor complexRCE50 Concentration of a drug–receptor complex that produces 50% a system’s

maximum effectS Salt factor; stimulatory effectS0 Status of a disease at time zeroSmax Maximum stimulatory effect of a drugSss Terminal disease status of a progressive diseaseSC50 Drug concentration that results in 50% of the maximum stimulatory action of a

drugSt Status of a disease at time tt Time or time after a dose� Transduction ratio; dosing interval; intercompartmental transit timet0 Absorption lag timet1/2 Half-lifeTmax Time of maximum observed plasma concentrationTRm Time of maximum responseV Rate of an enzymatic processV� Volume of distribution in the post-distribution phase

Page 433: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCgloss JWBS062-Rosenbaum February 16, 2011 9:30 Printer Name: Yet to Come

GLOSSARY OF ABBREVIATIONS AND SYMBOLS 413

V1 Volume of the central compartmentV2 Volume of the peripheral compartmentVmax Maximum rate of an enzymatic processVp Volume of plasmaVd Volume of distributionVdss Volume of distribution at steady stateVt Volume outside the plasma into which a drug distributes

Page 434: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCgloss JWBS062-Rosenbaum February 16, 2011 9:30 Printer Name: Yet to Come

Page 435: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX

AAG, see �1-Acid glycoproteinAbbreviations used, 409ABC superfamily, see Transporters, ABC

superfamilyAbsorption, drug

absorption half-life, 128, 188, 385absorption lag time, 192absorption phase, 187, 385absorption rate constant (ka), 55, 128, 184defined, 11–12, 11f, 20–21, 21fdiffusion controlled, 55dissolution controlled, 54nonlinear absorption, 280–281paracellular diffusion, 26–27, 47rate of absorption, 53–55, 128transcellular diffusion, 24–26, 46transporters involved in absorption, 30f, 31,

32, 33, 40, 47–50typical Cp-time profile, 183f, 185

Accumulation, 213, 231, 232f, 243–244, 243ffactor (r), 243

Acetaminophen, assessment of stomachemptying time, 53, 54f

Acetone, 104Acetylcholine, 5Acetyl salicylic acid, see AspirinAcute phase reactant protein, 72

Acylcovir, 29t, 47, 98Adefovir, 29t, 31t, 98, 99, 100Administration drug

local and systemic, 37routes, 37–40

Adverse events in intestinal lumen, 44–46Affinity

plasma protein, 74receptor, 7, 9, 301, 305, 306, 306f, 307freceptor, assessment in vivo, 327–329

Africans and CYP3A5 expression, 105Agonist, 6, 7. See also Full agonist and Partial

agonistAlbumin, 72, 72t, 98Albuterol, 4Alcohol dehyrogenase, 103Aldehyde dehydrogenase, 103Alendronate, 38, 247Alfentanil, 107Alkylating agents, 3, 4�1-Acid glycoprotein (AAG), 72, 72tAlprazolam, 51t, 115Alzheimer’s disease, biomarkers, 299Aminoglycosides, 27, 47, 63, 69t

nonlinear pharmacokinetics, 281three-compartment model, 132, 172

para-Aminohippurate, 108

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook and Computer Simulations,First Edition. By Sara Rosenbaum.© 2011 John Wiley & Sons, Inc. Published 2011 by John Wiley & Sons, Inc.

415

Page 436: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

416 INDEX

Amiodarone, 32t, 69t, 96, 107, 385Ammonium chloride to decrease urinary pH,

100, 101Amoxicillin, 2t, 44Amphetamine, urinary excretion, 101Angiotensin-converting enzyme inhibitors,

47Antacids

complexation, 45gastric pH, 44mechanism of action, 5

Antagonist, 7Antihistamines, 34Antipyrene, 71tApparent volume of distribution, see Volume of

distributionApple juice, see Fruit juiceArea under the curve (AUC)

assessment of bioavailability, 196–198changes for restrictively and nonrestrictively

cleared drugs, 113t, 113, 114determination after an IV injection,

one-compartment model, 151determination after an IV injection,

two-compartment model, 166determination after oral administration, 190determination using NCA, 115–117, 207,

389–392, 399–402determination using the trapezoidal rule,

388–392multiple injections, 240multiple oral doses, at steady state, 271, 275

Area under the first moment curve (AUMC )definition, 204, 205fdetermination using the trapezoidal rule,

206, 207tAriens, 300Aspirin, 4, 98Astemizole, 105Atenolol, 2t, 25t, 26, 47, 69t, 102t, 314Atorvastatin, 29t, 32t, 47, 50, 51t, 51, 52, 107AUC, see Area under the curveAUMC, see Area under the first moment curveAverage amount of drug in the body at steady

state, 243Average steady-state plasma concentration

multiple injections, 240, 240fmultiple oral doses, 271

Baseline effects, 310Bendroflumethiazide, 98Benzylpenicillin, 29t. See also Penicillin G

�2-Adrenergic receptors, 4�-Blockers, 45�-Lactams, 98�-Receptors subtypes, 6Bile salt export pump (BSEP), 30, 30f, 31,

108–109, 108fBiliary excretion, 89, 103, 107, 108f, 108Bioavailability. See also Bioavailability factor

(F)absolute, 197assessment, 195–198, 278assessment after multiple oral doses,

269–272, 275bioequivalence, 198definition, 195–198relative, 198simulation exercise, 199

Bioavailability factor (F)definition, 41–42determinants of, 43–53

adverse events in intestinal lumen, 44–46disintegration, 43dissolution, 43excipients, 43membrane diffusion, 46–47presystemic hepatic metabolism, 52–53presystemic intestinal metabolism, 50–52transporters, 47–50

effective dose modifier, 127–128individual components, 42–43influence on steady state plasma

concentration, 219, 240Bioequivalence, 198Biological factor, turnover model, 329Biological half-life, 164Biomarkers, 15, 298–299

required qualities, 298–299Biopharmaceutics, 11Biopharmaceutics Classification System

(BCS), 55–56, 55tBiopharmaceutics Drug Disposition

Classification System (BDDCS), 56Biophase, 12, 12t, 317Biotransformation, see Hepatic metabolismBlack and Leff, see Operational model of

agonismBlood brain barrier, 21, 39, 63

biological component, 84drug distribution, 83–85structural component, 84transporters, 84–85, 84f

Blood-cerebrospinal fluid barrier, 83

Page 437: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 417

Blood clearance versus plasma clearance,114–115

Brain, see Central nervous systemBreast cancer resistance protein (BCRP), 28f,

30f, 31, 31tabsorption, 48f, 49t, 50CNS distribution, 84–85, 84fhepatic elimination, 108, 108f

Bromide (Br−), 71tBuccal drug administration, 38Buprenorphine, 9Burns, protein concentration, 74Buspirone, 51t, 51, 52, 96

Calcitonin, 39Calcium carbonate, 2tCalcium sulfate, 44Cancer, protein concentration, 74Capacity limited metabolism, 281–284

characteristics, 283–284determination of Km, 294–295determination of Vmax293estimation of Cp, 288estimation of dose and rate of

administration, 287individualization of doses, 292–295influence of Km and Vmax289pharmacokinetic model, 285–287plasma protein binding effects, 289simulation exercise, 288, 289time to eliminate a dose, 290time to reach steady state, 291

Capacity limited processesdrug response, 6, 302–307, 308–310, 326,

327metabolism, 109, 281–284protein binding, 73receptor binding, 301

Captopril, 47Carbamazepine, induction of metabolism, 281Carbidopa, 83–84, 83fCefazolin, 100Ceftizoxime, 100Central compartment, 130Central nervous system

distribution, see Distribution, drug, to centralnervous system

epithelial membrane, 27, 82transporters, 84–85, 84f

Cephaloridine, 33, 99Cephalosporins, 29t, 47Chitosan, 26

Chloroquine, 2t, 69t, 96Cholestasis, 109Chymotrypsin, 44Cidofovir, 33, 98, 99Cimetidine, 28, 29t, 32t, 99, 106t, 108Ciprofloxacin, 102tCisapride, 105Cisplatin, 28, 29t, 33, 99Clarithromycin, 107Clinical practice, parameter determination,

153, 176after multiple short infusions, 259–264

Clark, 300Clearance. See also Hepatic clearance and

Renal clearancecomponent clearances, 91, 94–95definition and determinants, 91–94determination after oral administration,

189–190determination in a one-compartment model,

151, 152–153determination in a two-compartment model,

165determination using NCA and the

trapezoidal rule, 115–117, 205,387–392

effect on steady state plasma concentrations,219, 241

effect on time to steady state, 220influence on t1/2 and k, 91total body clearance, 94–95

Cmax, see Peak plasma concentrationCompartments

application to pharmacokinetics, 126–138compartment versus body characteristics,

141tdefinition, 130metabolite models, 136–137

Competitive inhibitors, 106Complexation, 45Computer simulation models, see Simulation

modelsConstant continuous drug administration, see

Intravenous infusionCorticosteroid-binding protein, see TranscortinCounter-regulatory force tolerance model,

345–348Coupling, 5. See also Signal transductionCpn, see Multiple IV injectionsCpmax,n, see Multiple IV injectionsCpmax,ss, see Multiple IV injections or Multiple

intermittent infusions

Page 438: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

418 INDEX

Cpmin,n, see Multiple IV injectionsCpmin,ss, see Multiple IV injections or Multiple

intermittent infusionsCpss, see Multiple IV injections or Multiple

intermittent infusionsCreatinine clearance, 119–120Curve stripping, see Method of residualsCyclosporine

bioavailability, 48, 49t, 49, 51t, 51, 52Neoral, 49, 128plasma blood ratio, 115Sandimmune, 49, 128therapeutic range, 17ttransporters, 29t, 31t, 32t, 33, 48, 49t, 50, 85,

109Cytochrome P450, 103, 104–105

CYP1A2, 104tCYP2A6, 104tCYP, 3A, 104CYP3A4, 30, 49, 50, 51, 104–105, 104tCYP3A5, 104CYP2B6, 104t, 106tCYP2C8, 104t, 106tCYP2C9, 50, 77, 104, 104t, 106tCYP2C19, 104, 104t, 106tCYP2D6, 104, 104t, 106tCYP2E1, 104, 104tdrug–drug interactions, 105–107

Daunorubicin, 31tDecay function, 373–375Deep tissue compartment, 132, 132fDerived pharmacokinetic parameters, see

Secondary pharmacokinetic parametersDesimipramine, 106t, 108Desmopressin, 39Detoxification, drug, 103Diazepam, 69t, 113tDidanosine, 44, 96Diffusion, passive, 22–27

diffusion-controlled absorption, 55diffusion controlled distribution, 82–83of free, unbound drug, 10, 62, 64paracellular passive diffusion, drug

absorption, 47paracellular passive diffusion, general, 24f,

26–27transcellular passive diffusion, drug

absorption, 46transcellular passive diffusion, general,

24–26, 24fDiflunisal, 77

Digoxin, 2t, 90, 105, 178absorption, 45BCS classification, 55distribution phase, 18, 164, 179, 386effect compartment, 318therapeutic range, 17tthree-compartment model, 132, 172transporters, 29t, 32t, 33, 48, 49t, 49, 51,

99–100Diltiazem, 32t, 96, 107, 113, 113tDiluents, 44Disease progression models, 356–360

exponential decay models, 358–359exponential model, 359–360linear models, 357–358nonzero maximum disease status models,

359–360zero asymptotic models, 358–359

Disintegration, 11, 12t, 40, 41f, 43Disolvprazole

clinical pharmacology, 407determination of Cl, 122pdetermination of Clr, 118–119distribution characteristics, 86pdrug-drug interaction study, 211pfood effects, 210pintravenous infusion regimen, 228pmultiple injection regimen, 253ppharmacodynamic model, 351–352, 363ppharmacokinetic and pharmacodynamic

properties, 408tpharmacokinetic analysis after IV injection,

156psimulation model, 351, 363

Disopyramidedose dependent binding, 74effect compartment, 318

Disposition, 10, 11fDisposition half-life, 164Dissolution, 11, 12t, 40, 41f, 43

controlled absorption, see Absorption rateDissociation constant (Kd), 301, 303. See also

AffinityDistribution clearance (Cld), 166–167Distribution coefficient (D), 25, 25t, 46Distribution delays to the site of action,

315–318Distribution, drug

assessment of the extent of distribution, 65defined and described, 11f, 12, 12t, 21, 21f,

61–62, 62fdiffusion controlled, 82–83

Page 439: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 419

distribution clearance, 166–167distribution half-life, 163, 386distribution phase, 62, 62f, 79–82, 159–160,

164, 179, 386distribution volumes, 62–64extent of distribution, 61–79in a two-compartment model, 159–162nonlinear, 280–281perfusion controlled, 80–82plasma protein binding effects, 64–65postdistribution phase, 62, 62frate of distribution, 79–83, 129tissue binding and distribution, 64–65to central nervous system, 83–85

Distribution phase, see Distribution, drugDocetaxel, 49t, 50, 85

model for hematological toxicty, 354Dopa decarboxylase, 84, 83fDopamine, 83–84, 83fDosing interval calculation

multiple bolus injections, 235multiple intermittent infusions, 264

Dosing regimen design, pharmacokineticbased, 246–251

Doxorubicin, 32tDrug definition, 1Drug-drug interaction(s)

metabolism-mediated, 105–107studies, 208–210transporter-mediated, 30

Drug-receptor concentration at, 50% Em (Ke),327

Drug-specific properties, 299, 305, 324Duration of action, 15, 16f

pharmacodynamic influence, 314, 319, 336,337, 338, 341t, 350

EC50

defined, 303, 307, 308, 309f, 309relationship to Cp, 313–314relationship to parameters of OMA,

328Effect compartment, 315–318Effect, drug, 3Effective dose, 127–128Efficacy, 304, 305, 306f, 307Efficacy in OMA, 327Efficacy, intrinsic, see Intrinsic efficacyEfficacy model, 303–307Efflux transporters, see Transporters, ABC

superfamilyElacridar, 31t, 49t, 50, 85

Elimination, drug, 88–121defined and described, 11f, 12, 12t, 21, 21f,

89, 90felimination phase after oral absorption, 185,

187elimination phase in a two-compartment

model, 159–160half-life, see Elimination half-lifeinfluence of drug distribution, 61first-order, 90–91nonlinear, 280–281. See also Capacity

limited metabolismrate constant, see Elimination rate constantrate of elimination, 90, 92, 95routes, 90f

Elimination half-life, 90determination, 150, 152, 164, 205, 206, 209,

260, 394determination in patients, 154, 176–177determination using NCA, 205, 206, 209factors affecting for restrictively and

nonrestrictively cleared drugs, 113trelationship to clearance and volume of

distribution, 91, 95–96two-compartment model, 164

Elimination rate constant, 90determination, 150, 152, 190, 205, 259, 391,

394determination using NCA, 205relationship to clearance and volume of

distribution 91, 95–96terminal, see Terminal elimination rate

constantEm, see Maximum response of systemEmax, see Maximum response of drugEmax model, 308–310

comparison to OMA, 327disadvantages, 324–325

Empirical pharmacodynamic models, 307Endoxifen, 106Enteric coating, 44Enterohepatic recirculation, 44–45, 45fEnzymatic degradation, gastrointestinal tract,

44Enzyme(s)

enterocytes of small intestine, 50–52gastrointestinal flora, 44–45hepatic, 103–107kinetics, 109within gastrointestinal lumen, 44

Epinephrine, 4Erythromycin, 105, 107

Page 440: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

420 INDEX

Estradiol glucuronide, 99Ethanol, 71t, 101, 104

nonlinear pharmacokinetics, 281Ethinyl estradiol, 39, 44Etoposide, 32t

model for hematological toxicity, 354Etorphine, relative efficacy, 328Evans blue, 71tExcipients, 43Excretion, see Renal clearance and Biliary

excretiondefinition, 11f, 12

Exponential decay, 373Exponential decay disease progression models,

358–359Exponential model with nonzero maximum

disease status 359–360Exponents, 368–373Extent of absorption, see Bioavailability factor

(F)Extracellular fluid, 63, 63f, 69tExtraction ratio (E), 52, 53t, 93, 94t, 111–112,

113tExtravascular drug administration, 38,

182–199Exubera, 39

Famotidine, 25, 25t, 99Feathering, see Method of residualsFeedback for neutrophil proliferation, 352Felodipine, 25, 25t, 51t, 52, 69t, 96Fentanyl, 26, 39

effect compartment, 318relative efficacy, 328

Fexofenadine, 29t, 32t, 47, 49, 51, 98, 99Fick’s law

absorption, 55diffusion, 22distribution, 82

First-order absorption modelsone-compartment, 183–199two-compartment, 135

First order processes, 380–386fraction of completion, 384–385, 385thalf-life, 381–382, 383

First-order rate constant for absorption (ka),see Absorption, rate constant

First-order rate constant for elimination (k), seeElimination rate constant

First-order rate constant for excretion (ke), 90First-order rate constant for metabolism (km),

90

First pass hepatic metabolism, 11f, 12, 40, 41f,52, 112, 113. See also Hepaticbioavailability(Fh)

nonlinear pharmacokinetics, 281Flavin monooxygenase, 103Flip flop model

oral absorption, 192–193, 199two compartment model, 172

Fluctuation, 213, 231, 232f, 242consideration for dosing regimens, 246–247

Fluoxetine, 2tFluvastatin, 107Food effects, 46, 56, 56tFormulation excipients, see ExcipientsFraction absorbed (Fa), 41f, 42–43Fraction excreted unchanged (fe), 120Fraction of completion of a first-order process,

384–385, 385tFraction of drug in the plasma or in the tissues,

69Fraction of drug unbound

and the therapeutic range, 77–79in plasma (fu), 70, 72–79in tissues (fut), 70

Fraction of steady state at any time, 244Fruit juice, 29t, 47Full agonist, 8, 8f, 9, 302, 303f, 307, 308, 310Furchgott, 300Furosemide, 25t, 29t, 98, 100

Gastric pH, 44Gastrointestinal pH, 44Generic products, 198Genetic polymorphism

BCRP, 50CYP isozymes, 104–105, 104tOATP, 108transporters, 32, 33UGTs, 105

Gentamicindistribution phase, 386three-compartment model, 172

Glomerular filtration rate, 98Glucuronidation, see

UDP-Glucuronosyltransferases (UGT)Glucuronide conjugates, 31t, 99, 103. See also

UDP-Glucuronosyltransferases (UGT)and Enterohepatic recirculation

Glutathione conjugates, 31t, 103Glyburide, 31t, 109G-protein, 5f, 6

coupled receptors (GPCR), 6

Page 441: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 421

Grapefruit juice, 29t, 47, 51Growth function, 374Gut microflora, 44

Half-life, in first order processes defined381–382, 383. See also Absorption,absorption half-life; Distribution,distribution half-life; or Eliminationhalf-life

Heparin, 69tHepatic bioavailability (Fh), 40, 41f, 42,

52–53, 53t, 107, 112–114. See alsoFirst pass hepatic metabolism

Hepatic blood flow, 111effects on restrictively and nonrestrictively

cleared drugs, 112–114Hepatic clearance

and hepatic bioavailability, 112blood clearance, 111blood versus plasma clearance, 114–115defined, 111–114, 111fdeterminants of, 111–112nonrestrictive clearance, 112–113relationship to Michaelis-Menton

parameters, 110restrictive clearance, 114

Hepatic extraction ratio, see Extraction ratio(E)

Hepatic first pass metabolism, see First passhepatic metabolism and Hepaticbioavailability (Fh)

Hepatic metabolismdefined and described, 11f, 12, 89, 90f,

102–107, 103ffirst order metabolism, 90kinetics, 109–110, 282nonlinear, 281–295purpose, 102–103

Hepatic transporters, 107–109, 108fHigh density lipoproteins, 72, 72tHigh extraction drugs, see Nonrestrictive

hepatic clearanceHMG-CoA reductase inhibitors, 6, 33, 47,

107H2-receptor blockers, 29t, 44Human ether a-go-go related gene (hERG),

105Hybrid rate constant for distribution (�), 163,

165Hybrid rate constant for elimination (�),

163–164, 165Hydrochlorothiazide, 100

Hydrocodone, relative efficacy, 328Hydromorphone, relative efficacy, 328Hysteresis, 316, 316f

Ibuprofen, 2t, 25tImatinib, 31t, 85Indinavir, 31t, 32tIndirect effect models, 331–341

application to precursor pool model oftolerance, 348

effect of dose, 334f, 336effect of IC50, 338effect of Imax, 338influence of k, 338influence of kin, 337–338influence of kout, 337–338maximum achievable response of a drug

(Rmax), 336, 340, 341tmaximum response to a dose (Rm), 334, 336,

338model I (inhibition of kin), 332–341model II (inhibition of kout), 332–333, 340,

341tmodel III (stimulation of kin), 332–333, 340,

341tmodel IV (stimulation of kout), 332–333,

340, 341tsummary of model characteristics, 341ttime course of response, 335–336time of maximum response (TRm), 336, 337,

338, 341ttime to steady state, 338–339

Individualization of dosing regimensinfusions, 224–227multiple injections, 247–251multiple intermittent infusions, 264phenytoin and nonlinear pharmacokinetics,

292–295selection of IV equations for multiple oral

doses, 272–274single intravenous doses, 147

Indomethacin, 50, 98Inducers of metabolism, 106t, 107

effects on restrictively and nonrestrictivelycleared drugs, 113, 114

Inducers of P-gp, 32t, 33, 49, 49tInhibitors of metabolism, 107

effects on restrictively and nonrestrictivelycleared drugs, 113, 114, 113t

Inhibitors of transporters, 29t, 31t, 32t. See alsoindividual transporters

Insulin, 6, 27, 39, 44

Page 442: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

422 INDEX

Integrated PK-PD models, 298, 312–318.See also Mechanism-based integratedmodels

direct link model, 312–314direct link simulation exercise, 314–315effect compartment link, 315–318effect compartment simulation exercise,

318–319Intercompartmental transit time (� ), 341Interindividual variability

2D6 and tamoxifen, 104BCRP, 50bioavailability, 51CYP2C9 and warfarin’s dose, 77, 104intestinal CYP3A4, 50OATP1B1, 33OCT1, 33pharmacokinetic, 153phenytoin’s pharmacokinetics, 285

Intestinal metabolism, extraction andbioavailability (Fg), 40, 41f, 42–43,50–52

Intestinal pH, 44Intraarterial drug administration, 37Intracellular fluid, 63Intramuscular drug administration, 38Intranasal drug administration, 26, 39Intravascular drug administration, 37Intravenous drug administration, 37, 140Intravenous infusion

basic equation, 214–216, 218definition, 140factors controlling Cpss, 218–219individualization of dosing regimens,

224–227infusion challenge, 229ploading dose, 221–222model, 212–227simulation exercise Part 1, 216simulation exercise Part 2, 220simulation exercise Part 3, 223steady state plasma concentration, 217termination of infusion, 223–224time to steady state, 219

Intravenous injection in a one-compartmentmodel, 133, 139–155

derivation of equations, 140–143determination of AUC, 151determination of Cl, 151determination of k and t1/2, 150determination of Vd, 150–151estimating a dose, 147

estimating a loading dose, 147estimating plasma concentrations, 145–146estimating the duration of action, 146–147pharmacokinetic analysis, 148–155simulation exercise, 144worksheet for parameter determination,

393–394, 395fIntravenous injection in a two-compartment

modelbasic equation, 162–163clearance, 165–166determination of parameters, 173–176distribution clearance (Cld), 166–167pharmacokinetic analysis, 176, 394–397simulation exercise, 170–173volume of central compartment (V1),

167–168, 168fvolume of distribution at steady state (Vdss),

169–170volume of distribution in � phase (V�),

168–169, 168fvolumes of distribution, 167–170worksheet for parameter determination,

394–397Intrinsic activity (�), 302, 303, 303f, 307Intrinsic activity model, 300, 302–303, 307,

308Intrinsic clearance

definition, 111effect on restrictively and nonrestrictively

cleared drugs, 112–114Intrinsic efficacy, 7, 9, 305, 307, 307fIon channels, 5, 5fIrinotecan transporters, 31, 31t, 32t, 48, 50, 108Irreversible action, 4Irreversible effects

hematological toxicity of anti cancer drugs,352–356

model for proton pump inhibitors, 350–351pharmacodynamic models, 350–356turnover model, 350–351

Irreversible enzyme inhibitors, 107Isomers, 4, 77Isoniazid, 45Itraconazole, 32t, 40, 105

Ke, drug-receptor concentration at, 50% Em,327–328

Ketamine, 85Ketoconazole, 32t, 85, 105

dissolution and nonlinear pharmacokinetics,281

Page 443: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 423

ke0 for an effect compartment, 317–318, 318fkin, in turnover model, 329kout in turnover model, 329

Lactose, 44Levodopa, 83–84, 83fLevofloxacin, 84Lidocaine, 52, 223Linear disease progression models, 357–358Linear pharmacodynamic model, 310–311Linear pharmacokinetics, defined, 277–280Lipoamide

clinical pharmacology, 407determination of Cl, 122pdistribution characteristics, 86pdrug interaction study, 208–210food effects, 210pintravenous infusion regimen, 228pmultiple injection regimen, 250pharmacodynamic model, 362ppharmacokinetic analysis after IV injection,

156ppharmacokinetic and pharmacodynamic

properties, 408tsimulation model, 362

Lipophilicity, 24Lipoproteins, 72, 72tLithium, 17tLiver disease, protein concentration, 74Loading dose, 147

infusion, 221–222multiple injections, 245two compartment model, 177

Local drug administration, 37Logarithmic pharmacodynamic model, 311,

312fLogarithms, 369–373Loop of Henle, 97f, 100Loperamide, 34, 63, 85Losartan, 32tLovastatin, 51, 52Low density lipoproteins, 72, 72tLow extraction drugs, see Restrictive hepatic

clearance

Macro rate constants, 163Maximum plasma concentration (Cmax), see

Peak plasma concentrationMaximum response of a drug (Emax), 303

relationship to OMA parameters, 328Maximum response of the system (Em),

302–303in operational model of agonism, 327

Maximum tolerated concentration (MTC),15–18, 16f, 17f

Mean absorption time ((MAT), 207Mean residence time (MRT)

definition, 202determination, 204

Mean transit time (MTT)extravascular administration, 207neutropenia model, 353transit compartment models, 341

Mechanism based inhibitors, see Irreversibleenzyme inhibitors

Mechanism-based integratedpharmacodynamic models, 323–360

Membranes, epithelial structure, 21, 22fMeperidine, 53, 113tMetabolism, drug, 11f. See also Hepatic

metabolism and Intestinal metabolismMetabolite compartment model, 136–137Metformin, 6, 28, 29t, 33, 63, 84, 99,

108Methadone

accumulation, 244relative efficacy, 328

Method of residualsoral absorption, 187–189two compartment model, 173–175

Methotrexatenonlinear pharmacokinetics, 281transporters, 31t, 84, 98, 99, 100, 102t,

108Methylphenidate, 39Metoclopramide, effect on stomach emptying,

53, 54fMetoprolol, 96Mibefradil, 105Michaelis-Menton constant (Km), 109, 282.

See also Capacity limited metabolismdetermination in patients, 295factors affecting, 289

Michaelis-Menton equation, 109, 282. See alsoCapacity limited metabolism

similarity to Emax model, 308Microflora, gut, 44Micronization, 43Micro rate constants, 163Midazolam, 51t, 115Minimum effective concentration (MEC),

15–18, 16f, 17fMitoxantron, 31tModel selection, 137–138Molecular size, 26, 27, 46, 47, 63

Page 444: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

424 INDEX

Monitoring therapyinfusions, 224–227phenytoin, 292–295two-compartment model, 179, 179f

MOP agonist, see �-Opioid agonistsMorphine, 26, 38, 39, 96, 113t

relative efficacy, 328Multidrug resistance-associated protein

(MRP), 30, 30f, 31, 47, 48f, 50,98–100, 99f, 107, 108, 108f

Multidrug resistance protein (MDR), seePermeability glycoprotein (P-gp)

Multiple dosing, general equation, 245, 246fMultiple intermittent infusions, 254–265

calculation of a dosing interval, 264determination of half-life, 259–260determination of peak, 261determination of plasma concentrations,

257–259determination of trough, 261determination of Vd, 261individualization of regimens, 264peak steady state plasma concentration

(Cpmax,ss), 256, 256fplasma concentration at any time during a

steady state dosing interval (Cpss), 257plasma concentration-time profile during a

dosing interval, 255–256, 256fsimulation exercise, 265steady state equations, 256–257trough steady state plasma concentration

(Cpmin,ss), 256, 256fMultiple IV doses, 230–253

accumulation, 231, 232f, 243assumptions, 233average amount of drug in the body at steady

state (Abav,ss), 244average steady state plasma concentration

(Cpav,ss), 240–241basic equations, 236–237, 403–406, 245,

246fcalculation of dosing interval, 235calculation of plasma concentrations, 239equation derivation, 403–406fluctuation, 231, 232f, 242fraction of steady state at any time, 244,

245tindividualization of regimens, 247–251loading dose, 245peak plasma concentration during the nth

dosing interval (Cpmax,n), 233, 233f,234, 237

peak steady state plasma concentration(Cpmax,ss), 233, 233f, 235, 238

plasma concentration during a steady statedosing interval (Cpss), 233, 235, 238

plasma concentration during the nth dosinginterval (Cpn), 233, 234, 237

simulation exercise, 251–253steady state equations, 238–242, 406symbols, 232–233time to reach steady state, 244trough plasma concentration during the nth

dosing interval (Cpmin,n), 233, 233f,234, 237

trough steady state plasma concentration(Cpmin,ss), 233, 233f, 235, 238

Multiple oral doses, 267–274AUC, 270, 275Cmax,ss, 270, 275selection of appropriate IV equations for

clinical use 272–274, 273fsimulation exercise, 274steady-state equations, 268–269Tmax, 269, 275

�-Opioid (MOP) agonists, 328, 329tolerance, 329, 347

Myocardial infarction, protein concentration,74

Naloxone, 39, 52Natural ligands, 4Natural logarithms, 369Nefazodone, 109Nelfinavir, 32tNephron, 97, 97fNeutropenia classification system, 355tNeutropenia pharmacodynamic model,

352–356. See also Irreversible effects,pharmacodynamic models

Neutrophil count, 352Neutrophil proliferation model, 353Nexium, 4Nicotine, 5

tolerance model, 345–348Nicotinic receptors, 5Nifedipine, 51tNitroglycerin, 15, 38, 52

tolerance model, 347Noncompartmental analysis (NCA), 201–210

benefits over compartmental analysis, 202mean residence time (MRT), 202use in clinical studies, 208–210worksheet for analysis, 399–402

Page 445: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 425

Nonlinear pharmacokinetics, 277–295.See also Capacity limited metabolism

Nonlinear processes in ADME, 280–281, 281tNonrestrictive hepatic clearance, 112–113Nonsteroidal antiinflammatory drugs, 29t, 98,

108Nonzero maximum disease status disease

progression models 359–360Norethindrone, 39Nortriptyline, 69tNosolatol

clinical pharmacology, 407determination of clearance, 122pdistribution characteristics, 86pdrug interaction study, 210pfood effects, 210pintravenous infusion regimen, 228pmultiple injection regimen, 253ppharmacodynamic-based dosing regimen,

319ppharmacokinetic analysis after IV injection,

156ppharmacokinetic and pharmacodynamic

properties, 408tNoyes–Whitney equation, 54Number of last dose (n), 232

Olmesartan, 29t, 31tOmeprazole, 4Ondansetron, 25t, 32tOne-compartment model, 130–131

multiple intermittent infusions, see Multipleintermittent infusions

multiple intravenous doses, see Multipleintravenous doses

multiple oral doses, see Multiple oral dosessingle intravenous injections, see Intravenous

injection in a one-compartment modelsingle oral doses, see Oral absorption in a

one-compartment modelOnset of action, 15, 16fOperational model of agonism (OMA),

325–329, 360pOral absorption in a one-compartment model,

183–199determination of parameters, 186–192peak plasma concentration (Cmax), 195simulation exercise, 198time of peak plasma concentration (Tmax),

194–195worksheet for parameter determination,

398–399

Oral clearance, 189Orange juice, see Fruit juiceOrder of process, 378Organic anion transporter (OAT), 28, 28f, 29t,

30f, 34hepatic elimination, 107–108, 108frenal excretion, 98–99, 99f

Organic anion transporting polypeptide(OATP), 28, 28f, 29t, 30f, 33

absorption, 47, 48fCNS distribution, 84, 84fhepatic elimination, 107–108, 108f

Organic cation transporter (OCT), 28, 28f, 29t,30f, 33

distribution, 63hepatic elimination, 107–108, 108frenal excretion, 98–99, 99f

Osmotic laxatives, 5Overall elimination rate constant, see

Elimination rate constantOxycodone, relative efficacy, 328

Paclitaxel, 32t, 48, 49t, 50, 85model for hematological toxicity, 354

Paracellular diffusion, see Diffusionparacellular

Parameterizations of models, 170Parenteral route of drug administration, 38Parkinson’s disease, use of levodopa, 83–84,

83fPartial agonist, 7–9, 8f, 302, 303f, 304, 305,

306f, 307, 308, 310Partition coefficient, n-octanol (P), 25, 25t,

46Partition coefficient, tissue to blood (Kp), 80

impact on duration of distribution phase,82

Passive diffusion, see Diffusion, passivePeak plasma concentration (Cmax), 194–195

after multiple oral doses, 270–271, 275changes for restrictively and nonrestrictively

cleared drugs, 113tinfluence of F, 195, 199, 270influence of ka, 195, 199

Penicillin, 44, 47, 99penicillin G (benzyl penicillin), 44, 99penicillin V, 44

Pentazocine, 26, 52Pepsin, 44Peptide growth factors, 6Peptide transporter (PEPT), 28, 29t, 30f, 47Perfusion controlled distribution, 80–82

Page 446: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

426 INDEX

Peripheral compartment, 131, 131fPermeability glycoprotein (P-gp), 28f, 29, 30f,

30, 31t, 32t, 33, 34absorption, 48, 48f, 49, 49t, 50, 51, 52distribution, 63, 84–85, 84fhepatic clearance, 108, 108frenal excretion, 98–100, 99f

Permeability, membrane, 23–24Perpetrator drug, 105pH, see Gastric pH; Gastrointestinal pH;

Intestinal pH; or Renal tubular pHPharmacodynamic

definition, 3modeling difficulties, 14–15, 298models, 299–319. See also

Mechanism-based pharmacodynamicmodels

phase, 2, 3fPharmacokinetic

compartment models, 126–138definition, 9phase, 2, 3f

Pharmacokinetic analysisclinical practice, 153–155, 176, 259–264,

292–295IV injection in one-compartment model,

148–153, 156p, 393IV injection in two-compartment model,

173–176, 181p, 394noncompartmental analysis, 208–210,

399–402oral absorption, 186, 199p, 398

Pharmaceutical equivalents, 198Phase I metabolism, 103–105Phase II metabolism, 103–105Phase III metabolism, 107Phenobarbital, 96, 101, 106t, 107Phenylbutazone, 77Phenytoin, 106, 106t, 113t

adverse effects, 285bioavailability, 43, 44, 45determination of Km in patients, 294–295determination of Vmax in patients, 292–295estimation of Cp, 288estimation of dose and rate of

administration, 287individualization of doses, 292–295influence of Km and Vmax, 289nonlinear pharmacokinetics, 281, 284–288pharmacokinetic model, 285–287plasma protein binding, 77–79, 78tsimulation exercise, 288, 289

therapeutic range, 17t, 78, 284time to eliminate a dose, 290time to reach steady state, 291

Physicochemical effects of drugs, 5Physiological turnover model, 329–331Pindolol, 9, 25tPitavastatin, 31tPlacenta, epithelial membrane, 27Plasma clearance versus blood clearance,

114–115Plasma concentration, 10Plasma protein binding, 10, 72–79

clinical consequences of altered binding,75–79

displacement, 75effect of drug concentration, 74effect of protein concentration, 74–75effect of renal and hepatic disease, 75effect on drug elimination, 76effect on extent of distribution, 64–71effect on glomerular filtration, 98effect on hepatic clearance, 111–114effect on restrictively and nonrestrictively

cleared drugs, 112–114, 113teffect on therapeutic range, 77–79factors controlling, 73–75interpreting Cp, 77–79nonlinear pharmacokinetics, 281warfarin, 77

Plasma water, 62, 63f, 69tPolarity, 25, 46Polar surface area, 46Polyethylene glycol, 5Positron-emission tomography, 85Postdistribution phase, 159–160Potassium citrate, to increase urinary pH, 100Potency, 7, 306, 307Pravastatin, 29t, 31t, 107, 108Precursor pool tolerance model, 348–350Pregnancy, protein concentration, 74Presystemic extraction, see Intestinal

metabolism and First pass hepaticmetabolism

Prilosec, 4Primary pharmacokinetic parameters, 91, 96Principle of superposition, 403Probenecid, 29t, 99Procainamide, 28Prochlorperazine, 39Propantheline, effect on stomach emptying, 53,

54fPropofol, 85

Page 447: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 427

Propranolol, 53, 113tnonlinear pharmacokinetics, 280

Protease inhibitors, 31tProteresis, 346f, 347Protective drug action, 356, 357, 358, 360

definition, 356Protein and peptide drugs, 39, 44, 46, 47Protein binding, see Plasma protein bindingProtein kinase, 5f, 6Prolonged release, see Sustained releaseProton pump inhibitors, 4, 31t, 44

pharmacodynamic model, 350–352Pulmonary drug administration, 39–40

Quinidine, 32t, 34, 49t, 51t, 85Quinolones, 31t, 45, 99

Ramipril, 2tRanitidine, 32tRate constant for distribution (k12), 131, 161,

164Rate constant for redistribution (k21), 131, 161,

164Rate of drug absorption, 53–55, 128

assessment in bioavailability studies195–196, 269–271

Rate of drug administrationeffect in nonlinear pharmacokinetics

283–284, 284feffect on steady state plasma concentrations,

218, 241, 278, 280fRate of drug distribution, 79–83, 129. See also

Distribution, drugand compartment models, 130–133

Rate of drug elimination, 90, 92, 95. See alsoElimination

Rates of processes, 377–386comparison of zero- and first-order

processes, 382RCE50 drug-receptor concentration at, 50% Em

(Ke), 327–328Rebound effect, 349Receptor, 3

concentration (RT), 301, 305Receptor theory of drug action, 299–307

in vivo models, 325Rectal drug administration, 39Redistribution, 161, 162fRenal blood supply, 97Renal clearance, 91, 94–95, 97–102

glomerular filtration, 97–98measurement, 117–120

nonlinear, 280–281, 281tputting meaning into the value, 101–102tubular reabsorption, 100–101tubular secretion, 98–100

Renal excretion. See also Renal clearancedefined and described, 9f, 12, 89, 90f,

97–102first-order excretion, 90

Renal filtrate flow, 101Renal filtrate pH, 100–101Renal transporters, 98–100, 99fResponse, 3Response chain, 299, 300f. See also Signal

transductionResponse variable

indirect effect models, 331physiological turnover model, 329

Restrictive elimination, and protein binding, 76Restrictive hepatic clearance, 114–115Retina, epithelial membrane, 27Riboflavin, nonlinear absorption, 280Rifampin, 29t, 32t, 33, 45, 49t, 49, 100, 106t,

107Ritonavir, 29t, 32t, 63, 85, 106t, 107, 108Rosuvastatin, 31, 31t, 50, 106t, 107Rotigotine, 39Routes of drug administration, 37–40

Saint John’s wort, 32t, 49t, 49Salicylates, dose-dependent binding, 74Salt factor, 127Salts, 43Saquinavir, 29t, 32t, 47Secondary pharmacokinetic parameters, 91, 96Second messengers, 6Serotonin, 6Serum concentration, 10Sigmoidal Emax model, 308–310

comparison to OMA, 327disadvantages, 324–325

Signal transduction, 5, 15, 299, 300f, 304,307f, 308, 316

modeled using transit compartments, 340Simulation exercises

hematological toxicity: mirotecan, 355indirect effect model I, 335, 340indirect effect model II, 340indirect effect model III, 340indirect effect model IV, 340infusion challenge, 229intravenous infusion model, 216, 220, 223,

224

Page 448: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

428 INDEX

Simulation exercises (Continued )intravenous injection in a one-compartment

model, 144intravenous injection in a two-compartment

model, 162, 170irreversible effects: disolvprazole 351, 363lipoamide, 363multiple intermittent infusions, 265multiple iv bolus injections, 251multiple oral doses, 274nonlinear pharmacokinetics, 288, 289operational model of agonism, 329oral absorption in a one-compartment model,

198sigmoidal Emax model, 314sigmoidal Emax model with an effect

compartment, 318tolerance: counter-regulatory force model,

347tolerance: precursor pool model, 349transit compartment model, 343transporter model, 30, 32

Simulation models, 409Simvastatin, 51t, 51, 52, 108Sirolimus, 51Site of action

drug action, 3–5drug concentration, 298, 299, 312, 315–318,

316f, 317fSLC superfamily, see Transporters, SLC

superfamilySlope factor (n), 309, 310fSmall intestine surface area, 23, 24f, 46, 47SN38, 108SN38 glucuronide, 108Sodium bicarbonate to increase urinary pH,

100Spare receptors, 7, 303Statins, see HMG-CoA reductase inhibitorsSteady state, 147, 148f, 215Stephenson, 300Steroids, 6Stimulus, 3, 304Stomach emptying

effect of food, 53rate, 53

Subcutaneous route, 38Subject drug, 105Sublingual route, 38Sulfasalazine, 31t, 37, 50Sulfate conjugates, 99Sulindac, 109

Sumatriptan, 39Surface area, membrane and diffusion, 23,

24fSurgery, protein concentration, 74Sustained release, 192Symbols used, 410Symptomatic drug action, 356, 357, 358, 360

definition, 356Systemic clearance, see Clearance, total bodySystemic drug administration, 37–40

Tacrolimusbioavailability, 51, 51tCYP, 3A5 and toxicity, 105therapeutic range, 17ttransporters, 32t, 48, 49t

Talinolol, 32t, 48, 49, 51Talwin, 52Tamoxifen, 31t, 104, 106, 107Tau, dosing interval, 232Tenofovir, 100Terbutaline, 26Terfenadine, 105Terminal elimination rate constant (�), 206

determination, 402Testes, epithelial membrane, 63, 82Tetracylines, complexation, 45Theophylline, 17t, 25t, 106, 113t, 228p

nonlinear pharmacokinetics, 281Therapeutic index (TI), 18Therapeutic range, 14–18, 16f, 17f

and unbound plasma concentration, 77–78examples, 17t

Therapeutic ratio, see Therapeutic indexThiopental, 85Three-compartment model, 131–133, 172Tight junctions, 26, 47Time of peak plasma concentration (Tmax),

194–195influence of k, 194f, 195influence of ka, 194–195, 194ffor multiple oral doses, 269–270

Time to eliminate a dose, 143Time, t in multiple dosing, 232Tissue binding, 64, 65f, 70

effects on extent of distribution, 64–71, 65fTissue blood partition coefficient (Kp), see

Partition coefficient, tissue to bloodTissue concentration, 65, 80–82Tissue perfusion, 79–82, 79tTissue specific properties, 299, 305, 307, 325Tmax, see Time of peak plasma concentration

Page 449: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

INDEX 429

Tolerance, 344–345counter-regulatory force, 345–347MOP agonists, 329OMA, 329precursor pool model, 348–350simulation exercise, 347, 350

Topotecan, 31, 31t, 49t, 50, 85model for hematological toxicity, 354

Torsades de pointes, 105Total body water, 62, 63f, 69tTotal plasma concentration (Cp), 10Transcellular diffusion, see Diffusion

transcellularTranscellular passive diffusion, see Diffusion,

transcellularTranscortin, 73Transdermal drug administration, 39Transduction ratio (� ), 327–328Transduction, see Signal transductionTransit compartment models, 340–343, 353

effect of number of compartments, 342–343,343f

model for neutropenia, 352–356simulation exercise, 343transfer of signal through the compartments,

342, 342fTranslational research, 298, 325, 329Transporters

ABC superfamily, efflux transporters, 29–31,31t, 32t

characteristics, 31–32efflux transporters and absorption, 48–50,

48f, 49tefflux transporters and CNS distribution,

84–85, 84fefflux transporters and hepatic elimination,

107–109, 108fefflux transporters and renal excretion,

98–100, 99fexamples of clinical effects, 33–34general, 27–34, 28f, 30fsimulation exercise, 32–33SLC superfamily, uptake transporters,

28–29, 29tuptake transporters and absorption, 47–48,

48fuptake transporters and CNS distribution,

84, 84fuptake transporters and renal excretion,

98–100, 99fuptake transporters hepatic elimination,

107–109, 108f

Trapezoidal rule, 388Triazolam, 51tTroglitazone, 31t, 109Trough plasma concentration after IV

administration, see Multipleintravenous doses; and Multipleintermittent infusion

Trypsin, 44d-Tubocurarine, and effect compartment, 318Tubular reabsorption, 100

lipophilicity, 100pH, 100

Turnover model, 329–331use in disease progression models, 360

Two-compartment model, 131, 158–179.See also Intravenous injection in atwo-compartment model

evaluation of response and Cp, 179half-life determination in patients, 176–177relationship among parameters, 164,

165–170

Unbound drug. See also Plasma protein bindingplasma, 10, 70tissues, 70

UDP-Glucuronosyltransferases (UGT), 103,105

Uptake transporters, see SLC superfamily

Valacyclovir, 29t, 47Valproic acid

dose-dependent protein binding, 74, 281hepatic clearance, 113tlog P and log D, 25t

Valsartan, 29tValspodar, 32t, 50Vancomycin, 2tVenous equilibrium model, 111Verapamil, 32t, 48, 53, 85, 113tVilli and microvilli, 23, 24fVinblastine, 31t, 32tVmax, 109–110. See also Capacity limited

metabolismdetermination in patients, 292–295factors affecting, 289

Volume of distributiondefinition, 65–71determination after oral administration, 189determination in one-compartment model,

150different Vds of a two-compartment model,

167–170

Page 450: Basic Pharmacokinetics and Pharmacodynamics - S. Rosenbaum (Wiley, 2011) WW

P1: OTA/XYZ P2: ABCind JWBS062-Rosenbaum February 23, 2011 7:45 Printer Name: Yet to Come

430 INDEX

Volume of distribution (Continued )effect on time to steady state, 220effects of tissue and plasma protein binding,

70–71estimation after multiple intermittent

infusions, 261–262examples, 69tin a one-compartment model, 141influence on the half-life and elimination

rate constant, 91, 96lack of influence on average steady state

plasma concentrations, 219, 241Volume of distribution at steady state (Vdss)

definition, 169–170determination using compartmental analysis,

170determination using NCA, 205

Volume of distribution in the elimination phase(Vd�), 168–169

definition, 168–169

dependency on elimination, 168, 169fdetermination using compartmental analysis,

169determination using NCA, 208

Volume of central compartment (V1), definitionand determination, 167–168

Warfarin, 4, 69t, 104, 105, 113tplasma protein binding, 77

Xanthine oxidase, 103

Zero asymptotic disease progression models,358–359

Zero order drug administration, 213. See alsoIntravenous infusion

Zero order processes, 378–380Zidovudine, 98, 105Zolmitriptan, 39