throbber
VOLUME 15
`
`RLUk
`
`second Edition
`Revised and Expanded
`
` DRUGS AND THE PHARMACEUTICAL SCIENCES
`
`informa
`
`Milo Gibaldi
`
`Delile msleatae
`
`CELGENE 2148
`APOTEX v. CELGENE
`IPR2023-00512
`
`healthcare
`
`
`
`

`

`

`

`Pharmacokinetics
`
`

`

`DRUGS AND THE PHARMACEUTICAL SCIENCES
`
`A Series of Textbooks and Monographs
`
`Edited by
`dames Swarbrick
`School of Pharmacy
`University of North Carolina
`Chapel Hilti, North Carolina
`
`Volume 1,
`
`Volume 2.
`
`PHARMACOKINETICS, Milo Gibaldi and Donald Perrier
`fout ofprint}
`
`GOOD MANUFACTURING PRACTICES FOR
`PHARMACEUTICALS: APLAN FOR TOTAL QUALITY
`CONTROL,Sidney H. Willig, Murray M. Tuckerman, and
`William S. Hitchings (V fout ofprint)
`
`Volume 3.
`
`MICROENCAPSULATION, edited by J. A. Nixon {out of print}
`
`Volume4.
`
`DRUG METABOLISM: CHEMICAL AND BIOCHEMICAL
`ASPECTS, Bernard Testa and Peter Jenner
`
`Volume 5.
`
`Volume 6.
`
`NEW DRUGS: DISCOVERY AND DEVELOPMENT,
`edited by Alan A, Rubin
`
`SUSTAINED AND CONTROLLED RELEASE DRUG DELIVERY
`SYSTEMS, édited by Joseph A. Robinson
`
`Volume 7.
`
`MODERN PHARMACEUTICS,edited by Gilbert S.
`Banker and Christopher T. Rhodes
`
`Volume 8.
`
`Volume 9.
`
`PRESCRIPTION DAUGS IN SHORT SUPPLY: CASE
`HISTORIES, Michael A, Schwartz
`
`ACTIVATED CHARCOAL: ANTIDOTAL AND OTHER
`MEDICAL USES, David 0. Cooney
`
`Volume 10.
`
`CONCEPTS IN DRUG METABOLISM {in two parts), edited
`by Peter Jenner and Bernard Testa
`
`Volume 11.
`
`PHARMACEUTICAL ANALYSIS: MODERN METHODS
`(in two parts}, edited by Jarnes W. Munson
`
`Volume 12.
`
`TECHNIQUES OF SOLUBILIZATION OF DRUGS,
`edited by Samuel H. Yalkowsky
`
`Volume 13.
`
`ORPHAN DRUGS, edited by Fred £. Karch
`
`Volume 14.
`
`NOVEL DRUG DELIVERY SYSTEMS: FUNDAMENTALS,
`DEVELOPMENTAL CONCEPTS, BIOMEDICAL ASSESSMENTS,
`Yie W. Chien
`
`Volume 15.
`
`PHARMACOKINETICS, Second Edition, Revised and Expanded,
`Milo Gibatdi and Donald Perrier
`
`Other Volumes in Preparation
`
`

`

`Pharmacokinetics
`
`SECOND EDITION, REVISED AND EXPANDED
`
`Milo Gibaldi
`University of Washington
`School of Pharmacy
`Seattle, Washington
`
`Donald Perrier
`School of Pharmacy
`University of Arizona
`Tucson, Arizona
`
`informa
`
`healthcare
`
`New York London
`
`

`

`Informa Healthcare USA, Inc.
`52 Vanderbilt Avenue
`New York, NY 10017
`
`© 2007 by Informa Healthcare USA, Inc.
`Informa Healthcare is an Informa business
`
`No claim to original U.S. Government works
`Printed in the United States of America on acid-free paper
`30 29 28 27 26 25 24 23 22 21
`
`International Standard Book Number-10: 0-8247-1042-8 (Hardcover)
`International Standard Book Number-13: 978-0-8247-1042-2 (Hardcover)
`
`This book contains information obtained from authentic and highly regarded sources. Reprinted material
`is quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable
`efforts have been made to publish reliable data and information, but the author and the publisher cannot
`assume responsibility for the validity of all materials or for the consequences of their use.
`
`No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,
`mechanical, or other means, now known or hereafter invented, including photocopying, microfilming,
`and recording, or in any information storage or retrieval system, without written permission from the
`publishers.
`
`For permission to photocopy or use material electronically from this work, please access www.copyright.
`com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood
`Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and
`registration for a variety of users. For organizations that have been granted a photocopy license by the
`CCC, a separate system of payment has been arranged.
`
`Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are
`used only for identification and explanation without intent to infringe.
`Visit the Informa Web site at
`www.informa.com
`and the Informa Healthcare Web site at
`www.informahealthcare.com
`
`

`

`Preface
`
`Pharmacokinetics is the study of the time course of drug absorption,
`distribution, metabolism, and excretion.
`It also concerns the relation-
`ship of these processes to the intensity and time course of pharma-
`cologic (therapeutic and toxicologic) effects of drugs and chemicals.
`Pharmacokinetics is a quantitative study that requires a preexisting
`competence in mathematics at least through calculus.
`It is also a
`biologic study and can be very useful to the biomedical scientist.
`At a fundamental level, pharmacokinetics is a tool to optimize the
`design of biological experiments with drugs and chemicals, All bio-
`logists would benefit from some knowledge of pharmacokinetics when-
`ever they engage in data analysis.
`It has become increasingly impor-
`tant in the design and development of new drugs and in the reass-
`essment of old drugs. Clinical applications of pharmacokinetics have
`resulted in improvements in drug utilization and direct benefits to
`patients,
`There is consensus that the origin of pharmacokinetics can be
`traced to two papers entitled "Kinetics of distribution of substances
`administered to the body" written by Torsten Teorell and published
`in the International Archives of Pharmacodynamics in 1937. Since
`this unheralded beginning, the study of pharmacokinetics has matured
`rapidly; undoubtedly growth has been stimulated by major break-
`throughs in analytical chemistry, which permit us to quantitatively
`detect minute concentrations of drugs and chemicals in exceedingly
`small volumes of biological fluids, in data processing, and by the bril-
`liant insights of many scientists. Dost, Kruger-Theimer, Nelson,
`Wagner, Riegelman, and Levy are among those scientists and must be
`reserved a special place in the history of the development of phar-
`macokinetics.
`Our goals in preparing this revision were similar to those that
`prompted us to undertake the initial effort. The need for revision
`was amply clear to us each time we looked at ourfiles, bulging with
`research papers and commentaries on pharmacokinetic methods and
`
`iii
`
`

`

`iv
`
`Preface
`
`applications published since 1975, The buzz words today are clearance
`concepts, noncompartmental models, and physiologic pharmacokinetics.
`Again, we strived to present the material in an explicit and detailed
`manner, We continue to believe that Pharmacokinetics can be used in
`formal courses, for self-study, or for reference purposes.
`We thank our colleagues for their work and publications, our
`staffs for their labors and support, and our families for their love
`and understanding.
`
`Milo Gibaldi
`Donald Perrier
`
`

`

`Contents
`
`Preface
`
`iil
`
`i.
`
`2.
`
`1
`
`One-Compartment Model
`Intravenous Injection
`Intravenous Infusion
`33
`First-Order Absorption
`Apparent Zero-Order Absorption
`References
`42
`
`2
`27
`
`40
`
`45
`
`Multicompartment Models
`Intravenous Injection
`Intravenous Infusion
`81
`First-Order Absorption
`Determination of Pharmacokinetic Parameters
`References
`109
`
`48
`63
`
`84
`
`113
`Multiple Dosing
`Intravenous Administration
`Intravenous Infusion
`128
`132
`First-Order Absorption
`Determination of Pharmacokinetic Parameters from
`Multiple-Dosing Data
`143
`References
`143
`
`113
`
`Absorption Kinetics and Bioavailability
`Absorption Rate
`146
`167
`Extent of Absorption
`Statistical Considerations in Comparative Bioavailability
`Studies
`185
`Sustained Release
`References
`195
`
`145
`
`188
`
`

`

`vi
`
`Contents
`
`199
`Apparent Volume of Distribution
`Relationship Between Volume of Distribution, Drug Binding
`and Elimination, and Anatomic Volume
`200
`Tissue Binding
`209
`Estimation of Apparent Volumes of Distribution
`References
`218
`
`211
`
`221
`Kinetics of Pharmacologic Response
`Kinetics of Directly Reversible Pharmacologic Response
`Kinetics of Indirect Pharmacologic Response
`245
`254
`Kinetics of Irreversible Pharmacologic Response
`Appendix: Solutions for Cs, Cp, and Cy for Cell Systems
`Sensitive to Drugs That are Cell Cycle Specific
`265
`References
`267
`
`221
`
`271
`Nonlinear Pharmacokinetics
`271
`Michaelis-Menten Kinetics
`Some Pharmacokinetic Characteristics of Michaelis-Menten
`Processes
`272
`277
`In Vivo Estimation of Km and Vy,
`Clearance, Half-Life, and Volume of Distribution
`Drug Concentration at Steady State
`289
`Time to Steady State
`290
`Area Under the Curve and Bioavailability
`Composition of Urinary Excretion Products
`Other Nonlinear Elimination Processes
`301
`Enzyme Induction
`303
`Nonlinear Binding
`307
`Some Problems in Quantifying Nonlinear Pharmacokinetics
`References
`315
`
`294
`297
`
`287
`
`313
`
`319
`
`Clearance Concepts
`319
`Organ Clearance
`321
`Total Clearance
`322
`Hepatic Clearance
`Hepatic Clearance and Drug Binding in Blood
`Drug Binding and Free Drug Concentration
`Half-Life, Intrinsic Clearance, and Binding
`First-Pass Effect
`332
`Gut Wali Clearance
`336
`338
`Lung Clearance
`341
`Renal Clearance
`Clearance Concepts Applied to Metabolites
`Physical Models of Organ Clearance
`347
`Blood Clearance Versus Plasma Clearance
`References
`351
`
`327
`330
`331
`
`344
`
`349
`
`

`

`Contents
`
`vii
`
`355
`
`9.
`
`364
`
`Physiological Pharmacokinetic Models
`Blood Flow Rate-Limited Models
`358
`Experimental Considerations
`Blood Clearance
`366
`Lung Clearance
`368
`Apparent Volume of Distribution
`Nonlinear Disposition
`370
`372
`Membrane-Limited Models
`Species Similarity and Scale-Up
`References
`382
`
`369
`
`375
`
`10. Application of Pharmacokinetic Principles
`Multiple Dosing
`385
`Dose Adjustments in Renal Failure
`Hemodialysis
`397
`Methods for Determination of Individual Patient Parameters
`References
`405
`
`385
`
`393
`
`401
`
`410
`
`11. Noncompartmental Analysis Based on Statistical Moment
`Theory
`409
`Statistical Moments
`Bioavailability
`411
`Clearance
`411
`Half-Life
`412
`413
`Absorption Kinetics
`Apparent Volume of Distribution
`Fraction Metabolized
`414
`Predicting Steady-State Concentrations
`Predicting Time to Steady State
`415
`Conclusions
`416
`References
`416
`
`413
`
`414
`
`Appendix A Method of Laplace Transforms
`References
`423
`
`419
`
`Appendix B Method for Solving Linear Mammillary Models
`References
`431
`
`425
`
`Appendix C Method of Residuals
`
`433
`
`Appendix D Estimation of Areas
`Reference
`449
`
`445
`
`Appendix E Prediction of Drug Concentrations on Multiple Dosing
`Using the Principle of Superposition
`451
`References
`457
`
`

`

`vil
`
`Contents
`
`Appendix F Estimation of Rates
`Reference
`463
`
`459
`
`Appendix G Selective Derivations
`465
`Michaelis-Menten Equation
`Time To Reach a Fraction of Steady State for a Drug Eliminated
`by Parallel First-Order and Capacity-Limited Processes
`467
`Reference
`473
`
`465
`
`Computer Programs
`Appendix H
`References
`476
`
`475
`
`Author Index
`Subject Index
`
`479
`489
`
`

`

`7 N
`
`onlinear Pharmacokinetics
`
`At therapeutic or nontoxic plasma concentrations, the pharmacokinetics
`of most drugs can be adequately described by first-order or linear
`processes. However, there are a small number of well-documented
`examples of drugs which have nonlinear absorption or distribution
`characteristics [e.g., ascorbic acid [1] and naproxen [2,3], re-
`spectively], and several examples drugs that are eliminated from
`the body in a nonlinear fashion.
`
`MICHAELIS-MENTEN KINETICS
`
`Drug biotransformation, renal tubular secretion, and biliary secretion
`usually require enzyme or carrier systems. These systems are rel-
`atively specific with respect to substrate and have finite capacities
`(i.e., they are said to be capacity Himited). Frequently, the kinetics
`of these capacity-limited processes can be described by the Michaelis-
`Menten equation:
`
`_ ac mo
`dt K,te
`
`(7.1)

`
`where —dC/dt is the rate of decline of drug concentration at time t,
`Vm the theoretical maximum rate of the process, and Km the Michaelis
`constant.
`It is readily seen by determining C when —dC/dt = (1/2)Vm
`that Kp is in fact equal to the drug concentration at which the rate
`of the process is equal to one-half its theoretical maximum rate.
`Equation (7.1) can be derived based on the following scheme (see
`Appendix G for derivation):
`
`Ky
`k
`E+Cc => EC ye ESM
`Ky
`
`271
`
`

`

`272
`
`Pharmacokinetics
`
`In this scheme C is the concentration of drug, E the concentration of
`enzyme, EC the concentration of the enzyme-drug complex, and M the
`concentration of metabolite. The constants kg and k_, are first-order
`rate constants, and kj is a second-order rate constant. The Michaelis-
`Menten equation is of value for describing in vitro and in situ as well
`as certain in vivo rate processes. For in vivo systems the constants
`Vm and Kp are affected by distributional and other factors and there-
`fore must be viewed as functional, model-dependent constants.
`
`SOME PHARMACOKINETIC CHARACTERISTICS OF
`MICHAELIS-MENTEN PROCESSES
`
`There are two limiting cases of the Michaelis-Menten equation.
`is much larger than C, (7.1) reduces to
`
`If Ky
`
`dC _om
`-z Cc
`
`_
`
`(7.2)
`
`This equation has the same form as that describing first-order elimina-
`tion of a drug:
`(1) after intravenous administration in a one-compart-
`ment model, (2) in the postabsorptive phase after some other route of
`administration in a one-compartment model, or (3) in the postabsorp-
`tive, postdistributive phase in a multicompartment model. Assuming
`apparent first-order elimination of a drug which confers one-compart-
`ment characteristics to the body and which is eliminated by a single
`biotransformation process, the first-order rate constant K is actually
`Vm/Km. As shown in (7.2), if treatment with an enzyme inducer
`causes an increase in the amount of enzyme (and therefore of V,,), the
`apparent first-order rate constant of the process will also be increased.
`Given the fact that drug elimination is so frequently observed to follow
`apparent first-order kinetics, one must conclude that the drug con-
`centration in the body (or, more correctly, at the site of an active
`process) resulting from the usual therapeutic dosage regimens of
`most drugs is well below the Km of the processes involved in the dis-
`position of these drugs.
`There are some notable exceptions to this generalization and among
`them are ethanol [4], salicylate [5,6], and phenytoin [7]. The elim-
`ination kinetics of phenytoin [8] and ethanol [9] appear to be ade-
`quately described by a single Michaelis-Menten expression, while
`salicylate elimination [6] may be described by two capacity-limited
`and three linear processes. Marked deviations from apparent first-
`order drug elimination have also been noted frequently in cases of
`drug intoxications.
`In the latter situation there is often some ambigu-
`ity as to whether the deviations are due to capacity-limited biotrans-
`formation of the high drug levels in the body [described by (7.1)] or
`due to some toxicologic effect of the drug.
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`273
`
`Another limiting case of the Michaelis-Menten equation is that
`which results when the drug concentration is considerably greater
`than Ky,. Equation (7,1) then reduces to
`
`dc
`dt
`
`m
`
`(7.3)
`
`Under these conditions, the rate is independent of drug concentra-
`tion, so that the process occurs at a constant rate Vy,. The kinetics
`of biotransformation of ethanol [4] have been observed to approach
`the condition described by (7.3) even at drug levels in the body
`that are appreciably lower than those considered to be toxic.
`Based on the discussion above, if ~dC/dt is plotted as a function
`of plasma concentration, —dC/dt would initially increase linearly
`with concentration, indicating first-order kinetics (Fig. 7.1). As the
`concentration increases further, —dC/dt would increase at a rate less
`
`than proportional to concentration, and eventually asymptote at a S
`
`
`
`
`
` 0 190 150 200
`
`
`
`CONCENTRATION (g/m?)
`
`Fig. 7.1 Relationship between drug elimination rate -dC/dt and drug
`concentration C for a Michaelis-Menten process.
`In this particular
`example the Michaelis constant K,, is equal to 10 ug/ml and the max-
`imum rate V,, is equal to 2ug/m))h-1,
`
`

`

`274
`
`Pharmacokinetics
`
`(ug/ml) 0
`Logscale—plasmaconcentrationofdiphenythydantoin
`
`
`50
`
`joo
`
`150
`
`Time measured from !2h after last dose (h)
`
`Fig. 7.2 Phenytoin (diphenylhydantoin) concentration in plasma 12h
`after the last dose of a 3 day regimen of the drug at three different
`daily doses. The data are described by Eq. (7.9). O:
`17.9 mg/kg;
`A: 4.7 mg/kg; O: 2.3 mg/kg.
`(From Ref. 10, © 1972 PJD Publica-
`tions Ltd., reprinted with permission.)
`
`rate equal to V,, which would be independentof concentration (i.e.,
`a zero-order rate).
`The time course of drug plasma concentration after intravenous
`injection of a drug that is eliminated only by a single capacity-limited
`process can be described for a one-compartment system by the inte-
`grated form of the Michaelis-Menten equation. Rearrangement of
`(7.1) yields
`
`_
`ac
`C (C+K) =v, dat
`
`or
`
`K dc
`
`-ac -—— =v at
`Cc
`m
`
`(7.4)
`
`(7.5)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`Integration of this equation gives the expression
`
`-C -K_ mnC=Vit+ti
`m
`m
`
`275
`
`(7.6)
`
`where iis an integration constant. Evaluating i at t = 0, where
`C=Cq, yields
`i=-C)- Ki. in Cy
`
`(7.7)
`
`Substituting this expression for iin (7.6) and rearranging terms gives
`
`(7.8)
`
`1.0
`
`a3-=:
`
`c.,-c¢
`- 8,
`t =
`7
`m
`
`Cc
`K
`_m,, 2
`+ 7 in c
`m
`
`100
`
`o
`Te) 250=500
`a
`TIME (min)
`
`
`
`°c
`
`o = _z>S
`
`o=<
`
`4
`
`Of
`
`o
`
`200
`
`400
`TIME (min)
`
`600
`
`800
`
`Fig. 7.3 Amount of drug in the body following intravenous adminis-
`tration of 1, 10, and 100 mg doses of a drug that is eliminated by a
`single Michaelis-Menten process. A one-compartment system is as-
`sumed; Km = 10 mg and V,, = 0.2 mg/min. The inset shows a plot of
`amount of drug in the body divided by administered dose versus time
`to show that the principle of superposition does not apply.
`
`

`

`276
`
`3000
`
`2000
`
`Pharmacokinetics
`
`
`
`Amountunexcreted(mgospirinequivolent)
`
`
`
`
`
`500
`
`200
`
`100
`
`re,°
`
`ore.‘as
`
`1000
`

`
`"a,
`~e
`™
`
`e
`
`“a,
`ese
`“e
`Beginning of
`N\ ~
`LN Gem first-order
`*
`
`teo
`
`Time
`
`(h)
`
`Fig. 7.4 Elimination of salicylate after oral administration of 0.25,
`1.0, and 1,5 g doses of aspirin. Vertical arrows on the time axis
`indicate t5qg, the time to eliminate 50% of the dose.
`(From Ref. 5,
`reprinted with permission.)
`
`Unfortunately, it is not possible to solve this equation explicitly for
`Cc. Rather, one must determine the time t at which the initial concen-
`tration Cg has decreased to C. A modified form of (7.8), that is,
`
`c,-cC kK
`Cc
`0
`m
`0
`t-th=—y_ +-——In >
`m
`m
`
`(7.9)
`
`has been used to fit phenytoin levels in the plasma as a function of
`time 12 h after administration of the last of several oral doses to human
`subjects (see Fig. 7.2).
`In this case Cg represents the phenytoin
`
`plasma concentration at 12 h after the last dose, tg = 12h, andCis
`the phenytoin plasma concentration at time t, where t > ty.
`Conversion of (7.8) to common logarithms (In x = 2.303 log x)
`and solving for log C yields
`Via
`Cy 7
`log C = s-a59x «+ 1°06 Cg ~ Frg0gK t
`2.303K
`2.303K
`m
`m
`
`(7.10)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`277
`
`Figure 7.3 shows the time course of elimination, as described by (7.10),
`of three different doses of a drug that is eliminated by a process with
`Michaelis-Menten kinetics. The lowest dose represents the case where
`Ky, >> C. At this dose the decline in plasma concentrations is first
`order with a slope of —V,,/2.303K,,. On the other hand, the highest
`dose yields initial concentrations which are considerably above Ky,
`so that drug levels decline initially at an essentially constant rate (see
`inset to Fig. 7.3). The curves show that the time required for an
`initial drug concentration to decrease by 50% is not independent of
`dose, but, in fact, increases with increasing dose. This particular
`pharmacokinetic property may present considerable clinical difficulty
`in the treatment of drug intoxications. Figure 7.3 also shows that re-
`gardless of the initial dose, when the plasma concentration becomes
`significantly less than Kp, elimination is describable by first-order
`kinetics and the slope of this linear portion of the curve is inde-
`pendent of dose. Semilogarithmic plots of plasma concentration or
`amount unexcreted versus time after administration of phenytoin (Fig.
`7.2) or salicylate (Fig. 7.4) show characteristics that are remarkably
`similar to those described by the curves in Fig. 7.3.
`To assess whether or not a drug possesses nonlinear kinetic prop-
`erties, a series of single doses of varying size should be administered.
`If a plot of the resulting plasma concentrations divided by the ad-
`ministered dose are superimposable, the drug in question has linear
`kinetic properties over the concentration range examined.
`If, how-
`ever, the resulting curves are not superimposable (see inset to Fig.
`7.3), the drug behaves nonlinearly.
`
`IN VIVO ESTIMATION OF K,, AND Vy,
`
`For a drug that is eliminated by a single capacity-limited process,
`there are a number of general methods which permit the initial estima-
`tion of apparent in vivo Km and V,, values from plasma concentration-
`time data in the postabsorptive-postdistributive phase. Such estimates
`require the determination of the rate of change of the plasma concentra-
`tion from one sampling time to the next, AC/At, as a function of the
`plasma concentration Cy, at the midpoint of the sampling interval (see
`Appendix F). The data are usually plotted according to one of the
`linearized forms of the Michaelis-Menten equation, such as the Line-
`weaver-Burk expression,
`
`K
`
`1
`1
`m
`=
`+-~—
`AC/At
`Viaem Vin
`
`(7,11)
`
`so that a plot of the reciprocal of AC/At versus the reciprocal of Cm
`yields a straight line with intercept 1/V,, and slope Km/Vm. Two
`sometimes more reliable [11,12] plots are the Hanes-Woolf plot [13] and
`
`

`

`278
`
`Pharmacokinetics
`
`the Woolf-Augustinsson-Hofstee plot [13]. They are based on the
`relationships
`Cay
`Kn
` Cn
`AC/At VV
`m
`m
`
`(7,12)
`
`and
`
`SC iy _ OCHOKa(7,13)
`At
`m
`Cc.
`,
`
`respectively. Based on (7.12), a plot of C,,/(AC/At) versus Cy
`should yield a straight line with a slope of 1/V, and an intercept of
`Ky/Vm. Equation (7.13) indicates that a plot of AC/At versus
`(4C/At)/C,, gives a straight line with a slope of —Km and an inter-
`cept of Vy.
`A method for estimating V,, and K,, directly from log C versus time
`data, obtained following the intravenous administration of a drug that
`can be adequately described by a one-compartment system, is also
`available [14]. Extrapolation of the terminal log-linear portion of the
`log C versus time plot, where the plot is described by (7.10), would
`yield a zero-time intercept of log Ch (see Fig. 7.5). The resulting
`straight line can be described by
`
`=
`
`log C = log C,
`
`Vv
`‘mS
`
`ra0aK_*
`
`(7.14)
`
`At low plasma concentrations (7.10) and (7.14) are identical. By
`setting the right-hand sides of these two equations equal to each
`other, the following expression is obtained:
`
`c.-c
`
`Vv
`
`—9_siongcc —- —™_ t=log cc. - ——2-
`2.303K
`0
`2.303K
`0
`2.308K
`
`Vv
`
`(7.15)
`
`Cancellation of the common term, Vpt/2.303K,,, and rearrangement
`yields
`
`*
`
`Cc
`c.-c
`0 =loo—2
`
`7.303K.
`~ 8
`(7.16)
`
`m
`
`0
`
`Since the equality given by (7.15) is valid only at low concentrations,
`Cy can be assumed to be significantly greater than C, and therefore
`C,—-C Cg. Making this simplification in (7.16) and solving the re-
`sulting expression for K,, gives
`Cg
`K, ” 2,308 log (C#IE))
`
`(7.17)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`279
`
`SLOPE = “V2 -303K,,
`
`(ug/ml) 4
`CONCENTRATIONINPLASMA
`
`8
`
`le
`
`16
`
`20
`
`24
`
`TIME (h)
`
`Fig. 7.5 Graphical method for estimating Km and V,, after intravenous
`administration of a drug that is eliminated by a single Michaelis-
`Menten process. The solid line is described by Eq. (7.10). The
`terminal slope gives an estimate of the ratio of Vy, to K,,, and the
`ratio of Ch to Cg is used to estimate K,, [see Eq. (7.17)].
`
`Since C* and Cg can be estimated from a log C versus time plot, an
`estimate of K,, is possible employing (7.17). V,, can be calculated
`from the slope of the terminal log-linear segment of the concentration
`versus time curve. Since slope = —-V_y/2.303K,, Vin = —2.303(slope)Km.
`It is plausible to consider that drug elimination may involve a
`eapacity-limited process in parallel with one or more first-order proc-
`esses. Under these conditions, the foregoing methods for estimating
`Vm and Ky do not apply. When capacity-limited and first-order elim-
`ination occur in parallel, the rate of decline of drug levels in the
`plasma after intravenous administration in a one-compartment system
`is given by
`
`VC
`dC Liaw pm
`at
`RCT RSS
`
`(7.18)
`
`where K' is the rate constant characterizing the various parallel
`first-order processes. The time course of drug levels under these
`conditions may be determined by integration of (7.18) as follows.
`Expansion of (7.18) yields
`
`

`

`280
`
`Pharmacokinetics
`
`K'C(K +C)+V_C K'K C+vV C+K'C?
`-e =2. (7.19)
`K +C
`~
`K +C
`dt ~
`'
`m
`m
`
`Further simplification gives rise to
`
`CCK'K + KV_
`+ K'C)
`d
`t
`~d@ omms Ca + K'C)
`at
`K +C
`K +¢
`m
`m
`
`(7.20)
`
`where a = K'K,, + Vm.
`
`Inversion and rearrangement of (7.20) yields
`
`m
`m
`dat
`dC C(a+K'C)” Clat+K'C)
`
`1
`a+kK'C
`
`This equation is separable and can be rewritten as
`
`_ —Kde
`
`C(a + K'C)
`
`_
`
`dt
`
`dc
`
`a+ K'C
`
`(7.22)
`
`(1.22)
`
`.
`
`The two terms in this equation are of the form 1/x(a + bx) and
`1/(a + bx), respectively, the integrals of which are (—1/a) In [(a +
`bx)/x] and (1/b) In (a + bx) [15]. Therefore, integration of (7.22)
`gives
`
`.
`;
`atKiC 1
`_om,
`tes ins
`xin (a+ KIC) +3
`
`(7.23)
`
`Evaluating i at t = 0, where C = Cg, yields
`Ka
`at KiCy
`1
`eoMy '
`(7,24)
`a
`In c
`tim In (a+ K'C))
`i
`Substituting this value of iin (7.23) and simplifying the resulting
`expression yields
`
`at Kc)
`a
`Cy
`at=K In | + x” ¥, nMoyKC
`
`(7.25)
`
`Since a = K'Ky, + Vm;
`
`(K'K +Vo)t=Ko
`m
`m
`
`m
`
`{KK+V
`Cc,
`In—+| —S"-xk }]1
`Ki
`Cc
`m
`
`KK +V_ +K'C,
`m
`TKK +V +K'C
`m
`m
`
`(7.26)
`
`or
`
`
`
`cv (C_+K )K'+V
`
`
`_ _0,m1 0 m m
`
`m
`m
`™m
`m
`
`aa s,m e+ In aK OKT |
`
`
`
`(7,27)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`281
`
`{0
`
`100
`
`4K
`
`y
`
`|
`
`Dose
`
`12
`
`tso%,
`
`oa
`

`
`001
`
`0.1
`
`Fig. 7.6 Comparison of dose dependence of t599 (time for elimination
`of 50% of the dose) after intravenous administration of a drug that is
`eliminated by a single Michaelis-Menten process (upper curve) or
`one that is eliminated by a single Michaelis-Menten process in parallel
`with a first-order process (lower curve).
`In each case, Kp = 1.0
`and Vm = 0.2. The rate constant K' for the first-order process is
`equal to 0.1.
`(Data from Ref, 16.)
`
`Equation (7.27), like (7.8), does not permit an explicit solution for
`C. Both (7.8) and (7.27) indicate that the time required to reduce
`an initial drug concentration by 50% is indeed dependent on the ad-
`ministered dose. Examples of this dependency are shown in Fig. 7.6.
`Expanding (7.27) and solving for In C gives
`Via
`Vv
`(Co +Kme + Via
`oOmm ! —_
`in C nC * igiK "CrkVV K *K t
`
`(7.28)
`
`=
`
`which in terms of common logarithms is
`Via
`(C,+K. )K! tv K' +V/K
`eC =e Cot eK ME (Ce KOK TV 2.308
`
`(7.29)
`
`

`

`282
`
`Pharmacokinetics
`
`At low concentrations (i.e., Ky, >> C), (7.29) becomes
`Via
`(Cy + KK + Vin
`K' + Vin Km
`log C= 108 Cy * ii~ log——"FetyVy-~— yon ~—t
`m
`m
`
`or
`
`log C = log Cy~ at
`
`K'+V_/K
`
`where
`
`(7.30)
`
`(7.31)
`
`Via
`(Cy + Ke + Va
`log Co =log Co*KKaeKKAV (7.32)
`
`As can be seen from (7.31), the slope of the terminal linear portion of
`a semilogrithmic plot of plasma concentration versus time at low plasma
`concentrations (i.e., K,, >> C) will yield an estimate of the first-order
`elimination rate constant of a drug, K' + Vm/K,, (see Fig. 7.7). The
`extrapolated intercept of this terminal linear phase wiil be log Cp.
`For certain drugs that exhibit parallel capacity-limited and first-
`order elimination, it may be possible to administer sufficiently high
`doses intravenously so that initial drug concentrations are substan-
`tially larger than K,. Under these conditions and where a one-com-
`partment model applies, the initial segment of a semilogarithmic plot
`of plasma concentration versus time will be linear (see Fig. 7.7).
`The slope of this linear segment will be —K'/2.303 [14]. This can be
`demonstrated by assuming C to be much greater than K,, in (7.18)
`and solving for C. Therefore, estimates of both K' and K' + Vm/Ky,
`ean be obtained directly from a semilogarithmic plot of plasma concen-
`tration versus time and the ratio V,,/K, can be calculated for the case
`where there is one capacity-limited process in parallel with one or
`more first-order processes. The following approach can then be used
`to obtain initial estimates of Ky, and Vp for this model. Expansion of
`the logarithmic term of (7.32) and rearrangement of this equation
`yields
`K'K|
`
`
`0
`
`m
`
`m
`
`Division of the numerator and denominator of this logarithmic term by
`K,, gives
`K'K
`
`m
`Vv,
`
`*
`
`Cc
`0
`Cy
`
`log —— = oe fe (7.34)
`
`Cc oh
`K(kh+V7K)
`
`*
`Cy
`
`Cok!
`
`log & = log (eg
`
`v
`
`m
`
`(7.33)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`283
`
`400
`
`200
`
`100
`
`10
`
`=
`ad
`oOo
`
`=<
`
`x 5<
`
`x
`amd
`a
`
`Go=
`o
`<=
`x=
`i
`wy
`
`So
`
`40
`
`20
`
`TIME (h)
`
`Fig. 7.7 4-Hydroxybutyric acid (4-HBA) concentration in plasma
`after intravenous administration. The compound appearsto be elim-
`inated by a Michaelis-Menten process in parallel with a first-order
`process. The initial slope gives an estimate of K' and the terminal
`slope provides an estimate of the ratio of V,, to K,,. K,, may be de-
`termined from Eq. (7.35). The deviation from theory for a short
`time after administration probably reflects drug distribution and the
`lack of strict adherence to a one-compartment model.
`(Data from
`Ref. 14.)
`
`A solution for K,, based on this equation is
`t
`'
`_— COKU(K' + V_/K.)
`m
`*
`K'K/V
`(Co/Co)
`-1
`
`(7.35)
`
`

`

`284
`
`Pharmacokinetics
`
`Since Cy and C4 can be obtained directly from a semilogarithmic plasma
`concentration-time plot, and K' and Vp/K,;, can be estimated as de-
`scribed above, an estimate of K,, is possible employing (7.35). Once
`K,, is known, V,, can be readily determined.
`Although this is an interesting approach for the estimation of K',
`Vm» and Km where there are parallel first-order and nonlinear elimina-
`tion pathways, caution must be exercised.
`Initial plasma concentrations
`have to be sufficiently high (i.e., C >> K,,) to yield a semilogarithmic
`plasma concentration-time curve which is truly linear.
`If these con-
`
`23.0
`
`21.0
`
`19.0
`
`4.0
`
`3.0
`
`2.0
`
`0.0
`
`(-dInCidt)x102
`
`J K+
`
`m= SLOPE = Vm
`
`0.1
`
`0.2
`
`Hatt
`10
`30
`50
`70
`90
`110 130
`0.3
`1/C x 107
`
`Fig. 7.8 Graphical method for estimating K' (designated K, in the
`plot), Vm and Km based on Eqs. (7.36) to (7.39). At high concentra-
`tions, a plot of -A In C/At versus 1/C will be linear with a slope of
`Vin and an intercept equal to K' [see Eq. (7.38)], whereas at low con-
`centrations the plot will asymptotically approach a limiting value equal
`to K' + (Vi,/Km) [see Eq. (7.39)].
`(From Ref. 17, © 1973 Plenum
`Publishing Corp.)
`
`

`

`7 / Nonlinear Pharmacokinetics
`
`285
`
`centrations are not attained, an overestimate of K' will result. This,
`in turn, will produce errors in the estimates of Vm and Ky.
`A different approach can also be used for the estimation of Ky
`and V,, where nonlinear and linear processes of drug elimination occur
`in parallel. At high plasma concentrations (i.e., C >> Km), (7.18)
`reduces to
`
`dt
`
`(7.36)
`- de = xtc tv,
`Division of both sides of (7.36) by C and recognition that (—dC/dt)/C
`equals —d In C/dt gives
`
`Vv
`_dinc _,,, m
`aC K' + C
`
`At low plasma concentrations (i.e., Ky >> C), (7.18) becomes
`
`Vv
`aC Lia,
`at “Ke +R
`and, therefore, the analogous expression to (7.37) is
`
`a ame =KI+ om
`
`Vv
`
`m
`
`(7.37)
`
`(7.38)
`
`(7.39)
`
`A plot of — A In C/At versus 1/C will consequently be linear with a
`slope of V,, and an intercept of K' at high plasma concentrations [Eq.
`(7.37)], but will reach an asymptotic value of K' + V,)/K,, at low con-
`centrations [Eq. (7.39)] from which K,, can be calculated (see
`Fig. 7.8).
`This method for estimating K', V,,, and K,, has limitations which
`are similar to those noted for the previous approach. Sufficiently
`high plasma concentrations are required to yield a straight line from
`the -A In C/At versus 1/C plot to permit accurate estimates of V,,
`and K'.
`Urine data can also be used to estimate V,, and K,,. Consider
`the following scheme:
`
`XE
`
`Kt
`
`M—>M
`mu
`
`

`

`286
`
`Pharmacokinetics
`
`where X is the amount of drug in the body, Xp the amount of drug
`eliminated by the linear or first-order processes, M the amount of
`metabolite in the body which is formed by a capacity-limited process,
`and M,, the amount of this metabolite present in the urine. All of
`these amounts are time dependent. The constants K' and ky are
`first-order rate constants, Vin is the maximum rate of metabolite forma-
`tion in units of amount per time, and Kin is the Michaelis constant in
`units of amount. Assuming that the urinary excretion rate of the
`metabolite (4M,,/At) is rate limited by its formation, and therefore
`reflects the rate of formation, the following relationship for AMy/dt
`‘ean be written:
`
`AM,
`
`Vin em
`
`m
`
`= 100
`
`980
`
`2<c z
`
`660
`
`2BR
`Lu
`co
`2
`peo
`Qga
`Ri
`a!
`°a
`Zz
`gs
`
`20
`
`0.5
`
`4.0
`
`1.5
`
`2.0
`
`2.5
`
`SA IN BODY (g}
`
`Fig. 7.9 Plot of linearized form of the Michaelis-Menten equation to
`describe the formation of salicy] phenolic glucuronide (SPG) after a
`single dose of salicylic acid (SA). According to Eq. (7.40) and the
`co

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket