throbber
Clinical Pharmacokinetics
`
`
`
`,
`iliird eolilion
`
`
`
`MALCOLM ROWLAND, PhD.
`‘
`
`
`
`
`
`
`
`
`Contep’rs and Applications
`
`Department ol Pharmacy
`
`Universily ol Manchester
`
`Manchester, England
`
`THOMAS N. TOZER, PhD.
`
`School of Pharmacy
`
`University ol Calilornia
`
`San Francisco, California
`
`
`
`A, Lea & Febiger Book
`
`
`
`
`
`é‘g LiPPINCOTTWILLlAMS a; WILKINS
`I, ' A Wolters Kluwér Company
`, Philadelphia ' Baltimore - New York ' London
`Buenos Aires - Hong Kong - Sydney - Tokyo
`
`BDSI V. RB PHARMACEUTICALS LTD
`IPR2014-00325
`
`Page 1
`
`Page 1
`
`RB Ex. 2026
`BDSI v. RB PHARMACEUTICALS LTD
`IPR2014-00325
`
`

`

`AVAVv
`V
`
`Executive Editor: Donna Balaa’o
`Developmental Editors: Frances Klass, Lisa Stead
`Production Manager.- laurz’e Forsytb
`Project Editor.- Roberl D. Magee
`
`Copyright © 1995
`Lippincott Williams 5c Wilkins
`530 Walnut Street
`Philadelphia, Pennsylvania l9l06-3621 USA
`
`
`
`All rights reserved. This book is protected by copyright, No part of this book may be reproduced in any
`form or by any means, including photocopying, or utilized by any information storage and retrieval system
`without written permission from the copyright owner.
`
`Accurate indications, adverse reactions, and dosage schedules for drugs are provided in this book, but it is
`possible they may change. The reader is urged to review the package information data of the manufacturers
`of the medications mentioned.
`
`Printed in the United States of America
`
`First Edition 1980
`
`Library of Congress Cataloging—in-Publication Data
`
`1213141516171819 20
`
`Rowland, Malcolm.
`Clinical Phannacokinetics : concepts and applications / Malcolm
`Rowland, Thomas N. Tozer. — 3rd ed.
`p.
`cm.
`“A Lea & Febiger Book.”
`Includes bibliographical references and index.
`ISBN 978—0-683-07404-8
`ISBN 0—683—07404—0
`
`1. Pharmacokinetics.
`11. Title.
`
`2. Chemotherapy.
`
`I. Tozer, Thomas N.
`
`2. Drug Therapy.
`
`QV 58 R883c 1994]
`
`[DNLM: 1. Pharmacokinetics.
`RM301.S.R68
`1994
`615.7—dc20
`DNLM/DLC
`
`for Library of Congress
`
`94—26305CIP
`
`7776 Publishers have made every eflort to trace the copyright boldersjor borrowed material. If they have in.—
`acluerterttly overlooked any, they will be pleased to ma/ee the necessary arrangements at tbefirst opportunity.
`
`Page 2
`
`Page 2
`
`

`

`
`
`VARIABILITY
`
`
`
`I
`
`itcrtVEs
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Th1e reader will beable to:I
`
`List six motor sources 0fvariabilityIn drUQ response
` 2 Evaluate whether variabilityIn drug response iscaused by a variabilityIn pharmacokinet'Ics
`
`pharmacodynamiIcs, or both, given response and pharmacokineticdata.
`
` 3. State why variability around the mean and shape of thefrequency distribution histogram of
`
`a parameter are as important as the mean itsell.
`
`
` 4, Explain how variability'In hepatic enzyme activity manifests itselfIn variabilityIn both phar-
`
`macokinetic parameters and plateau plasma drug concentrations for drUgs of highand low
`
`
`hepatic extraction ratios
`
`
` 5. Suggestan approach for initiating a dosage regimen foran individual patient, given patient '
`
`population pharmacokinetic data and the individualsmeasurable characteristics.
`
`
`
`
`
`Thus far, the assumption has been made that all people are alike. True, as a species, humans
`
`
`are reasonably homogeneous, but differences among people do exist including their re-
`
`
`sponsiveness to drugs. Accordingly, there is a frequent need to tailor drug administration
`
`to the individual patient. A failure to do so can lead to ineffective therapy in some patients
`and toxicity in others.
`This section of the book is devoted to individual drug therapy. A broad overview of the
`subject is presented in this chapter. Evidence for and causes of variation in drug response,
`and approaches toward individualizing drug therapy are examined. Subsequent chapters
`deal in much greater detail with genetics (Chap. 14), age and weight (Chap. 15), disease
`(Chap. 16), interactions between drugs within the body (Chap. 17), and monitoring of
`plasma concentration of a drug as a guide to individualizing drug therapy (Chap. 18).
`Before proceeding, a distinction must be made between an individual and the popula—
`tion. Consider, e.g., the results of a study designed to examine the contribution of an acute
`disease to variability in drug response. Suppose, of 30 patients studied during and after
`recovery, only 2 showed a substantial difference in response; in the remainder the differ—
`ence was insignificant. Viewed as a whole, the disease would not be considered as a sig—
`nificant source of variability, but to the two affected patients it would. Moreover, to avoid
`toxicity, the dosage regimen of the drug may need to be reduced in these two patients
`during the disease. The lesson is clear: Average data are useful as a guide; but ultimately,
`information pertaining to the individual patient is all—important.
`On a similar but broader point, substantial differences in response to most drugs exist
`among patients. Such z‘nten‘ndividual variability is often reflected by a variety of marketed
`dose strengths of a drug. Because variability in response within a subject (immindividual)
`is generally much smaller than inte'rindividual variability, once well—established, there is
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`203
`
`Page 3
`
`
`
`
`Page 3
`
`

`

`204
`
`VARIABILITY
`
`CHAPTER I 3
`
`usually little need to subsequently adjust an individuals dosage regimen Clearly, il‘intmin-
`dividual variability were large and unpredictable, trying to titrate dosage for an individual
`would be an extremely difficult task, particularly for drugs with narrow therapeutic win—
`dows. Stated differently, a drug that exhibits a high intmindividual variability in pharma—
`eokinetics can be prescribed only if it has a wide therapeutic window.
`
`EXPRESSIONS 0F INDIVIDUAL DIFFERENCES
`
`Evidence for interindividual differences in drug response comes from several sources.
`Variability in the dosage required to produce a given response is illustrated in Figure 17 5
`(Chap. 1), which shows the wide range in the daily dose of warfarin needed to produce a
`similar degree of anticoagulant control. Variability in the intensity of response with time
`to a set dose is seen with the neuromuscular agent doxacurium (Fig. 13—1). As illustrated
`in Figs. 13—2 and 13—3, which show frequency distribution histograms of the plateau plasma
`concentration of the antidepressant drug nortriptyline, to a defined daily dose of the drug
`and the plateau unbound plasma concentration of S—warfarin required to produce a similar
`degree of anticoagulant control, variability exists in both pharmacolcinetics and pharma—
`codynamics. Variability in pharmaeokinetics was also illustrated by the wide scatter in the
`plateau plasma concentration of phenytoin seen following various daily doses of this drug
`(see Fig. 1—6, Chap. 1).
`
`The Need for Models
`
`The magnitude and relative contribution of pharmacokinetics and pharmacodynamics to
`variability in response to a given dosage within a patient population vary with the drug and,
`to some extent, the condition being treated. For example, with a nonsteroidal anti—inflame
`matory drug, the relative contribution of pharmacodynamic variability may be different
`when the endpoint is the relief of a headache than when it is the relief from chronic aches
`and pains associated with inflamed joints. In clinical practice, attempts to assign the relative
`contribution to pharmacokinetics and pharmacodynamies may be made based on direct
`observations of plasma concentration and response. The assignment could be strongly in--
`fluenced, however, by the timing of the observations and the magnitude of the response,
`
`
`
`
`
`NumberofPatients
`
`
`
`Fig. 13—1. The degree of neuromus-
`cular block with time after an iv. bolus
`dose of 0.01 mg/kg doxacurium to pa-
`tients varies widely. (1 mg/L = 0.97 HM)
`(Modified from Sehmith, V.D., Fiedler-
`Kelly, l, AboueDonia, M., Huffman,
`(3.8., and Grasela, T.H.: Population
`pharmacodynamics of doxacumin. Clin.
`Pharmacol. Ther., 525287536, 1992.)
`
`_,
`
`
`
`PercentBlock
`
`I
`
`80
`
`F
`
`160
`Minutes
`
`‘I
`
`240
`
`,
`
`320
`
`Page 4
`
`Page 4
`
`

`

`CHAPIER 13
`
`CHAPTER 1 3
`
`VARlABlLITY
`
`205
`
`as illustrated in Fig. 13—4. Here, a drug that displays little inteipatient variability in C"m,
`tum and in maximum effect, but large variability in half—life and concentration needed to
`produce 50% maximum response, is given orally at two doses, one that achieves close to
`maximal response in all patients and one that does not. At the higher dose, observations
`made at Cum would suggest little variability in either concentration or pharmaeodynamicS,
`with perhaps a greater assignment of variability to the former, as variation in plasma con—
`centration produces relatively little change in response. At later times after this higher
`dose, substantial variability is observed in both concentration and response. In contrast, for
`
`[BI]. Clearly, ifz'ntrain-
`sage For an individual
`.‘I'OVV therapeutic Win~
`variability in pharma—
`V.
`
`From several sources.
`
`rstrated in Figure 1—5
`needed to produce a
`)l‘ response with time
`. 13—1). As illustrated
`
`of the plateau plasma
`:laily close ol‘ the drug
`d to produce a similar
`kinetics and pharma—
`16 Wide scatter in the
`
`Lily doses of this drug
`
`)harmaeodynamies to
`try with the drug and,
`isteroidal anti—inflam—
`
`lity may be different
`if from chronic aches
`
`; to assign the relative
`iade based on direct
`
`could be strongly in—
`:udc of the response,
`
`
`
`
`
`g
`g
`E
`Z
`
`,
`
`
`
`
`
`
`11
`
`
`W
`
`[j
`,
`0.3
`0.2
`0.1
`0
`Plasma Nortriptyline Concentration (mg/L)
`
`A
`
`99 —
`
`A 90 —
`
`.'
`
`.
`
`.x
`
`B
`
`.
`
`_ '
`
`.
`

`
`g 50 -
`g
`“g 10 1
`8
`5‘:
`
`M
`
`1
`
`1
`I
`0.1
`0.05
`0.01
`Plasma Nortrlptyline Concentration
`(mg/L, log scale)
`
`r
`0.5
`
`Fig. 13—2. A, The plateau plasma concentration of nortriptyline varies widely in 263 patients receiving a regimen
`of 25 mg nortriptyline orally three times daily. B, The concentrations are logenormally distributed, as seen from
`the straight line, when the percentiles of the cumulative number of patients are plotted on probit scale against
`the logarithm of the concentration. (1 mg/l, = 3.8 MM) (Redrawn and calculated from Sjoqvist, F., Borga, 0.,
`and Orme, M.L.E.: Fundamentals ofclinical pharmacology. In Drug Treatment. Edited by CS. Avery. Edinburgh,
`Churchill Livingstone, l976, pp. 1—42.)
`
`
`
`
`
`0
`
`2
`
`4
`
`6
`
`Unbound Plasma S-Warfarin
`Concentration (ug/L)
`
`240
`
`320
`
`y active S—warfarin associated with a similar
`Fig. 13—3. The unbound plateau concentration of the predominatel
`ely among a group of 38 patients receiving racemic warfaiin. (1 mg/L = 3.3
`degree of anticoagulation, varies Wid
`uM) (Adapted from Chan, E., McLachlan, A.]., Pegg, M., Mackay, AD, Cole, R. B., and Rowland, M.: Disposition
`of‘ warfarin enantiomers
`and metabolites in patients during multiple dosing. Br. ] Clin. Pharrnacol., 37563—569,
`l 994.
`
`/ .
`
`/
`
`tes
`
`
`
`Page 5
`
`Page 5
`
`

`

`VARIABIl ITY
`
`CHAPTER l 3
`
`
`
`PlasmaDrugConcentration
`
`53 m
`
`.0 M on
`
`O '__.L 01
`
`O ;A
`
`
`
`Response/(MaximumResponse)
`
`18
`
`24
`
`12
`Hours
`
`18
`
`24
`
`12
`Hours
`
`therapy. To illustrate this statement consider the frequency distributions in clearance oil
`
`
`Fig. 13—4. The interindividual variability in concentration and response varies with dose and time of observation.
`Shown are plasma concentrations (left) and responses (right) following large and small doses of a drug that displays
`little intcrpatient variability in Cmax, tum and maximum response, but large inter-patient variability in halfelife and
`concentration needed to produce 50% maximum response. High dose (top): at tum, the maximum response in all
`patients is produced with little variability in either Cum or response. Greater variability in concentration and
`response is seen at later times. Low dose (bottom): at tmm, variability in CW“. is still low, but that in response is
`now considerable.
`
`the lower dose, at tum there is still little interpatient variability in Cum, but now there is
`considerable variability in response. This dependence on dose and time in the assignment
`of variability is minimized by expressing variability not in terms of observations but rather
`in terms of the parameter values defining pharmacokinetics and pharmacodyniaxnics, that
`is, in F, kn, CL, and V for pharmacokinetics, and in maximal response, concentration to
`achieve 50% of the maximum response, and the factor defining the steepness of the con—
`centration~response relationship for pharmacodynamics (Chap. 20, Pharmacologic Re—
`sponse). Once variability in these parameters is defined, the expected variability in con
`centration and response within the patient population associated with given dosage
`regimens can be estimated. The accuracy of the models defining pharmacokinetics and
`pharmacodynamics is obviously critical to an understanding of variability in patient re—
`sponse. Where appropriate, these models should incorporate such factors as protein bind—
`ing, active metabolites, and tolerance.
`
`DESCRIBING VARIABILITY
`
`Knowing how a particular parameter varies within the patient population is important in
`
`Page 6
`
`Page 6
`
`

`

`
`CHAPl [R l 3
`VARIABILITY
`207
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`CHAPTER 13
`
`\ LargeDose
`
`18
`
`'
`
`24
`
`SmaH
`Dose
`
`l//
`
`18
`
`24
`
`and time of observation.
`scs of a drug that displays
`variability in half—life and
`maximum response in all
`ity in concentration and
`v, but that in response is
`
`u, but now there is
`e in the assignment
`arvations but rather
`
`nacodynamics, that
`:e, concentration to
`
`eepness of the con—
`Pharmacologic Re—
`i variability in con—
`with given dosage
`armacokinetics and
`
`aility in patient re—
`ors as protein bind—
`
`the three hypothetical drugs shown in Fig. 13—5. The mean, or central tendency, for all
`three drugs is the same, but the variability about the mean is very different. For Drugs A
`and B, the distribution is unimodal and normal; here the mean represents the typical value
`of clearance expected in the population. As variability about the mean is much greater for
`Drug B than for Drug A, one has much less confidence that the mean of Drug B applies
`to an individual patient. For Drug C, distribution in clearance is bimodal, signifying that
`there are two major groups within the population: those with high and low clearances.
`Obviously, in this case, the mean is one of the most unlikely values to be Jfound in this
`population.
`Generally, distributions of pharmacokinetic parameters or observations are unimodal
`rather than polymodal, and they are often skewed rather than normal, as seen, e.g., in the
`frequency distribution of plateau plasma concentrations of nortriptyline (Fig. 1372A). A
`more symmetrical distribution is often obtained with a logaiitlnnic transformation of the
`parameter; such distributions are said to be log—normal. A common method of examining
`for log—normal distribution is to plot the cumulative frequency, or percentile, on a probit
`scale against the logarithm of the variable The distribution is taken to be log—normal it the
`points lie on a straight line, As can be seen in Fig. 13—2B, this is the ease for the plateau
`plasma concentration of nortriptyline. In such cases the median, or value above and below
`which there are equal numbers, (litters from the mean. For nortriptyline, examination of
`Fig. 13—213 indicates that the median concentration is 0.05 mg/L, which is less than the
`average value of 0.069 mg/L.
`A comment on the quantitation of variability is needed here. Variance is a measure of
`the deviations of the observations about the mean; it is defined as the sum of the squares
`of these deviations. While useful to convey variability within a particular set of observations,
`variance does not allow ready comparison of variability across sets of observations of dif—
`ferent magnitude. Suppose, e.g., clearance in an individual is 50 mL/min and the mean is
`
`100 mL/min; the squared deviation is 2500 (1nL/min)2. If instead clearance had been
`
`quoted in L/min, the squared deviation would be (0.05 — 0.1)2, or 0.0025 (L/min)2. Coeffie
`
`cient of variation, which expresses variability with respect to the mean value, overcomes this
`
`problem, Specifically, it is the square root of variance (the standard deviation) normalized to
`
`the mean. In the example above, the deviation normalized to the mean is 0.5 and is independ-
`ent of the units ofclearance. Furthermore, a large coellieient oi‘variation now always signifies
`
`a high degree ot'variability. Subsequently, in the book, high and low variability refer to dis—
`
`tributions that have high and low coefficients of variation, respectively.
`
`
`
`A
`Fig. 13—5. As the frequency distributions for the
`clearance of three hypothetical drugs (A, B, C) show,
`
`
`it is as important to define variability around the mean
`and the shape of the frequency distribution curve as
`
`it is to define the mean itself.
`
`
`
`
`Frequency
`
`
`
`
`
`tion is important in
`ons in clearance of
`
`
`
`Clearance (arbitrary units)
`
`
`Page?
`
`Page 7
`
`

`

`the country.
`
`,
`,
`Noncomplionce
`Route of administration
`7', H
`7
`,
`,
`;
`’y
`'
`
`,3
`
`Food 5
`'
`I
`,
`,
`'
`_
`r
`'
`VPOllD’to'nts'
`I
`,
`,r
`,
`Time/of day an seoson
`'
`" :
`" '
`' " '
`.
`,,
`,
`
`208
`
`VARlABlLI iY
`
`CHAPTER l 3
`
`WHY PEOPLE DIFFER
`
`The reasons why people differ in their responsiveness to drugs in medicinal products are
`manifold and include, in general order of importance, genetics, disease, age, drugs given
`concomitantly, and a variety of environmental factors. Although inheritance accounts for a
`substantial part of the differences in response among individuals, much of this variability
`is largely unpredictable. increasingly, however, this source of variability, particularly that
`related to drug metabolism, is being understood and made more predictable using the tools
`of molecular biology (Chap. 14, Genetics).
`Disease can be an added source of variation in drug response. Usual dosage regimens
`may need to be modified substantially in patients with renal function impairment, hepatic
`disorders, congestive cardiac failure, thyroid disorders, gastrointestinal disorders, and other
`diseases. The modification may apply to the drug being used to treat the specific disease
`but may apply equally well to other drugs the patient is receiving. For example, to prevent
`excessive accumulation and so reduce the risk of toxicity, the dosage of the antibiotic
`gentamicin used to treat a pleural infection of a patient must be reduced if the patient also
`has compromised renal function. Similarly, hyperthyroidic patients require higher than
`usual doses of digoxin, a drug used to improve cardiac efficiency. Moreover, a modification
`in dosage may arise not only from the direct impairment of a diseased organ but also from
`secondary events that accompany the disease. Drug metabolism, e. g., may be modified in
`patients with renal disease; plasma and tissue binding of drugs may be altered in patients
`with uremia and hepatic disorders.
`Age, weight, and concomitantly administered drugs are important because they are
`sources of variability that can be taken into account. Genderdinked differences in hormonal
`balance, body composition, and activity of certain enzymes manifest themselves in differ-
`ences in both pharmacokinetics and responsiveness, but overall, the effect of gender is
`small.
`
`Table 13—1 lists examples of additional factors known to contribute to variability in drug
`response. Perhaps the most important factor is noncompliance. Noncompliancc includes
`the taking of drug at the wrong time, the omission or supplementation of prescribed dose,
`and the stopping of therapy, either because the patient begins to feel better or because of
`development of side—effects that the patient considers unacceptable. Whatever the reason,
`these problems lie in the area of patient counselling and education. Occasionally, plasma
`concentration data are used as an objective measure of noncompliance.
`Pharmaceutical formulation and the process used to manufacture a product can be
`important as both can affect the rate of release, and hence entiy, into the body (Chap. 9).
`
`Table I 3-1 . Addilionul Facials Known to Coniribuie Io Vuriubilily
`in Drug Response
`
`FACTORS OBSERVATIONS AND REMARKS
`
`'
`
`.
`
`,
`
`,
`
`,
`
`.
`,
`tQCOtion ,
`_
`'
`,
`
`f
`
`,
`
`,
`
`,
`
`,
`
`.1
`
`xi
`,
`
`.'
`
`,
`
`'
`
`,,
`
`A motor problem in clinical practice; solution lies in ootient education.
`Patient response con vary on changing the route of administration. Not only
`phormocokinetics of drug but also metabolite concentrations can chonge.
`Rote ond occosionolly extent of absorption ore effected by eating. Effects
`depend on composition of food. Severe protein restriction may reduce the
`rote of drug metabolism.
`Drug effects ore often less in smokers and workers occupationally exposed to
`pesticides; o resultolenhoncecl drug metabolism.
`Diurnol voriotions ore seen in phormocokinetics ond in drug response. These
`effects have been sufficiently important to lead to the development of 0 new
`subiect, chronophormocology.
`Dose requirements of some drugs differ between patients living in town and in
`
`Page 8
`
`Page 8
`
`

`

`
`
`
`
`
`
`CHAPTER 13
`
`VARIABILITY
`
`
`209
`
`A well—designed formulation diminishes the degree of variability in the release character—
`istics of a drug in nine. Good manufacturing practice, with careful control of the process
`variables, ensures the manufacture of a reliable product. Drugs are given enterally, topi—
`cally, parenterally, and by inhalation, Route of administration not only can affect the con—
`centration locally and systemically but also can alter the systemic concentration of metab—
`olite compared with that of drug (Chap. 21). All these factors can profoundly affect the
`response to a given dose or regimen.
`Food, particularly fat, slows gastric emptying and so decreases the rate of drug absorp
`tion. Oral bioavailability is not usually affected by food, but there are many exceptions to
`this statement. Food is a complex mixture of chemicals, each potentially capable of inter—
`acting With drugs. Recall from Chap. 9, e.g., that the oral bioavailability of tetracycline is
`reduced when taken with milk, partly because of the formation of an insoluble complex
`with calcium. Recall also that a slowing of gastric emptying may increase the oral bioavail—
`ability of a sparingly soluble drug, such as griseofulvin. Diet may also affect drug metab—
`olism. Enzyme synthesis is ultimately dependent on protein intake. When protein intake
`is severely reduced for prolonged periods, particularly because of an imbalanced diet, drug
`metabolism may be impaired. Conversely, a high protein intake may cause enzyme induc—
`tion.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`CHAPTER l3
`
`dicinal products are
`se, age, drugs given
`tance accounts for a
`
`Ch of this variability
`ity, particularly that
`table using the tools
`
`ial dosage regimens
`mpairment, hepatic
`disorders, and other
`the specific disease
`example, to prevent
`ge of the antibiotic
`id if the patient also
`'equire higher than
`over, a modification
`organ but also from
`may be modified in
`2 altered in patients
`
`t because they are
`:rences in hormonal
`1emselves in differ,
`
`effect of gender is
`
`0 variability in drug
`ompliance includes
`of prescribed dose,
`)etter or because of
`'hatever the reason,
`tceasionally, plasma
`
`= a product can be
`the body (Chap. 9).
`
`Iv
`
`itieni education.
`ministration. Not only
`nirotions con Change,
`i by eating. Effects
`then may reduce the
`
`Jpotionolly exposed to
`
`drug response. These
`development of or new
`
`its living in town and in
`
`Chronopharmacology is the study of the influence of time on drug response. Many
`endogenous substances, e.g., hormones, are known to undergo cyclic changes in concen—
`tration in plasma and tissue with time. The amplitude of the change in concentration varies
`among substances. The period of the cycle is often diurnal, approximately 24 hr, although
`there may be both shorter and longer cycles upon which the daily one is superimposed.
`The menstrual cycle and seasonal variations in the concentrations of some endogenous
`substances are examples of cycles with a long period. Drug responses may therefore change
`with time of day, day of the month, or season of the year. Particular note of this phenom—
`enon is taken in cancer chemotherapy. Many chemotherapeutic agents have very narrow
`margins of safety and are given in combination. Appropriate phasing in the timing of ad—
`ministration of each drug during the day can improve the margin of safety.
`Cigarette smoking tends to reduce clinical and toxic effects of some drugs, including
`chlordiazepoxide, diazepam, and theophylline, The drugs affected are extensively metab—
`olized by hepatic oxidation; induction of the drug~metabolizing enzymes is the likely cause.
`Many environmental pollutants exist in higher concentrations in the city than in the country;
`
`they can also stimulate synthesis of hepatic metabolic enzymes.
`
`
`identiiying the Sources of Variability
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`In practice, all the above-mentioned factors can contribute to observed variability in re—
`sponse, and care must be taken to ensure an appropriate conclusion is reached when trying
`to assign causes of variability. Consider, e.g., the data displayed in Fig. 13—6 which show
`the half—lives of phenylbutazone, a once widely used drug, in healthy subjects and in patients
`with hepatic disease (primarily cirrhosis). initially, no difference was revealed between the
`two groups, except for a greater variability in the half—life among the patients with hepatic
`disease (Fig. 13—6A). When, however, both groups were further subdivided on the basis
`of whether they received other drugs, a clearer picture emerged (Fig. 1376B). Of those
`receiving no other drugs, patients with hepatic disease handled phenylbutazone more
`slowly than did healthy subjects. Evidently, some of the other drugs received hasten phen-
`ylbutazone elimination.
`Various strategies can be employed to identify sources of variability in response. The
`Classic design is one in which as many of the variables as possible are fixed, apart from the
`one of interest. For example, to test if renal disease affects the pharmacokinetics of a drug,
`
`
`
`Page 9
`
`Page 9
`
`

`

`VARIABILlTY
`
`CHAPTER l 3
`
`Normal
`No Drugs Drugs
`
`B
`
`Hepatic
`Disease
`No Drugs Drugs
`
`Hepatic
`Disease
`
`. 197
`,- 162
`144
`ago 0
`
`
`
`PhenylbutazoneHalf-life
`
`(hf)
`
`U‘I O
`
`aspire-5;:
`1:0..0‘9'00
`
`o
`
`3:»
`
`
`
`PhenylbutazoneHalt-lite
`
`(hr)
`
`01 O
`
`
`
`I
`
`siderable overlap in the plasma concentrations among the groups. Thus, individuals from
`
`
`0.09
`
`(O01
`
`36
`
`19
`
`34
`
`61
`
`Number of Subjects
`
`Number of Subjects
`
`Fig. 13—6 A, No difference is seen between the average half—life of phenylbutazone (horizontal line) in normal
`subjects (black points) and that in patients with hepatic disease (colored points). 13, After separating those who
`take other drugs (light points) from those who do not (dark points), the prolonged elimination of phenylbutazone
`in patients with hepatic disease becomes evident. In both groups the half—life tends to be shorter when other
`drugs are taken concurrently. (Redrawn from Levi, A.]., Sherlock, S., and Walker, D.: Phenylbutazone and iso—
`niazid metabolism in patients with liver disease in relation to previous drug therapy. Lancet, 1127571279, 1968.)
`
`all other factors such as age, gender, other drugs, and diet should be held constant. The
`ideal would be a longitudinal cross—over design in which each patient acts as his or her own
`control. This design is often not possible, however. The patient with renal disease is gen-
`erally not available for study prior to the disease, and renal disease is generally irreversible.
`The penalty for deviating from such a design is greater variability with loss of efficiency,
`such that many more patients are needed to allow a firm conclusion to be made about the
`contribution of a factor to variability. The benefit of loosening the design, however, is that
`many patients who might otherwise be excluded can be a part of the study. In this category,
`e.g, are elderly patients suffering from several diseases and requiring many drugs, including
`the one of interest. Care must still be taken, however, to ensure that a sufficient number
`of patients are included with each of the attributes or conditions of interest.
`
`DEFINING THE DOSE—RESPONSE RELATIONSHIP
`
`Variability has an important bearing on the estimation of doseeresponse relationships in
`clinical trials. A common procedure is to divide patients into several groups, each group
`receiving a different dose of drug such as 5, 10, or 20 mg. An attempt to establish a dose;
`response relationship is then made on the mean data for each group, using variability within
`groups to test for levels of statistical significance. A problem arises when much of the
`variability between dose and response resides in pharmacolcinetics such that there is con—
`
`Page 10
`
`Page 10
`
`

`

`
`
`
`
`
`
`
`c; tAP’IER 13
`VARIABiLITY
`21 1
`
`
`
`the high- and low—dose groups can have the same plasma concentration (and response),
`namely, those in the low—dose group with a low Clearance and those in the high—dose group
`
`
`With a high clearance of the drug. The overall effect, by increasing variability within each
`
`
`group, is to weaken the ability to detect a dose—response relationship.
`
`
`7 One solution is to increase the number of subjects in each group to reduce the uncer—
`
`
`tainty of estimating the mean response at each dose level. Here, the problem is often one
`
`
`of not knowing in advance how many subjects would be needed in the trial, as well as the
`
`
`added expense of an increased number of subjects. Another solution is to expose each
`
`
`patient to several dose levels of the drug. This last solution has the distinct advantage of
`
`
`not only increasing the chances of establishing a dose—response relationship, but also of
`
`
`providing an estimate of interpatient variability in the relationship. Unfortunately, in prac—
`
`
`tice, this design is not always possible, especially for drugs for which the full effect only
`
`
`occurs after several months or longer into drug administration. A third solution is the
`concentration—controlled clinical trial. In this approach, the pharmacokinetics of the drug
`is first evaluated in the patient cohort and then, based on this information, doses are
`adjusted so that the plasma concentration in each patient lies in one of several tightly
`defined bands. This more elaborate, and sometimes more expensive, design enables much
`
`
`clearer statements to be made about the concentrationresponse relationship and about
`
`
`interpatient variability in pharmacokineties. However, it may have limited utility for dose
`
`
`recommendations, if a poor correlation is found between plasma drug concentration and
`
`
`response. Many other designs, varying in complexity, each with advantages and disadvan—
`tages, can be envisaged. In all cases, variability is a central issue.
`
`
`iiiNETIC MANIFESTATIONS
`
`
`Considerable variability in enzymatic activity and, to a lesser extent, in plasma and tissue
`binding exists even among healthy individuals. How such variability manifests itself, in
`
`
`pharmacokinetic parameters and in such measurements as plateau plasma concentration,
`
`
`depends on the hepatic extraction ratio and route of administration of the drug. For ex—
`
`
`ample, the large interindividual variability in half—life of theophylline (Fig. 13—7) can be
`
`
`explained primarily by variations in hepatic enzyme activity, probably associated with var—
`
`
`iations in the amounts of the enzymes responsible for metabolism of this compound. This
`conclusion is based on theophylline being predominantly metabolized in the liver, having
`
`
`a low extraction ratio, and being only moderately bound to plasma and tissue components.
`
`
`In contrast, such a high degree of variability in enzymatic activity is expected to be masked
`
`
`in the clearance of a drug having a high hepatic extraction ratio, because clearance tends
`
`
`to be perfusion rate—limited and hepatic blood flow is relatively constant among healthy
`
`
`individuals. Moreover, unless plasma and tissue binding are highly variable, volume of
`
`
`distribution, and hence disposition kinetics, of such a drug are much the same for all healthy
`
`
`individuals. This is so for propranolol (Fig. 13—8) a drug of high hepatic clearance.
`
`
`As described in Chap. 11., when considering induction and inhibition, changes in hepatic
`enzyme activity result in variations in oral bioavailability for a drug with a high hepatic
`
`
`extraction ratio. Accordingly, with subsequent disposition being controlled by hepatic per—
`fusion, a series of similarly shaped plasma drug concentration-time profiles, but reaching
`
`
`different peak concentrations, should be seen among individuals with varying enzyme ac—
`tivity receiving

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