throbber
third edition
`
`Concepts and Applications
`
`
`
`MALCOLM ROWLAND
`
`THOMAS N. TOZER
`
`Apotex v. Novartis
`lPR2017-00854
`
`NOVARTIS 2105
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2105
`
`1
`
`

`

`
`
`Clinical Pharmacokinetics
`
`Concepts and Applications
`
`third edition
`
`MALCOL/Vl ROWLAND, Ph . D.
`
`Department of Pharmacy
`
`University of Manchester
`
`Manchester, England
`
`THOMAS N. TOZER, PhD.
`
`School of Pharmacy
`
`University of California
`
`San Francisco, California
`
`A Lea & Febiger Book
`
`fib LIPPINCOTT WILLIAMS e \X/lLKlNS
`' A Wolten Kluwer Company
`Philadelphia . Baltimore . New York - London
`Buenos Alres . Hong Kong . Sydney . Tokyo
`
`2
`
`

`

`AV;V
`
`V '
`
`-
`‘
`do
`.
`Executne Editor. Donna Bala
`Developmental Editors: Frances Klass. Lisa Stead
`qer: Laurie Forsyth
`Production Mona
`Project Editor.- Robert D. Magee
`
`Copyright © 1995
`Lippincott Williams & \Vllkins
`530 Walnut Street
`Philadelphia, Pennsylvania 19106-3621 USA
`
`
`
`pyright. No part of this book may be reproduced in any
`All rights reserved. This book is protected by CO or utilized by any information storage and retrieval system
`form or by any means, including photocopying,
`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
`
`Rowland, Malcolm.
`Clinical Pharmacokinetics : concepts and applications / Malcolm
`Rowland, Thomas N. Tozer. — 3rd ed.
`p.
`cm.
`“A Lea & Febiger Book."
`Includes bibliographical references and index.
`ISBN 0-683-07404-0
`1. Pharmacokinetics
`11. Title.
`[DNLM-. 1. Pharmacokinetics
`RM301.5.R68
`1994
`615.7—dc20
`DNLM/DLC
`
`~
`
`-
`2 Chemoth
`
`.
`
`e a
`r W
`
`1- Tozer. Thomas N.
`
`2
`
`'
`
`"Drug Therapy'
`
`QV 38 R883c1994]
`
`for Library of Congress
`
`94-26505
`CIP
`
`7799 Publishers have m d
`adve
`a e every effort to trace the copyright holdersfor borrow d
`l f b
`b
`.
`e materia . I t ey We m—
`tb/
`l
`I
`.
`over oo/eed any hey w be 0 eased 0 make be necessary arrangements (1
`e
`75
`rten
`
`i
`
`04
`
`78910
`
`3
`
`

`

`
`
`CONTENTS
`
`............................................................................ xi
`Definitions of Symbols
`1 . Why Clinical Pharmacokinetics?
`............................................................... 1
`
`SECTION I. ABSORPTION AND DISPOSITION KINETICS
`2. Basic Considerations
`......................................................................... 1 1
`3.
`Intravenous Dose ............................................................................... 18
`4. Extravascular Dose ............................................................................ 34
`
`SECTION II.
`THERAPEUTIC REGIMENS
`5. Therapeutic Response and Toxicity ........................................................ 53
`(D. Constant-Rate Regimens ...................................................................... ()0
`7. Multiple-Dose Regimens ...................................................................... 83
`
`PNYSIOLOGIC CONCEPTS AND KINETICS
`SECTION III.
`8. Movement Through Membranes
`......................................................... 109
`9. Absorption ..................................................................................... 119
`10. Distribution
`.................................................................................... 137
`1 1. Elimination ..................................................................................... 156
`12.
`Integration With Kinetics
`................................................................... 184
`
`INDIVIDUALIZATION
`SECTION IV.
`13. Variability ...................................................................................... 203
`14. Genetics
`....................................................................................... 220
`15. Age and Weight
`............................................................................ 230
`16. Disease ......................................................................................... 248
`17.
`Interacting Drugs
`............................................................................. 267
`18. Concentration Monitoring ................................................................. 290
`
`SELECTED TOPICS
`SECTION V.
`19. Distribution Kinetics .......................................................................... 313
`20. Pharmacologic Response .................................................................. 340
`21 . Metabolite Kinetics
`.......................................................................... 367
`22. Dose and Time Dependencies
`........................................................... 394
`23. Turnover Concepts
`.......................................................................... 424
`24. Dialysis
`......................................................................................... 443
`
`SELECTED READING .................................................................................... 463
`
`APPENDIX I. ADDITIONAL CONCEPTS AND DERIVATIONS
`
`A. Assessment of AUC ......................................................................... 469
`B. Estimation of Elimination Hall-lite From Urine Data .................................. 473
`
`ix
`
`4
`
`

`

`x
`
`CONTENTS
`
`......i...... 478
`C. Estimation of Absorption Kinetics From Plasma Concentration Data
`D. Mean Residence Time ...................................................................... 485
`E. Amount of Drug in Body on Accumulation to Plateau ............................... 490
`F. Distribution of Drugs Extensively Bound to Plasma Proteins
`........................ 494
`G. Blood to Plasma Concentration Ratio ................................................... 502
`H. Estimation of Creatinine Clearance Under Nonsteady-State Conditions ....... 504
`
`APPENDIX ll. ANSWERS T0 PROBlEMS ......................................................... 507
`
`INDEX ......................................................................................................... 58b
`
`5
`
`

`

`as...
`
`..
`
`f
`
`‘(
`
`WHY CLINICAL PHARMACOKINETICS?
`
`Those patients who suffer from chronic ailments such as diabetes and epilepsy may have
`to take drugs every day for the rest of their lives. At the other extreme are those who take
`a single dose of a drug to relieve an occasional headache. The duration of drug therapy is
`usually between these extremes. The manner in which a drug is taken is called a dosage
`regimen. Both the duration of drug therapy and the dosage regimen depend on the ther-
`apeutic objectives, which may be either the cure, the mitigation, or the prevention of
`disease. Because all drugs exhibit undesirable effects, such as drowsiness, dryness of the
`mouth, gastrointestinal irritation, nausea, and hypotension, successful drug therapy is
`achieved by optimally balancing the desirable and the undesirable effects. To achieve op—
`timal therapy, the appropriate “drug of choice” must be selected. This decision implies an
`accurate diagnosis of the disease, a knowledge of the clinical state of the patient, and a
`sound understanding of the pharmacotherapeutic management of the disease. Then the
`questions How much? How often? and How long? must be answered. The question How
`much? recognizes that the magnitudes of the therapeutic and toxic responses are functions
`of the dose given. The question How often? recognizes the importance of time, in that the
`magnitude of the effect eventually declines with time following a single dose of drug. The
`question How long? recognizes that a cost (in terms of side effects, toxicity, economics) is
`incurred with continuous drug administration. In practice, these questions cannot be di-
`vorced from one another. For example, the convenience of giving a larger dose less fre-
`quently may be more than offset by an increased incidence of toxicity.
`In the past, the answers to many important therapeutic questions were obtained by trial
`and error. The dose, interval between doses, and route of administration were selected,
`and the patient’s progress fOIIOWed. The desired effect and any signs of toxicity were care-
`fully noted, and if necessary, the dosage regimen was adjusted empirically until an accept—
`able balance between the desired effect and toxicity was achieved. Eventually, after con—
`siderable experimentation on a large number of patients, reasonable dosage regimens were
`established (Table 1—1), but not without some regimens producing excessive toxicity or
`proving ineffective. Moreover, the above empirical approach left many questions unan-
`swered. Why, for example, does tetracycline have to be given every 6 to 8 hours to be
`effective, while digoxin can be given once daily? Why must oxytocin be infused intrave-
`nously? Why is morphine more effective given intramuscularly than when given orally?
`Furthermore, this empirical approach contributes little, if anything, toward establishing a
`safe, effective dosage regimen of another drug. That is, our basic understanding of drugs
`has not been increased.
`To overcome some of the limitations of the empirical approach and to answer some of
`the questions raised, it is necessary to delve further into the events that follow drug ad-
`ministration. In vitro and in vivo studies show that the magnitude of the response is a
`function of the concentration of drug in the fluid bathing the site(s) of action. From these
`observations the suggestion might be made that the therapeutic objective can be achieved
`by maintaining an adequate concentration of drug at the site(s) of action for the duration
`
`6
`
`

`

`WHY CtINICAL PHARMACOKINETICS
`
`2
`
`CHA
`
`PTER I
`
`1e
`
`1
`
`'
`
`-
`
`'
`
`'
`
`ration to the site 0 .
`
`_
`
`.
`
`nnu aneo .
`
`. .
`
`.
`
`'
`
`'
`
`'
`
`'
`
`m the bod .
`
`at its site of action. Indeed, most drugs are
`.
`_
`'
`.
`-
`l
`laced
`of therapy. However. rarely is atldl'tlfr-En on the heart, at the neuromuscular lunchon, 0r
`‘ t thev act in
`,
`'
`'
`'
`'
`glven orally. and ye
`J
`l
`l
`A dru must therefore move from the Slte Ofladfiltnslfltes includjng thOSefGale“.
`3881:ere- uslv filowever
`the dmg distributes to all 0t ler lS
`Organs,
`notably the liver and the kidneys, that elunmate 1t frod se of drylg is administered or 11
`Figure 1—1 illustrates the events occurring after a 0
`a y_
`,
`.
`.
`.
`bod exceeds its rate of elimination; the Con.
`ElirhfiznZtof’lllfilgiglohndilhlcllyoillgrtl5:131;as;oftenStifficieéitlylligijllto(5111?:thedFsired
`therapeutic effects and sometimes even to produce tonglty. thfgolicefitrafio:(if: drug
`elimination exceeds the rate of its absorption, and therea ter,
`d _
`. t
`d
`.I’Ug In
`both blood and tissues declines and the efffecltfsl Sllb}::i:-m:00: deIgHZESI‘or-Stliisnogfifiggl
`32::eigfellllrllllzliggnefiitn:1; ofthe lantics ofthese processes: that IS, phamcpkmetics.
`'
`dednot on
`o t emec
`.
`.
`-
`,
`
`The application of pharmacokinetic principles to the therapeutic management 0 Patlents
`is clinical phamwcokinetics.
`
`Table 1-1 . Empiricully Derived Usual Adult Dose 0 ll. imons of Some
`Roprosonlullvo Drugs Before "l0 Inmduciion of C inica Pharmacokinolics-
`DRUG
`INDICATED USE
`ROUTE
`DOSAGE REGIMEN
`Tetracycline
`Treatment of infections
`Oral
`250 mg every 6-8 hr
`Digoxin
`Amelioration of congestive
`Oral
`l .5—2 mg initially over 24
`cardiac failure
`hr, thereafter 025-05
`mg once a day
`Intravenous
`induction and maintenance
`0.2-4 milliunits/min by
`of labor
`infusion
`Intramuscular
`Relief of severe pain
`lO mg when needed
`OFGl
`Not recommended because
`0 reduced effectiveness
`”Taken from American Medical Association: Drug Evaluations. 2nd Ed, Publishers Science Group, Acton MA 1973
`
`Oxyiocin
`Morphine sulfate
`
`6
`
`5
`
`Fig. 1—1. Plasma concentration of
`theophylline in a subject following an
`oral dose of a GOO-mg controlled-re-
`lease formulation. Before the peak is
`reached, the rate of absorption ex-
`ceeds that of elimination. At the
`a) A
`peak, the two rates are equal; there- E S,
`after, the rate ofelimination exceeds 3 E
`that of absorption. (Redrawn from
`8" E
`Sauter, R., Steinijans, V.W., Diletti, 3 log
`E., Bohm, A., and Schulz, H.U.:
`'— g
`Presentation ofresults in bioequival-
`E 5
`ence studies. Int. ], Clin. Pharmacol.
`g g 2
`Ther. Toxicol., 305740, 1992.)
`CT- 8
`
`4
`
`3
`
`l
`
`0
`
`0
`
`12
`
`24
`Hours
`
`36
`
`43
`
`7
`
`

`

`CHAPTER 1
`
`WHY CLINICAL PHARMACOKINETICS?
`
`3
`
`The events following drug administration can be divided into two phases, a pharmaco-
`kinetlc phase, in which the adjustable elements of dose, dosage form, frequency, and route
`of administration are related to drug level—time relationships in the body, and a pharma-
`codynamic phase, in which the concentration of drug at the site(s) of action is related to
`the magnitude of the effect(s) produced (Fig. 1—2). Once both of these phases have been
`defined, a dosage regimen can be designed to achieve the therapeutic objective. Despite
`the greater amount of information required with this approach, it has several advantages
`over the empirical approach. First, and most obvious, distinction can be made between
`pharmacokinetic and pharmacodynamic causes of an unusual drug response. Second, the
`basic concepts of pharmacokinetics are common to all drugs; information gained about the
`pharmacokinetics of one drug can help in anticipating the pharmacokinetics of another.
`Third, understanding the pharmacokinetics of a drug often explains the manner of its use;
`occasionally such an understanding has saved a drug that otherwise may have been dis-
`carded or has suggested a more appropriate dosage regimen. Lastly, knowing the phar—
`macokinetics of a drug aids the clinician in anticipating the optimal dosage regimen for an
`individual patient and in predicting what may happen when a dosage regimen is changed.
`A basic tenet of clinical pharmacokinetics is that the magnitudes of both the desired
`response and toxicity are functions of the drug concentration at the site(s) of action. Ac-
`cordingly, therapeutic failure results when either the concentration is too low, giving in—
`effective therapy, or is too high, producing unacceptable toxicity. Between these limits of
`concentration lies a region associated with therapeutic success; this region may be regarded
`as a “therapeutic window.” Rarely can the concentration of the drug at the site of action
`be measured directly; instead the concentration is measured at an alternative and more
`accessible site, the plasma.
`Based on the foregoing considerations, an optimal dosage regimen might be defined as
`one that maintains the plasma concentration of a drug within the therapeutic window. For
`many drugs, this therapeutic objective is met by giving an initial dose to achieve a plasma
`concentration within the therapeutic window and then maintaining this concentration by
`replacing the amount of drug lost with time. One popular and convenient means of main—
`tenance is to give a dose at discrete time intervals. Figure 1—3 illustrates the basic features
`associated with this approach by depicting the concentrations that follow the administration
`of two regimens, A and B. The dosing interval is the same but the dose given in regimen
`B is twice that given in regimen A. Because some drug always remains in the body from
`preceding doses, accumulation occurs until, within a dosing interval, the amount lost equals
`the dose given; a characteristic saw-toothed plateau is then achieved. With regimen A,
`
`Pharmacokinetics
`
`Pharmacodynamics
`
`Dosage
`
`Plasma
`Concen-
`tration
`
`Regimen
`
`Fig. 1—2. An approach to the design of a dosage regimen. The phammcokinetics and the phamlacodmamics of
`the drug are first defined. Then, either the plasma drug concentration-time data or the effects produced are used
`via phannacoldnetics as a feedback (dashed hues) to modify the dosage regimen to achieve optimal therapy.
`
`8
`
`

`

`4
`
`WHY CLINICAL PHARMACOKINET
`
`ICSZ
`
`CHAR“)
`
`I
`
`- nt to produCe a th J
`.
`ulatlon was suffime
`.
`Cr.
`several doses had to be given before drug accum
`then, the drug mlght have been
`ed before
`-
`apeutic concentrationaHadltherZIIZZ’IIEJSSSeZtZIgmaturely. AltematJvely, larger doses might
`thought ineffective an per laps
`'
`onse would haVe b
`.
`h a therapeutlc resp
`.
`.
`.
`66“
`if
`e. “ re men BI Althoug
`‘
`tinued admlnlstratlonW
`Ezhfievlefiefaillyegromitly, gxicitv would have ensued W'ltll con
`hen
`.
`.
`/ a
`the concentration exceeded the upper 'hmlligfatld:$l:rped during World War II to substi-
`The synthetic antimalarial agent, qumac
`,
`.
`-
`was either ineffecti
`I
`’
`‘
`'
`z
`example. Qulnacrme
`Ve
`.
`th
`relatlvel
`scarce quinine, IS in
`.
`.
`h n a
`_
`tute for
`?
`al y or eventually produced unacceptable tox101ty w e
`dOSIng rate
`acutely against m aria
`ned. Only after its pharmacokmetics
`‘
`'
`as maintai
`.
`.
`'
`sufficrently high to be effective acutely W
`Quinacl‘ine is ehmmated slowly and
`'
`full
`.
`h d b
`d fined was this drug used success
`.
`'
`.y
`adcumzelgteseextensively with repeated daily admimstratlon. The answer was to give large
`doses over the first few days to rapidly achieve therapeutic success, followed by small daily
`doses to maintain the plasma concentration within the ther-aIEEUtfi$132:- eutic
`. d
`The plateau situation in Fig. 1—3 shows that both the w1d . 0
`e
`d P
`d :th fpw
`and the speed of drug elimination govern the size of the maintenance l.Os'e and e. e-
`quency of administration. When the window is narrow and the drug IS e 1m11nate
`rapidly,
`small doses must be given often to achieve therapeutic success. .Both cyc osporine and
`digoxin have a narrow therapeutic window, but because cyclosporine lS eliminated much
`more rapidly than digoxin, it has to be given more frequently. OxytoCIn IS an extreme
`example; it also has a narrow therapeutic window but is eliminated mthln mlnutes. The
`only means of adequately ensuring a therapeutic concentration of oxytocm therefore is to
`infuse it at a precise and constant rate directly into the blood. This degree of control is not
`possible with other modes of administration. Besides, had oxytocin been given orally, this
`polypeptide hormone would have been destroyed by the proteolytic enzymes in the gas-
`trointestinal fluids. Morphine, given orally, is also destroyed substantially before entering
`the general circulation, but for a reason different from that of oxytocin. Morphine is ex-
`tensively metabolized on passage through the liver, an organ lying between the gastroin-
`testinal tract and the general circulation.
`Awareness of the benefits of understanding pharmacokinetics and concentration—r6-
`sponse relationships has led in recent years to the extensive application of such information
`by the pharmaceutical industry to drug design, selection, and development. For example.
`
`concentration is ultimately too high.
`
`Fig. 1-3. When a drug is given in
`a fixed dose and at fixed time inter-
`vals (denoted by the arrows), it ac-
`cumulates within the body until a
`plateau is reached. With regimen A
`therapeutic success is achieved al:
`though not initially. With regimen B
`the therapeutic objective is achieved
`more quickly, but the plasma dmg
`
`9
`
`

`

`CHAPTER I
`
`WHY CLINICAL PHARMACOKINETICS?
`
`5
`
`titative framework improves the chances of selecting not only the most promising
`compounds but also the correct range of safe doses to first test in humans. Incorporation
`of a pharmacokinetic element with these early Phase I studies, usually in healthy subjects,
`together with assessment of any side effects produced, helps to define candidate dosage
`forms and regimens for evaluation in Phase II studies conducted in a small number of
`patients. These Phase II studies are aimed at defining the most likely safe and efficacious
`dosage regimens for use in the subsequent larger Phase III clinical trials, often involving
`many thousands of patients. Ultimately, some compounds prove to be of sufficient benefit
`and safety to be approved for a particular clinical indication by drug regulatory authorities.
`Even then the drug undergoes virtually continuous postmarketing surveillance to further
`refine its pharmacotherapeutic profile. This sequence of events in drug development and
`evaluation is depicted schematically in Fig. 1—4.
`Figure 1—5 illustrates an important problem identified during drug development and
`therapy, variability. There is a wide range of daily dose requirements of the oral antico-
`
`PRECLINICAL
`TESTING
`
`Toxicity
`
`
`CLINICAL (HUMAN) TESTING
`Dose (Conc)
`Population PK/PD
`Post-
`Response Trials
`Characteristics in
`Large Efficacy Trials
`.
`4:9 PK-guided fl [LIII
`<2 Market“
`In vitro PK/PD
`Dose escalation
`Efficacy
`:> Surveillance
`Animal PK/PD
`Safety
`Dosage
`Q37 Assessment
`Selection
`Patient Variables
`
`IZZ>
`PK/PD in Special
`POpulatlons
`
`Animal Testing
`
`I
`
`Fig. 1—4. The development and subsequent marketing of a drug. The prehuman data helps to identify promising
`compounds and to suggest useful doses for testing in humans. Phases 1, II, and III of human assessment generally
`correspond to the first administration to humans, early evaluation in selected patients, and the larger trials,
`respectively. Pharmacokinetic (PK) and pharmacodynamic (PD) data gathered during all phases of drug devel-
`opment help to efficiently define safe and effective dosage regimens for optimal individual use. Postmarketing
`surveillance helps to refine the PK/PD information.
`
`Fig. 1—5. The daily dose ofwarfarin required to produce similar
`prothrombin times in 200 adult patients varies widely. (1 mg/L =
`3.3 pM). (Redrawn from Koch-Weser, ].: The serum level ap-
`proach to individualization of drug dosage. Eur. ]. Clin. Pharma-
`co]. 9:1—8, 1975.)
`
`
`
`Qééwémmmwmm
`VT??? Twweg
`NC‘OV‘LD nobdaéA
`
`Daily Dose (mg)
`
`10
`
`m 25
`g
`1g 20
`E
`§ 15
`
`O “
`
`5 10
`
`5
`
`E e
`
`a.)
`m
`
`10
`
`

`

`5
`
`WW am am “Vii/Hf]? 57632
`
`ME? 1
`
`agulant warfarin needed to roducc a similar prothromhin time/an, index of hlood mag.
`ulabilitv). Sources of variability in dmg resptmsc include the patient 3 age, Wight degree
`of obesity. tvpe and degree of severity of the disease. the patient 3 genetic rlrli
`eup, 0th“
`drugs concurrently administered, and environmental factms. The resnlt 13 t at a standaJ-d
`dosage regimen of a drug may prove therapeutic in some patients, Ineffective in others,
`and toxic in still others. The need to adjust the dosage regimen of a drug for an indmdUa]
`patient is evident; this need is clearly greatest for drugs that have a narrow Ithe‘W’GUtic
`window, that exhibit a steep concentratitm—resymse curve, and that are critical to drug
`therapy. Examples are digoxin, used to treat some cardiac disorders; W011! used to
`prevent epileptic convulsions; theophylline, used to diminish chronic airway resistance in
`asthmatics; and cyclosporine, an immunosuppressant used In organ transplantation With
`these drugs, and with many others, variability in pharmacoldnetlcs Is a major source of total
`variabilityindru re
`use.
`'
`..
`It is becoming in:ll')(:)asingly common to gain as much information on vanablllty as P05-
`sible during drug development by gathering, albeit limited, individual plasma concentratio“
`and response data in a large population of patients during Phase III clinical trials. Attempts
`are then made to account for this variability in terms of such patient characteristics as age
`and weight. These population phannacoldnetidphannacodynamic studies form a basis for
`dosage regimen recommendations in clinical practice.
`Coadministration of several drugs to a patient, prevalent in clinical practice, can pose
`problems. Although the response produced by each drug alone may be predictable, that
`produced by the combination may be less certain and ocmsionally unpredictable. Ketc—
`conazole, for example, devoid of immunosuppressant activitv, potentiates the effect of
`cyclosporine. Possible causes of this kind of effect are many. In this instance, as in many
`others, the interaction involves a change in pharmacoldnetics. Some drugs stimulate drug-
`
`Fig. 1—6. Although the average plateau plasma
`concentration of phenytoin tends to increase with
`the dosing rate, there is considerable variation in
`the individual Values. (One mg/L = 3.97 pM.) (Re-
`drawn from Lund. L: Eflects of phenytoin in pa-
`tients with epilepsy in relation to its concentration
`in plasma. In Biological Eflects of Drugs in Relation
`to Their Plasma Concentration. Edited by D.S, Da-
`vies and B.;\’.C. Prichard. Macmillan, London and
`Basingstoke, 1973, pp. 227—238.)
`
`11
`
`50
`
`‘5
`
`(mg/L)
`
`
`PlasmaPhenytoinConcentration
`
`B8
`
`_s 0
`
`11
`
`

`

`WHY CLINICAL PHARMACOKINETICS?
`
`7
`
`be coadministered.
`Figure]: 10—6 lllufitrates a Situation in which monitoring of the drug concentration may be
`bene C1
`' vert e narrow range 0f the daily dose of the antiepileptic drug phenytoin, the
`plateau plasma drug concentration varies markedly within the patient population. Yet the
`therapeuth WlndOW of phenytoin is narrow, 7 to 20 mg/L; beyond 20 mg/L, the frequency
`and the. degree 0f tOXiCity increase progressively with concentration. Here again, pharma-
`cokinetlcs IS the major source of variability. A pragmatic approach to this problem would
`be to adjust the dosage until the desired objective is achieved. Control on a dosage basis
`alone, however, has proved difficult. Control is achieved more readily and accurately when
`plasma drug concentration data and the pharmacokinetics of the drug are known.
`Drug selection and therapy have traditionally been based solely on observations of the
`effects produced. In this chapter, the application of pharmacokinetic principles to decision
`making in drug therapy has been illustrated. Both approaches are needed to achieve optimal
`drug therapy. This book emphasizes the pharmacokinetic approach. It begins with a con-
`sideration of kinetic concepts basic to pharmacokinetics and ends with a section containing
`selected topics.
`
`12
`
`12
`
`

`

`
`
`THERAPEUTIC RESPONSE AND TOXICITY
`
`OBJECTIVES
`
`The reader will be able to:
`
`I . Explain why effect (desired or toxic) of a drug is often better correlated with plasma con'
`centration than with dose.
`
`2. Define the terms: graded response, all-or-none response, therapeutic concentration range,
`utility curve, and tolerance.
`3. List the range of plasma concentrations associated with therapy for any of the drugs given
`in Table 5—2.
`
`4. Discuss briefly situations in which poor plasma drug concentration—response relationships
`are likely to occur.
`5. Explain briefly why modality of administration of a given daily dose can affect therapeutic
`outcome.
`
`The rational design of safe and efficacious dosage regimens is now examined. In this section,
`fundamental aspects of dosage regimens are covered primarily from the point of view of
`treating a patient population with a given disease. It is realized, of course, that individuals
`vary in their responses to drugs, and subsequently, in Section Four, focus is turned toward
`the establishment of dosage regimens in individual patients.
`A therapeutic dosage regimen is basically derived from the kinds of information shown
`in Table 5—1. One consideration includes those factors that relate to both efficacy and
`safety of the drug, that is, its pharmacodynamics and toxicology. Another consideration is
`how the body acts on the drug and its dosage form, the essence of pharmacokinetics. A
`third consideration is that of the clinical state of the patient and his or her total therapeutic
`regimen. A fourth category includes all other factors such as genetic differences, tolerance,
`and drug interactions. All of these determinants are, of course, interrelated and interde—
`pendent.
`Dosage regimens are designed to produce a therapeutic objective. This objective may
`be achieved by various modalities of drug administration, extending from a single occasional
`dose to continuous and constant input. An example of the former is the use of aspirin to
`treat an occasional headache; the continuous iv. infusion of heparin to maintain a desired
`degree of anticoagulation is an example of the latter. More commonly, drugs are admin-
`istered repeatedly in discrete doses. The frequency and duration vary with the condition
`being treated. Some drugs are administered relatively infrequently, producing large fluc—
`tuations in the plasma concentration. Reasons for this approach include the development
`of tolerance to the drug and the need to produce high concentrations for short periods of
`time, as occurs in some antibiotic and anticancer chemotherapies. In other situations, main—
`
`53
`
`13
`
`13
`
`

`

`54
`
`THERAPEUTIC RESPONSE AND
`
`IOXILH Y
`
`C H
`
`“Pitts
`
`Pts are
`
`eded. In all cases, attem
`'
`.
`‘ tion of drug ‘5 ne
`‘neffectlve thera
`.
`' el
`(30115“:Int concentra
`23:32:: Omfihirriliivungesirable and toxic effects and prevent l
`py
`lasma concentr
`,
`‘
`'
`Evidence exists that response is often better correlated With P
`at1011 than
`.
`-
`to be most appropriate to aPpl
`with dose administered. Accordingly, 't would seerrrthens. Thus, given pharmacokineli phat
`macoldnetic principles to the design ofdos:g:fi1:nglor amount of drug in the body fol]
`.
`_
`1 d e, the lasma concen
`$30525;:(lllllirie(:m be egtimated. Ultimately, however, thce; vaél‘tiie1:);:ngfglfi reglmen
`0 nehcs fa.
`must be assessed by the therapeutic and toxic responses pro uc
`.
`‘
`cilitates the achievement of an approprlate dosage reglmen and se
`useful
`mEans
`of evaluatin existin dosa e regimens.
`.
`.
`.
`In this chapter gariousg elements of the concentration—response relationship are ex-
`plored. Principles for attaining and maintaining a therapeutic level 0f drug in the body are
`discussed in the subsequent two chapters of this SCCUOH-
`
`(Min
`.
`
`g
`
`.
`rves as a
`
`RESPONSE AND CONCENTRATION
`
`Response may be as vague as a general feeling of improvement or as prec15e as a lowering
`of the diastolic blood pressure by 30 mm Hg.
`.
`.
`Information relating concentration to response is obtained at three levels: m mtro ex-
`periments, animal studies, and investigations in human volunteers and patients. The last
`level is the most relevant to human drug therapy but, unfortunately, only limited infor-
`mation is often obtainable here about the nature of the drug—receptor interaction. In vitro
`experiments, which include studies of the action of drugs on enzymes, receptors, micro-
`organisms, and isolated tissues and organs, serve this purpose best. However, in isolating
`the variables, many of the complex interrelationships that exist in vivo are destroyed. An-
`imal studies bridge much of the gap between in vitro experimentation and human inveS-
`tigation. Studies in animals introduce both the variable time, with all that it connotes, and
`
`Table 5-1 . Dolomincnh of a Dos-go Regimen
`
`ACTTVTWOXTCIW
`PHARMACOKINET‘CE
`Therapeutic window
`Absorption
`glde ifeds
`Distribution
`OXICI
`Metabolism
`Concentration—response relationships
`Excretion
`
`
`
`CllNlCAl FACTORS
`
`MANAGEMENT
`STATE OF PATIENT
`OF THERAPY
`
`A e, we,- h,
`. W
`anditiongbein treated
`MUlhple~drug therapy
`R
`t
`t d '
`islration
`E .
`t
`t
`Convenience ot regimen
`oueo 0 min
`hQ d'
`XlS ence a at er
`Isease states
`Compliance of patient
`golsage Loam
`d
`ce
`epen en
`oeranc
`PhcrmOCogenetics-idiosyncroSY
`DrUg interactions
`
`14
`
`14
`
`

`

`CHAPTER 5
`
`THERAPEUTIC RESPONSE AND TOXICITY
`
`55
`
`the elements of absorption and disposition as well as the feedback control systems that
`operate to maintain homeostasis. Animal studies are often most useful for evaluating the
`pliannacologic spectrum of activity of a potential therapeutic agent and for determining
`aspects 0f its tOXiCity profile. Irrespective of the level of information, however, the conclu-
`sion is the same: A relationship, although sometimes complex, exists between the concen-
`tration of active agent at the site of measurement and the response.
`The majority of drugs used clinically act reversibly in that the effect is reversed upon
`reducing concentration at the site of action. Many responses produced are graded, so called
`because the magnitude of the response can be scaled or graded. An example of a graded
`response, shown in Fig. 5—1, is the improvement of pulmonary function produced by the
`bronchodilator terbutaline, after its s.c. administration. The intensity of the response varies
`with the drug concentration in plasma. Many other pharmacologic and toxic responses do
`not occur on a continuous basis; these are known as quantal or all-or-none responses. An
`obvious but extreme example is death. Another is the suppression of an arrhythmia. The
`arrhythmia either is or is not suppressed. Sometimes, a limit is set on a graded response
`below which an effect is said not to occur clinically. For example, a potentially toxic effect
`of antihypertensive therapy is an excessive lowering of blood pressure. The lowering of .
`blood pressure produced by the antihypertensive agents is a graded response, but hypo—
`tensive toxicity is said to occur only if the blood pressure falls to too low a value. Here the
`clinical endpoint is all-or-none, but the pharmacologic response is graded.
`Returning to terbutaline, Fig. 5~1 is a plot of the fo

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