throbber
Concepts and Applications
`
`third edition
`
`Clinical Pharmacokinetics
`
`
`
`MALcoiM ROWLAND
`: THOMAS N. 102ch
`
`'
`
`DRL EXHIBIT 1022 PAGE 1
`
`DRL EXHIBIT 1022 PAGE 1
`
`

`

`--~~~~~------------------------------------------------
`
`C I i n i c o I P h o r m o c o k i n e ·t i c s
`
`Coricepts and Applications
`
`third edition
`
`MALCOlM ROWlAND, Ph .D.
`Department of Pharmacy
`
`University of Manchester
`
`Manchester, England
`
`THOMAS N. TOZER, Ph .D.
`School of Pharmacy
`
`University of California
`
`San Francisco, California
`
`A Lea & Febiger Book
`
`'~LIPPINCOTT WILLIAMS & WILKINS
`
`•
`
`A Wolters Kluwer Company
`Philadelphia • Baltimore • New York • London
`Buenos Aires • Hong Kong • Sydney • Tokyo
`
`DRL EXHIBIT 1022 PAGE 2
`
`

`

`F.xecutive Editor: Donna Balado
`Developmental Editors: Frances Klass, Lisa Stead
`Production Manager: Laurie Forsy th
`Project Editor: Robert D. Magee
`
`Copyright© 1995
`Lippincott W ill iams & Wi lkins
`530 Walnut Street
`Philadelphia, Pe nnsylvania l 9 l 06-362 l USA
`
`All rights reserved. This book is protected by copyright. No part of this book may be re produced in any
`form or by any means, including photocopying, or utilized by any information storage and retrieval system
`without w ritten pe rmissio n from the copyright owner.
`
`Accurate indications , adverse reactions, and dosage schedules fo r 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 I Malcolm
`3rd ed.
`Rowland, Tho mas N. Tozer. -
`p .
`cm.
`"A Lea & Febiger Book. "
`Includes b ibliographical references and index.
`ISBN 978-0-683-07404-8
`ISBN 0-683-07404-0
`1. Pharmacokinetics. 2. Chemothe rapy.
`II. Title.
`[DNLM: 1. Pharmacokinetics.
`RM301.5 R68
`1994
`615 7-<lc20
`DNLM/ DLC
`fo r Library of Congress
`
`7be Publishers have made every effort to trace the copyright holders for borrowed material. If they have in(cid:173)
`advertently overlooked any, they will be pleased to make the necessary arrangements at the first opportunity .
`
`l l l 2 13 14 15 16 17 18 19 20
`
`94-26305
`CIP
`
`I. Tozer, Thomas N.
`
`2. Drug Therapy.
`
`QV 38 R883c 1994]
`
`DRL EXHIBIT 1022 PAGE 3
`
`

`

`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(cid:173)
`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(cid:173)
`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(cid:173)
`vorced from one another. For example, the convenience of giving a larger dose less fre(cid:173)
`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 t1ial
`and error. The dose, interval between doses , and route of administration were selected,
`and the patient's progress followed. The desired effect and any signs of toxicity were care(cid:173)
`fu lly noted, and if necessary, the dosage regimen was adjusted empi1ically until an accept(cid:173)
`able balance between the desired effect and toxicity was achieved. Eventually, after con(cid:173)
`siderable experimentation on a large number of patients, reasonable dosage regimens we re
`established (Table 1- 1), but not without some regimens producing excessive toxicity or
`proving ineffective. Moreover, the above empirical approach left many questions unan(cid:173)
`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(cid:173)
`nously? Why is morphine more effective given intramuscularly than when given orally?
`Fmihermore, 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(cid:173)
`ministration. In vitro and in vivo studies show th at 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 ach ieved
`by maintaining an adequate concentration of drug at the site(s) of action for the duration
`
`DRL EXHIBIT 1022 PAGE 4
`
`

`

`2
`
`WHY CLINICAL PHARtv\ACOKINETICS2
`
`CHAPTER l
`
`of therapy. However, rarely is a drug placed at its site of action. Indeed, most drugs are
`given orally, and yet they act in the brain, on the heart, at the neuromuscular junction, or
`elsewhere. A drug must therefore move from the site of administration to the site of action.
`Simultaneously, however, the drug distributes to all other tissues including those organs,
`notably the liver and the kidneys, that eliminate it from the body.
`Figure 1- 1 illustrates the events occurring after a dose of drug is administered orally.
`The rate at which drug initially enters the body exceeds its rate of elimination; the con(cid:173)
`centrations of drug in blood and other tissues rise, often sufficiently high to elicit the desired
`therapeutic effects and sometimes even to produce toxicity. Eventually, the rate of drug
`elimination exceeds the rate of its absorption, and thereafter, the concentration of drug in
`both blood and tissues declines and the effect(s) subsides. To administer drugs optimally,
`therefore, knowledge is needed not only of the mechanisms of drug absorption, distribu(cid:173)
`tion , and elimination but also of the kinetics of these processes, that is, pharmacokinetics.
`The application of pharmacokinetic principles to the therapeutic management of patients
`is clinical pharmacokinetics.
`
`Table 1-1 • Empirically Derived Usual Adult Dosage Regimens of Some
`Representative Drugs Before the Introduction of Clinical Pharmacoklnetics•
`
`DRUG
`
`Tetracycline
`
`Digoxin
`
`INDICATED USE
`
`Treatment of Infections
`Amelioration of congestive
`cardioc failure
`
`ROUTE
`
`Oral
`
`Oro!
`
`DOSAGE REGIMEN
`
`250 mg every 6-8 hr
`
`l 5- 2 mg initiolly over 24
`hr, thereafter 0.25-0.5
`mg once a day
`
`0 2-4 milliunits/ min by
`infusion
`
`Oxytocin
`
`Morphine sulfate
`
`Induction a nd mo intenance
`of labor
`Relief of severe pain
`
`Intravenous
`
`Intramuscular
`
`l 0 mg when needed
`
`Oral
`
`Not recommended because
`of reduced effectiveness
`
`"Token from American Medical Associolon. Drug Evoluolrons. 2nd Ed., Publishers Science Group, Acton , MA. 1973.
`
`Fig. 1-1. Plas!lla concentration of
`theophylline in a subject following an
`oral dose of a 600-!llg controlled-re(cid:173)
`lease forlllulation. Before the peak is
`reached, the rate of absorption ex(cid:173)
`cee ds that of eli lll in ati on. At the
`peak, the two rates are equal ; there(cid:173)
`after, the rate of elilllination exceeds
`that of absorption. (Redrawn from
`Sauter, R. , Steinijans, V.W., Oiletti ,
`E .. Bohlll , A. , and Sc hul z, H.U.:
`Presentation of results in bioequival(cid:173)
`ence studies. Int. J. Clin. Phar!llacol.
`The r. Toxicol. , .30:S7-30, 1992. )
`
`6
`
`5
`
`4
`
`3
`
`2
`
`Q) - - -
`
`c: ~ = en
`>- E
`..c: -
`Cl.. c:
`0
`0
`Q) · (cid:173)
`I- e
`..c: -
`~-E §3
`CJ)
`(.)
`~ c
`-
`0
`a.. u
`
`0
`
`0
`
`12
`
`24
`Hours
`
`36
`
`48
`
`DRL EXHIBIT 1022 PAGE 5
`
`

`

`CHAPTER l
`
`WHY CLINICAL PHARMACOKINETICS?
`
`3
`
`The events following drug administration can be divided into two phases, a pharrnaco(cid:173)
`kinetic 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 pharrna(cid:173)
`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(cid:173)
`carded or has suggested a more appropriate dosage regimen. Lastly, knowing the phar(cid:173)
`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(cid:173)
`cordingly, therapeutic failure results when either the concentration is too low, giving in(cid:173)
`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(cid:173)
`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
`Regimen
`
`Plasma
`Concen-
`tration
`
`.-- --- ---
`I
`Site
`I
`I
`of
`I
`I Action
`[ ________
`
`I
`
`Effects
`
`.)
`
`+
`...... ___ ____ ___ __ ;
`
`\
`
`I
`I
`I
`I
`I
`
`+
`...... ____ ________ ;
`
`F ig. 1- 2. An approach to the design of a dosage regimen. The pharmacokinetics and the pharmacodynamics of
`the drug are first defined. Then, either the plasma drug concentration-time data or the effects produced are used
`i ia pharm acoki netics as a feedback (dashed lines) to modify the dosage regimen to achieve optimal therapy.
`
` DRL EXHIBIT 1022 PAGE 6
`
`

`

`4
`
`W HY CLINICAL PHARMACOKINETICS2
`
`CHAPTER l
`
`several doses had to be given before drug accumulation was sufficient to produce a ther(cid:173)
`apeutic concentration. Had therapy bee n stopped before then, the drug might have been
`thought ineffective and perhaps abandoned prematurely. Alternatively, larger doses might
`have been tried, e.g., regimen B. Although a therape utic response would have been
`achieved fairly promptly, toxicity would have ensued with continued administration when
`the concentration exceeded the upper limit of the therapeutic window.
`The synthetic antimalarial agent, quinacrine, developed during World War II to substi(cid:173)
`tute for the relatively scarce quinine, is an example. Quinacrine was either ineffective
`acutely against malaria or eventually produced unacceptable toxicity when a dosing rate
`sufficiently high to be effective acutely was maintained. Only after its pharmacokinetics
`had been defined was this drug used successfully. Quinacrine is eliminated slowly and
`accumulates extensively with repeated daily administration. 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 therapeutic window.
`The plateau situation in Fig. 1-3 shows that both the width of the therapeutic window
`and the speed of drug elimination govern the size of the maintenance dose and the fre(cid:173)
`quency of administration. When the window is narrow and the drug is eliminated rapidly,
`small doses must be given often to achieve therapeutic success. Both cyclosporine and
`digoxin have a narrow therapeutic window, but because cyclosporine is 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 within minutes. The
`only means of adequately ensuring a therapeutic concentration of oxytocin 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(cid:173)
`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(cid:173)
`tensively metabolized on passage through the liver, an organ lying between the gastroin(cid:173)
`testinal tract and the general circulation.
`Awareness of the benefits of understanding pharmacokinetics and concentration-re(cid:173)
`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,
`a potent compound found to be poorly and unreliably absorbed and intended for oral
`administration may be shelved in favor of a somewhat less potent but more extensively and
`reliably absorbed compound. Also, many of the basic processes controlling both pharma(cid:173)
`cokinetics and response are similar across mammalian species such that data can be ex(cid:173)
`trapolated from animals to predict quantitatively the likely behavior in humans . This quan-
`
`Regimen B
`
`Fig. 1-3. When a drug is given in
`a fixed dose and at fixed time inter(cid:173)
`vals (denoted by the arrows ), it ac(cid:173)
`cumu lates within the body until a
`plateau is reached. With regimen A,
`therapeutic success is achieved al(cid:173)
`though not initially. With regimen B,
`the therapeutic objective is achieved
`more quickly, but the plasma drug
`concentration is ultimately too high.
`
`Therapeutic
`Failure
`
`Therapeutic
`Success
`
`}
`
`Therapeutic
`Failure
`
`--- -------- --- -
`
`Time
`
`·-=-------------------------------------
`
`DRL EXHIBIT 1022 PAGE 7
`
`

`

`CHAPTER 1
`
`WHY CLINICAL PHARMACOKINETICS?
`
`s
`
`titative framework improves the chances of selecting not only the most prom1smg
`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 regulat01y 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
`
`CLINICAL (HUMAN) TESTING
`
`Dose (Cone)
`Population PK/PD
`Response Trials
`Characteristics in
`/----; (';. n
`Large Efficacy Trials
`G==:v PK-guided ~ U v ~ <=
`Efficacy ~ =>
`In vitro PK/PD
`Dose escalation
`PK/PD in Special
`Safety ~ Dosage
`Animal PK/PD
`Populations
`~ Assessment
`Selection
`Patient Variables
`
`Toxicity
`I Animal Testing)
`
`I Phase I
`
`)
`.
`
`~P_h_ase_ll ~> l~P_hase_l_ll __.)
`
`Post(cid:173)
`Marketing
`Surveillance
`
`Fig. 1-4. The development and subseque nt marketing of a drug. The prehuman data helps to ide ntify promising
`compounds and to suggest useful doses for testing in humans. Phases I, II , and III of human assess ment gene rally
`correspond to the first adm inistration to hum ans, early evaluation in selected patients, and the larger trials,
`respectively. Pharmacokinetic (PK ) and pharmacodynamic (PD ) data gathered du1ing all phases of drug devel(cid:173)
`opment help to efficiently de fin e safe and effective dosage regimens for optimal individual use. Postmarke ting
`surveillance helps to refin e the PK/PD information .
`
`(/) 25
`c
`0
`:;::::; 20
`~ > '-
`CV 15
`
`(/)
`..0
`
`0 - 10
`
`0
`.......
`c
`CV 5
`(.;)
`'-
`CV
`CL 0
`
`Fig. 1-5. The daily dose of warfarin required to produce si milar
`prothrombin times in 200 adult patients varies widely. (1 mg!L =
`3.3 µM ). (Redrawn from Koch-Weser, J.: The serum level ap(cid:173)
`proach to individualization of drug dosage. Eur. J. Clin. Pharma(cid:173)
`col. 9:1-8, 1975.)
`
`N O'l O'l O'l O'l O'l O'l O'l O'l O'l O'l
`
`V N c0 -.:t ir:i c.O ,.....: a:i m ci ci
`I
`I
`I
`I
`I
`I
`I
`I ..--..-(cid:173)
`-.::t" UJ C.0 f'- 00 O'l 6 f\
`N M
`
`Daily Dose (mg)
`
`DRL EXHIBIT 1022 PAGE 8
`
`

`

`6
`
`WHY CLINICAL PHARfv\ACOKINETICS?
`
`CHAPTER l
`
`agulant warfa1in needed to produce a similar prothrombin time (an index of blood coag(cid:173)
`ulability). Sources of variability in drug response include the patient's age, weight, degree
`of obesity, type and degree of severity of the disease, the patient's genetic makeup, other
`drugs concurrently administered, and environmental factors. The result is that a standard
`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 individual
`patient is evident; this need is clearly greatest for drugs that have a narrow therapeutic
`window, that exhibit a steep concentration-response curve, and that are critical to drug
`therapy. Examples are digoxin, used to treat some cardiac disorders; phenytoin, 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 pharmacokinetics is a major source of total
`variability in drug response .
`It is becoming increasingly common to gain as much information on variability as pos(cid:173)
`sible during drug development by gathering, albeit limited, individual plasma concentration
`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 pharmacokinetidpharmacodynamic 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 occasionally unpredictable. Keto(cid:173)
`conazole, for example, devoid of immunosuppressant activity, 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 pharmacokinetics. Some drugs stimulate drug-
`
`F ig. 1- 6. Although the average plateau plasma
`concentration of phenytoin tends to increase with
`the dosing rate, the re is considerable variation in
`the individual values. (One mg!L = 3.97 µM .) (Re(cid:173)
`drawn from Lund, L.: Effects of phenytoin in pa(cid:173)
`tients \vith epilepsy in relation to its concentration
`in plasma. In Biological Effects of Drugs in Relation
`to Their Plasma Concentration. Edited by D.S. Da(cid:173)
`vies and B.N.C. P1ichard, Macmillan, London and
`Basingstoke, 1973, pp. 227-238.)
`
`E
`c
`0
`
`__J --0>
`~ ...... -c
`
`Q.)
`
`'-' c
`0 u
`c
`0
`>.,
`c
`Q.)
`..c
`D._
`
`50
`
`40
`
`30
`
`20
`
`•
`
`•
`
`•
`
`•
`• •
`• • • •
`• •
`
`•
`•
`
`• •
`
`~
`
`E
`~ 10
`
`D._
`
`0
`
`0
`
`10
`5
`Daily Dose (mg/kg)
`
`15
`
`DRL EXHIBIT 1022 PAGE 9
`
`

`

`CHAPTER 1
`
`WHY CLINICAL PHARtv\ACOKINETICS?
`
`7
`
`metabolizing enzymes and hasten drug loss; others inhibit these enzymes and slow elimi(cid:173)
`nation. Still others interfere with drug absorption. Such interactions are graded; the change
`in the pharmacokinetics of a drug varies continuously with the plasma concentration of the
`interacting drug and hence with time. Indeed, given in sufficiently high doses, almost any
`drug can interact with another. It is always a question of degree. Understanding the quan(cid:173)
`titative elements of interactions ensures the more rational use of drugs that may need to
`be coadministered.
`Figure 1- 6 illustrates a situation in which monitoring of the drug concentration may be
`beneficial. Over the narrow range of the daily dose of the antiepileptic drug phenytoin, the
`plateau plasma drug concentration varies markedly within the patient population. Yet the
`therapeutic window of phenytoin is narrow, 7 to 20 mg!L; beyond 20 mg!L, the frequency
`and the degree of toxicity increase progressively with concentration. Here again, pharma(cid:173)
`cokinetics 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 pharrnacokinetics 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 pharrnacokinetic 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(cid:173)
`sideration of kinetic concepts basic to pharrnacokinetics and ends with a section containing
`selected topics.
`
`DRL EXHIBIT 1022 PAGE 10
`
`

`

`(
`
`.. '
`
`':'--
`
`-~··
`~ ' ..
`
`'
`
`.
`
`4
`
`EXTRAVASCULAR DOSE
`
`OBJECTIVES
`
`The reader will be able to:
`l . Describe the characteristics of, and the differences between, first-order and zero-order ab(cid:173)
`sorption processes .
`
`2. Determine w hether absorption or disposition rate limits drug elimi nation, given plasma con(cid:173)
`centration-time data following different dosage forms or routes of administration.
`
`3. Anticipate the effect of altering rate of absorption , extent of absorption , clearance, or volume
`of distribution on the plasma concentration and amount of drug in the body following extra(cid:173)
`vascular administration.
`
`4. Estimate the bioavailability of a drug, given ei ther plasma concentration or urinary excretion
`data following both extravascular and intravascular administration.
`
`5 . Estimate the relative bioavailability of a drug , given either plasma concentration or urinary
`excretion data following different dosage forms or routes of administration .
`
`6. Estimate the renal clearance of a drug from plasma concentration and urinary excretion
`data following extravascular administration.
`
`For systemically acting drugs , absorption is a prerequisite for therapeutic activity when
`they are administered extravascularly. The factors that influence drug absorption are con(cid:173)
`sidered in Chap. 9, Absorption. In this chapter the following aspects are examined: the
`impact of rate and extent of absorption on both plasma concentration and amount of drug
`in the body; the effect of alterations in absorption and disposition on body level-time
`relationships; and the methods used to assess pharmacokinetic parameters from plasma
`and urinary data following extravascular administration.
`The term bioavailability is commonly applied to both rate and extent of drug input into
`the systemic circulation. Throughout this book the term will be limited to the extent of
`drug input and can be considered as the fraction, or percent, of the administered dose
`absorbed intact.
`
`KINETICS OF ABSORPTION
`
`The oral absorption of drugs often approximates first-order kinetics, especialiy when given
`in solution. The same holds true for the absorption of drugs from many other extravascular
`sites including subcutaneous tissue and muscle. Under these circumstances, absorption is
`characterized by an absorption rate constant, ka , and a corresponding half-life. The half(cid:173)
`lives for the absorption of drugs administered orally in solution or in a rapidly disintegrating
`dosage form usually range from 15 min to 1 hr. Occasionally, they are longer.
`
`34
`
`DRL EXHIBIT 1022 PAGE 11
`
`

`

`CHAPTER 4
`
`EXTRAVASCUlAR DOSE
`
`35
`
`Sometimes, a drug is absorbed at essentially a constant rate. The absorption kinetics are
`then called zero order. Differences between zero-order and first-order kinetics are illus(cid:173)
`trated in Fig. 4-1. For zero-order absorption, a plot of amount remaining to be absorbed
`against time yields a straight line, the slope of which is the rate of absorption (Fig. 4-lA).
`Recall from Chap. 3 that the fractional rate of decline is constant for a first-order process;
`the amount declines linearly with time when plotted semilogarithmically. In contrast, for
`a zero-order absorption process, the fractional rate increases with time, because the rate
`is constant but the amount remaining decreases. This is reflected in an ever-increasing
`gradient with time in a semilogarithmic plot of the amount remaining to be absorbed (Fig.
`4-lB ). A graphical method of examining the kinetics of absorption from plasma data fol(cid:173)
`lowing extravascular administration is given in Appendix 1-C.
`For the remainder of this chapter, and for much of the book, absorption is assumed to
`be first order. If absorption is zero order, then the equations developed in Chap. 6 (Con(cid:173)
`stant-Rate Regimens ) apply.
`
`A. Regular Plot
`
`8. Sem ilogarithmic Plot
`
`-0 100
`Q.)
`.....
`..Cl
`0 en
`..Cl
`<l:
`Q.) ro
`
`80
`
`0 - 60
`
`40
`
`First Order
`
`Ol
`c
`c
`ro
`E
`
`Q.) a: - 20
`
`c
`Q.)
`u .....
`
`Q.)
`Q_
`
`0
`
`100
`50
`
`10
`
`5
`
`Zero Order
`
`First Order
`
`-0
`Q.)
`.....
`..Cl
`0 en
`..Cl
`<l:
`Q.) ro
`
`0 -Ol
`
`c
`c
`ro
`E
`
`Q.) a: -c
`
`Q.)
`
`u .....
`
`Q.)
`Q_
`
`Time
`
`Time
`
`Fig. 4- 1. A comparison of zero-order and first-order absorption processes. Depicted are: A , regular and B,
`semilogarithmic plots of the percent remaining to be absorbed against time.
`
`BODY LEVEL-TIME RELATIONSHIPS
`
`Comparison With an Intravenous Dose
`
`Absorption delays and reduces the magnitude of the peak compared to that seen following
`an equal i.v. bolus dose. These effects are portrayed for aspirin in Fig. 4- 2. The rise and
`fall of the drug concentration in plasma are best understood by remembe1ing (Chap. 2,
`Eq. 2, p. 16) that at any time
`
`dAa
`dt
`Rate of
`absorption
`
`k ·A
`
`Rate of
`eli mination
`
`dA
`dt
`Rate of
`change of
`drug in
`body
`
`DRL EXHIBIT 1022 PAGE 12
`
`

`

`36
`
`EXTRAVASCUlAR DOSE
`
`CHAPTER 4
`
`where Aa is the amount of drug at the absorption site remaining to be absorbed. When
`absorption occurs by a first-order process, the rate of absorption is given by ka ·Aa .
`Initially, with all drug at the absorption site and none in the body, rate of absorption is
`maximal and rate of elimination is zero. Thereafter, as drug is absorbed, its rate of absorp(cid:173)
`tion decreases, whereas its rate of elimination increases. Consequently, the difference be(cid:173)
`tween the two rates diminishes. However, as long as the rate of absorption exceeds that of
`elimination the plasma concentration continues to rise. Eventually, a time t,,,aX' is reached
`when the rate of elimination matches the rate of absorption; the concentration is then at
`a maximum, C,,,ac Subsequently, the rate of elimination exceeds the rate of absorption and
`the plasma concentration declines.
`The peak plasma concentration is always lower following extravascular administration
`than the initial value following an equal i.v. bolus dose. In the former case, at the peak
`time some drug remains at the absorption site and some has been eliminated, while the
`entire dose is in the body immediately following the i.v. dose. Beyond the peak time, the
`plasma concentration exceeds that following i.v. administration of the same dose because
`of the continual entry of drug into the body.
`Frequently, the rising portion of the plasma concentration-time curve is called the ab(cid:173)
`sorption phase and the declining portion, the elimination phase. As will be seen, this de(cid:173)
`scription may be misleading. Also, if bioavailability is low, the drug concentration may
`remain lower than that observed after i.v. administration at all times.
`Lag time, the delay between drug administration and the beginning of absorption, may
`be particularly important when a rapid onset of effect is desired. The lag time can be
`anywhere from a few minutes to many hours. Long lag times have been observed following
`ingestion of ente1ic-coated tablets. The coating is resistant to the gastric environment, thus
`protecting an acid-labile drug or preventing gastric irritation by a drug. Factors contributing
`to the lag time are the delay in emptying the product from the stomach and the time taken
`for the protective coating to dissolve or to swell and release the inner contents into the
`intestinal fluids. Once absorption begins, however, it may be as rapid as with uncoated
`tablets. Clearly, enteric-coated products should not be used when a prompt and predictable
`response is desired. A method for estimating lag time is discussed in Appendix I-C.
`Bioavailability and area are also important factors. As discussed more fully in Chaps. 7
`and 9, the completeness of absorption is of primary importance in therapeutic situations.
`
`c
`0
`·.;::::;
`ro ,_
`
`-c
`
`(/)
`<(
`ro
`E
`(/) ro
`0....
`
`CJ.)
`u
`c
`__J -- 6
`o -u
`c Ol
`·;::::: E
`·a.- 4
`
`10
`
`8
`
`2
`
`0
`
`0
`
`20
`
`40
`
`60
`Minutes
`
`80
`
`100
`
`120
`
`Fig. 4-2. Aspi1in (650 mg) was administered as an intravenous bolus (• ) and as an oral solution (o) on separate
`occasions to the same individual. Absorption causes

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