throbber
AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Clinical Pharmacokinetics
`Concepts and Applications
`
`third edition
`
`MA.LCOLM 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
`
`Williams & Wilkins
`
`BALIIMORE • PHilADELPHIA • HONG KONG
`lONDON • MUNICH • SYDNEY • TOKYO
`A WAVE RLY COMPANY
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Executive Editw·: Donna Balado
`Developmental Editors: Frances Klass, Lisa Stead
`Production Manage~· Laurie Forsyth
`Project Editor: Robert D. Magee
`
`Copyright © 1995
`Williams & Wilkins
`Rose Tree Corporate Center
`1100 North Providence Road
`Building IT, Suite 5025
`Media, PA 19063-2043 USA
`
`All rights reserved. This book is protected by copyright. No patt 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 n:~ader 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 an d applications I Malcolm
`Rowland, Thomas N. Tozer. - 3rd eel.
`em.
`p.
`"A Lea & Fcbiger Book."
`Includes bibliographical references and index.
`ISBN 0-683-07104-0
`1. Phannacokinetics. 2. Chemotherapy.
`ll. Title.
`IDNL.l\1: 1. Pharmacokinetics. 2. Drug Therapy.
`RM301.5.R68
`1994
`615.7-clc20
`DmM/ DLC
`for Library of Congress
`
`I. Tozer, Thomas
`
`QV 38 R883c 1991]
`
`94-26305
`ClP
`7be Publishers have made every ef!ort to trace the copyright holders for borrowed material. If they have in-
`advertently overlooked any, they will be pleased to make tbe necessmy an·angemenls at the .first opportunity.
`
`Reprints of cl1apters may be purchased from Williams & Wilkins in quantities of 100 or more. Call
`Isabella Wise, Special Sales Department, (BOO) 358-3583.
`
`96 97 98
`2 3 4 5 6 7 8 9 10
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`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 exb·eme are those who take
`a single dose of a drug to relieve ao occasional headache. The duration of drug therapy i'>
`usually between these exhemes. The manner in which a drug is taken is called a dosage
`regim.en. 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 appropliate "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 djsease. Then the
`questions How much? How often? and How long? must be answered. The question Hm.v
`much? recognizes that the magnitudes of the therapeutic and toxic responses m·e 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 m1other. 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 tlial
`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-
`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 expe1irn.entation on a large number of patients, reasonable dosage regimens \·Ver e
`established (Table 1-1), but not without some regimens producing excessive toxicity or
`proving ineflective. Moreover, the above empirical approach left many questions unan-
`S\Vered. Why, for example, docs 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? vVhy 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 batl1ing the site(s) of action. From these
`observations the suggestion might be made that tl1e therapeutic objective can be achieved
`by maintaining an adequate concentration of drug at the site(s) of action for the duration
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`J
`
`CHAPTER l
`
`WHY CLINICAL PHARMACOKINETICS?
`
`2
`of therapy. However, rarely is a drug placed at its site of action. IndeP.d, 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-
`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 tl1ereafter, the concentration of dmg in
`both blood and tissues declines and the effect(s) subsides. To administer drugs optimally,
`d1erefore, knowledge is needed not only of the rnecbanisms of drug absorption, distribu-
`tion, and elimination but also of the kinetics of these processes, that is, pharmacokinetics.
`The application of pharmacokinetic principles to tl1e therapeutic management of patients
`is clinical pharmacokinetics.
`
`DRUG
`Tetracycline
`Digoxin
`
`ROUTE
`Oral
`Oral
`
`Intravenous
`
`lntra muscula·
`Oral
`
`Table 1-1. Empirically Derived Usual Adult Dosage Regimens of Some
`Representative Drugs Before the Introduction of Clinical Pharmacokineticsa
`DOSAGE REGIMEN
`INDICA TED USE
`25 0 mg every 6-8 hr
`Treatment of Infections
`1.5-2 mg initially over 24
`Amelioration of congestive
`hr, thereafter 0. 25-0.5
`cardiac failure
`mg once a day
`0.2-4 milliunits/min by
`infusion
`1 0 mg when needed
`Not recommended because
`of reduced effectiveness
`"Token from American Medical Association: Drug Evoluolions. 2nd Ed .• Publishers Science Group, Acton, 11/\A, 1973.
`
`O xytocin
`
`Morphine sulfate
`
`Ind uction and maintenance
`of labor
`Relief of severe pain
`
`Fig. 1-l. Pla~ma concentratio n of
`theophylline in a subjectfollowing an
`oral dose of a 600-mg controlled-re-
`lease formu lation. Before the peak is
`reached , the rate of absorption ex-
`ceed s that of e limination. At the
`peak, the two rates arc eCJual; the re-
`after, the rate of elimination exceed~
`that of absorption. (Redrawn f1·om
`Sauter, R., Steinijans, V.W., D iletti,
`E., Bt'l lu.n, A., and Sch ulz, H.U.:
`P resentation of results in b ioequival-
`ence studies. Int . J. Clin. Pharmacal.
`Ther. Toxicol., 30:57-30, 1992.)
`
`6
`
`Q ) -
`
`5
`·= ~ 4
`3
`
`>-E
`.c--
`C..c
`0
`0
`Q) · -
`~"§
`ct:l +-'
`E li5 U ) u
`~§ o..o
`
`2
`
`1
`
`0
`
`0
`
`12
`
`36
`
`48
`
`24
`Hours
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`CHAPTER l
`
`W HY CLINICAL PrARMACOKINETICS?
`
`3
`
`The events following drug administration can be divided into two phases, a pharnwco-
`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 pharnw-
`codynarnic phase, in which the concentration of drug at the site(s) of action is related to
`the magnitude of the effect(s) produced (Fig. l-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 seveml advantages
`over the empilical approach. First, and most obvious, distinction can be made between
`pharmacok:inetic 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, knovving 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. Betw·een 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 dmgs, this therapeutic objective is met by giving an initial dose to achieve a plasma
`concentration witllin 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 l-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. \Vith regimen A,
`
`Pharmacokinetics
`
`Pharmacodynamics
`
`Dosage
`Regimen
`
`Plasma
`Concen-
`tration
`
`,...-------
`I
`Site
`I
`I
`of
`I
`I Action
`[ ________
`I
`
`Effects
`
`+ l
`+ l
`...... ___________ _
`-------------
`Fig. 1-2. An approach to tbe design of a dosage regimen. The pharmacokinetics and the pharmacodynamics of
`the dmg are first defined. Then, either the plasma drug concentration-time data or the effects produced are used
`via pharmacokinetics as a feedback (dashed lines) to rttoclil)r the dosage regimen to achieve optimal therapy.
`
`l
`/
`
`I
`I
`I
`
`I
`/
`
`I
`I
`I
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`4
`
`WHY CUNICAL PHARMACOKINETICS?
`
`CHAPTER l
`
`several doses had to be given before drug accumulation was sufficient to produce a ther-
`apeutic concentration. Had therapy been stopped before then, the drug might have been
`thought ineffective and perhaps abandoned prematw-ely. Alternatively, larger doses might
`have been tried, e.g., regimen B. Although a therapeutic response would have been
`achieved fairly promptly, toxicity would have ensued with continued adminisb·ation when
`the concentration exceeded the upper limit of the therapeutic window.
`The synthetic antimala1ial agent, quinacrine, developed during World vVar II to substi-
`tute for the relatively scarce quinine, is an example. Quinacline was either inefTective
`acutely against malalia or eventually produced unacceptable toxicity when a dosing rate
`sufficiently high to be eflective acutely vvas maintained. Only after its phannacokinetics
`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 tl1erapeutic success, followed by small daily
`doses to maintain the plasma concenb·ation within the therapeutic window.
`The plateau situation in Fig. l-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-
`quency of administration. \Vhen the window is nan-ow and tl1e dmg 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-
`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 metaLollL.ed on passage through the liver, an organ lying between thP- gastroin-
`testinal tract and tb.e general circulation.
`Awareness of the benefits of understanding pharmacokinetics and concentration-re-
`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-
`cokinetics and response are similar across mammalian species such that data can be ex-
`b"apolated froin animals to predict quantitatively the likely behavior in humans. This quan-
`
`F ig. 1-3. When a drug is given in
`a fixed dose and at fixed lime inter-
`vals (denoted by the arrows), it ac-
`cumulates within the body until a
`plateau is reached. With mgimen A,
`therapeutic success is achjeved al-
`though not initially. With regimen B,
`th.e therapeutic objective is acllievcd
`more quickly, but the plasma drug
`concentration is ulti111ately too high.
`
`c:
`0
`
`~ -c:
`
`Q)
`
`(.) c:
`0
`(.)
`0>
`,_
`:::l
`Cl
`m
`E en
`m
`Q..
`
`Regimen B
`
`- ---- ------ ----
`
`+
`
`+
`
`+ + +
`Time
`
`Therapeutic
`Failure
`
`Therapeutic
`Success
`
`}
`
`Therapeutic
`Failure
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`CHAPTER 1
`
`WHY CLINICAL PHARMACOKINETICS?
`
`5
`
`titative framework improves the chances of selecting not only the most prmmsmg
`compounds but also the correct range of safe doses to first test in humans. Incorporation
`of a pharmacokinetic element vvith 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 dming drug development and
`therapy, variability. There is a wide range of daily dose requirements of the oral antico-
`
`PRECLINICAL
`TESTING
`
`G=v PK-guided ? D .Q,
`
`CLINICAL (HUMAN) TESTING
`Population PK/PD
`Dose (Cone)
`Characteristics in
`Response Trials
`Large Efficacy Trials
`¢=::1
`c::::>
`PK/PD in Special
`Populations
`
`Post-
`Marketing
`Surveillance
`
`Efficacy
`Dose escalation
`In vitro PK/PD
`Safety ~ Dosage
`Animal PK/PD
`Selection
`~Assessment
`Patient Variables
`
`Toxicity
`I Animal Testing)
`
`I
`
`Phase I
`
`> Phase II
`
`)
`~----~ ~~ -P-h-as-e-11-1~)
`
`Fig. l-4. The development and subsequent marketing of a drug. The prehmnan data helps to identify promising
`compounds and to suggest usefill doses for testing in humans. Phases I, II, and III of human assessment generally
`correspond to the first administration to hmnaus, early evaluation in selected patients, and the hu·ger trials,
`respectively. l)hannacokinetic (P K) 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. l'ostrnarketing
`smveillance helps to refine the PK/PD information.
`
`(/)
`
`(/) 25
`c
`0
`~ 20
`:;:::::;
`> ..... Q) 15
`0 - 10
`.c
`0 ...... c
`Q) 5
`..... Q) a.. 0
`
`(..)
`
`Fig. l -5. The daily dose of warfarin required to produce similar
`prothrombin times in 200 adult patients vaJies widely. (J mg!L =
`3.3 J.tM). (Redrawn from Kocb-\~1eser, J.: The serum level ap-
`proach to individualization of drug dosage. Eur. J. Clin. Pharrna-
`col. 9:1- 8, 1975.)
`
`-
`
`-
`
`1-
`
`,.--
`
`-_ 1-
`
`-
`
`-
`
`r--
`
`1-
`
`NO">O">O'>O'>O'>O'>O'>O'>O'>O'>
`. . . . . . . . . .
`V NC"J'<:j"L.(')C.OI"-000'>00
`I
`I
`I
`I
`I
`I
`I
`I T""T""
`NC"J-::I"L.(')C.OI"-OOO'>c!,/\
`
`Daily Dose (mg)
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`6
`
`WHY CUNICAL PHARMACOKINETICS?
`
`CHAPTER I
`
`agulant warfm·in needed to produce a similar prothrombin time (an index of blood coag-
`ulability). Sources of vruiability 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 otl1ers,
`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 arc 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. \Vitl1
`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 vru·iabiJity as pos-
`sible during drug development by gathering, albeit }jmited, individual plasma concentration
`and response data in a large population of patients during Phase III cliillcal trials. Attempts
`are then made to account for this variability in terms of such patient characte1istics as age
`and weight. These population phannacokinetic/pharmacodynamic 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-
`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-
`•
`
`Fig. 1- 6. Although the average plateau plasma
`concentration of phenytoin tends to increase with
`t.he dosing rate, there is considerable variation in
`the individual values. (One mg!L = 3.97 J.1M.) (Re-
`drawn from Lund, L.: EH'ects of phenytoin in pa-
`tients with epilepsy in relation to its concentration
`in plasma. In Biological Effects ofDmgs in Hclation
`to Their Plasma Concentration. Edited by D.S. Da-
`vies and B.N.C. Prichard, Macmillan, London and
`Basingstoke, 1973, pp. 227- 238.)
`
`50
`
`40
`
`c::
`0
`~ 30
`,._..
`c::
`<P
`(..) c::
`0 <.:>
`c::
`0 20
`>, c::
`<P .c a..
`E
`~ 10
`([
`
`~
`
`0
`
`0
`
`•
`
`•
`• • •
`•
`.........
`• • ••
`• •
`•
`••••••
`..........
`••
`• •
`• •
`• ••••••••
`. ,·:ft-'~···
`•
`•• -··1
`..... a ....
`\~:\.
`. . :,. :-·~( ~.
`••
`fO:i• ••
`
`•
`
`• •
`
`10
`5
`Daily Dose (mg/kg)
`
`15
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`CHAPTER l
`
`WHY CLINICAL PHARMACOKINETICS2
`
`7
`
`metabolizing enzymes and hasten drug loss; others inhibit these enzymes and slow elimi-
`nation. Still others interfere with drug absorption. Such interactions are graded; the change
`in the pharmacokinetics of a drug varies continuously vv:ith 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-
`titative elements of interactions ensures the more rational use of drugs tl1at may need to
`be coadministered.
`Figure l-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 tl1.e 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 witl1 concentration. Here again, pharma-
`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. Contwl 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 tl1e drug are known.
`Dmg 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 pharmacoki.nctic approach. It begins with a con-
`sideration ofldnetic concepts basic to pharmacokinetics and ends with a section containing
`selected topics.
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`.
`
`--
`--
`
`'L
`
`2
`
`BASIC CONSIDERATIONS
`
`OBJECTIVES
`The reader w ill be able to:
`l . Define the following terms:
`Pharmacokinetics, intravascular and extravascular administration, absorption, d ispo-
`sition, distribution, metabolism, excretion, first-pass effect, enterohepatic cycling, com-
`partment
`2. Discuss the limitations to interpretation of pharmacokinetic data imposed by assays that fail
`to distinguish between compounds administered (e.g., R-and $-isomers) or between drug
`and metabolite.
`3. Show the general contribution of mass balance concepts to drug absorption and drug and
`metabolite disposition.
`
`Pharmacokinetics has many useful applications that stem from basic concepts. These con-
`cepts are developed in this section of the book. This chapter specifically defines terms and
`describes a basic model for drug absorption and disposition.
`
`ANATOMIC AND PHYSIOLOGIC CONSIDERATIONS
`Measurement of a drug in the body is limited usually to blood or plasma. Nonetheless, the
`information obtained has proved very useful. Such usefulness can be ex-plained by anatomic
`and physiologic features that affect a drug following its administration.
`Blood or plasma, in addition to being a practical and convenient site of measurement,
`is the most logical one for determining drug in the body. Blood receives drug from the site
`of' administration as well as carries it to all the organs, including those in which the drug
`acts and those in which it is eliminated. This movement of drug is depicted schematically
`in Fig. 2-1. This scheme forms a basis for physiologic 1nodeling in pharrnacoki·netics. Such
`modeling has applications not only in clinical pharmacokinetics but in drug development,
`veterinary medicine, and in assessing risk associated with exposures to environrnental and
`occupational substances.
`
`Sites of Administration
`There are several sites at which drugs are commonly administered. These sites may be
`classified as either intravascular or extravascular. Intravascular admi.njstration refers to the
`placement of a drug directly into the blood, either intravenously or intra-arterially.
`Extravascular modes of administration include the oral, sublinguaL buccal, intranms-
`cular, subcutaneous, dermal, pulmonary, and rectal routes. To enter the blood, drug ad-
`
`11
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`12
`
`BASIC CONSIDERATIONS
`
`CHAPTER2
`
`ministered extravascularly must be absorbed: No absorption step is required when a drug
`is administered intravascularly.
`Drug may also be administered regionally, e.g., into the pleural or peritoneal cavities or
`into the cerebrospinal fluid. Regional administration includes intra-arterial injection into
`the vesselleadin.g to a tissue to be treated, e.g., one containing a cancerous tumor. It is a
`potential means of gaining a selective therapeutic advantage. This advantage, in comparison
`with other routes of administration, comes about by increasing drug exposure locally, where
`it is needed, and decreasing or producing little or no change in exposure throughout the
`rest of the body, where it is not wanted.
`
`Disposition
`Once absorbed, a drug is distributed to the various organs of the body. Distribution is
`influenced by how well each organ is perfused with blood, organ size, binding of drug
`within blood and in tissues, and permeability of tissue membranes.
`The two principal organs of elimination, the liver and the kidneys, are shown separately
`in Fig. 2-l. The kidneys are the primary site for excretion of the chemically unaltered, or
`unchanged, drug. The liver is the usual organ for drug metabolism; however, the kidneys
`and other organs can also play an important metabolic role for certain drugs. The metab-
`olites so formed are either further metabolized or excreted unchanged. The liver may also
`secrete unchanged drug into the bile. The lungs are, or may be, an important route for
`
`Fig. 2-1. Once absorbed from any of the many sites
`of administration, drug is conveyed by blood to aU sites
`within the body including the eliminating organs. Sites
`of admiojstration include: a, artery; b, peripheral vein;
`c, muscle and subcutaneous tissue; d, lung; and e, gas-
`lrointestiual tract. The dark- and light-colored lines
`V\~th arrows refer to the mass movement of dm g in
`blood and in bile, respectively. The movemc11t of vir-
`tually any drug can be followed from site of adininis-
`tration to site of elimination.
`
`* Site of Administration
`D Enterohepatic Cycle
`
`•
`
`Route of Elimination
`
`* d
`
`-
`
`-
`
`Tissues
`
`* b
`
`+
`
`CB Heart D
`C o~:,
`....
`· c;J
`
`'0
`0
`0
`a:J
`en
`:::J
`0
`c
`C1) >
`
`J
`
`Renal Excretion
`Gut
`Wall
`
`* e
`Gut Q Lumen
`
`Fecal Excretion Metabolism
`and Decomposition
`
`AMN1082
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`CHAPTER 2
`
`BASIC CONSIDERATIONS
`
`13
`
`eliminating volatile substances, for example, gaseous anesthetics. Another potential route
`of eHmination is via a mother's milk. Although an insignificant route of elimination in the
`mother, drug in the milk may be consumed in sufficient quantity to affect the suckling
`infant.
`
`CHEMICAL PURITY AND ANALYTIC SPECIFICITY
`
`A general statement needs to be made about the chemical purity of prescribed medicines
`and the specificity of chemical assays.
`Over the years, a major thrust of the pharmaceutical industry has been to produce
`therapeutic agents that are not only as safe and eflective as possible but also arc well
`characte1izcd to ensure reproducible quaHties. The majority of administered drugs today
`are therefore essentially pure mate1ials, and coupled with specific analytic techniques for
`their determination in biologic :Buids, definitive information about their pharmacokinetics
`can be gained. However, a large number of drug substances are not single chemical entities
`but rather mixtures. This particularly appHcs to stereoisomers and proteins. The most
`common stcrcoisomers found together in medicines are optical isomers, or compounds for
`which their structures are mirror images; the drug substance is usually a racemate, a 50:.50
`mixture of the R- and S-isomers. Some dmg substances contain geometric isomers, and
`still others, especially proteins of high molecular weight derived from natural products or
`through fermentation, may be a mixture of structurally related, but chemically distinct,
`compounds. Each chemical entity within the drug substance can have a different phar-
`macologic, toxicologic, and pharmacokjnetic profile. Sometimes these differences are small
`and inconsequential, other times the differences can be therapeutically important. For
`example, dextroamphetamine (S-isomer) is a potent central nervous stimulant, whereas the
`R-enantiomer is almost devoid of such activity. Despite such differences, many commonly
`employed chemical assays do not distinguish between stereoisomers. Obviously, under
`these circumstances, attempting to quantifY the various processes and to relate plasma
`concentration to response has many problems with no simple solutions. Notwithstanding
`these problems, specific information about each chemical entity should be sought whenever
`possible. Increasingly, stereoisomers are being produced as single chemical entities, such
`as S-naproxyn, which avoid these problems. In contrast, many new protein and polypeptide
`drugs are being introduced that may, in many instances, lack purity. Fmthermore, these
`substances arc often measured by assays that lack specificity.
`An added problem exists following drug administration, namely, the formation of me-
`tabolites. To be of value, an analytic procedure must distinguish between drug and mctab-
`olite(s). Today, most assays have this desired specificity, except for some of those used to
`measure many proteins and polypeptides.
`A potential problem exists vvhen

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