throbber
Page 1 of 44
`
`Astraleneca Exhibit 2087
`Mylan v. Astraleneca
`IPR2015-01340
`
`Page 1 of 44
`
`AstraZeneca Exhibit 2087
`Mylan v. AstraZeneca
`IPR2015-01340
`
`

`
`%
`Burger’s Medicinal
`Chemistry and Drug Discove
`
`Page 2 of 44
`
`

`
`BURGER’S MEDICINAL
`
`CHEMISTRY
`AND DRUG DISCOVERY
`
`Fifth Edition
`
`Volume I: Principles and Practice
`
`Edited by
`
`Manfred E. Wolff
`
`lmm
`San
`
`'
`
`harmaceutics. Inc.
`0, California
`
`A WILEY-INTERSCIENCE PUBLICATION
`
`JOHN WILEY & SONS, Inc., New York - Chichester - Brisbane - Toronto - Singapore
`
`Page 3 of 44
`
`

`
`Notice Concerning Trademark or Patent Rights.
`The listing or discussion in this book of any drug in
`respect to which patent or trademark rights may exist
`shall not be deemed, and is not intended as
`a grant of, or authority to exercise, or an
`infringement of, any right or privilege protected by
`such patent or trademark.
`
`This text is printed on acid—free paper.
`
`Copyright © 1995 by John Wiley & Sons, Inc.
`
`All rights reserved. Published simultaneously in Canada.
`
`Reproduction or translation of any part of this work beyond
`that permitted by Section 107 or 108 of the 1976 United A
`States Copyright Act without the permission of the copyright
`owner is unlawful. Requests for permission or further
`information should be addressed to the Permissions Department,
`John Wiley & Sons, 605 Third Avenue, New York, NY
`10158-0012.
`
`Library of Congress Cataloging in Publication Data:
`Burger, Alfred, 1905-
`[Medicinal chemistry]
`Burger’s medicinal chemistry and drug discovery. -- 5th ed. /
`edited by Manfred E. Wolff.
`p.
`cm.
`“A Wiley-Interscience publication.”
`Contents: v. 1. Principles and practice
`Includes bibliographical references and index.
`ISBN 0-471-57556-9
`1. Wolff, Manfred E.
`1. Pharmaceutical chemistry.
`III. Title: Medicinal chemistry and drug discovery.
`RS403.B8
`1994
`615'. 19--dc20
`
`II. Title.
`
`94-12687
`
`Printed in the United States of America
`
`10987654321
`
`Page 4 of 44
`
`

`
`CHAPTER FIVE
`
`Drug Absorption, Distribution
`and Elimination
`
`LESLIE Z. BENET and
`BEATRICE Y. T. PEROTFI
`
`Department of Pharmacy, University of California
`San Francisco, California, USA
`
`CONTENTS
`
`114
`
`1 Overview of Product Development Issues,
`2 Defining Drug Absorption, Disposition and
`Elimination Using Pharmacokinetics,
`114
`3 Important Pharmacokinetic Parameters,Used in
`Defining Drug Disposition,
`115
`J
`3.1 Clearance, 116
`3.2 Bioavailability, 119
`3.3 Volume of distribution, 120
`
`4 Important Pharmacokinetic Parameters Used in
`Therapeutics,
`121
`4.1 Dosing rate, 121
`4.2 Dose adjustment, 122
`4.3 Dosing interval, 122
`4.4 Loading dose, 125
`5 How is Phannacokinetics Used in the
`Development Process?,
`1216
`5.1 Pre-clinical development, 126
`5.2 Pharmacokinetics in initial phase I human
`studies, 126
`5.3 Pharmacokinetics in phases II and III
`studies, 127
`5.4 Late phase I pharmacokinetic studies, 127
`5.5 Pharmacokinetics in phase IV, 128
`6. Applications in Areas Other Than Drug
`Discovery,
`128
`
`Burger’s Medicinal Chemistry and Drug Discovery,
`Fifth Edition, Volume 1: Principles and Practice,
`Edited by Manfred E. Wolff.
`ISBN 0-471-57556-9 © 1995 John Wiley & Sons, Inc.
`
`Page 5 of 44
`
`

`
`114
`
`Drug Absorption, Distribution and Elimination
`
`1 OVERVIEW OF PRODUCT
`DEVELOPMENT ISSUES
`
`Although many compounds come through
`drug discovery every year, only a rare few
`can actually make their way through prod-
`uct development and pass the scrutiny of
`the United States Food and Drug Adminis-
`tration (U.S. FDA). With some ups and
`downs in marketing, these newly approved
`pharmaceutical products eventually land on
`the shelves in pharmacies where they are
`dispensed to patients upon presentation of
`the proper prescriptions.
`Why are so many compounds “killed” in
`the product development stage? Since the
`bottom line question in drug development
`asks, “Is this drug safe and effective in the
`human body?” many of these compounds
`simply do not prove to be safe and effective
`in the human body.
`In order to answer this safety and effica-
`cy question satisfactorily, development sci-
`entists must address
`issues
`from many
`areas. Some of the common questions are:
`What route of administration will be used
`
`to deliver the drug? How much of the drug
`gets into the systemic circulation and to the
`site of action? Where in the body will the
`drug distribute? How long does the drug
`remain intact in the body? How does the
`drug get eliminated from the body? Is the
`administered compound metabolized into
`other entities? Is the pharmacological effect
`derived from the administered compound,
`or/ and from metabolites? IIow much drug
`in the body is needed to elicit the pharma-
`cological effect? Can patients develop aller-
`gic or toxic reactions toward the adminis-
`tered compound or its metabolites? How
`much drug in the body is needed to initiate
`such adverse reactions? What is the mecha-
`
`nism for such allergic reactions? Are these
`reactions reversible? Is
`the drug or
`the
`metabolites carcinogenic,
`teratogenic, or
`mutagenic? If so, what
`is the underlying
`mechanism?
`
`2 DEFINING DRUG ABSORPTION,
`DISPOSITION AND ELIMINATION
`USING PHARMACOKINETICS
`
`in product development
`Initial progress
`rests heavily on a thorough understanding
`of the absorption, distribution and elimina-
`tion characteristics of a drug. Absorption,
`distribution
`and
`elimination
`processes
`begin when a dose is administered, and
`may govern the appearance of any thera-
`peutie effect (Fig. 5.1). Pharmacokinetics is
`used to quantitate these processes. Apart
`from its usefulness in explaining the safety
`and toxicity assessment data which will be
`discussed later in the chapter, pharmaco-
`kinetic analysis is used primarily to design
`appropriate dosing regimens, and also to
`quantitatively define drug disposition
`The term pharmacokinetics can be de-
`fined as what the body does to the drug.
`The parallel term, pharmacodynamics, can
`be defined as what the drug does to the
`body. A fundamental hypothesis of phar-
`macokinetics is
`that a relationship exists
`between a pharmacologic or toxic effect of
`a drug and the concentration of the drug in
`a readily accessible site of the body (e.g.,
`blood). This hypothesis has been docu-
`mented for many drugs (3, 4), although for
`some drugs no clear relationship has been
`found between pharmacologic effect and
`plasma or blood concentrations.
`This chapter will focus on the conceptual
`approach to pharmacokinetics, its use as a
`tool
`in therapeutics and in defining drug
`disposition. The more common mathema-
`tical modeling-exponential
`equation-data
`analysis approach to pharmacokinetics will
`not be discussed here. These mathematical
`
`techniques are necessary so as to be able to
`determine the important pharmacokinetic
`parameters that are to be discussed; how-
`ever, for medicinal chemists who need to
`interact with other pharmaceutical scien-
`tists in the drug development process, a
`conceptual approach to pharmacokinetics
`
`Page 6 of 44
`
`

`
`3 Important Pharmacokinetic Parameters Used in Defining Drug Disposition
`
`Dose of drug
`administered
`
`"' " BIOAVAILAHILITY
`
`Drug concentration
`in systemic circulation
`
`Drug metabolized
`M exuemd
`
`Pharmacokrnellcs
`
`r
`CLEARANCE
`
`Ar
`
`Il
`
`Drug in tissues
`oi distribution
`
`DISTRIBUTION
`
`Pharmacodynamics
`
`Drug concentration
`at site of action
`
`Pharmacologic effect
`l
`Clinical response
`
`\E
`
`fficacy
`
`/
`Toxicity
`
`\ /
`Fig. 5.1 A schematic representation of the doseresponse relationship of a drug (1). Reproduced courtesy of the
`author and Appleton & Lange.
`
`_I
`
`will be more useful. Nevertheless, to even
`gain a conceptual understanding of phar-
`macokinetics and its application to drug
`development, some basic simple equations,
`as discussed in this chapter, are necessary.
`This approach should allow the medicinal
`chemist to appreciate why modification of a
`drug molecule may -lead
`to important
`changes in drug disposition.
`
`3 IMPORTANT PHARMACOKINETIC
`PARAMETERS USED IN DEFINING
`DRUG DISPOSITION
`
`Table 5.1 Ten Critical Pharmacokinetic and
`Pharmacodynamic Parameters in Drug
`Development in Order of Importance"
`
`Clearance
`Effective concentration range
`Extent of availability
`Fraction of the available dose excreted
`unchanged
`Blo0d/ Plasma concentration ratio
`Half—life
`Toxic concentrations
`Extent of protein binding
`Volume of distribution
`Rate of availability
`
`Among the many pharmacokinetic parame-
`ters (Table 5.1)
`that can be determined
`
`“Ref. 5. Reproduced courtesy of L.Z. Benet and
`Plenum Press.
`
`Page 7 of 44
`
`

`
`116
`
`when delining a new drug substance, clear-
`ance. (CL) is the most important for defin-
`ing drug disposition. Bioavailability (F)
`and volume of distribution (V) are also of
`primary in1poi'tant:e when pharm:u:okin-
`ctics are used to deline drug disposition.
`
`3.1 Clearance
`
`the
`the measure of
`is
`Clearance (CL)
`ability of the body to eliminate a drug.
`Initially, clearance will be looked at from a
`physiological point of view. Figure 5.2
`depicts how a drug is removed from the
`systemic circulation when it passes through
`an eliminating organ. The rate of presenta-
`tion of a drug to a drug elimination organ is
`the product of organ blood [low (Q) and
`the concentration of drug in the arterial
`blood entering the organ (CA). The rate of
`exit of a drug from the drug eliminating
`organ is the product of the organ blood
`flow (Q) and the concentration of the drug
`in the venous blood leaving the organ (CV).
`By mass balance, the rate of elimination (or
`extraction) of a drug by :1 drug eliminating
`organ is the difference between the rate of
`presentation and the rate of exit.
`
`Drug Absorption, Distribution and Elimination
`
`Rate of presentation = Q - CA
`
`Rate of exit = Q - CV
`
`(5.1)
`
`(5.2)
`
`Rate of elimination = Q - CA — Q - CV
`
`= Q ' (CA — CV)
`
`(5.3)
`
`Extraction ratio (ER) of an organ can be
`defined as the ratio of the rate of elimina-
`tion to the rate of presentation.
`
`Extraction ratio = (Rate of elimination/
`
`Rate of presentation)
`
`(5.4a)
`
`ER:AQ'(CA'CV)/QICA
`
`ER = (CA - CV)/CA
`
`(5.4b)
`
`The maximum possible extraction ratio
`is 1.0 when no drug emerges into the
`venous blood upon presentation to the
`eliminating organ (i.e., CV = 0). The low-
`est possible extraction ratio is zero when all
`the drug passing through the potential drug
`eliminating organ appears in the venous
`blood (i.e., CV = CA). Drugs with an ex-
`traction ratio of more than 0.7 are by
`convention considered as high extraction
`ratio drugs, while those with an extraction
`ratio of less than 0.3 are considered as low
`extraction ratio drugs.
`‘
`
`-u—|ntravenous
`dose
`
`Remainder
`of the body
`
`Fig. 5.2 A schematic representation of the concentration—clearance relationship.
`
`Page 8 of 44
`
`

`
`117
`3 Important Pharmacokinetic Parameters Used in Defining Drug Disposition
`occurring in the liver, the kidney. and other
`The product of organ blood [low and
`organs. The total systemic clearance will be
`extraction ratio of an organ represents a
`the sum of the individual organ elezirauccs.
`rate at which a certain volume ol' blood is
`completely cleared of a drug. This expres-
`sion defines the organ clearance (CTLUWH)
`of a drug.
`
`CLtotal : CLhepatic + CLrenal + Cl‘other
`
`(5.10)
`
`CL
`
`organ
`
`=Q-ER=Q-(CA—C\,)/CA
`
`(5.5)
`
`From equations 5.3 and 5.5, one can see
`that clearance is a proportionality constant
`between rate of elimination and the arterial
`drug concentration.
`At steady ‘state, by definition, rate in
`equals rate out. Rate in is given by the
`dosing rate (Dose/1-, i.e., dose divided by
`dosing interval 1-) multiplied by the drug
`availability (F), whereas rate out is the rate
`of elimination (clearance multiplied by the
`systemic concentration (C)).
`Rate In = Rate Out
`
`(5.6)
`
`F-Dose/~r=CL-C
`
`(5.7)
`
`When equation 5.7 is integrated over time
`from zero to infinity, equation 5.8 results.
`
`F-Dose = CL-AUC
`
`(5.8)
`
`where AUC is the area under the con-
`centration time curve, and F is the fraction
`of dose available to the systemic circulation
`(see Bioavailability in section 3.2).
`the
`Clearance may be calculated as
`available dose divided by the area under
`the systemic drug concentration curve.
`
`-
`
`CL = F-Dose/AUC
`
`(5.9)
`
`The maximum value for organ clearance is
`limited by the blood flow to the organ, i.e.,
`extraction ratio is 1. The average blood
`flow to the kidneys and the liver is approxi-
`mately 66 L/ h and 81 L/h,
`respectively.
`Clearance can occur in many sites in the
`body and is generally additive. Elimination
`of a drug may occur as a result of processes
`
`Among the many organs that are ca-
`pable of eliminating drugs,
`the liver,
`in
`general, has the highest metabolic capa-
`bility. Drug molecules in blood are bound
`to blood cells and plasma proteins such as
`albumin and alpha‘-acid glycoprotein. Yet
`only unbound drug molecules can pass
`through hepatic membranes into hepato-
`cytes where
`they are metabolized by
`hepatic enzymes or transported into the
`bile. Thus in order to be eliminated, drug
`molecules must partition out of the red
`blood cells, and dissociate from plasma
`proteins to become unbound or free drug
`molecules. Because unbound drug mole-
`cules are free to partition into and out of
`blood cells and hepatocyles,
`there is an
`equilibrium of free drug concentration be-
`tween the blood cells, the plasma, and the
`hepatocytes. The ratio between unbound
`drug concentration and total drug concen-
`tration constitutes the fraction unbound
`(f\l)'
`Fraction unbound = Unbound drug con-
`centration/ Total drug concentration
`
`f.=C;/C
`
`(5.11)
`
`Since an equilibrium exists between the
`unbound drug molecules in the blood cells
`and the plasma,
`the rate of elimination of
`unbound drugs is the same in the whole
`blood as
`in the plasma at steady state.
`Thus,
`
`(5.12)
`CLP -.cp : CL, - C, = CL“ - cu
`where the subscripts p, b, and u refer to
`plasma, blood and unbound, respectively.
`From the material ]’Jl‘I3SC1llBl'_l so far, one
`may intuitively imagine that hepatic drug
`clearance will be
`inlhienced by hepatic
`
`Page 9 of 44
`
`

`
`118
`
`blood flow, fraction unbound, and intrinsic
`clearance; that is, the intrinsic ability of the
`organ to clear unbound drug. The simplest
`model
`that describes hepatic clearance in
`terms of these pliysiolngic parameters is the
`well-stirred mode] ('6'). Assuming instanta-
`neous and complete mixing, the well-stirred
`model states that hepatic clearance (with
`respect to blood concentration) is
`CLhep = Qhep . fu I CLint/(Qhep +fu . CLint)
`(5.13)
`
`Note that f“ is calculated from unbound
`and total concentration in whole blood.
`Equation 5.5 advises that hepatic clear-
`ance is the product of hepatic blood ilow
`and hepatic extraction ratio. 'l'herefore_. as
`shown in equation 5.13, hepatic extraction
`ratio is
`,
`
`ERhep =f.. ' CLim/(Qiep +fu ' CLint)
`(5.14)
`
`Exainining equations 5.13 and 5.14, one
`finds that for drugs with a high extraction
`ratio (i.e., ER approaches 1.0), f“ -CLIM is
`much greater
`than QM, and clearance
`approaches QM. In other words. the clear-
`ance for a high extraction ratio drug,
`imi-
`pramine for example,
`is perfusion rate
`limited. For drugs with a low extraction
`ratio (i.e., ER approaches zero), QM,
`is
`much greater than fu - (TIMI, and clearance
`is approximated by fu - CLIM. An example
`of a low extraction ratio drug is acetamino-
`phen.
`The intrinsic ability of an organ to clear
`a drug is directly proportional to the activi-
`ty of the metabolic enzymes in the organ.
`Such metabolic processes. both in vi'.'ro and
`in vivo, are characterized by Michaelis—
`Menten kinetics:
`
`Rate of metabolism = VMAX - C/(KM + C)
`
`(5.15)
`
`the maximum rate at
`in which VMAX,
`which metabolism can proceed, is propor-
`
`Drug Absorption, Distribution and Elimination
`
`tional to the total concentration of enzyme.
`KM is the Michaclis-Menten constant corre-
`sponding to the drug concentration which
`yields 112 of the maximum rate of meta-
`bolism. Dividing both sides of equation
`5.15 by the systemic concentration (C)
`yields:
`
`Rate of metabolism/C = CL,nm,m,i5m
`
`= VMAX/(KM + C)
`
`(5.16)
`
`Since ‘identification of a saturable process
`only occurs for low extraction ratio com-
`pounds (1.e., CL.mm,m“5_“ = -__-(..‘Li,,,),
`the
`relationship
`between
`classical
`enzyme
`kinetics and pharmacokinetics is revealed:
`
`fu . CLint : MAX/(KM +
`
`As VMAX and KM can be obtained from
`in vitro metabolism experiments, develop-
`ment scientists may reasonably predict the
`in vivo clearance parameter of a low or
`intermediate extraction ratio drug from in
`vi'.n'o data. using equations 5.13 and 5.17.
`By using appropriate scaling factors, the in
`viva clearance parameter in humans can be
`approximated from in virm metabolism
`data from other species (7, 8).
`The kidneys are also important drug
`eliminating organs. Renal clearance (CL,)
`is a proportionality term between urinary
`excretion rate and systemic concentration.
`Integrating equation 5.18a over time from
`zero to infinity, renal clearance is the ratio
`of the total amount of drug excreted un-
`changed to the area under the systemic
`concentration curve. The total amount of
`drug excreted unchanged can be measured
`experimentally or can be calculated from
`the close if the fraction of the available dose
`excreted unchanged (fe) is known.
`
`(5.18a)
`CL, = Urinary excretion rate/ C
`= Total amount of drug excreted
`
`unchanged/AUC (5.18b)
`
`=fe - F-Dose/AUC
`
`(5.18c)
`
`Page 10 of 44
`
`

`
`3 Important Pharmacokinetic Parameters Used in Defining Drug Disposition
`
`119
`
`It is obvious above that the conceptual
`approach to clearance requires measure-
`ments
`of
`blood
`concentrations. Yet,
`bioanalytical measurements are often car-
`ried out
`in plasma due to the ease of
`sample handling. However, measuring the
`blood to plasma ratio allows one to convert
`clearance values determined in plasma, to
`their corresponding blood values using
`equation 5.12.
`
`3.2 Bioavailability
`
`The bioavailability of a drug product via
`various routes of administration is defined
`as the fraction of unchanged drug that is
`absorbed intact and then reaches the site of
`action; or the systemic circulation following
`administration by any route. For an in-
`travenous dose of a drug, bioavailability is
`defined as unity. For drug administered by
`other routes of administration, bioavail-
`‘ability is often less than unity. Incomplete
`bioavailability may be due to a number of
`factors
`that
`can
`be
`subdivided
`into
`categories of dosage-form effects, mem-
`brane effects and site of administration
`
`effects. Obviously, the route of administra-
`tion that offers maximum bioavailability is
`the direct
`input at the site of action for
`which the drug is developed. This arrange-
`ment may be difflcult to achieve because
`the site of action is not known for some
`
`disease states, and in other cases, the site
`of action is completely inaccessible even
`4 when the drug is placed into the blood-
`stream. The most commonly used route is
`oral administration. However, orally ad-
`ministered drugs may decompose in the
`fluids of the gastrointestinal lumen, or be
`metabolized as they pass through the gas-
`trolntestinal membrane. In addition, once a
`drug passes into the hepatic portal vein, it
`{nay be cleared by the liver before entering
`Into the general circulation. The loss of
`drug as it passes through drug eliminating
`
`organs for the first time is known as the
`first pass effect.
`The fraction of an oral dose available to
`the systemic circulation considering both
`absorption and the first pass effect, can be
`found by comparing the ratio of AUCs
`following oral and intravenous dosing.
`
`F = Aucm,/A UC,_,_
`
`(5.19)
`
`If an assumption is made that all of a drug
`dose is absorbed through the gastrointesti-
`nal tract intact, and that the only extraction
`takes place at the liver, then the maximum
`bioavailability (Fmx) is
`
`Fm, = 1 — ER
`
`hep
`
`(5.20)
`
`From equations 5.5 and 5.20, one can
`derive the following relationship for Fmax:
`
`Fmax = 1 _ (CLhep/Qhep)
`
`(5.21)
`
`For high extraction ratio drugs, where
`CLhep approaches Qhep, Fm“ will be small.
`For
`low extraction» ratio drugs, Qhep is
`much greater than CLMP, and Fm“ will
`approximate one.
`Recently, bioavailability and clearance
`data obtained from a cross-over study of
`cyclosporine
`kinetics before
`and after
`rifampin dosing,
`revealed a new under-
`standing of drug metabolism and disposi-
`tion of this compound (9). Healthy vol-
`unteers were given cyclosporine, intraven-
`ously and orally, before and after their
`CYP3A (P450 3A) enzymes were induced
`by rifampin. As expected, the blood clear-
`ance of cyclosporine increased from 0.31 to
`0.42 L/h/kg due to the induction of the
`drug’s metabolizing enzymes (i.e., an in-
`crease in VMAX in equation 5.15). There
`was no change in volume of distribution,
`but there was a dramatic decrease in bio-
`availability from 27% to 10% in these
`individuals.
`A decrease in bioavailability is to be
`expected, because cyclosporine undergoes
`some
`first—pass metabolism as
`it goes
`
`Page 11 of 44
`
`

`
`120
`
`through the liver following oral dosing. But
`if one predicts on the basis of pharma-
`cokinetics what
`the maximum bioavail-
`ability (as calculated by Eq. 5.21) would be
`before and after rifampin dosing, the maxi-
`mum bioavailability would decrease from
`77% to 68%. Thus,
`there would be an
`expected cyclosporine bioavailability de-
`crease of approximately 12% just on the
`basis of
`the clearance changes resulting
`from inducing CYP3A enzymes in the liver.
`In fact, there was a bioavailability decrease
`of 60%. Furthermore, bioavailability was
`significantly less than would be predicted at
`maximum. While some of that lower bio-
`availability was due to formulation effects,
`the discrepancy between the theoretical
`maximum bioavailability and the achiev—
`able bioavailability of cyclosporine
`re-
`mained a question.
`However, on the basis of new findings
`during the last several years about the high
`prevalence of CYP3A isoxymcs in the gut,
`significant metabolism of cyclosporine in
`the gut as well as in the liver was specu-
`lated. This hypothesis can consistently ex-
`plain the sigmfzcrmtly lower bioavailability
`than would be predicted even if all of the
`drug could be absorbed into the blood
`stream. This finding, particularly quantifi-
`cation of the magnitude of gut metabolism
`(more than 2/ 3s of the total metabolism for
`an oral dose of cyclosporine occurs in the
`gut), would not have been realized had
`pharmacokinetics not been utilized in the
`analysis of the given data.
`
`3.3 Volume of Distribution
`
`The volume of distribution (V) relates the
`amount of drug in the body to the con-
`centration of drug i11 the blood or plasma,
`depending upon the fluid nteasured. At its
`simplest,
`this
`relationship is defined by
`equation 5.22:
`
`V: Amount of drug in body/C (5.22)
`
`Drug Absorption, Distribution and Elimination
`
`the plasma
`For a normal 7U-kg man,
`volume is 3L. the blood volume is 5.5 L.
`the extracellular linid outside the plasma is
`12L and the total body water is approxi-
`mately 42 L. However. many classical drugs
`exhibit volumes of distribution far in excess
`of these known liuid volumes. The volume
`of distribution for digoxin is about 700 L,
`which is ztpproximately "ill
`times greater
`than the total body volume of a 7t)—kg man.
`This example serves to emphasize that the
`volume of distribution does not represent :1
`real volume. Rather.
`it
`is an apparent
`volume which should be considered as the
`size of the pool of body fluids that would he
`required if the drug were equally distribut-
`ed throughout all portions of the body.
`in
`fact, the relatively hydrophobic digoxin has
`a high apparent volume of distribution
`because it distributes predominantly into
`muscle and adipose tissue,
`leaving only a
`very small amount of drug in the plasma in
`which the concentration of drug is mea-
`sured.
`a
`the distribution of
`At equilibrium,
`drug within the body depends upon binding
`to blood cells, plasma proteins, and tissue
`components. Only the unbound drug is
`capable of entering and leaving the plasma
`and tissue compartments. A memory aid
`for this relationship can be summarized by
`the expression:
`
`V: Vp + V'rw ' fu/fu,T
`
`(5.23)
`
`where VP is the volume of plasma, VTW is
`the aqueous volume outside the plasma, f“
`is the fraction unbound in plasma, and fu‘T
`is the fraction unbound in tissue. Thus, a
`drug that has a high degree of binding to
`plasma proteins (i.e., low fa) will generally
`exhibit
`a
`small volume ol‘ distribut'io_n.
`Unlike plasma protein binding, tissue bind-
`ing of a drug cannot be nteasurerl directly.
`Generally, this parameter is assumed to be
`constant unless indicated otherwise.
`in equation 5.22. the body is considered
`:1 single homogeneous pool of body
`
`as
`
`Page 12 of 44
`
`

`
`4 Important Pharmacokinetic Parameters Used in Therapeutics
`V55 = Dose“ - AUMC/AUC2
`
`121
`(5.24)
`
`fluids as described above for digoxin. For
`most drugs, however, two or three distinct
`pools of distribution space appear to exist.
`This condition results in a time—dependent
`decrease in the measurable blood or plasma
`concentration, which reflccts distribution
`into other body pools independent of the
`body’s ability to eliminate the drug. Figure
`5.3 describes mean serum IFN-oz concen-
`trations after a 40 min intravenous infusion
`as well as after intramuscular and subcuta-
`neous injections of the same dose. Note the
`logarithmic biphasic nature of the mean
`plasma concentration time curve following
`the intravenous infusion. This biphasic na-
`ture represents both the distribution and
`elimination processes.
`Generally, comparisons of volume of
`distribution are made using a parameter
`designated as the volume of distribution at
`steady state (V55), which refleets the sum of
`the volumes of all the pools into which the
`drug may distribute. V55 can be calculated
`from the area under the moment curve
`(AUMC) and the area under the curve
`(AUC) as defined by Benet and Galeazzi
`(11):
`
`AUMC can be calculated from areas
`under a plot of concentration - time vs time.
`Both AUC and AUMC may be calculated
`from the coeilicients and exponents of the
`equations utilized to described the multi-
`eompartnient nature of drug kinetics as
`depicted in Figure 5.3. These concepts will
`be revisited following a discussion of halt‘-
`life.
`
`4 IMPORTANT PHARMACOKINETIC
`PARAMETERS USED IN
`'1‘HERAPEUTICS
`
`In addition to the three parameters, clear-
`ance (CL), bioavailability (F), and volume
`of distribution (V) discussed previously, a
`fourth parameter, half-life (rm),
`is also
`crucial
`in therapeutics. The decreasing
`order of importance of these four parame-
`ters are clearance, bioavailability, half-life
`and volume. Clearance defines the dosing
`rate, bioavailability defines dose adjust-
`ment, and half-life defines the dosing inter-
`val. Volume of distribution defines
`the
`loading dose.
`
`4.1 Dosing Rate
`
`As discussed in equation 5.6 in section 3. I,
`rate of presentation equals rate of exit at
`steady state. Whereas rate of presentation
`is the product of bioavailability and dosing
`rate, rate of exit is the product of clearance
`and average (steady state} concentration.
`By replacing the average concentration
`with the target concentration,
`the dosing
`rate can be computed with known values of
`bioavailability and clearance.
`
`Rate in = Rate out
`
`(5.6)
`
`
`
`Meanplasmaeoncn(pg/mt)
`
`
`
`
`
`I._ _I_
`10
`Time tn)
`
`l"ll£- 5.3 Mean serum lFN—n concentrations after a
`"l"R.|e 3:‘) r. til" l_| dose as an inlravenntts infusion (O)
`III‘ an itttrantttscttliir (X) or subeutilneotts (A) injection
`UU). Reproduced eotlrtcsy of the author and Clin.
`Pharmacol. Ther.
`
`Bioavailability >< Dosing rate = Clearance
`
`>< Average concentration
`
`(5.7a)
`
`F - Dosing rate = CL - Cmge,
`
`(5,7b)
`
`Page 13 of 44
`
`

`
`122
`
`Drug Absorption, Distribution and Elimination
`
`4.2 Dose Adjustment
`
`Table 5.2 Percentages of Dose Lost as a Func-
`tion of Number of Half-Lives
`
`There is often a therapeutic concentration
`range of drug in the blood that is necessary
`to elicit a clinical effect without causing
`drug toxicity. The boundaries of this range
`are set by the minimum effective concen-
`tration (MEC) and the minimum toxic
`concentration (M TC ). For example,
`in
`order to maintain a steady rate of presenta-
`tion of 100 mg/min, one can either ad-
`minister a completely bioavailable i.v. infu-
`sion at 100 mg/min, or a sustained release
`oral dosage form with 50% bioavailability
`at 200 mg/min. As shown in equations 5.7a
`and 5.7b, the actual dosing rate depends on
`the bioavailability of the dosage form. For
`proper dosing adjustment,
`the bioavail-
`ability (F) of a given dosage form stands as
`a must-know parameter in therapeutics.
`
`4.3 Dosing Interval
`
`Half-life (rm) is an extremely useful kinetic
`parameter in terms of therapeutics, since
`this parameter,
`together with therapeutic
`index, helps define the dosing interval at
`which drugs should be administered. By
`definition, half—life is the time required for
`50% of the drug remaining in the body to
`be eliminated. In three and one third half-
`lives, 90% of the dose would have been
`eliminated. Table 5.2 shows the percentage
`of dose lost in different numbers of half-
`lives. If the dosing interval is long relative
`to the half—life,
`large fiuctuations in drug
`concentration will occur. On the other
`hand, if the dosing interval is short relative
`to half—life,
`significant accumulation will
`occur. The half—life parameter also allows
`one to predict drug accumulation within the
`body and
`quantifies
`the
`approach to
`plateau that occurs with multiple dosing
`and constant rates of infusion. Convention-
`ally, three and one third half-lives are used
`as the time required to achieve steady state
`under constant infusion. The concentration
`
`Number of
`Half-Lives
`
`Dose Lost, %
`
`50
`75
`87.5
`90
`93.8
`96.9
`
`level achieved after this time is already
`90% of the steady state concentration, and
`clinically, it is difficult to distinguish a 10%
`difference in concentrations.
`After determining the extent of oral
`drug bioavailability, half—life is probably
`the next most important parameter in terms
`of deciding the appropriateness of a drug
`for further development. Drugs with very
`short half—lives create problems in main-
`taining steady-state concentrations in the
`therapeutic range. Thus, a successful drug
`product with a short half—life will require a
`dosage form which allows a relatively con-
`stant prolonged input. Drugs with a very
`long half—life are favored in terms of effica-
`cy considerations; however, this long half-
`life may be a negative characteristic in
`terms of toxicity considerations. Figure 5.4
`illustrates the importance of half—life in
`defining the dosing interval.
`This figure depicts the relationship be-
`tween the frequency of theophylline dosing
`and the plasma concentration time course
`when a steady-state theophylline plasma
`level of 15 p.g/mL is desired. The smoothly
`rising curve shows the plasma concentra-
`tions achieved with an intravenous infusion
`of 43.2 mg/h to a patient exhibiting an
`average theophylline clearance of 0.69 mL/
`min/ kg. The steady-state theophylline plas-
`ma level achieved is midway within the
`therapeutic concentration range of 10-
`20 ,u.g/mL. The figure also depicts the time
`courses
`for 8-hourly administration of
`
`
`
`
`
`
`
`
`
`
`
`....-.—._-i._.-_.=.-‘sag—".1' ',::;-L“'—--;'4a-3=I'Ii.._4:._-.-.-:~_«.F-;_-—--‘-+‘:-:.._.—r-.:Jo—-£-
`
`Page 14 of 44
`
`

`
`4 Important Pharmacokinetic Parameters Used in Therapeutics
`
`4'0
`
`0.) O
`
`R)O
`
`24~h dosage
`
`8—h dosage
`
`3E\b
`
`N-
`
`e1\—
`:...
`3-e
`3‘
`§“\Q
`2'Q
`'43o
`
`1:eUQ:QQ
`(JoN
`9.:
`
`oE‘
`
`8
`
`16
`
`24
`
`32
`
`40
`Time (h)
`Fig. 5.4 Relationship between frequency of dosing and maximum and minimum plasma concentrations when a
`steady—state plasma level of 15 uglinl.
`is desired. The time course of plasma concentrations for 43.2 mg/h
`intravenous infusion, an Bvltourly 34l.l mg oral dose, and a 24 hourly 1020 mg oral dose are depicted. Reproduced
`courtesy of the author and Appleton S: Lange.
`
`48
`
`56
`
`64
`
`72
`
`80
`
`340 mg and 24-hourly administration of
`1020 mg, assuming that
`these doses are
`administered in an immediate release dos-
`age l’orm. in each case the mean steady-
`state concentration is l5 ugfml... However,
`the peak to trough ratio and the concert-
`trations acliicved with the once daily dosing
`(i.e., 1.020 mg) of a rapidly released formu-
`lation would result in concentrations in the
`toxic range,’ exceeding 20 ,u.gfmL for cer-
`tain periods of time, as well as concen-
`trations expected to not yield eflicaey, i.e.,
`less than 1U p.glmL, for signilicant periods
`during each dosing interval. In contrast, the
`8-hourly dosing, which is approximately
`equivalent
`to the half—lit'e of theophylline.
`shows :1 2-fold range in peak to trough, and
`theophylline levels stay within the thera-
`peutic plasma concentration range.
`Although half-life is a very important
`p{lt':.tlTtClCl‘
`in therapeutics for detining the
`dosing interval,
`half—|il'e can he
`a very
`inislcnding parameter when one is attempt-
`
`in
`ing to use pharmacokinetics as a tool
`defining drug disposition. As depicted in
`equation 5.29, half-life varies as a function
`of the two physiologic-related parameters,
`volume and clearance.
`
`t1,2 = 0.693 ~ V/CL
`
`(5.29)
`
`Half-life has little value as an indicator
`of

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