throbber
w.
`
`
`
`
`
`$
`
`E
`
`
`InnOSharma Exhibit 1022.0001
`
`

`

`
`
`éeerzegeir
`
`{filler
`
` :i'
`3‘
`
`.-
`i‘ilzei 3: ME
`
`1.
`
`Prafessor and Executive Dean, South Carolina College of Pharmacy, The
`University of South Carolina, Columbia, Medical University of South Carolina,
`Charleston, South Carolina
`
`
`
`Prefessor, Department of Clinical and Administrative Pharmacy at the
`University of Georgia College of Pharmacy, Athens, Georgia
`
`
`
`Professor, Department of Clinical and Administrative Pharmacy at the
`University of Georgia College of Pharmacy, Athens, Geergia
`
`Professor and Dean, School of Pharmacy, University of North Carolina at
`Chapel Hill, Chapel Hill, North Carolina
`
` «Eerie Mil
`
`Associate Professor of Clinical Pharmacy Practice, University of Cincinnati
`College of Pharmacy, Cincinnati, Ohio
`
`American Society of Health-System Pharmacists®
`
`InnoPharma Exhibit 1022.0002
`
`

`

`Any correspondence regarding this publication should be sent to the publisher, American
`Society of Health-System Pharmacists, 1272, Wisconsin Avenue, Bethesda, MD 20814, atten-
`tion: Special Publishing.
`
`The information presented herein reflects the opinions of the contributors and advisers. It
`should not be interpreted as an official policy of ASH? or as an endorsement of any product.
`
`Because of ongoing research and improvements in technology, the information and its appli-
`cations contained in this text are constantly evolving and are subject to the professional judg-
`ment and interpretation of the practitioner due to the uniqueness of a clinical situation. The
`editors, contributors, and ASH? have made reasonable efforts to ensure the accuracy and
`appropriateness of the information presented in this document. However, any user of this
`information is advised that the editors, contributors, advisers, and ASHP are not responsible
`for the continued currency of the information, forany errors or omissions, andr’or for any
`consequences arising from the use of the information in the document in any and all practice
`settings. any reader of this document is cautioned that ASH? makes no representation, guar-
`antee, or irra'rranty, express or implied, as to the accuracy and appropriateness of the informer
`tion contained in this document and specifically disclaims any liability to any party for the
`accuracy and?or completeness of the material or for any damages arising out of the use or
`non-use of any of the information contained in this document.
`
`Director; Special Publishing: Jack Bruggeman
`Acquisition Editor: l-lal Pollard
`Senior Editorial Project Manager: Dana Battaglia
`Production: Silverchair Science + Communications, Inc.
`
`[et all. -- 5th ed.
`
`Library of Congress Cataloging-in-Publication Data
`Concepts in clinical pharmacoltinetics .3" Joseph T. DiPiro
`p. ; cm.
`includes bibliographical references and index.
`ISBN 9?8—l—58528-241—8 {alk. paper}
`1. Pharmacoliinetics.
`l. DiPiro, Joseph T. H. American Society of Health—System
`Pharmacists.
`[DNLMz l. Pharmacolrinetics~Programmed Instruction. 2. Pharmaceutical
`Preparations—-administration 8 dosage-Programmed Instruction. QV 18.2 C744 2010]
`RMBOl.5.CSS 2010
`615‘3wd022
`
`2009043338
`
`© 2010, American Society of Health—System Pharmacists, inc. All rights reserved.
`
`No part of this publication may be reproduced or transmitted in any form or by any means,
`electronic or mechanical, including photocopying, microfilming, and recording, or by any
`information storage and retrieval system, Without written, permission from the American
`Society of Health—System Pharmacists.
`
`ASH? is a service mark of the American Society of Health-System Pharmacists, Inc; regis-
`tered in the us. Patent and Trademark Office.
`
`InnoPharma Exhibit 1022.0003
`
`

`

`LESSON
`
`1 I
`
`ntroduction to Pharmacokinetics
`
`and Pharmacodynamics
`
`E OBJECTIVES
`
`After completing Lesson 1, you should be able to:
`
`1. Define and differentiate between pharmacokinetics
`and clinical pharmacokinetics.
`
`5. Describe situations in which routine clinical phar-
`macokinetic monitoring would be advantageous.
`
`2. Define pharmacodynamics and relate it to pharma-
`cokinetics.
`
`3. Describe the concept of the therapeutic concentra-
`tion range.
`
`4.
`
`Identify factors that cause interpatient variability in
`drug disposition and drug response.
`
`6. List the assumptions made about drug distribution
`patterns in both one- and two-compartment models.
`
`log of
`7. Represent graphically the typical natural
`plasma drug concentration versus time curve for a
`one-compartment model
`after
`an
`intravenous
`dose.
`
`Pharmawkinetics is currently defined as the study of the
`time course of drug absorption, distribution, metabo-
`lism, and excretion. Clinical pharmawkinetics is the
`application of pharmacokinetic principles to the safe
`and effective therapeutic management of drugs in an
`individual patient.
`Primary goals of clinical pharmacokinetics include
`enhancing efficacy and decreasing toxicity of a patient's
`drug therapy. The development of strong correlations
`between drug concentrations and their pharmacologic
`responses has enabled clinicians to apply pharmacoki—
`netic principles to actual patient situations.
`A drug's effect is often related to its concentration at
`the site of action, so it would be useful to monitor this
`concentration. Receptor sites of drugs are generally inac-
`cessible to our observations or are widely distributed in
`the body, and therefore direct measurement of drug con-
`centrations at these sites is not practical. For example, the
`
`receptor sites for digoxin are thought to be within the
`myocardium. Obviously we cannot directly sample drug
`concentration in this tissue. However, we can measure
`drug concentration in the blood or plasma, urine, saliva,
`and other easily sampled fluids (Figure 1-1). Kinetic
`h0m0geneity
`describes
`the predictable
`relationship
`between plasma drug concentration and concentration at
`the receptor site where a given drug produces its thera-
`peutic effect (Figure 1-2). Changes in the plasma drug
`concentration reflect changes in drug concentrations at
`the receptor site, as well as in other tissues. As the con-
`centration of drug in plasma increases, the concentration
`of drug in most tissues will increase proportionally.
`Similarly,
`if the plasma concentration of a drug is
`decreasing,
`the concentration in tissues will
`also
`decrease. Figure 1-3 is a simplified plot of the drug con-
`centration versus time profile after an intravenous drug
`dose and illustrates this concept.
`
`InnoPharma Exhibit 1022.0004
`
`

`

`2
`
`Concepts in Clinical Pharmacokinetics
`
`DTUQ in
`Blood
`
`I
`I
`
`Drug in
`Tissue
`
`Sample Removed for
`Drug Concentration
`Determination
`
`
`
`Kidney
`Plasma
`Receptor
`
`Time—>
`
`:o
`
`.g
`g’:
`*5no:
`
`0U
`
`FIGURE 1-3.
`
`Drug concentration versus time.
`
`BASIC PHARMACODYNAMIC CONCEPTS
`
`Pharmacodynamics refers to the relationship between
`drug concentration at the site of action and the resulting
`effect, including the time course and intensity of thera-
`peutic and adverse effects. The effect of a drug present
`at the site of action is determined by that drugs binding
`with a receptor. Receptors may be present on neurons in
`the central nervous system (i.e., opiate receptors) to
`depress pain sensation, on cardiac muscle to affect the
`intensity of contraction, or even within bacteria to dis-
`rupt maintenance of the bacterial cell wall.
`For most drugs, the concentration at the site of the
`receptor determines the intensity of a drug's effect (Fig-
`ure 1-4). However, other factors affect drug response as
`well. Density of receptors on the cell surface, the mech-
`anism by which a signal is transmitted into the cell by
`second messengers (substances within the cell), or regu-
`latory factors that control gene translation and protein
`production may influence drug effect. This multilevel
`
`FIGURE 1-1.
`
`Blood is the fluid most often sampled for drug concentration
`determination.
`
`The property of kinetic homogeneity is important
`for the assumptions made in clinical pharmacokinet-
`ics. It is the foundation on which all therapeutic and
`toxic plasma drug concentrations are established. That
`is, when studying concentrations of a drug in plasma,
`we assume that these plasma concentrations directly
`relate to concentrations in tissues where the disease
`
`process is to be modified by the drug (e.g., the central
`nervous system in Parkinson's disease or bone in
`osteomyelitis). This assumption, however, may not be
`true for all drugs.
`
`[I CLINICAL CORRELATE
`
`Drugs concentrate in some tissues because of physi-
`cal or chemical properties. Examples include digoxin,
`which concentrates in the myocardium, and lipid-
`soluble drugs, such as benzodiazepines, which con—
`centrate in fat.
`
`:
`mo
`
`DJ::
`mm
`EuW:
`'200.0
`
`Concentration of Drug in
`Tissues
`
`
`
`l
`Cell Signal
`(2nd Messenger)
`
`Altered
`Receptor
`Expression
`\ Gene Regulation
`i
`Regulation of Protein Production
`
`\l Cellular
`Event
`
`FIGURE 1-2.
`
`Relationship of plasma to tissue drug concentrations.
`
`FIGURE 1-4.
`
`Relationship of drug concentration to drug effect at the recep-
`tor site.
`
`lnnoPharma Exhibit 1022.0005
`
`

`

`Lesson 1: Introduction to Pharmacokinetics and Pharmacodynamics
`
`3
`
`Maximum Effect (Emax
`
`
`20
`
`Plasma Drug Concentration (mg/L)
`
`(log scale)
`
`10
`
`100
`
`Plasma Drug Concentration (mg/L)
`
`FIGURE 1-5.
`
`Relationship of drug concentration at the receptor site to
`effect (as a percentage of maximal effect).
`
`regulation results in variation of sensitivity to drug
`effect from one individual to another and also deter-
`
`mines enhancement of or tolerance to drug effects.
`In the simplest examples of drug effect, there is a rela-
`tionship between the concentration of drug at the receptor
`site and the pharmacologic effect. If enough concentra-
`tions are tested, a maximum effect (Ema can be deter-
`mined (Figure 1-5). When the logarithm of concentration
`is plotted versus effect (Figure 1-5), one can see that there
`is a concentration below which no effect is observed and a
`
`concentration above which no greater effect is achieved.
`One way of comparing drug potency is by the concen-
`tration at which 50% of the maximum effect is achieved.
`
`This is referred to as the 50% eflecn’ve concentration or ECSO.
`When two drugs are tested in the same individual, the
`drug with a lower EC50 would be considered more potent.
`This means that a lesser amount of a more potent drug is
`needed to achieve the same effect as a less potent drug.
`The EC50 does not, however, indicate other important
`determinants of drug response, such as the duration of
`effect. Duration of effect is determined by a complex set
`of factors, including the time that a drug is engaged on
`the receptor as well as intracellular signaling and gene
`regulation.
`
`Later Doses
`AFirst Dose
`
`
`
`
`
`
`Plasma Drug Concentration (log scale)
`
`FIGURE 1-6.
`
`Demonstration of tolerance to drug effect with repeated dosing.
`
`For some drugs, the effectiveness can decrease with
`continued use. This is referred to as tolerance. Tolerance
`
`may be caused by pharmacokinetic factors, such as
`increased drug metabolism, that decrease the concen-
`trations achieved with a given dose. There can also be
`pharmacodynamic tolerance, which occurs when the
`same concentration at the receptor site results in a
`reduced effect with repeated exposure. An example of
`drug tolerance is the use of opiates in the management
`of chronic pain.
`It
`is not uncommon to find these
`patients requiring increased doses of the opiate over
`time. Tolerance can be described in terms of the dose—
`
`response curve, as shown in Figure 1-6.
`To assess the effect that a drug regimen is likely to
`have,
`the clinician should consider pharmacokinetic
`and pharmacodynamic factors. Both are important in
`determining a drug's effect.
`
`I] CLINICAL CORRELATE
`
`Tolerance can occur with many commonly used drugs.
`One example is the hemodynamic tolerance that occurs
`with continued use of organic nitrates, such as nitroglyc—
`erin. For this drug, tolerance can be reversed by inter-
`spersing drug-free intervals with chronic drug use.
`
`[I CLINICAL CORRELATE
`
`One way to compare potency of two drugs that are in
`the same pharmacologic class is to compare EC5O. The
`drug with a lower EC5O is considered more potent.
`
`InnoPharma Exhibit 1022.0006
`
`

`

`4
`
`Concepts in Clinical Pharmacokinetics
`
`Response
`
`I/
`
`Toxicity /’
`
`Drug Concentration (mg/LI
`
`01O
`
`"o‘
`35
`>
`.t
`Ec;
`.0o._
`Q
`
`IO
`
`FIGURE 1-7.
`
`Relationship between drug concentration and drug effects for
`a hypothetical drug. Source: Adapted with permission from
`Evans WE, editor. General principles of applied pharmaco-
`kinetics. In: Applied Pharmacokinetics, 3rd ed. Vancouver, WA:
`Applied Therapeutics; 1992. pp.1—3.
`
`THERAPEUTIC DRUG MONITORING
`
`Therapeutic drug monitoring is defined as the use of
`assay procedures for determination of drug concentra-
`tions in plasma, and the interpretation and application
`of the resulting concentration data to develop safe and
`effective drug regimens. If performed properly, this pro-
`cess allows for the achievement of therapeutic concen-
`trations of a drug more rapidly and safely than can be
`attained with empiric dose changes. Together with
`observations of the drug's clinical effects, it should pro-
`vide the safest approach to optimal drug therapy.
`The usefulness of plasma drug concentration data is
`based on the concept that pharmacologic response is
`closely related to drug concentration at the site of action.
`For certain drugs, studies in patients have provided infor-
`mation on the plasma concentration range that is safe
`and effective in treating specific diseases—the therapeu-
`tic range (Figure 1-7). Within this therapeutic range, the
`desired effects of the drug are observed. Below it, there is
`greater probability that the therapeutic benefits are not
`realized; above it, toxic effects may occur.
`No absolute boundaries divide subtherapeutic, thera-
`peutic, and toxic drug concentrations. A gray area usu-
`ally exists for most drugs in which these concentrations
`overlap due to variability in individual patient response.
`Numerous pharmacokinetic characteristics of a drug
`may result in variability in the plasma concentration
`achieved with a given dose when administered to vari-
`ous patients (Figure 1-8). This interpatient variability is
`primarily attributed to one or more of the following:
`
`FIGURE 1-8.
`
`Example of variability in plasma drug concentration among
`subjects given the same drug dose.
`
`0 Differences in an individual's ability to metabolize
`and eliminate the drug (e. g., genetics)
`0 Disease states (renal or hepatic insufficiency) or
`physiologic states (e.g., extremes of age, obesity) that
`alter drug absorption, distribution, or elimination
`0 Drug interactions
`
`Therapeutic monitoring using drug concentration data
`is valuable when:
`
`1. A good correlation exists between the pharmaco-
`logic response and plasma concentration. Over at
`least a limited concentration range, the intensity of
`pharmacologic effects should increase with plasma
`concentration. This relationship allows us to pre-
`dict pharmacologic effects with changing plasma
`drug concentrations (Figure 1-9).
`2. Wide intersubject variation in plasma drug concen-
`trations results from a given dose.
`
`'.T
`
`7.
`2
`
`Plasma Drug ('nrii wili'w‘iian
`
`0 Variations in drug absorption
`0 Variations in drug distribution
`
`When pharmacologic effects relate to plasma drug concentra-
`tions, the latter can be used to predict the former.
`
`FIGURE 1-9.
`
`InnoPharma Exhibit 1022.000?
`
`OJ
`
`Concentration
`
`D1
`:5h
`
`Da
`
`s Ew 2mHma
`
`)
`.r:
`.9
`
`I
`
`C110
`
`.L—LMMU10
`
`C 0
`
`1
`
`

`

`Lesson 1: Introduction to Pharmacokinetics and Pharmacoclynamics
`
`5
`
`3. The drug has a narrow therapeutic index (i.e., the
`therapeutic concentration is close to the toxic
`concentration).
`4. The drugs desired pharmacologic effects cannot be
`assessed readily by other simple means (e.g., blood
`pressure measurement for antihypertensives).
`
`The value of therapeutic drug monitoring is limited
`in situations in which:
`
`1. There is no well-defined therapeutic plasma con-
`centration range.
`2. The formation of pharmacologically active metabo-
`lites of a drug complicates the application of plasma
`drug concentration data to clinical effect unless
`metabolite concentrations are also considered.
`
`3. Toxic effects may occur at unexpectedly low drug
`concentrations as well as at high concentrations.
`4. There are no significant consequences associated
`with too high or too low levels.
`
`Theophylline is an excellent example of a drug in
`which significant interpatient variability in pharmacoki—
`netic properties exists. This is important from a clinical
`
`..
`
`Co Eg
`
`?._N
`“3“a:we
`area:m
`o1:
`=
`
`0 O
`
`a O
`
`N O
`
`EI!
`E0a
`>"in
`IL
`
`Z.
`
`_ZI
`
`“IL
`
`U>OKA Eo“
`
`JNI<280Z
`
`THEOPHYLLINE CONCENTRATION (mg/liter)
`
`10
`
`20
`
`30
`
`FIGURE 1-10.
`
`Relationship between plasma theophylline concentration and
`change in forced expiratory volume (FEV) in asthmatic patients.
`Source: Reproduced with permission from Mitenko PA, Ogilvie
`Rl. Rational intravenous doses of theophylline. N Engl J Med
`1973;289:600—3. Copyright 1973, Massachusetts Medical
`Society.
`
`
`
`standpoint as subtle changes in serum concentrations
`may result in marked changes in drug response. Figure
`1-10 shows the relationship between theophylline con-
`centration (x—axis, on a logarithmic scale) and its
`pharmacologic effect, (changes in pulmonary function
`[y-axisD. This figure illustrates that as the concentration
`of theophylline increases, so does the intensity of the
`response for some patients. Wide interpatient variability
`is also shown.
`
`Figure 1-11 outlines the process clinicians may
`choose to follow in making drug dosing decisions by
`using therapeutic drug monitoring. Figure 1-12 shows
`the relationship of pharmacokinetic and pharmacody-
`namic factors.
`
`Examples of therapeutic ranges for commonly used
`drugs are shown in Table 1-1. As can be seen in this
`table, most drug concentrations are expressed as a unit
`of mass per volume.
`
`[l CLINICAL CORRELATE
`
`A drug’s effect may also be determined by the
`amount of time that the drug, is present at the site of
`action. An example is with beta-lactam antimicrobials.
`The rate of bacterial killing by beta-lactams (the bac-
`terial cell would be considered the site of action) is
`usually determined by the length of time that the
`drug concentration remains ab0ve the minimal con-
`centration that inhibits bacterial growth.
`
`A diagnosis is made
`
`i
`A drug is selected
`l
`Dosage schedule is
`designed to reach a
`target plasma
`concentration
`
`i
`Drug is administered
`
`/ \
`
`Patient assessments
`are pertormed
`
`Drug concentrations
`are determined
`
`\/
`
`A pharmacokinetic
`model is applied and
`clinical judgment is
`used
`
`FIGURE 1-11.
`
`Process for reaching dosage decisions with therapeutic drug
`monitoring.
`
`InnoPharma Exhibit 1022.0008
`
`

`

`6
`
`Concepts in Clinical Pharmacokinetics
`
`FIGURE 1-12.
`
`Relationship of pharmacokinetics and
`pharmacodynamics and factors that
`affect each.
`
`Pharmacokinatics
`
`Pharmacodynamlcs
`
`
`
`- (.umpliant U
`- liming: .mcl rnmlimiinn (Irmrs
`-
`.*\|J~‘.IJI|i:iuz’i
`-
`iissuin am] hurli- iluirl
`mass and walLimU
`- Drug; inivmi llilll‘,‘
`-
`||ii'|'|ii1.ilinn
`» Drug milmimlism
`
`A Drug rm cpinr siiilus
`- (il‘m‘llt.
`Int hm
`» Iliint mm.” [inns
`lilli'l'i'i‘lit'l’
`
`PHARMACOKINETIC MODELS
`
`The handling of a drug by the body can be very complex,
`as several processes (such as absorption, distribution,
`metabolism, and elimination) work to alter drug concen-
`trations in tissues and fluids. Simplifications of body pro-
`cesses are necessary to predict a drug's behavior in the
`body. One way to make these simplifications is to apply
`mathematical principles to the various processes.
`To apply mathematical principles, a model of the
`body must be selected. A basic type of model used in
`pharmacokinetics is the compartmental model. Com-
`partmental models are categorized by the number of
`
`TABLE 1-1.
`
`Therapeutic Ranges for Commonly Used Drugs
`
`Drug
`
`Digoxin
`
`Lidocaine
`
`Lithium
`
`Phenobarbital
`
`Phenytoin
`
`Quinidine
`
`Cyclosporin
`
`Valproic acid
`
`Range
`
`0.5—2.0 ng/mL
`
`1.5—5.0 mg/L
`
`0.6—1.4 mEq/L
`
`15—40 mg/L
`
`10—20 mg/L
`
`2—5 mg/L
`
`150—400 ng/mL
`
`50—100 mg/L
`
`Carbamazepine
`
`4—12 mcg/mL
`
`Ethosuxamide
`
`40—100 mg/L
`
`Primidone
`
`5—12 mg/L
`
`Source: Adapted with permission from Bauer LA. Clinical phar-
`macokinetics and pharmacodynamics. In: DiPiro JT, Talbert RL,
`Yee GC, et al., editors. Pharmacotherapy: a Pathophysiologic
`Approach, 7th ed. New York: McGraw-Hill; 2008. p. 10.
`
`compartments needed to describe the drug's behavior in
`the body. There are one-compartment,
`two-compart-
`ment, and multicompartment models. The compart-
`ments do not represent a specific tissue or fluid but may
`represent a group of similar tissues or fluids. These
`models can be used to predict the time course of drug
`concentrations in the body (Figure 1-13).
`Compartmental models are termed deterministic
`because the observed drug concentrations determine the
`type of compartmental model required to describe the
`pharmacokinetics of the drug. This concept will become
`evident when we examine one- and two-compartment
`models.
`
`To construct a compartmental model as a representa-
`tion of the body, simplifications of body structures are
`made. Organs and tissues in which drug distribution is
`similar are grouped into one compartment. For example,
`distribution into adipose tissue differs from distribution
`into renal tissue for most drugs. Therefore, these tissues
`may be in different compartments. The highly perfiised
`organs (e.g., heart, liver, and kidneys) often have similar
`drug distribution patterns, so these areas may be consid-
`ered as one compartment. The compartment
`that
`includes blood (plasma), heart, lungs, liver, and kidneys is
`usually referred to as the central compartment or the
`highly blood—perfused compartment (Figure 1-14). The
`other compartment that includes fat tissue, muscle tissue,
`
`Drug
`Dose + Compartment
`
`
`
`l
`
`Elimination
`
`FIGURE 1-13.
`
`Simple compartmental model.
`
`InnoPharma Exhibit 1022.0009
`
`

`

`Lesson 1: Introduction to Pharmacokinetics and Pharmacoclynamics
`
`7
`
`Central
`Compartment
`
`Heart
`
`Liver
`
`Lungs
`
`Kidney
`
`Blood
`
`
`
`Examples of
`Peripheral
`Compartments
`
`Fat Tissue
`
`Muscle
`Tissue
`
`Cerebrospinal
`Fluid
`
`FIGURE 1-14.
`
`Typical organ groups for central and peripheral compartments.
`
`and cerebrospinal fluid is the peripheral compartment,
`which is less well perfused than the central compartment.
`Another simplification of body processes concerns
`the expression of changes in the amount of drug in the
`body over time. These changes with time are known as
`rates. The elimination rate describes the change in the
`amount of drug in the body due to drug elimination over
`time. Most pharmacokinetic models assume that elimi-
`nation does not change over time.
`The value of any model is determined by how well it
`predicts drug concentrations in fluids and tissues. Gener-
`ally, it is best to use the simplest model that accurately
`predicts changes in drug concentrations over time. If a
`one-compartment model is sufficient to predict plasma
`drug concentrations (and those concentrations are of most
`interest to us), then a more complex (two-compartment or
`more) model is not needed. However, more complex mod-
`els are often required to predict tissue drug concentrations.
`
`[l CLINICAL CORRELATE
`
`Drugs that do not extensively distribute into extravascu-
`lar tissues, such as aminoglycosides, are generally well
`described by one-compartment models. Extent of dis-
`tribution is partly determined by the chemistry of the
`agents. Aminoglycosides are polar molecules, so their
`distribution is
`limited primarily to extracellular water.
`Drugs extensively distributed in tissue (such as lipophilic
`drugs like the benzodiazepines) or that have extensive
`intracellular uptake may be better described by the
`more complex models.
`
`COM PARTMENTAL MODELS
`
`K
`
`rate constant
`
`Where: X0: Dose of drug
`X1 = Amount of drug
`in body
`K = Elimination
`
`FIGURE 1-15.
`
`One-compartment model.
`
`ment is represented by an enclosed square or rectangle,
`and rates of drug transfer are represented by straight
`arrows (Figure 1-15). The arrow pointing into the box
`simply indicates that drug is put into that compartment.
`And the arrow pointing out of the box indicates that
`drug is leaving the compartment.
`This model is the simplest because there is only one
`compartment. All body tissues and fluids are considered
`a part of this compartment. Furthermore, it is assumed
`that after a dose of drug is administered, it distributes
`instantaneously to all body areas. Common abbrevia-
`tions are shown in Figure 1-15.
`Some drugs do not distribute instantaneously to all
`parts of the body, however, even after intravenous
`bolus administration. Intravenous bolus dosing means
`administering a dose of drug over a very short time
`period. A common distribution pattern is for the drug
`to distribute rapidly in the bloodstream and to the
`highly perfused organs, such as the liver and kidneys.
`Then, at a slower rate, the drug distributes to other
`body tissues. This pattern of drug distribution may be
`represented by a two-compartment model. Drug moves
`back and forth between these compartments to main-
`tain equilibrium (Figure 1-16).
`Figure 1-17 simplifies the difference between one-
`and two-compartment models. Again, the one-compart-
`ment model assumes that the drug is distributed to tissues
`very rapidly after intravenous administration.
`
`Intravenous
`Administration
`
`Elimination
`
`
`
`Peripheral
`
`is the most frequently
`The one-compartment model
`used model
`in clinical practice.
`In structuring the
`model, a visual representation is helpful. The compart-
`
`FIGURE 1-16.
`
`Compartmental model representing transfer of drug to and
`from central and peripheral compartments.
`
`InnoPharma Exhibit 1022.0010
`
`

`

`8
`
`Concepts in Clinical Pharmacokinetics
`
`FIGURE 1-17.
`
`Drug distribution in one- and two-compartment
`models.
`
`One-compartment model
`belore administration
`
`One-compartment model
`immediately alter
`administration
`
`equilibrium
`
`Two-compartment model
`belore administration
`
`Two-compartment model
`immediately after
`administration
`
`Two-compartment model
`after distributive
`
`The two-compartment model can be represented as
`in Figure 1-18, where:
`
`X0 = dose of drug
`X1 = amount of drug in central compartment
`X2 = amount of drug in peripheral compartment
`K = elimination rate constant of drug from central
`compartment to outside the body
`K12 = elimination rate constant of drug from central
`compartment to peripheral compartment
`K21 = elimination rate constant of drug from periph-
`eral compartment to central compartment
`
`[l CLINICAL CORRELATE
`
`is an
`Digoxin, particularly when given intravenously,
`example of a drug that is well described by two-
`compartment pharmacokinetics. After an intravenous
`dose is administered, plasma concentrations rise and
`then rapidly decline as drug distributes out of plasma
`and into muscle tissue. After equilibration between
`
`drug in tissue and plasma, plasma concentrations
`decline less rapidly (Figure 1-19). The plasma would
`be the central compartment, and muscle tissue would
`be the peripheral compartment.
`
`Volume of Distribution
`
`Until now, we have spoken of the amount of drug (X) in
`a compartment. If we also consider the volume of the
`
`
`
`F|GURE 1-18,
`Two-compartment model.
`
`InnoPharma Exhibit 1022.0011
`
`

`

`Lesson 1: Introduction to Pharmacokinetics and Pharmacoclynamics
`
`9
`
`TABLE 1-2.
`
`Approximate Volumes of Distribution
`of Commonly Used Drugs
`
`Drug
`
`Volume of Distribution (L/kg)
`
`Amlodipine
`
`Ganciclovir
`
`Ketorolac
`
`Lansoprazole
`
`Montelukast
`
`Sildenafil
`
`Valsartan
`
`16.0 i 4
`
`1.1 i 0.2
`
`0.21 i 0.04
`
`0.35 i 0.05
`
`0.15 i 0.02
`
`1.2 i 0.3
`
`0.23 i 0.09
`
`Source: Brunton LL, Lazo JS, Parker KL (editors). The Pharma—
`cologic Basis of Therapeutics, 11th edition. New York: McG raw-
`Hill; 2006. pp. 1798, 1829, 1839, 1840, 1851, 1872, 1883.
`
`the body is primarily composed of water. To calculate
`the volume of the tank, we can place a known quantity
`of substance into it and then measure its concentration
`
`in the fluid (Figure 1-20). If the amount of substance (X)
`and the resulting concentration (C) is known, then the
`volume of distribution (V) can be calculated using the
`simplified equations:
`
`X=VC
`
`or C:K
`V
`
`X
`
`X = amount of drug in body
`V = volume of distribution
`
`C = concentration in the plasma
`As with other pharmacokinetic parameters, volume of
`distribution can vary considerably from one person to
`another because of differences in physiology or disease
`states. Something to note: The dose of a drug (X0) and
`
`
`
`FIGURE 1-20.
`The volume of a tank can be determined from the amount of
`
`substance added and the resulting concentration.
`
`InnoPharma Exhibit 1022.0012
`
`_L O
`
`01
`
`mEi
`
`n
`L“D.
`.E><
`o
`.E’
`
`
`
`DConcentration(ng/mL)
`
`18
`
`12
`Time (hr)
`
`24
`
`FIGURE 1-19.
`
`Plasma concentrations of digoxin after an intravenous dose.
`
`compartment, we can describe the concept of drug con-
`centration. Drug concentration in the compartment is
`defined as the amount of drug in a given volume, such
`as mg/L:
`
`A
`
`1'1
`
`
`.
`amount of drug in body X
`concentration —
`.
`.
`_
`volume in which
`V
`drug is distributed
`
`Volume of distribution (V) is an important indicator of
`the extent of drug distribution into body fluids and tis-
`sues. V relates the amount of drug in the body (X) to the
`measured concentration in the plasma (C). Thus, V is
`the volume required to account for all of the drug in the
`body if the concentrations in all tissues are the same as
`the plasma concentration:
`
`volume of distribution 2
`
`amount of drug
`concentration
`
`A large volume of distribution usually indicates that the
`drug distributes extensively into body tissues and fluids.
`Conversely, a small volume of distribution often indi-
`cates limited drug distribution.
`Volume of distribution indicates the extent of distri-
`
`bution but not the tissues or fluids into which the drug
`distributes. TWo drugs can have the same volume of dis-
`tribution, but one may distribute primarily into muscle
`tissues, whereas the other may concentrate in adipose
`tissues. Approximate volumes of distribution for some
`commonly used drugs are shown in Table 1-2.
`When V is many times the volume of the body, the
`drug concentrations in some tissues should be much
`greater than those in plasma. The smallest volume in
`which a drug may distribute is the plasma volume.
`To illustrate the concept of volume of distribution, let
`us first imagine the body as a tank filled with fluid, as
`
`

`

`10
`
`Concepts in Clinical Pharmacokinetics
`
`
`
`FIGURE 1-21.
`
`K
`
`rate constant
`
`Where: X0: Dose of drug
`X1 = Amount of drug
`in body
`K = Elimination
`
`Drug elimination complicates the determination of the "vol-
`ume" of the body from drug concentrations.
`
`FIGURE 1-22.
`
`One-compartment model.
`
`the amount of drug in the body (X) are essentially the
`same thing because all of the dose goes into the body.
`In this example, important assumptions have been
`made: that instantaneous distribution occurs and that it
`
`occurs equally throughout the tank. In the closed tank,
`there is no elimination. This example is analogous to a
`one-compartment model of the body after intravenous
`bolus administration. However, there is one complicat-
`ing factor—during the entire time that the drug is in the
`body, elimination is taking place. So, if we consider the
`body as a tank with an open outlet valve, the concentra-
`tion used to calculate the volume of the tank would be
`
`constantly changing (Figure 1-21).
`We can use the relationship given in Equation 1-1 for
`volume, amount of drug administered, and resulting
`concentration to estimate a drug's volume of distribu-
`tion in a patient. If we give a known dose of a drug and
`determine the concentration of that drug achieved in
`the plasma, we can calculate a volume of distribution.
`However,
`the concentration used for this estimation
`must take into account changes resulting from drug
`elimination, as discussed in Lessons 3 and 9.
`For example:
`If 100 mg of drug X is administered intravenously and
`the plasma concentration is determined to be 5 mg/ L
`just after the dose is given, then:
`
`volume of
`distribution:
`dose
`2 X0 = 100 mg 2 20 |_
`(V)
`resulting
`C
`5 mg/L
`concentration
`
`I] CLINICAL CORRELATE
`
`The volume of distribution is easily approximated for
`many drugs. For example, if the first 80—mg dose of
`gentamicin is administered intravenously and results
`in a peak plasma concentration of 8 mg/L, volume of
`distribution would be calculated as follows:
`
` volume of
`distribution:
`dose
`2 X0 2 80 m9 = 10 |_
`(V)
`resulting
`C
`8 mg/L
`concentration
`
`[l CLINICAL CORRELATE
`
`Drugs that have extensive distribution outside of
`plasma appear to have a large volume of distribu-
`tion. Examples include digoxin, diltiazem, imipramine,
`labetalol, metoprolol, meperidine, and nortriptyline.
`
`PLASMA DRUG CONCENTRATION
`
`VERSUS TIME CURVES
`
`With the one-compartment model (Figure 1-22), if we
`continuously measure the concentration of a drug in the
`plasma aft

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