throbber
APPLIED
`BIDPHARMACEUTICS
`
`&PHARMACOK|NET|CS
`
`FIFTH EDITION
`
`LEON SHARGEL, PhD, RPh
`Vice President, Biopharmaceutics
`Eon Labs, Inc.
`Wilson, North Carolina
`
`Adjunct Associate Professor
`School of Pharmacy
`University of Maryland
`Baltimore, Maryland
`
`SUSANNA WU—PDNG PhD. RPh
`Associate Professor
`
`Department of Pharmaceutics
`Medical College of Virginia Campus
`Virginia Commonwealth University
`Richmond, Virginia
`
`ANDREW B.C. YU PhD. RPh
`Registered Pharmacist
`Gaithersburg, MD
`Formerly Associate Professor of Pharmaceutics
`Albany College of Pharmacy
`Present Affiliation: HFD—520, CDER, FDA*
`
`*The content of this book represents the personal views of the authors and not that of the FDA.
`
`McGraw-Hill
`
`Medical Publishing Division
`
`New York Chicago San Francisco Lisbon London Madrid Mexico City Milan
`New Delhi Sanjuan Seoul Singapore Sydney Toronto
`
`Page 1
`
`RB Ex. 2025
`BDSI v. RB PHARMACEUTICALS LTD
`IPR2014-00325
`
`

`
`The McGraw-Hill Companies
`
`Applied Biopharmaceutics and Pl1a1macokinefics, Fifth Edition
`
`Copyright © 2005 by The McGraw—Hill Companies, Inc. Copyright © 1999, 1993 by Appleton
`8c Lange; copyright © 1985, 1980 by Appleton~Century-Crofts. All rights reserved. Printed
`in the United States of America. Except as permitted under the United States copyright’
`Act of 1976, no part of this publication may be reproduced or distributed in any form or
`by any means, or stored in a data base or retrieval system, without the prior written per-
`mission of the publisher.
`
`23456'7‘?890 DOC/DOC 098765
`
`ISBN 0-07—l37550—3
`
`This book was set in New Baskerville by TechBooks.
`The editors were Michael Brown and Christie Naglieri.
`The production service was TechBool<s.
`The production supervisor was Phil Galea.
`The cover designer was Kelly Parr.
`RR Donnclley was printer and binder.
`
`This book is printed on acid-free paper.
`
`Library of Congress Cataloging-in-Publication Data
`Shargel, Leon, 1941-
`Applied biopharmaceutics 8c pharmacokinetics/Leon Shargel, Susanna Wu-Pong,
`Andrew B.C; Yu. ;—-5th ed.
`p. ; cm.
`Includes bibliographical references and index.
`ISBN O-(J7-l 37550-3
`I. Title: Applied biopharmaceutics and
`2. Pharmacokinetics.
`‘* 1. Biopharmaceutics.
`pharmacokirieucs.
`ll. Wu-Pong, Susanna.
`III. Yu, Andrew B. C.,
`1945‘ IV. Title.
`
`2. Models, Chemical.
`1. Biopharrnaceutics.
`[DNLM:
`QV 38 S531a 2004] RM301.4.S52 2004
`615‘/7-dc22
`
`3. Pharmacokinetics.
`
`2004044993
`
`Please tell the authors and publisher what you think of this book by sending your
`comments to pharmacy@mcgraw—hill.com. Please put the author and title of the
`book in the subject line.
`
`Page 2
`
`

`
`RELATIONSHIP
`
`BETWEEN
`
`PHARMACOKINETICS
`AND
`
`PHARMACODYNAMICS
`
`PHARMACODYNAMICS AND PHARMACOKINETICS
`
`Previous chapters in this book have discussed the importance of using pharmaco-
`kinetics to develop dosing regimens that will result in plasma concentrations in the
`therapeutic window and yield the desired therapeutic or pharmacologic response.
`The interaction of a drug molecule with a receptor causes the initiation of a se-
`quence of molecular events resulting in a pharmacodynamic or pharmacologic
`response. ifhe term pha-rmacodynamics refers to the relationship between drug con-
`ccntrations at the site of action (receptor) and pharmacologic response, including
`the biochemical and physiologic effects that influence the interaction of drug with
`the receptor. Early pharmacologic research demonstrated that the pharmacody-
`namic response produced by the drug depends on the chemical structure of the
`drug molecule. Drug receptors interact only with drugs of specific chemical struc-
`ture, and the receptors were classified according to the type of pharmacodynamic
`response induced.
`Since most pharmacologic responses are due to noncovalent interaction between
`the drug and the receptor, the nature of the interaction is generally assumed to
`be reversible and conforms to the Law of Mass Action. One or several drug mole-
`cules may interact simultaneously with the receptor to produce a pharrnacologic
`response. Typically, a single drug molecule interacts with a receptor with a single
`binding site to produce a pharrnacologic response, as illustrated below.
`
`Page 3
`
`

`
`Page 4
`
`

`
`RELATIONSHIP l5L”l'WEEN PHARMACOKINETICS AND PHARMACQDYNAQAICS CHAPTER19.
`
`577
`
`Both theories are consistent with the observed saturation (siginoidal) drug—dose
`response relationships, but neither theory is sufficiently advanced to give a detailed
`description of the “locl<-and-key” or the more recent “induced-fit” type of drug in-
`teractions with enzymatic receptors. Newer theories of drug action are based on
`in-z/itro studies on isolated tissue receptors and on observation of the conforma-
`tional and binding changes with different. drug substrates. These in-vitro studies
`show that other types of interactions between the drug molecule and the receptor
`are possible. However, the results from the in—1/itro studies are dillicult to extrapo-
`late to in—v~ivo conditions. The pharmacologic response in drug therapy is often a
`product of physiologic adaptation to a drug response. Many drugs trigger the phar-
`macologic response through a cascade of enzymatic events highly regulated by the
`body
`Unlike pharmacokinetic modeling, pharmacodynamic modeling can be more
`complex because the clinical measure (change in blood pressure or clotting time)
`is often a surrogate for the drug’s actual pharmacologic action. For example, after
`the drug is systemically absorbed, it is then transported to site of action where the
`pharmacologic receptor resides. Drug—receptor binding may then cause a second-
`ary response, such as signal transduction, which then produces the desired elfect.
`Clinical measurement of drug response may only occur after many such biologic
`events, such as transport or signal transduction (an indirect effect), so pl1armacody-
`namic modeling must account for biologic processes involved in eliciting drug-
`induced responses.
`The complexity of the molecular events triggering a pharmacologic response is
`less difficult to describe using a pharmacokinetic approach. Pharmacokinetic mod-
`els allow very complex processes to be simplified. The process of pharmacokinetic
`modeling continues until a model is found that describes the real process quanti-
`tatively. The understanding of drug response is greatly enhanced when pharma-
`cokinetic modeling techniques are combined with clinical pharmacology, resulting
`in the development of pharmacokineticfiharmacodynanzic models. Pharmacol<inetiC—
`pharmacodynamic models use data derived from the plasma drug concentration-
`versus-time profile and from the time course of the pharmacologic effect to predict
`the pharmacodynamics of the drug. Pharmacokinetic—pharmacodynamic models
`have been reported for antipsychotic medications, anticoagulants, neuromuscular
`blockers, antihypertensives, anesthetics, and many antiarrhythmic drugs (the phar-
`macologic responses of these drugs are well studied because of easy monitoring).
`
`RELATION OF DOSE TO
`PHARMACOLOGIC EFFECT
`
`The onset, intensity, and duration of the pharmacologic effect depend on the dose
`and the pharmacokinetics of the drug. As the dose increases, the drug concentra-
`tion at the receptor site increases, and the pharmacologic response (effect) increases
`up to a maximum effect. A plot of the pharmacologic effect to dose on a linear
`scale generally results in a hyperbolic curve with maximum effect at the plateau
`(Fig. 19-1). The same data may be compressed and plotted on a log~linear scale
`and results in a sigmoid curve (Fig. 19-2).
`
`Page 5
`
`

`
`S78
`
`CHAPTER19. RELATIONSI-llP BETWEEN PHARM/\COl<lNEIICS7_AND PHARMACODYNAMICS
`
`response
`
`A small increase in
`response occurs by
`c: given dose change
`
`
`
`Pharmczcologicresponse
`
`A large increase in
`response occurs by
`0 given close change
`in this region
`
`DW9 ‘i059
`
`Figure 19-1. Plot of pharmacologic re
`sponse versus dose on a linear scale.
`
`For many drugs, the graph of log dose~response curve shows a linear relation-
`ship at a dose range between 20% and 80% of the maximum response, which typ-
`ically includes the therapeutic dose range for many drugs. For a drug that follows
`one-compartment pharmacokinetics, the Volume of distribution is constant; there-
`fore, the pharmacologic response is also proportional to the log plasma drug con-
`centration within a therapeutic range, as shown in Figure 19-3.
`Mathematically, the relationship in Figure 19-3 may be expressed by the follow-
`ing equation, where m is the slope, e is an extrapolated intercept, and E is the drug
`effect at drug concentration C:
`
`E = mlogC + e
`
`Solving for log C yields
`
`(j:
`log ,
`
`E—e
`
`m
`
`
`
`Phormcicologicelieci
`
`Log dose
`
`log dose versus
`Figure 19-2. Typical
`pharmacologic response curve.
`
`(19.1)
`
`
`
`Phcrmucologiceilecl
`
`Log drug concentration
`
`Figure 19-3. Graph of log drug con-
`centration versus pharmacologic effect.
`Only the linear portion of the curve is
`shown.
`
`Page 6
`
`

`
`RELATIONSHIP BETWEEN l’llARMACOKlNETlCS AND PHARMACODYNAMICS CHAPTER,
`
`579
`
`However, after an intravenous dose, the concentration of a drug in the body in a
`onecompartment open model is described as follows:
`kt
`logC“—’ logC0 — 23
`
`(19.3)
`
`By substituting Equation 19.2 into Equation 19.3, we get Equation 19.4, where
`E0 '—" effect at concentration C0:
`Eee
`m __E0-e
`m
`
`kt
`2.3
`
`E
`
`E0
`
`km
`2.3
`
`(19.4)
`
`The theoretical pharmacologic response at any time after an intravenous dose
`of a drug may be calculated using Equation l9/L. Equation 19.4 predicts that the
`pharmacologic effect will decline linearly with time for a drug that follows a one-
`compartment model, with a linear log dose—pharmacologic response. From this
`equation, the pharmacologic effect declines with a slope of km/2.3. The decrease
`in pharmacologic effect is affected by both the elimination constant k and the
`slope m. For a drug with a large m, the pharmacologic response declines rapidly
`and multiple doses must be given at short inteivals to maintain the pharmacologic
`effect.
`
`The relationship between pharmacokinelics and pharmacologic response can
`be demonstrated by observing the percent depression of muscular activity after an
`IV dose of (+)-tubocurarine. The decline of pharmaeologic effect is linear as a
`function of time (Fig. 19-4). For each dose and resulting pharmacologic response,
`the slope of each curve is the same. Because the values for each slope, which in-
`clude km (Eq. 19.4), are the same, the sensitivity of the receptors for (+)—tubocu—
`rarine is assumed to be the same at each site of action. Note that a plot of the log
`concentration of drug versus time yields a straight line.
`A second example of the pharmacologic effect declining linearly with time was
`observed with lysergic acid dicthylamide, or LSD (Fig. 196). After an IV dose of
`the drug, log concentrations of drug decreased linearly with time except for a brief
`distribution period. Furthermore, the pharmacologic effect, as measured by the
`performance score of each subject, also declined linearly with time. Because the
`
`Figure 19-4. Depression of normal muscle activity
`as a function of time after lv administration of O. l -0.2
`mg l+}—tubocurarine per kilogram to unanesthetized
`volunteers, presenting mean values of 6 experiments
`on 5 subjects. Circles represent head lift; squares,
`hand grip; and triangles, inspiratory flow.
`(Adapted from Johansen et al, 1964, with permission.)
`
`é (
`
`DO
`
`OO
`
`3:-0
`
`ix)0
`
`00
`
`15
`10
`5
`Time (minutes)
`
`20
`
`
`
`
`
`Depressionofnormalactivity(percent)
`
`Page 7
`
`

`
`580
`
`CHAPTER 19. RELATIONSHIP BETWIEEN l’HARMAL'()KlNljTlCS AND PMARMACODYNAMICS
`
`O
`
`i\)O
`
`
`
`Performance{percentofcontrol)> ooo~xx0O0
`
`
`
`
`
`PlasmaCOflCEfllVGlIO|"l(ng/mL)
`
`5
`
`()1
`
`—-to
`
`O
`
`A
`
`Time (hours)
`
`4
`
`Time (hours)
`
`Figure 19-5. Mean plasma concentrations of LSD and performance test scores as a function of time
`after IV administration of 2 pg LSD per kilogram to 5 normal human subjects.
`(Adapted from Aghajanian and Bing, I964, with permission.)
`
`slope is governed in part by the elimination rate constant, the pharmacologic effect
`declines mucl-.1 more rapidly when the elimination rate constant is increased as a
`result of increased metabolism or renal excretion. Conversely, a longer pharma»
`cologic response is experienced in patients when the drug has a longer halfilife.
`
`RELATIONSHIP BETWEEN DOSE AND DURATION
`
`OF ACTIVITY (teff), SINGLE IV BOLUS INJECTION
`
`The relationship between the duration of the pharmacologic effect and the dose
`can be inferred from Equation 19.3. After an intravenous dose, assuming a
`one—compartment model, the time needed for any drug to decline to a con-
`centration C is given by the following equation, assuming the drug takes effect
`immediately:
`
`2.3 1 re —1
`C
`
`z=_»9°52 0g )
`.1"
`,
`
`(19.5)
`
`Using Ceg to represent the minimum effective drug concentration, the duration of
`drug action can be obtained as follows:
`
`‘.3l
`
`1) V —l
`
`‘C
`
`lefi:
`
`(19_(;)
`
`Some practical applications are suggested by this equation. For example, a dou-
`bling of the dose will not result in a doubling of the effective duration of pharma—
`cologic action. On the other hand, a doubling of 231/2 or a corresponding decrease
`in k will result in a proportional increase in duration of action. A clinical situation
`is often encountered in the treatment of infections in which C53 is the bacteriocidal
`concentration of the drug, and, in order to double the duration of the antibiotic,
`a considerably greater increase than simply doubling the dose is necessary.
`
`Page 8
`
`

`
`581
`
`The minimum effective concentration (MEG) in plasma for a certain antibiotic is
`0.1 /veg/mL. The drug follows a one-compartment open model and has an apparent
`volume of distribution, VI), of 10 L and a first—order elimination rate constant of
`1.0 hr‘1.
`
`a. What is the Jeff for a single 100—mg IV dose of this antibiotic?
`b; What. is the new tag; or t’eg- for this drug if the dose were increased 10-fold, to
`1000 mg?
`i
`‘
`
`Solution 0
`a. The tcff for a 100-xiig dose is calculated as follows. Because VD = 10,000 mL,
`‘
`l00 mg
`=
`6° , 10,000 mL
`
`= 10
`
`L
`
`“g/I"
`
`For a lone-cornpartmem-model IV dose, C = C004“. Then
`0.1 = l0e“(1‘°"°”
`
`tdf
`
`hr
`
`téfffora}/1000-ing dose is calculated as follows.(prime refers to a new dose).
`Because V15 = 10,000 mL,
`’
`
`0
`
`.0- éfF§—,X.1cOO
`-
`teff '
`‘
`0,
`0
`_
`e "-6.91— .10
`entinc easefiri = L
`‘ -4 6146 L.>< 100
`7*Pe_rclente in reaseuin zeg %5o% 0
`0
`
`0
`
`am 1 shows hatac10¥foldincrease in the doseiinci~ease}Stl{1€s, agxramm of,‘
`rt.g;(:g;;>leil§ycie¢:11yji50%. *
`
`Page 9
`
`

`
`582
`
`CHAPTER 19. REL/\TliQNSHlP”l57ETWEEN PHARM/\COKlNETlCS AND PHARMACODYNAMICS
`
`\
`
`EFFECT OF BOTH DOSE AND ELIMINATION
`HALF-LIFE ON THE DURATION OF ACTIVITY
`
`A single equation can be derived to describe the relationship of dose (D0) and the
`elimination half—life (t1/2) on the effective time for therapeutic activity (tcff), This
`expression is derived below.
`
`ln Ccff -“ lnC0 —~ kteff
`
`Because C0 = D0/ VD,
`
`D
`
`ln(-9) ~ kleif
`
`Vn
`D
`
`ln<T/E) — ln Ce“
`1
`<D0/ VD)
`
`k
`
`Ct-eff
`
`Substituting 0.693/t1/2 for k,
`
`Arr
`
`):1.44 1 ——~
`
`51/2 n(VDCCff)
`
`(19.3)
`
`From Equation 19.8, an increase in £1/2 will increase the tsp; in direct propor-
`tion. However, an increase in the dose, D0, does not increase the teff in direct pro-
`portion. The effect of an increase in VD or Ceff can be seen by using generated
`data. Only the positive solutions for Equation 19.8 are valid, although mathemati-
`cally a negative tog can be obtained by increasing Ceff or VD. The effect of chang-
`ing dose on teff is shown in Figure 19-6 using data generated with Equation 19.8.
`A nonlinear increase in tcff is observed as dose increases.
`
`EFFECT OF ELIMINATION HALF—LIFE
`ON DURATION OF ACTIVITY
`
`Because elimination of drugs is due to the processes of excretion and metabolism,
`an alteration of any of these elimination processes will effect the t1/2 of the drug.
`In certain disease states, pathophysiologic changes in hepatic or renal function will
`
`Figure 19-6.
`
`Plot of reg versus dose.
`
`D059 ("'9/l<9l
`
`8
`
`12
`
`16
`
`Page 10
`
`

`
`RELATIONSHIP Btrwttn RHARMACOKINETICS AND PHARMACODYNAMICS CPTAPTERI9.
`
`583
`
`decrease the elimination ofa drug, as observed by a prolonged t1/2. This prolonged
`I1/2 will lead to retention of the drug in the body, thereby increasing the duration
`of activity of the drug (teff) as well as increasing the possibility of drug toxicity.
`To improve antibiotic therapy with the penicillin and cephalosporin antibi-
`otics, clinicians have intentionally prolonged the elimination of these’ drugs by
`giving a second drug, probenecid, which competitively inhibits renal excretion
`of the antibiotic. This approach to prolonging the duration of activity of antibi-
`otics that are rapidly excreted through the kidney has been used successfully for
`a number of years. Similarly, Augmentin is a combination of amoxicillin and
`clavulanic acid; the latter is an inhibitor of ,8-lactamase. This B—lactamase is a bac—
`terial enzyme that degrades penicillin-like drugs. The data in Table 19.1 illustrate
`how a change in the elimination ti/2 will affect the t,,n- for a drug. For all doses,
`a 100% increase in the m2 will result in a 100% increase in the teff. For example,
`for a drug whose I1/2 is 0.75 hour and that is given at a dose of 2 mg/kg, the tcff
`is 3.24 hours. If the tug is increased to 1.5 hours, the tcff is increased to 6.48 hours,
`an increase of 100%. However, the effect of doubling the dose from 2 to 4 mg/kg
`(no change in elimination processes) will only increase the teq to 3.98 hours, an
`increase of 22.8%. The effect of prolonging the elimination half—life has an
`extremely important effect on the treatment of infections, particularly in patients
`with high metabolism, or clearance, of the antibiotic. Therefore, antibiotics must
`be dosed with full consideration of the effect of alteration of the tug on the teff.
`Consequently, a simple proportional increase in dose will leave the patient’s blood
`concentration below the effective antibiotic level most of the time during drug
`therapy. The effect of a prolonged tefg is shown in lines a and c in Figure 19-7,
`and the disproportionate increase in teff as the dose is increased 10-fold is shown
`in lines a and b.
`
`1
`
`=
`
`*
`
`TABLE 19.1 Relationship between Elimination Ha1f»Life and Duration of Activity
`5 Dost-:50
`‘tj,"3 i=fo.7i15 hr
`“
`t”; = 1.5 hr
`(ma/K91
`1 mzihri ~
`tan ihri
`2.0
`3.24
`(3.48
`3.0
`3.67
`7.35
`4.0
`3.98
`7.97
`5.0
`4.22
`8.45
`6.0
`4.42
`8.84
`7.0
`4.59
`9.18
`8.0
`4.73
`9.47
`9.0
`4.86
`9.72
`10
`4.97
`9.95
`11
`5.08
`10.2
`12
`5.17
`10.3
`13
`5.26
`10.5
`14
`5.34
`10.7
`15
`5.41
`10.8
`16
`5.48
`11.0
`17
`5.55
`11.1
`18
`5.61
`11.2
`19
`5.67
`11.3
`20
`5.72
`11.4
`
`Page 11
`
`

`
`584
`
`CHAPTER 1‘). RELATIONSHIP BETWEEN PHARMACOKINETICS AND PHARMACODYNAMICS
`
`
`
`
`
`Logpiosrnoconcentration(1.19/mL)
`
`1 O
`
`I
`
`0,]
`
`00
`
`2
`
`A
`Time (hours)
`
`6
`
`Plasma |evel—time curves describing
`Figure 19-7.
`the relationship of both dose and elimination half—llfe
`on duration of drug action. Ceff = effective concentra-
`tion. Curve a= single lOO—mg IV injection of drug;
`k = 1.0 hr". Curve b = single lOOO—mg IV injection,‘
`k: 1.0 hr“. Curve C: single 100-mg lV injection;
`/<= 05 hr“? VD is 10 L.
`
`CW
`
`Pharmacokinetic/Pharmacodynamic Relationships
`and Efficacy of Antibiotics
`
`In the previous section, the time above the effective concentration, tag, was shown
`to be important in optimizing‘ the therapeutic response of many drugs. This con-
`cept has been applied to antibiotic drugs (Drusano, 1988; Craig, 1995; Craig and
`Andes, 1996; Scaglione, 1997). For example, Craig and Andes (1996) discussed the
`antibacterial treatment of otitis media. Using the minimum inhibitory antibiotic. con-
`centration (MIC) for the microorganism in serum, the percent time for the antibi-
`otic drug coiiceiitrationi to be above the MIC was calculated for several antibacterial
`classes, including cephalosporins, macrolides, and trimethoprim—sulfatnethoxazole
`(TMP/SMX) coinbination (Table 19.2). Although the drug concentration in the
`
`TABLE 19.2 Middle Ear F|uid—to—Serum Ratios for Common Antibiotics
`
`<
`
`Cephalosporins
`Cefaclor
`‘
`Cefuroxime
`Macrolide antibiotic
`Erythromycin
`Sulfa drug
`Sulfisoxazole
`
`From Craig and Andes (19%).
`
`0.184028
`0.22
`
`’
`
`0.49
`
`0.20
`
`Page 12
`
`

`
`
`
`
`
`Bocieriologiccure(percent)
`
`RELATIONSHIP BETWEEN7P7HKAi§r\/L/\COKlNETlCS AND PHARMACODYNAMICS CHAPTER19.
`
`585
`
`LEGEND:
`0 0 IHGCIGMS
`D I M°C”°”d9‘
`V ' WP/SMX
`
`60
`40
`Time above MIC (percent)
`
`80
`
`Figure 19-8. Relationship between the
`percent
`time above MlC9o of the dosing
`interval during therapy and percent of
`bacteriologic cure in otltis media caused by
`S. pneumoniae [open symbols) and ,8-
`lactamaseposltive
`and —negative
`H.
`inf/uenzae (closed symbols). (Circles, closed
`and open = ,8-lactams; squares, closed and
`open=macrolides;
`triangles, closed and
`open = TMP/SMX.}
`(From Craig and Andres,
`mission‘)
`
`I996, with per»
`
`middle ear fluid (MEF) is important, once the ratio (MEF/serum) is known, the
`serum drug level may be used to project MEF drug levels. The percent time above
`MIC of the dosing interval during therapy correlated well to the percent of bacte-
`riologic cure (Figure 19-8). An almost 100% Cure was attained by maintaining the
`drug concentration above the MIC for 60-70% of the dosing interval; an 80-85%
`cure was achieved with 40-50% of the dosing interval above MIC. When the per-
`cent of time above MIC falls below a critical value, bacteria will regrow, thereby
`prolonging the time for eradication of the infection. The pharmacokinetic model
`was further supported by experiments from a mouse infection model in which an
`infection in the thigh due to Pseudomonas aeruginosa was treated with ticarcillin and
`tobramycin.
`In another study, Craig (1995) compared the AUC/MIC, the time above MIC, and
`drug peak concentration over MIC and found that the best fit was obtained when
`colony-forming units (CFUS) were plotted versus time above MIC for cefotaxime in
`a mouse infection model (Fig. 19-9).
`Both Drusano (1988) and Craig (1995) reviewed the relationship of pharmaco-
`kinetics and pharmacodynamics in the therapeutic efficacy of antibiotics. For some
`antibiotics, such as the aminoglycosides and fluoroquinolones, both the drug con
`centration and the dosing interval have an influence on the antibacterial effect.
`For some antibiotics, such as the /3-lactams, vancomycin, and the macrolides, the
`duration of exposure (time-dependent killing) or the time the drug levels are main-
`tained above the MIC (tag) is most important for efficacy. For many antibiotics (cg,
`fluoroquinolones), there is a defined period of bacterial growth suppression after
`short exposures to the antibiotic. This phenomenon is known as the postantibiotic
`efiect (PAE). Other influences on antibiotic activity include the presence of active
`metabolite (s), plasma drug protein binding, and the penetration of the antibiotic
`into the tissues. In addition, the MIC for the antibiotic depends on the infectious
`microorganism and the resistance of the microorganism to the antibiotic. In the
`case of ciprofloxacin, a quinolone, the percent of cure of infection at Various doses
`was better related to AUIC, which is the product of area under the curve and the
`
`Page 13
`
`

`
`586
`
`CHAPTER19. RELA'l‘lONSHII’ BETWEEN PHARMACo1<iN_ET1cs AND Pl-MRMACODYNAMICST
`
`
`
`LogmCFUperlungat24hours
`
`l00 l000l0000
`l0
`Peak/MIC ratio
`
`l0 30 lOO 300l0OO3000
`24-Hour AUC/MIC ratio
`CFU = Colony-Forming unit
`
`0
`
`80 lO0
`60
`40
`20
`Time above MIC (percent)
`
`Figure 19-9. Relationship among three pharmacodynamic parameters and the number of K/ebsiel/a
`pneumoniae in the lungs of neurotroponic mice after 24-hour therapy with cefotaximc. Each point represents
`one mouse.
`
`(From Craig \)</A, i995, with permission.)
`
`211, 1993).
`reciprocal of minimum inhibition concentration, MIC (Forrest Ct
`Interestingly, quinolones inhibit bacterial DNA gyrase, quite different from the
`[3—lactam antibiotics, which involve darnage to bacterial cell walls.
`
`Relationship between Systemic Exposure and
`Response—Anticancer Drugs
`
`Plasma drug concentrations for drugs that have highly variable drug clearance
`in patients fluctuate widely even after intravenous infusion (Rodman and Evans,
`1991). For highly variable drugs, there is no apparent relationship between the
`therapeutic response and the drug dose. For example,
`the anticancer drug
`teniposide at three different doses give highly variable steady—state drug con-
`centrations and therapeutic response (Fig. 19-10). In some patients, single—point
`drug concentrations were variable and even higher with lower doses. Careful
`pharmacokinetic—pharmacodynarnic analysis showed that a graded response
`curve may be obtained when responses are plotted versus systemic exposure as
`
`(4)O
`
`ION)O01
`
`ou.oG
`
`
`
`Steady-stateconcentration
`
`OO
`O
`
`8
`
`LEGEND:
`0 Response
`g No response
`
`750
`600
`Dose level (mg/mzl
`
`Figure I9-10. Steady-state concentration and
`response after three levels of teniposide administered
`by intravenous infusion.
`(From Rodman and Evans, 1991, with permission.)
`
`Page 14
`
`

`
`RELATIONSHIP BETWEEN l’IlARMACOKlNETlC_S_ AND PHARlvtACODYN}§MlCS WCHAPTER19.
`
`587
`
`
`
`Responselpropofiion)
`
`Response
`(3/7)
`
`<5 500. 12oo—>18oo
`1200 1300
`I'M ' “W
`
`Figure 19-1 1. Relationship between systemic expo-
`sure for teniposide and toxicity and efficacy, shown as
`proportions of patients.
`{From Rodman and Evans, 199», with permission.)
`
`measured by “concentration >< time” (Fig. 19-11). This is one example showing
`that anticancer response may be better correlated to total area under the drug
`concentration curve (AUC), even when no apparent dose—response relation-
`ship is observed. Undoubtedly, the cytotoxic effect of the drug involves killing
`cancer cells with multiple-resistance thresholds that require different time ex-
`posures to the drug. The objective of applying pharmacokinetic—pharmacody—
`namic principles is to achieve therapeutic efficacy without triggering drug toxic-
`ity. This relationship is illustrated by the sigmoid curves for response and toxicity
`(Fig. 19-11), both of which lie close to each other and intensify as concentration
`increases.
`
`RATE OF DRUG ABSORPTION AND
`PHARMACODYNAMIC RESPONSE
`
`The rate of drug absorption influences the rate in which the drug gets to the re-
`ceptor and the subsequent pharmacologic effect. For drugs that exert an acute
`pharmacologic effect, usually a direct-acting drug agonist, extremely rapid drug ab-
`sorption may have an intense and possibly detrimental effect. For example, niacin
`(nicotinic acid) is a vitamin given in large doses to decrease elevated plasma cho-
`lesterol and triglycerides. Rapid systemic absorption of niacin when given in an im-
`mediate-release tablet will cause vasodilation, leading to flushing and postural hy-
`pertension. Exte_nded—release niacin products are preferred because the more
`slowly absorbed niacin allows the baroreceptors to adjust to the vasodilation and
`hypotensive effects of the drug. Phenylpropanolamine was commonly used as a
`nasal decongestant in cough and cold products or as an anorectant in weight-loss
`products. Phenylpropanolarnine acts as a pressor, increasing the blood pressure
`much more intensely when given as an immediate-release product compared to an
`extended-release product.
`
`Equilibration Pharmacodynamic Half-Life
`
`For some drugs, the half-time for drug equilibration has been estimated by ob-
`serving the onset of response. A list of drug half-times reported by Lalonde (1992)
`is shown in Table 19.3. The factors that affect this parameter include perfusion of
`
`Page 15
`
`

`
`S88
`
`CHAPTEl{19. REL/\TilONSl:l_lP BETWEEN PHARMACOKINETICS AND PH/\RMl\CODYNAMl7CS
`
`TABLE 19.3 Equllibration Half—Tlmes Deter/mined Using the Effect Compartment Method
`EOUILIBRATION
`PHARMACOLOGIC
`t,,2 (min)
`RESPONSE
`
`DRUG T
`
`d—Tubocurarine
`Dlsopyramide
`Ouinidine
`Digoxin
`Terbutaline
`Terhutaline
`Thcophylline
`Verapamil
`Nizatidine
`Thiopental
`Fentanyl
`Altentanil
`Ergotamine
`\/ercuronium
`/\l~/\cetylprocainamide
`
`From Lalonde ii992l,‘°\'lvith permission.
`
`4
`2
`8
`
`7 5
`
`1
`
`l
`2
`B3
`l.2
`6.4
`l
`l
`
`.
`
`Muscle paralysis
`OT prolongation
`OT prolongation
`LVET shortening
`FEV;
`Hypokalemia
`FFV.
`PR prolongation
`Gastric pH
`Spectral edge
`Spectral edge
`Spectral edge
`Vasoconstriction
`Muscle paralysis
`OT prolongation
`
`the effect compartment, blood—tissue partitioning, drug diffusion from capillaries
`to the effect compartment, protein binding, and elimination of thc drug from the
`effect compartment.
`
`Substance Abuse Potential
`
`The rate of drug absorption has been associated with the potential for substance
`abuse. Drugs taken by the oral route have the lowest abuse potential. For example,
`cocoa leaves containing cocaine alkaloid have been chewed by South American
`Indians for centuries (johanson and Fischman, i989). Cocaine abuse has become
`a problem as a result of the availability of cocaine alkaloid (“crack” cocaine) and
`because of the use of other routes of drug administration (intravenous, intranasal,
`or SII10klI1g,§‘_frtVl}§.>t allow a very rapid rate of drug absorption and onset of action
`(Gone, 1995); Studies on diazepam (deWit et al, 1993) and nicotine (Henningfield
`and Keenan, 1998) have shown that the rate of drug delivery correlates with the
`abuse liability of such drugs. Thus, the rate of drug absorption influences the abuse
`potential of these drugs, and the route of drug administrat.ion that provides faster
`absorption and more rapid onset leads to greater abuse.
`
`DRUG TOLERANCE AND PHYSICAL DEPENDENCY
`
`The study of drug tolerance and physical dependency is of particular interest in
`understanding the actions of abused drug substances, such as opiates and cocaine.
`Drug tolerance is a quantitative change in the sensitivity of the drug and is demon-
`strated by a decrease in pharmacodynamic effect after repeated exposure to the
`same drug. The degree of tolerance may vary greatly (Cox, 1990). Drug tolerance
`has been well described for organic nitrates, opioids, and other drugs. For example,
`
`Page 16
`
`

`
`Rl;'LATlONSlllP BETWEEN PHARMACOKINETICS AND PHARMACODYNAMICS CHAPTER 19.
`
`589
`
`the nitrates relax vascular smooth muscle and have been used for both acute angina
`(eg, nitroglycerin sublingual spray or transmucosal tablet) or angina prophylaxis
`(eg, nitroglycerin transdcrmal, oral controlled-release isosorbide dinitrate). VVell-
`controlled clinical studies have shown that tolerance to the vascular and antiangi—
`nal effects of nitrates may develop. For nitrate therapy, the use of a low nitrate or
`nitrate-free periods has been advocated as part of the therapeutic approach. The
`magnitude of drug tolerance is a function of both the dosage and the frequency
`of drug administration. Cross tolerance can occur for similar drugs that act on the
`same receptors. Tolerance does not develop uniformly to all the pharmacologic or
`toxic actions of the drug. For example, patients who show tolerance to the de-
`pressant activity of high doses of opiates will still exhibit “pinpoint" pupils and con-
`stipation.
`The mechanism of drug tolerance may be due to (1) disposition or pharmaco-
`kinetic tolerance or (2) pharmacodynamic tolerance. Pharmacakinetic tolerance is of-
`ten due to enzyme induction (discussed in earlier chapters), in which the hepatic
`drug clearance increases with repeated drug exposure. Pharmacodynawtic tolerance is
`due to a cellular or ‘receptor alteration in which the drug response is less than what
`is predicted in the patient given subsequent drug doses. Measurement of serum
`drug concentrations may differentiate between pharmacokinetic tolerance and
`pharmacodynamic tolerance. Acute tolerance, or taeltyphylaxifs, which is the rapid
`development of tolerance, may occur due to a change in the sensitivity of the re-
`ceptor or depletion of a cofactor after only a single or a few doses of the drug.
`Drugs that work indirectly by releasing norepinephrine may show tachyphylaxis.
`Drug tolerance should be differentiated from genetic factors which account for
`normal variability in the drug response.
`Physical dependency is demonstrated by the appearance of withdrawal symptoms af-
`ter cessation of the drug. Workers exposed to volatile organic nitrates in the work-
`place may initially develop headaches and dizziness followed by tolerance with con-
`tinuous exposure. However, after leaving the workplace for a few days, the workers
`may demonstrate nitrate withdrawal symptoms. Factors that may affect drug de-
`pendency may includc the dose or amount of drug used (intensity of drug effect),
`the duration of drug use (months, years, and peak use) and the total dose (amount
`of drug X duration). The appearance of withdrawal symptoms may be abruptly pre-
`cipitated in opiate-dependent subjects by the administration of naloxone (Narcan),
`an opioid antagonist that has no agonist properties.
`
`HYPERSENSITIVITY AND ADVERSE RESPONSE
`
`Many drug responses, such as hypersensitivity and allergic responses, are not fully
`explained by ph

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