throbber
Pharmacodynamics and
`Drug Development
`Perspectives in
`Clinical Pharmacology
`
`Editecf by
`
`Neal R. Cutler
`Caiifawzia Cfirzical Tz1}:z!s, 8%-s2:‘Iy Hilis, Ctmfomicz, U521
`
`John J. Sramek
`Caf£fa:'m’a Ciirzicczf Triafs, Beverb: Efiffs, Calsfomio, USA
`
`Prem K. Narang
`Pizcmnacia Acfricz, Cfiaical P§sar222acoZ0g}-jPfzamzaco:%i22e£2'es, Cofiunzfms, Ohio, USA
`
`IQHN WILEY 8: SONS
`Chichester - New York - Brisbane ° Toronto - Singapore
`
`|nnoPharma Exhibit 10290001
`
`

`

`Copyright
`
`1994 by Iohri Wiley 3: Sons Ltd,’‘
`Baffins Lane, Chiczhester,
`West Sussex PO19 IUD, England
`Telephone:
`Chichester {(3243} ?‘?9’???
`National
`International +44 243 ?'?9?"??
`
`"except Chapter 12 which is in the public domain.
`
`All rights rese1'ved.
`
`No part of this book may be reproduced by any means?
`or transmirtecl, or translated into a machine language
`without the written permission of the publisher.
`
`Otlzer ll’/Way Editorial Offices
`
`Iolm Wiley fir Sons, loo, 605 Thirtl iwenue,
`New York NY 101580012, USA
`
`Jacaranda Wiley Ltd, 33 Park Road, Milton,
`Queensland 4064, Australia
`
`john Wiley 3: Sons (Canada) Ltd, 22 Worcester Road,
`Rexclale, Ontario M9\X’ 1L1, Canada
`
`John Wiley 8: Sons {SBA} Pte Ltd, 3'?’ Jalan Pemimpin ;%0S-04
`Block B, Union Industrial Building, Singapore 2035?
`
`Library of Czmgress Catczirygirzg-irz-Paciviicatirm Data
`
`Pharmaeodynamiee and drug development : perspectives in clinical
`pharmacology} edited by Neal R. Cutler, john I. Sramek and Pram K.
`Narang.
`cm.
`p.
`Includes bibliographical references and index.
`ISBN 0 4'31 95052 1
`
`1. Drugsmlfiiysiological effect.
`Prem K.
`III. Sramek, John I.
`EDNLM: l. Pharrnarsology, Clinical. QV 38 P3318 1994}
`RM3G{).P48
`1994
`6lS'.?——clc20
`
`I. Cutler, Neal R.
`
`II. Narang,
`
`DNLMHDLC
`for Library of Congress
`
`946103
`{ZIP
`
`British Liizrary Gatafeguirrg in Pztbfieatioza Sam
`
`3‘; catalogue record for this book is available from the British‘ Library
`
`ISBN 0 4?} 950 521
`
`Typeset in 10;‘ 12 pt Plantirt by
`Mathematical Corttposition Setters Ltd, Salisbury, \5i"iltshi1‘e
`Printecl and bouncl in Great Britain by
`Bookeraft (Bath) Ltd, Midserner Norton, Avon
`
`|nnoPharma Exhibit 1029.0002
`
`

`

`
`Contents I
`
`
`
`List of Contriirgutors
`
`0
`
`0
`
`_
`
`Fareword t
`Lesfie Z. Benet
`
`’
`
`I OVERVIEW OF PHARMACODYNAMICS
`
`1 Basic Pharmacodynamic Concepts and Modem
`Raisers} ii’/E535
`
`2 Simultaneous i’harmacakinetic,{1’harmacociynamic Modeling
`Wayrze A. Coiinmz am? Mickaef A. Efdan
`
`3 Factors Influencing Variability in Kinetics and Dynamics
`Pram K. Ncmmg am!‘ Ronaia’ C. Li
`
`4 Populatiomfiased Approaches to the Assessment of
`Phatmacokinetics and Pharmacodynamics
`‘
`Eosepiz C. FZe2'332a}aez* and Ekiward 5'. zinicré
`
`S Generai Perspectives on the Role of Metabolites in
`Pharmacokinetics and Pharmacodynamics
`Rcmdaff D. Seéferz
`
`6 Enantioselectivity in Drug Action and Drug Metabolism:
`Influeace on Dynaanics
`Hey: K.
`I<'2‘0eme:*, Amzetze S. Grass med Micfzei }_3Iic?zeZ£:amrz
`
`’? Regulatory Perspective: The Role of Pharmacokinetics and
`Pharmacodynamics
`Lawrence Lesfea (ma? Roger L. Wéffiams
`
`1! APPLICATION OF PHARMACODYNAMICS IN SELECTED
`THERAPEUTIC DOMAINS
`
`8 Theoreticai Models for Developing Anxiolytics
`P. V. Nickefi and Wzamczs W’. Ufzcfe
`
`9 Pharmacodynamics of Antidepressants
`Karon Dawkém, Husseini K. Marzji and if/éifiam 2. Poster
`
`2:
`
`xiii
`
`3
`
`19
`
`45
`
`‘F3
`
`39
`
`103
`
`1 15
`
`133
`
`15?
`
`|nnoPharma Exhibit 10290003
`
`

`

`vi
`
`CONTENTS
`
`Pharmaccedynamics of Antipsychotic Drugs in
`Schizophrenia
`3033?: 3’. Smmeie and George M. Simpson
`
`11
`
`Pharmacodynamic Modeis Useful in the Evaluation of
`Drugs for Cognitive Impairment
`Micizaefi F. Murphy, Kiazsdizzs R. Sieg,f}*s'ec2’, F. facoia Hujffnnd
`Neai R. Cutie?
`
`181
`
`201
`
`Alzheimer’s Disease: Assessment of Chnlinomimetic Agents
`Scat: A. Reines
`
`225
`
`Antihypertensive Drugs
`L. Micfiznes? P:~is¢:m and Aiber-3 A. Cam‘
`
`Pharmacodynamics of Calcium Antagonist Drugs
`Dawefi R. Abemezizy czmi Nabii S. Andmrwis
`
`Agents in Congestive Heart Failure
`Edmund V. Capparefli
`
`Antiarrhythmic Drugs
`Eanice B. Sclzzuezrzz
`
`Antibiotic Pharmacodynamics
`30322: C. Rotscizafer, 1'<'cm’::z 3’. Waifzer, Kilt‘? 3’. K3555» cmci
`C}m'szop?2er ff. Szefiitscziz
`
`Pharmacodynamics of Antineoplastic Agents
`G233}: L. Rosszer and Mczzrfe 3'. Retain
`
`Controlling the Systemic Exposure of Anticancer Drugs:
`The Dose Regimen Design Problem
`Dania! Z. D’Args2rzio and fofzzvz H. Redraw:
`
`20
`
`Virology and Antiviral Drug Development
`Mficfzaef A. z"i??3€I?IE€<.Z,
`_§*::mzes R. Minor anti Stepizen E.’ Sirens
`
`Ill
`
`FRONTIERS IN PHARMACODYNAMICS: INSIGHT FROM
`MOLECULAR APPROACHES
`
`21
`
`ozvfidrencceptors and their Subtypes: Plaarmacological
`Aspects
`P. 2%. mm Zwieterz
`
`241
`
`253
`
`26?‘
`
`291
`
`315
`
`345
`
`363
`
`3??
`
`409
`
`|nnoPharma Exhibit 1029.0004
`
`

`

`CONTENTS
`
`22 Muscarinic Receptors: Pharmacologicai Subtypes,
`Stmcmrea Function and Regulation
`Lire Mei, Wi§§£a:;z R. Roesiee and‘ Hemy I. Ycmza;~mm:z
`
`23 Serotmiin Receptor Subtypes
`Doknz B. Pritcizest
`A
`
`Index
`
`Vii
`
`433
`
`4-57"
`
`475
`
`|nnoPharma Exhibit 10290005
`
`

`

`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`2 Simultaneous Pharmacokineticf
`Pharrnacodynamic Modeling
`
`warns A. cotsuan AND MICHAEL a. ELDON
`
`the use of
`Simultaneous pharmacolcineticfpharmacodynarnic modeling is
`integrated pharrnacolcinetic and pharrnacodynamic models to interpret and
`extrapolate the temporal relationship between some sampled drug concentration
`and observed drug effect. The basis for such modeling is the need to analyze
`and describe measurable concentration—effect data, as well as to make ciinicaiiv
`relevant extrapolations from experimental conditions to therapeutic conditions.
`Investigation of drug action using pharniacokinetic or dose-response models is
`well established in clinical pharmacology; the linking of these tools through
`specific mathematical models is relatively new.
`ing a
`Pharrnacokinetics contributes to clinical phartnacolog1g by provi
`means to characterize drug distribution and elimination.
`its usefulness is
`predicated on the assumption that measurable drug concentrations are related
`to drug effect in some manner,
`thereby forming the basis for deterr ining
`concentration-effect relationships {pharmacodynarnics) and employing thera-
`peutic drug monitoring. In recent years, significant advances have been made
`in technologies to measure drug and metabolite concentrations in bio ogical
`matrices, further advancing the use of pharrnacoltinetics as an adjunct
`to
`optimizing drug therapy. Concurrent advances in the abiiity to quantita e and
`understand drug effects have similarly promoted the stud}; and use of
`pharmacodynaniics.
`Pharmacokineticfphartnacodynamic relationships have been investiga ed in
`two general approaches. The first approach involves the determination of drug
`effect and concentration over a series of doses administered to a relatively large
`patient population. Correlation of concentration and effect
`is performed
`retrospectively, usually resulting in the determination of target plasma drug
`concentration ranges which are thought to provide some level of drug effect
`while zniniinizing the risk of toxicity (1). Unfortunately,
`this approach is
`relatively imprecise due to its
`sensitivity to inter--subiect variability in
`pharmacokinetic as well as pharmacological factors. It is the imprecision and
`non-specificity of this method which requires the study of large numbers of
`patients to determine a therapeutic dose range} and even then may lead to
`
`Pleczmmcmiwramirs and ilimg Deaelopmem: Perspectives in Clinical Pf:am:a::ofag;-
`Edited by N. R. Cutlery I. I. Sramek and I’. K. Narang
`'33) 1994 lohn Willey‘ 8t Sons Ltd
`
`|nnoPharma Exhibit 10290006
`
`

`

`20
`
`OVERVIEW OF PHARMACODYNAMICS
`
`inappropriate conclusions that drug effect and blood or plasma drug concen-
`trations are ‘not correlated’.
`The relevant question is not whether comtentration and effect are related for
`a given drug, but rather how are they reiated and what is necessary to elucidate
`the relationship. Answering these questions is the goal of the second approach,
`which involves correlation of graded pharmacological responses with circulating
`drug concentration in a smaller number of patients. This more specific approach
`allows investigation of the nature of drug effect and its relationship to drug
`concentration, while minirnizing the impact of phartnaeoirinetie and pharmaco-
`djrnantic inteosubject variability.
`This chapter is concerned with the latter approach and gives an overview
`of key developments during evolution of simultaneous pharrnaeoltineticf
`pharmacodynamic modeling, a review of contemporary methods, and goals for
`future reiinetnent of the topic. Detailed discussion of pharrnaeokinetic theory
`and practice will not be given here. The reader is directed to excellent references
`on the topic (2,3) for further information.
`
`EVOLUTION T0 THE PRESENT
`
`PHARMACOKINETIC EKPPROACI-{ES
`
`The evolution to simultaneous modeling was based on the desire to refine
`understanding of drug action. This was expressed in 196? by Brodie (4) when
`he observed that fewer patients were required to deterrnine antirnalarial activity
`if drug effect was correlated to plasma concentration rather than dose. In
`retrospect, this observation could most likely be attributed to the reduction
`of
`intersabject variability in the pharmacokinetic cornponent of
`the
`dose—response relationship. During this stage of evolution, Levy (5) proposed
`that for many drugs, the intensity of effect was linearly related to log eoncen-
`tration over the range of 20-80% of the maximum possible effect (Emx).
`He suggested the following equation to describe the cor1<:entration~effe»:t
`relationship after intravenous drug dosing:
`
`E-»m‘logA+a
`

`
`{1}
`
`where E is the effect intensity, A is the amount of drug present (which may he
`represented by concentration values}; in is the slope of the linear plot of E versus
`log 21, and e is the intercept of that plot. This equation is based on the assump-
`tion that effect is directly related to drug concentration at the site of action and
`is rapidly reversible. However; the log transformation is only pseudo-linear over
`the 2(}-m80% effect range} owing to the underlying sigrnoici nature of the
`dose—response relationship.
`
`|nnoPharma Exhibit 1029.000?
`
`

`

`PHARMACO Kl Ni;ITlC,? PHtXR3s’l:\CODYNAMIC MODKLING
`
`2 1
`
`the drug exhibited a oneconipartment pharrnacoltinetic
`Assuming that
`profile, Levy further proposed the following equation to describe the decline of
`effect after intravenous drug administration (5):
`
`§=Eg~(K'??1)[2.3°£
`
`(2)
`
`where B’; is the initial effect intensity, K is the apparent iirsvorder elimination
`rate constant} 2* is time, and all other parameters are as previously defined. These
`equations predict that the intensity of effect is linearly related to log concen-
`tration, and that effect declines linearly rather than exponentially following
`bolus drug administration. The practice of relating effect to log concentration
`data was a logicai extension of analysing dose—response relationships using the
`log transform. The log transform does compress the dose or concentration range
`and linearize the concentration—effect relationship over the inner 20-80% of the
`effect range. However, as discussed by Holford and Sheiner (6), this method of
`data analysis does not explain effect at the extremes of the concentration range
`(i.e., zero effect when no drug is present), provide a means to estimate Emmi,
`or accommodate the existence of baseline effect. While the log transformation
`may be appiicable for specific drugs,
`it
`is not a suitable substitute for
`characterizing the entire range of the dose or con<:entration—effect relationship
`as later ciescribed in the sections on parametric anci semi-parametric methods.
`Additionally, Equations 1 and 2 do not permit assessment of the deiay in onset
`of drug effect following administration by routes requiring drug absorption or
`distribution before reaching the effector site, or the persistence of effect when
`drug is no longer present in plasma. This delay in drug equilibration between
`the sampled biofluid and the responding tissue gives rise ‘to hysteresis in the
`effect versus concentration plot as shown in Figure 1.
`
`Concentration
`
`50
`
`80
`
`
`-«Mt.»O-O‘O 10
`
`12
`Time
`
`20
`30
`40
`Concentration
`
`(a) Theoretical -plasma concentration (solid line) and effect (dashed line)
`Figure I.
`profiles versus time following extravasculat drug aclzninistration. {la} Corresponding
`cottnterclockwise hysteresis plot of effect versus plasma concentration data front (a)
`
`|nnoPharma Exhibit 10290008
`
`

`

`22
`
`OVERVIEW’ OF PHARMACODYNAMICS
`
`Levy et ai. if’) addressed the problem of equilibration delay by extending the
`elationships described by Equations 1 and 2 to include multicompartment
`harmacokinetic models and empiricalijs comparing pharmacokinetic and drug
`effect profiles. This approach was used to investigate the relationship between
`ental performance test scores and predicted lysergic acid diethylamide (LSD)
`pharrnacokinetics from work by Aghaianian and Bing (8). Figure 2 shows
`harmacokinetic and effect profiles from this experiment. Based on a two
`compartment pharrnacokinetic model, effect {reduction in performance score}
`did not appear to be directly related to central compartment (plasma) concen-
`rations, but rather to the time course of drug in the second or tissue cotnpart~
`merit. However, counterclockwise hysteresis was stiil evident in the plot of
`erformance score versus fraction of dose in the tissue compartment, as shown
`in Figure 3. Accordingly, a third compartment representing slowly equi—
`lihrating tissue was added to the pharmacokinetic model. This modification of
`the model resulted in a linear plot of performance score versus fraction of dose
`in the slowly equilibrating compartment, shown in Figure 4, indicating that the
`observed equilibration delay between plasma LSD concentration and effect
`couid be explained by the effector compartment being pharmacokineticaliy
`distinct from the plasma compartment.
`The pharmacol-zinetic compartment approach is limited in that it is dependent
`on identifying a potentialiy complex pharinacokinetic mode} with concentrations
`
`28
`
`score
`
`
`
`Performancetest
`
`Fractionofdose
`
`EU
`
`80
`
`100
`
`Time {it}
`
`Figure 2. Observed {o} and predicted {upper curve} amounts of LSD in the central
`compartment, predicted amounts in the tissue cornpartnient
`(lower curve) of a
`twocompartment model, and performance test
`scores
`{0}
`following intravenous
`aclniinistration of LSD to normal subjects (From reference ?, with permission)
`
`|nnoPharma Exhibit 10290009
`
`

`

`PHAIIMACGKJNETIC,‘1’HAR£\=lACODYNAMIC MODELING
`
`23
`
`23
`
`Perforrnanee
`
`£1.05
`
`0.49
`0.20
`0.10
`Fraction of dose
`
`Figure 3. Relationship between performance scores and the fractional amount of LSD
`in the tissue compartment of the two-compartment phsrmaeolcinetie model (From
`reference '3, with permission}
`
`Performance
`
`30
`
`55£23
`
`so
`9 ,»
`.30
`
`./
`3/ 120
`
`K
`
`
`
`0.05
`
`0.015
`
`0.15
`0.10
`Fraction of dose
`
`0.20
`
`Figure 4. Relationship between péi1‘f(}l'I113£1C(2 scores and the fractional amount of LSD
`in the slowly equilibrating tissue compartment of a three~eompartment pharmacokinetis:
`model {From reference 3’, with permission}
`
`in at least one compartment correlstsble with the effect profile. In many cases,
`pharmaeokinetic cornpartments are not readily recognizable as distinct body
`tissues which may be of interest, and therefore may not contribute to any real
`understanding of the effector site. An extension of this concept will be
`addressed later in this chapter.
`
`|nnoPharma Exhibit 10290010
`
`

`

`24
`
`OVERVIEW OF PEIARMACODYNAMECS
`
`PHfiRMfiCOUYNAMIC APPROACHES
`
`In l968, Wagner (9) proposed using the Hill equation to model the hyperbolic
`relationship between drug effect and dose or concentration. This proposal has
`been widely adopted and the model has been paraineterized for analysis of
`in aim and in vino concentration-effect relationships as the sigmoid En.“ model
`shown below:
`
`E
`
`_ Ema:-; ' C?
`-I Ecsgl -1? CW
`
`{3}
`
`where E is intensity of effect, Em“ is the maximum possible effect in the system
`being studied, C is the drug concentration, EC” is the steadywstate drug concen-
`tration evoking 50% of Ema, and qr is the sigmoidieity parameter indicating
`the slope and shape of the curve. Note that when the value of ~,« is l, the
`c:oncentration——effect curve is a simple ll§;‘ps‘3rl3{}l8. and the model is termed the
`Em,“ model. A typical sigmoid effectmconeentration curve depicting parameters
`of this model is shown in Figure 5.
`Hyperbolic models have been used to describe Various binding phenomena
`such as Michaelis—Menten enzyme kinetics and protein binding,
`thereby
`linking the use of the Em“ models to receptor binding theory (10). Clark {l 1)
`also proposed the use of a similar equation to model dose-response relationship
`as an application of mass action theory. The use of hyperbolic models to
`represent biological processes is einpiricall§; reasonable since they describe the
`Widely observed phenomena that as
`the maximum response (effect)
`is
`approaelied, increasing levels of stinmlation (concentration) are required to
`teach the niaxitnnm. The Em“ models offer advantages over the logarithmic
`model suggested by Levy (5) in that they predict effect over the entire COI1C€I1~
`tration range, including zero effect when concentration equals zero, and the
`tnasirnutn possible effect (Emit).
`Wagner (9) also proposed inserting eoncentrationwtime data predicted from
`pharmacokinetie models into the Hill equation to predict the time course of in
`nine response, based on its similarity to in nine experiments where the concen-
`tration in the bath solution could be varied to study response. This approach
`has been expanded to simultaneously fitting phatmacoltinetic and pharmaco-
`djgmamic models to concentration--effect data as detailed in the Present methods
`section of this chapter.
`Several other pharniacodynamic models and modifications of the Emx model
`have been used to describe concentration—effeet relationships. Examples of
`these are as follows.
`
`The linear model
`
`E -~ 3 ~ C + £5};
`
`_
`
`{4}
`
`where S is the slope of the linear effect versus coiicentration plot, Es is the effect
`
`|nnoPharma Exhibit 1029.0011
`
`

`

`l3l~Ir’lRMAC{)KINETlC,?l“l"IARl‘vl:&(3()DYNAMlC l‘viODELIl‘~lG
`
`25
`
`Effect
`
`' o
`
`1 o
`
`3 0
`2 o
`Concentration
`
`ft 0
`
`5 0
`
`Figure 5. Plot of drug effect versus concentration simulated using the sigmoid Emx
`model given in Equation 3 (parameter values: Sam =0.}’9, EC“: = l0, and ~,« m3}
`
`intensity when no drug is present, and E and C are as previously defitled. The
`linear model has limited application, usually to defined segments of the true
`response curve,
`since it predicts
`that effect
`increases with increasing
`concentration without limit;
`
`The baseline subtraction model
`
`E~E::==
`
`«Emax ' CY
`EC50? + Q
`
`(5)
`
`This model is based on the assumption that E0 can be subtracted from the effect
`data, leaving the 0—l00°/E; response curve intact. This may not be the case when
`endogenous substances bind to the receptor or interaszt biochemically to main-
`tain the baseline effect. In this situation, the baseline effect should be included
`in the model as given below in Equation 6.
`
`|nnoPharma Exhibit 10290012
`
`

`

`Z6
`
`OVERVIEW OF PHARMACODYNAMlCS
`
`The baseline inclusion model
`
`Emmi - {C -1- Cg)"‘
`E :'?““.“j"""“""""'".
`Ecsgl + (C+ C9)?
`
`(6)
`
`where 8;} is the concentration of drug which would be required to generate
`baseline effect such that E includes the baseline effect. The concepts and appli-
`cations of the baseline subtraction and inclusion models have been previously
`described (12).
`
`The inhibitory Emx model
`
`E~—~i3o-
`
`Emax °
`lC5(;“’ 'i'
`
`('2?)
`
`where K353 is the drug concentration causing 50% inhibition of Emx. This
`model is useful for investigating the effects of inhibitory drugs without trans-
`forming the data. Its use will result in an inverted effect versus concentration
`plot with the tnaxiniutn and minimum effects occurring at zero and the
`maximum concentration value. Reviews of these and other pharmacodynannc
`models O5,1Z,l3) and examples of their application {6,l4,1S) have recently been
`published. In addition, Colburn (12) has discussed many considerations of
`phartnacolcineticfphartnacodynamic study design, including selection of dosing
`routes and regimens and corresponding pharmacodynatnic models. Alternative
`models including those for dealing with indirect effects and tolerance will be
`presented in the section on future developments.
`
`PRESENT
`
`The present state of simultaneous pharmacolcineticfpharmacodjgnainic modeling
`has drawn heavily on the foundations of relating effect to an accessible bioilnid
`as described in the preceding section. This too has evolved, beginning with fully
`pararneterized pharrnacoltinetic and pharmacodynaniic models linked by a
`parametric model. Recent advances have been made where both pharmaco-
`kinetic and pharmacodjznatnic data are analyzed non-pararnetrically, that is,
`without assuming that the correct nnderlying model and its parameters are
`known and/or identifiable. This latter approach is perhaps better termed semi—
`parametric since the parameters of the linking model are still estimated.
`Althottgh the term parametric was not originally applied to the first simul-
`taneous pharrnacokineticfpharmacodynannc models, it has come into use since
`the advent of the semi—pat‘ametric methods.
`
`|nnoPharma Exhibit 10290013
`
`

`

`3?l~IARMACOKlNETlCf I’I{ARNlACOD‘1’N£9sil=1lC MODELING
`
`2?
`
`PARAMETRIC AIWROACH
`
`Sheiner at of. (16) first proposed that the pharmaeokinetic model parameters
`could be substituted into the Hill equation such that concentration and effect
`profiles could be simultaneously modeled using nonvlinear regression. The
`novel aspect of their compartment model-based approach was the inclusion of
`a theoretical effect compartment related to the central (plasma) compartment,
`but not influencing the overall pharrnacokinetic profile due to its relatively small
`size. A schematic representation of the model is shown in Figure 6(a). Drug
`transfer into and loss from the effect compartment were controlled by first-order
`rate constants and drug effect was assumed to be directly related to the amount
`of drug in the effect compartment at any time. The plasma to effect compart-
`ment transfer rate constant, Em; and amount of drug transferred to the effect
`compartment were assumed to he so small that the pharznacokinetic profile
`would not be altered and that the negligible amount of drug in the effect
`compartment did not need to be returned to the central compartment. Under
`these conditions, the rate constant for drug loss from the effect compartment,
`Kgo, would control the temporal relationship between effect and the concen-
`tration profile in the plasma compartment.
`Sheiner et
`<22.
`(16) evaluated the model using concentrationmeffect data
`obtained following of-tubocurarine administration as a two-stage intravenous
`infusion to healthy patients and to patients with end~stage renal failure. They
`concluded that the method was robust and could predict the equilibrium delay
`between appearance of drug in plasma and onset of effect, shown in Figure 27.
`One of the main advantages of this approach is that it allows the characterization
`
`
`
`lb}
`
`KEG
`
`and peripheral
`{a}
`compartment
`representation of central
`Figure 6. Schematic
`compartment (lo) effect models used in pharmacokineticjpharmacodynamic modeling
`
`|nnoPharma Exhibit 10290014
`
`

`

`28
`
`OVERVIEW‘ OF PHEXRMACODYNAMICS
`
`DTC infusion rate (pg! kg I min}
`
`I2
`
`I68
`
`1
`I
`
`1.0
`
`
`
`x
`19

`
`I
`
`G
`
`‘<
`ix
`\0
`/
`'*
`a K
`X
`\
`Q
`(3 0
`G K '
`\
`I
`“M-W-”L'“‘
`o
`as
`Q
`0
`\\
`8
`
`
`
`\s"M
`
`.92
`E
`6;}.
`ad§O,6
`‘5
`E
`E
`1:0»:
`:2
`.23
`G
`
`0.2
`
`0
`
`‘%
`
`.v0
`5
`so
`as
`eosoaasoao
`BC!
`I00
`:20
`:40
`I60
`:30
`2:0
`220
`Time {min}
`
`Figure 3". Observed a‘-tubo:>cuz‘arine plasma concentrations {9} and effect Co) during and
`foliowing ixitraxrczmus infusion of the drug. Selid line: best fit of the pharmacnkinetief
`pha1~macod:mamic model to the data {From reference 16, with permission}
`
`relationship under nonwsteady-state conditions.
`of the c0ncent1‘ation~«effect
`Conversely, many facmrs of drug effect such as receptor binding and past-
`binding events are groupeci and represented in the model by aeingle firsmnrder
`rate constant.
`
`Calburn (1?) investigated the model proposed by Sheiner ex a2. (16) and found
`that it was able to represent a wide variety ofipliarmacokinetic and pharmaco»
`dynamic phenomena. He derived effect equations applicable to several classical
`COI1’1pa2‘t11’1{'2I1[ models and extended the approach to accommodate {he effect
`compamnem c0:1centra1;i<m being driven from 21 peripheral czempartment as
`shown in Figure 603). In the interest ofmaciel isrientifiability, he recomniended
`that C{il'1U‘£ii and peripheral compartment medeis be fit (.0 each data set 311:} that
`
`|nnoPharma Exhibit 10290015
`
`

`

`l’l~lARlt*lACOKINET1(3) i"l‘li’iRl‘vli*lCODYNr5ils‘liC M(}lf3i3l..iN(:‘s
`
`29
`
`drug be administered lay several routes of aclrninistration before extrapolating
`the concentration—effect relationship beyond observed data. rhdditioiially, a
`model selected from fitting to singlodose data should be tested for adequacy by
`studying the transition from single to multiple doses, since predicted and
`observed effects will
`sysiematically diverge when multiple doses
`are
`administered if an incorrect model has been chosen (l?}. Potential divergence
`due to inappropriate model selection is illustrated in Figure 8.
`The peripheral compartment effect model (Figure 64:13)} can be used to explain
`apparent changes in pharrnacolzineticfpharmacodynamic relationships as a
`function of route of administration, or other phenomena not explained by the
`central compartment effect model {Figure 63%) (14). The peripheral cornpart~
`merit effect model provides an additional tool for explaining nomparallclisrn
`between concentration and effect modeled using the central compartment effect
`model. Modeling the effect compartment as driven by a peripheral compart-
`ment may he more physiologically relevant if the effector tissue is believed to
`be a pharrnacokinetically identifiable tissue. More representative models could
`result if the pharmacokinetic compartment model is replaced with a physio»
`logical flow model where the target organ thought to be the receptor} effector site
`can be isolated (14).
`
`is
`Further refinement of the pharmacoltineticfpharmacodynanuc model
`possible using specially designed studies to isolate and identify the rate-limiting
`components of the proposed model {I2}. By using a varying iirst~order rate of
`intravenous administration,
`rate-limiting and] or controlling steps such as
`receptor binding can be isolated from the model. Alternatively, one may find
`that diffusion to the receptor is the slowest step, and construct the model to
`reflect this. Elucidation of a robust model that can predict drug effect under a
`variety of conditions will aid in selecting dosage regimens and optimizing
`therapy.
`
`SEMI-PARAMETRIC APPROACH
`
`the intrinsic
`thorough understanding of
`Parametric modeling requires
`pharmacokinetic and phartnacodynaniic models before combining them, as well
`as the ability to identify and reliably estimate each parameter of the combined
`model. This may often be difficult, depending on noise level of the pharmaco«
`kinetic and pharmacodynairiic data sets and the characteristics of the underlying
`models for a given drug. In an attempt to minimize these factors, Fnseau and
`Sheiner (18) proposed that the pharinacodynainie component of the combined
`model could be modeled non—pararnetrically using the relationship between
`observed effect and the effect compartment drug concentration (Ce) predicted
`using a parametric pliarrnacoltinetic model. To achieve this, it is necessary to
`assume that
`the relationship between Cs and effect
`is instantaneous and
`invariant with time, i.e.,i tolerance and sensitization do not occur. As in the
`parametric approach, the effect compartment is modeled as receiving negligible
`
`|nnoPharma Exhibit 10290016
`
`

`

`30
`
`
`
`....3......no
`
`conic):_
`
`9
`
`uu5uaswanI.
`
`uxnuxnnuxuxu,xxW»
`90m1&»
`
`0O00mctwat.
`
`xx._AMuux
`
`Ew3AvW
`
`.w.
`
`w.W.mnw
`
`.9Um.
`
`13mm.
`
`m.....tJ.J.l1.4$.:.JHk_.dHM__
`
`
`
`
`
`Ewfimwmo.&m_m«oo§S~:ooemwwfimo
`
`¢E..._.
`
`3mm..wEvmmovhmum
`
`$nE..Em.moo
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`any:3338mmA;Mmuommfiwm8GoHmbauumfifimsmmmnovmbwwmwqammwmowmxmfififimadamSmndoHmummomooambmamvowumuofim.wmrfimmm
`
`
`
`
`Eomwwnwuumfir.»wfiuumohmfiE85SufifimmfiooEumamfioaatS@838mmmfiummmwumwfi
`
`|nnoPharma Exhibit 1029.001?
`
`
`

`

`PHARi‘~élACOKlNE"l"lC{ Pllz’:Ri\=lACODYl\lAMIC MODELINC}
`
`31
`
`amounts of drug with the concentration profile determined by Kim and the
`plasma drt g concentration (Cp). In the non-parametric phartnacodynamic
`approach, ysteresis between effect and Cs is suppressed by choosing K50 such
`that the ascending and descending arms of the effect-—Ce plot are super-
`irnposable (18). The best estimate of K53 is determined using a univariate search
`method when minimizes the average squared difference between observed and
`interpolated effect Values from the hysteresis plot as shown in Figure 9.
`Fuseau and Sheiner (18) tested the nonparametric pharrnacodynamic method
`using simu ations based on hyperbolic and sigmoid Emit models as well as
`models of the g3—function {convex Ce-B relationship), tolerance, sensitization
`and non-equilibrium between Ce and the receptor, the latter three which violate
`the assump ions of the method. The proposed method was found to be accep-
`table for both Em“ models and the 6 function model when adequate numbers
`of data having minimal error were used. However, the non—parametric method
`could not provide accurate or precise estimates of Kg, when applied to data from
`the tolerance, sensitization or nonequilibrium simulation models. Additionally,
`performance was reduced for all simulation models when too few data or noisy
`data were used.
`
`Subsequently, Unadkat at of. (19) extended the nonparametric pharmaco-
`dynamic approach to include pliarmacolcinetic modeling such that pharn1aco~
`kinetics and pharmacodynamics could be simultaneously modeled non-
`parametrically with the link model still used to estimate the parameter K50,
`thereby allowing ‘semiwparametric’ simultaneous pharmacokinetic/pharmaeo—
`dynamic modelling. The advantages of this approach are that fewer assumptions
`about either the underlying pharmacokinetic or pharmacodynamic model are
`
`IIIIIIIIItIIIIIIIII!
`
`
`
`,..__.,....__l____l___.....l..__.
`Co“ Co;
`C932
`
`Figure 9. Application of the r;0n~parameI:rlc pharmacodynamie method of Fuseau and
`Sheiner (l8) to estimate K59 by minimizing the average squared difference between
`observed (Bi; and E32) and interpolated (E;m,;) effect data. Ce values are corresponding
`effect compartment concentration values estimated using a parametric pharmacoleinetic
`model {From reference 18, with permission’)
`
`|nnoPharma Exhibit 10290018
`
`

`

`32
`
`OVERVIEW OF PHARMACODYNAMICS
`
`(19) described this as a two-stage process where
`:12.
`required. Unadlcat st
`observed Cp values are used to model pharmacodynamics and determine the
`linlcing {<"_r;0 value. Simple linear interpolation is used to estimate Cp—time
`values if missing from the Cp-—effect data set as shown in Figure 10. The
`resultant Cp~time data set is used to estimate Ce as a function of time by
`numerically integrating the fnllowing equation for a given value of Km (19):
`
`dCe{dt = K, ~ (3,, —. Km Ce
`
`ts}
`
`Where K1 is effect compartment input rate constant (assumed to be equal to
`Kgo} and ail other parameters are as previousiy defined. A starting estimate of
`{€59 is selected and the parameter value is increased or decreased incrementally
`depending on the direction of hysteresis and area between the limbs of the
`effect-—Ce plot corresponding to each Km Value. The process is iterated until the
`Km value which minimizes the area within the hysteresis loop is found.
`This approach assumes that Ce and hence effect is a function of observed (and
`interpolated) C32: as determined by the value of K503 independent of intrinsic
`pharmacokinetics. Based on a series of simulations, the authors (19) suggested
`that this approach is nearly as efficient as the parametric approach even when
`
`Plasma
`
`concentration
`
`Time
`
`Figure 19. Example of non--parametric ‘fit’of plasma concentration ((33)) versus time
`data (0). If C}; was not observed at a pharmacodynamic observation time, it is estimated
`using linear interpolation between the nearest bracketing observed values {Catt-) and
`(Ipiz +}) (From reference 19, with permission)
`
`|nnoPharma Exhibit 10290019
`
`

`

`PHARMACOKlNl3TlCfPl~lARi’vlACOIi)YNAhlIC MODELING
`
`33
`
`the underlying models were known, but considerably more robust when the
`underlying models were rnis~speciiied.
`Shafer at tel. (20) reported a cornparison of the above method with parametric
`pharinacolcin

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