throbber
Pharmaeodynamics and
`Drug Development
`Perspectives in
`Clinical Pharmacology
`
`Ediiea‘ by
`
`Neal R. Cutler
`Cahfomia Clinical may, Beoei‘e‘y High, Cahfomia, {ISA
`
`John j. Sramek
`Cafflromia Ciirzim? fiiais, Bever‘bs E3535, Calsfomio} USA
`
`Prem K. Narang
`PSzmmacia Adria, Chaim! PfiannacoiogyjPharmacokizzesies, Cofiumfms, Ohio, USA
`
`ISBN WILEY 8: SONS
`Chiehester - New York - Brisbane ' Toronto - Singapore
`
`InnoPharma Exhibit 1029.0001
`
`

`

`Cepyright re 1994 by 30in: Wiley & Sons Leif
`Baffins Lane, Chiahestera
`West Sussex P019 lUD, England
`Telephone:
`Cliichester {0243} ??9???
`Naiienal
`Internatienal +44 243 39???
`
`*except Chapter 12 which is in the public demairi.
`
`All rights reserved.
`
`No part (if this back may be repmdueeel by any means?
`or transmitted, er translated into a machine language
`without the written permission of the pubiisher.
`
`Other Wife}? Editorial? Offices
`
`john Wiley fiz Sens, Inc.3 605 Third Avenue:
`New York NY 101580012, USA
`
`jacaranda Wiley Ltd, 33 Park Read, Milton,
`Queensland 4064. Australia
`
`John Wiley 3: Sons (Canada) Ltd: 22 Worcester Road,
`Rexdale, Ontario M93}? 1L1, Canada
`
`John Wiley :8: Sons (SBA) Pte Ltd, 3'? Jalan Pemimpin #GSMOEi
`Block B, Union Industrial Building, Singapere 205?
`
`Library of Ceagress Caraiegiizgwin-Pubfica55m: Data
`
`Pharmaeodynamiee and drug development : perspectives in clinical
`pharmacology! edited by Neal R. Cutler, Icahn I. Sramek and Prem K.
`Narang.
`cm.
`p.
`Includes bibliographical references and index.
`{SEN 0 4’31 95052 1
`
`1. DrugsmPliysielogical effect.
`Prem K.
`III. Sramek, John I.
`[DNLM1 l. Phermaeelogy) Clinical. QV 38 P3318 1994]
`RM300.P48 1994
`615'.?—dc20
`
`I. Cutler> Neal R.
`
`II. Nai‘ang,
`
`DNLMIDLC
`fer Library of Congress
`
`94’6103
`{ZIP
`
`British Liiirary Cateieguieg in Pubfieetien Date
`
`31 catalogue record for {his book is available from the British Library
`
`ISBN 0 4?} 950 521
`
`Typeset in 10; 12 pt Plantiri by
`Mathematical Compesitien Setters Ltd1 Salisbury, Wiitshire
`Printed and l'JGuI'lCi in Great Britain by
`Beekeraft (Bath) Ltd, Midsmner Norton, Avon
`
`InnoPharma Exhibit 1029.0002
`
`

`

`
`
`
`List of Contributors
`
`‘
`
`.5
`
`Foreword ,
`Lesfie Z. Benet
`
`’
`
`I OVERVIEW OF PHARMACODYNAMICS
`
`1 Basic Pharmacodynamic Concepts and Modem
`Raisers}. W535
`
`2 Simultaneous l’harmacakinetia!l’harmacodynamic Modeling
`Wayne A. Gallium (emf Mickaef A. 8355922
`
`3 Factors Influencing Variability in Kinetics and Dynamics
`Pram K. Narmg cam? Ronald C. Li
`
`4 Populatiomfiased Approaches to the Assessment of
`Pharmacokinetics and l’harmacodyflamics
`‘
`30362)}: C. Ffeislzakgr and Edward 5’. Anmé
`
`:3 General Perspectives on the Role of Metabolites in
`Pharmacokinetics and Pharmacodynamics
`Raizdalf D. 56932:?
`
`6 Enantioselectivity in Drug Action and Drug Metabolism:
`Influerzce on Dynamics
`Hey; K. Kroemer, Amaze 8. Grass and Miclzei Eidzeiéaam
`
`? Regulatory Perspective: The Role of Pharmacokinetics and
`Pharmacodynamics
`Lawreme 1283225: am? Roger L. Wie’fiams
`
`1! APPLICATION OF PHARMACODYNAMICS IN SELECTEI)
`THERAPEUTIC DOMAINS
`
`8 Theoretical Models for Developing Anxiolyfics
`P. V. Nz‘ckefi and “1320mm W. Ufche
`
`9 Pharmacodynamics of Antidepressants
`Karma Dawkim, Husseini K. Marzji and Wifiz'am Z. Posse?
`
`3.
`
`xiii
`
`3
`
`19
`
`45
`
`’33
`
`89
`
`103
`
`l 15
`
`133
`
`15?
`
`InnoPharma Exhibit 1029.0003
`
`

`

`10
`
`11
`
`12
`
`13
`
`14
`
`15
`
`16
`
`I?
`
`18
`
`19
`
`20
`
`Antihypertensive Drugs
`L. Miciznei Prisms: and A551??? 53. Cam
`
`Pharmaeodynamics of Calcium Antagonist Drugs
`Darrefi R. Aber‘neflzy emf Nabifi S. Andr‘awis
`
`Agents in Congestive Heart Failure
`Edmund V. Capparez’li
`
`Antiarrhythmie Drugs
`Ennice B. Sclzwm‘iz
`
`Antibiotic Pharmacodynamics
`303m C. Roiscizafer, Ken’n 3. Waiker, Km? 3’. K353}: and
`Ckrz'szopker ff. Suiiiwn
`
`Pharmacodynamics of Antineoplastie Agents
`Gm}! L. Renter and Maria 3'. Remix
`
`Controlling the Systemic Exposure of Anticancer Drugs:
`The Dose Regimen Design Problem
`Dania! Z. D’Argeizio and 30m; H. Redraw:
`
`Vimlogy and Antiviral Drug Bevelnpment
`Michele? A. Amamea, games R. Minor and 8:33)?an ES Swans
`
`Ill
`
`FRONTIERS IN PHARMACGDYNEXMICS: INSIGHT FROM
`MOLECULAR APPROACHES
`
`21
`
`aafidreneeeptors and their Subtypes: Pharmacological
`Aspects
`P. A. we Zwieten
`
`vi
`
`CONTENTS
`
`Pharmaeodynamies of Antipsychotic Drugs in
`Schizophrenia
`3033?: 3’. Smmefia and George M. Simpson
`
`’
`
`Pharmacodynamic Modeis Useful in the Evaluation of
`Drugs for Cognitive Impairment
`Me’ckaei F. Murphy, Kiazedizes R. Siegfl‘ied, F. 35:50.5 Huffand
`Neal? R. Cage?
`
`181
`
`201
`
`Alzheimer’s Disease: Assessment of Chelinamimetie Agents
`8501336. Reines
`
`225
`
`241
`
`253
`
`26?
`
`291
`
`315
`
`345
`
`363
`
`339
`
`409
`
`InnoPharma Exhibit 1029.0004
`
`

`

`CONTENTS
`
`22 Mumarinic Receptors: Pharmacologicai Subtypes,
`Structure.) Function and Regulation
`Lira Mei, Wifiz'am R. Roesé‘ee and Henry I. Yamamm‘a
`
`23 Serotonin Receptor Subtypes
`503m: B. Pritchest
`‘
`
`Index
`
`Vii
`
`433
`
`4-5?
`
`475
`
`InnoPharma Exhibit 1029.0005
`
`

`

`
` This material may be protected by Copyright law (Title 17 U.S. Code)
`
`
`
`2 Simultaneous Pharmacokinetic/
`Pharmaeodynamic Modeling
`
`
`warns A. COLBURN AND MICHAEL a. ELDON
`
`the use of
`Simultaneous pharmacoldnetic/pharmacodynatnic modeling is
`integrated pharmacolcinetic and pharmacodvnarnic 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 males clinically
`relevant extrapolations from experimental conditions to therapeutic conditions.
`Investigation of drug action using pharmacokinetie or dosewresponse models is
`well established in clinical pharmacology; the linking of these tools through
`specific mathematical models is relatively new.
`ing a
`Pharmacokinetics contributes to clinical pharmacology 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 {pharmacodynamics} 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
`matricesg further advancing the use of pharmacoitinetics as an adjunct
`to
`optimizing drug therapy. Concurrent advances in the ability to quantita e and
`understand drug effects have similarly promoted the stud}; and use of
`pharmacodynamics.
`Pharmacokineticfpharmacodynamic 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 minimizing the risk of toxicity (1). Unfortunately,
`this approach is
`relatively imprecise due to its
`sensitivity to inter-subject variability in
`pharmaeokinetic 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
`
`
`
`Phomacmiwmmirs and 3mg Development: Perspectives in Clinical Pharmacology-
`Edited by N. R. Cutlerg I. I. Sramek and P. K Narang
`'3”) 1994 lohn ’Wiley 8{ Sons Ltd
`
`InnoPharma Exhibit 1029.0006
`
`

`

`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 concentration and effect are related for
`a given drug> but rather how are they related 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 minimizing the impact of pharmacokinetic and pharmaco-
`dynamic inter~snhject variability.
`This chapter is concerned with the latter approach and gives an overview
`of key developments during evolution of simultaneous pharmacokinetic}
`pharmacodynamic modeling, a review of contemporary methods, and goals for
`future refinement of the topic. Detailed discussion of pharmacokinetic 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 nPPROhCl-IES
`
`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 determine antimalarial activity
`if drug effect was correlated to plasma concentration rather than dose. In
`retrospect, this observation could most likely he attributed to the reduction
`of
`intensabiect variability in the pharmacokinetic component of
`the
`dosewresponse relationship. During this stage of evolution: Levy {5) proposed
`that for many drugs, the intensity of effect was linearly related to log concen»
`tration over the range of 20-80% of the maximum possible effect (Emax).
`He suggested the following equation to describe the concentration~effect
`relationship after intravenous drug dosing:
`
`Ezm‘logA-I—e
`
`.
`
`(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 fl, 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 20430554} effect range3 owing to the underlying sigmoid nature of the
`dose-response relationship.
`
`InnoPharma Exhibit 1029.000?
`
`

`

`PHARMACO Kl NETIC; lirIrXRMACODYNAMIC MODKLING
`
`2 1
`
`the drug exhibited a oneacompartment phsrrnscohinetic
`Assuming that
`profile) Levy further proposed the following equation to describe the deciine of
`effect after intravenous drug administration {5):
`
`EngmiK-tny23't
`
`(2)
`
`where E; is the initial effect intensity, K is the apparent first-order elimination
`rate constant5 t 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 exponentiaiiy following
`bolus drug administration. The practice of relating effect to log concentration
`data was a logicsi 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
`(is, zero effect when no drug is present), provide a means to estimate 3mm,
`or accommodate the existence of baseline effect. While the log transformation
`may be applicable for specific drugs,
`it
`is not a suitable substitute for
`characterizing the entire range of the dose or concentration-effect relationship
`as later described in the sections on parametric and semi-parametric methods.
`Additionaliy, 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 site3 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 l.
`
`
`
`t
`
`1
`
`s
`
`5
`
`-.‘i
`
`.s
`
`.5
`
`‘4
`
`s
`«1“:
`L”
`
`2
`
`.2‘
`
`..(_._.
`
`V
`N we”:/
`
`/
`////
`
`/
`
`l
`
`//
`,/ /r
`
`an
`
`50
`
`c: (to
`3%
`{9230
`0
`5 ,
`<2 £0
`m
`
`{a} Q
`
`ta
`
`7
`
`M”“_o
`12
`24
`
`Time
`
`o
`(m o
`
`to
`
`so
`so
`20
`Concentration
`
`so
`
`so
`
`Figure I. {3) Theoretical plasma concentration (soiid line) and effect (dashed line)
`profiles versus time following extrsvascnlar drug administration.
`(is) Corresponding
`counterclockwise hysteresis plot of effect versus plasma concentration data from (2%}
`
`InnoPharma Exhibit 1029.0008
`
`

`

`22
`
`OVERVIEW OF PHARMACODYNAMICS
`
`
`
`levy at all. (3") addressed the problem of equilibration delay by extending the
`relationships described by Equations 1 and 2 to include mnlticompartment
`harmacokinetic models and empiricallj,T comparing pharmacokinetic and drug
`effect profiles. This approach was used to investigate the relationship between
`ental performance test scores and predicted lysetgic acid diethylamide (LSD)
`pharmacokinetics from work by Aghaianian and Bing (8}. Figure 2 shows
`harmacokinetic and effect profiles from this experiment. Based on a two«
`compartment pharmacokinetic 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 compattw
`meat. However, counterclockwise hysteresis was still evident in the plot of
`erformance score versus fraction of close in the tissue compartment, as shown
`in Figure 3. Accordingly, a third compartment representing slowly equi—
`librating tissne 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
`could be explained by the effector compartment being pharmacokinetically
`distinct from the plasma compartment.
`The phat‘macokinetic compartment approach is limited in that it is dependent
`on identifying a potentially complex pharmacokinetic model with concentrations
`
`28
`
`Fractionofdose
`
`Time (it)
`
`EU
`
`
`
`
`
`80
`
`100Periotmancetestscore
`
`Figure 2. Observed {o} and predicted {upper curve} amounts of LSD in the central
`compartment, predicted amounts in the tissue compartment
`(lower curve) of a
`twocompartment model, and performance test
`scores
`(0}
`following intravenous
`administration of LSD to normal subjects (From reference ’3) with permission)
`
`InnoPharma Exhibit 1029.0009
`
`

`

`PHARMACOKINETIC}PHAREVEACODYNAMIC MODELING
`
`23
`
`23
`
`Pattormance
`
`035
`
`0.49
`0.20
`9.13
`Fraction at dose
`
`Figure 3. Reiationship between performance scores and thc fractional amount of LSD
`in the tissue compartment of the nan-compartment pharmacokinetic model (From
`reference '5”, with permission}
`
`Performance
`
`20
`
`b‘23
`
`so
`C x
`.30
`
`./
`3/ 120
`
`_
`
`
`
`0.05
`
`0.025
`
`0.15
`0.10
`Fraction of dose
`
`{3.20
`
`Figure 4. Relationship between perfcxrmance acorns and the fractional amount of LSD
`in the slowly equilibrating tissue compartment of a three~cnmpartment pharmacokinetic
`model {From reference ’3, with permission)
`
`in at least one compartment correlatable with the effect profile. In many casres,
`pharmacokinetic compartments are not readily recognizable as distinct body
`tissues which may be of interest, and therefore may nnt contribute to any real
`understanding of the effector site. An extension of this concept will be
`addressed later in this chapter.
`
`InnoPharma Exhibit 1029.0010
`
`

`

`24
`
`OVERVIEW OF PHARMACODYNAMICS
`
`PHARMfiCODYNAMIC APPROACHES
`
`In 1968, 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 parameteriaed for analysis of
`in vino and it: also concentration—effeet relationships as the sigmoid Em“ model
`shown below:
`
`E
`
`_ Ema); ‘ {fr
`- ECSO’Y 'i' Cr};
`
`(3}
`
`Where E is intensity of effect, Emax is the maximum possible effect in the system
`being studied, C is the drug concentration, ECgs is the steady~state drug concen-
`tration evoking 50% of Email and ’y is the sigmoidicity parameter indicating
`the slope and shape of the curve. Note that when the value of y is 19 the
`concentration—effect curve is a simple hyperbole and the model is termed the
`Emx model A typical sigmoid effectmeoneenttation 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 Emax models to receptor binding theory (l0). Clark {l1}
`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 empirically reasonable since they describe the
`Widely observed phenomena that as
`the maximum response (effect)
`is
`approached} increasing levels of stimulation (concentration) are required to
`reach the maximum. The Emu models offer advantages over the logarithmic
`model suggested by Levy {5) in that they predict effect over the entire eoneen~
`tration range, including zero effect when concentration equals zero> and the
`maximum possible effect (Emax).
`Wagner (9) also proposed inserting concentrationwtime data predicted from
`pharmacokinetie models into the Hill equation to predict the time course of in
`also response, based on its similarity to in nine experiments where the coneenw
`tration in the bath solution could be varied to study response. This approach
`has been expanded to simultaneously fitting pharmacokinetie and pharmaco—
`dynamic models to concentration—effect data as detailed in the Present methods
`section of this chapter.
`Several other pharmaeodynatnie models and modifications of the Emax model
`have been used to describe concentration—effect relationships. Examples of
`these are as follows.
`
`The linear model
`
`a ~.~ s~ o + to
`
`,
`
`{4)
`
`where S is the slope of the linear effect versus concentration plot, Es is the effect
`
`InnoPharma Exhibit 1029.0011
`
`

`

`PHIKRMACQKINETIQPlifiRi‘tlfiCQDYNAMlC MODELING
`
`25
`
`Effect
`
`' e
`
`1 o
`
`3 0
`a e
`Coneenttation
`
`51 o
`
`5 0
`
`Figure 5. Plot of drug effect versus concentration simulated using the sigmoid Enmx
`model given in Equation 3 (parameter values: 8mm =0.79, RC” = 10, and «,e wB}
`
`intensity when no drug is present, and E and C are as previously defined. 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
`
`5-333:
`
`Emmi ' CW
`Elissa/Y + Cl
`
`(5)
`
`This model is based on the assumption that Ea can be subtracted from the effect
`data, leaving the O—lOG% response curve intact. This may not be the case when
`endegenous substances bind to the receptor or interact 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.
`
`InnoPharma Exhibit 1029.0012
`
`

`

`26
`
`OVERVIEW OF PHARMACODYNAMICS
`
`The baseline inclusion model
`
`Emax ° {C + Qt)?
`E : scat + (C+ can
`
`(6)
`
`where Ct} is the concentration of drug which would be required to generate
`baseline effect such that 15? includes the baseline effect. The concepts and appli—
`cations of the baseline subtraction and inclusion models have been previously
`described (12).
`
`The inhibitory Em“ model
`
`52333-
`
`Ema); ' G
`IC5(}Y 'i' C!"
`
`('2?)
`
`where 1850 is the drug concentration causing 50% inhibition of 5mm. 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 maximum and minimum effects occurring at zero and the
`maximum concentration value. Reviews of these and other pharmacodynamic
`models (6,12,13) and examples of their application {6,14,15) have recently been
`published. In addition, Colburn (12) has discussed many considerations of
`pharmacokinetic}pharmacodynamic study design, including selection of dosing
`routes and regimens and corresponding pharmacodynamic 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 pharmacokineticfpharmacodynarnic 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
`parameterized pharmacokinetic and pharmacodynamic models linked by a
`parametric model. Recent advances have been made where both pharmaco—
`kinetic and pharmacodynamic data are analyzed non-parametrically, that is,
`without assuming that the correct underlying 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.
`Although the term parametric was not originally applied to the first simul—
`taneous pharinacokinetic/pharmacodynamic models, it has come into use since
`the advent of the semi«pararnetric methods.
`
`InnoPharma Exhibit 1029.0013
`
`

`

`PHARMACOKINETIQ PHARMACODYNanC MODELING
`
`2?
`
`FARAMETRIC aaraoacn
`
`Sheiner at of. (16) first proposed that the pharmacokinetic model parameters
`could be substituted into the Hill equation such that concentration and effect
`profiles could he simultaneously modeled using nonlinear regression. The
`novel aspect of their compartment model~hased approach was the inclusion of
`a theoretical effect compartment related to the central (plasma) compartment,
`but not influencing the overall pharmacokinetic profile one 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, a”; and amount of drug transferred to the effect
`compartment were assumed to be so small that the pharmacokinetic 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,
`KEG, would control the temporal relationship between effect and the concen-
`tration profile in the plasma compartment.
`Sheiner et a2. {16) evaluated the model using concentrationmeffect data
`obtained following d-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 effecta shown in Figure 2?.
`One of the main advantages of this approach is that it allows the characterization
`
`
`
`(bl
`
`KEG
`
`and peripheral
`{a}
`compartment
`representation of central
`Figure 6. Schematic
`compartment in) effect models used in pharmacokineric/pharmacodynamic modeling
`
`InnoPharma Exhibit 1029.0014
`
`

`

`28
`
`OVERVIEW OF PHARMACODYNAMICS
`
`ETC infusion rate (pg! kg I min}
`
`12
`
`I68
`
`1
`I
`
`so
`
`
`
`r
`
`K \
`an
`/
`‘3
`Q \
`o
`3W0
`‘\:— *
`0

`° \
`
`a
`own—“me...

`.
`0
`\\
`8
`
`\
`
`f
`
`9
`
`1
`a
`
`a
`f
`
`G
`
`I
`
`\k
`
`0.2
`
`“a?
`a
`$06
`a
`‘5
`5
`g
`”:03;
`:3
`.3;
`m
`
`2 e
`
`o
`
`'j. OLWWWW
`
`5
`
`10
`
`t5
`
`no
`eosoaoeoao
`Time {min}
`
`mo
`
`:26 mo
`
`:60
`
`too
`
`am 220
`
`Figure ’1". Observed d—tubocurarine piaema concentrations {a} and effect (0) during and
`foliowing intravenous infusion of the drug. Solid line: best fit of the pharmacokinetief
`phatmacodynamic model to the data {From reference loj with permission}
`
`relationship under nonwsteadystate conditions.
`of the concentrationmeffect
`Conversely, many factors of drug effect such as receptor binding and post-
`binding events are grouped and represented in the model by aningle first-order
`rate constant.
`
`Colburn (I?) investigated the model proposed by Sheiner et a3. (16) and found
`that it was able to represent a wide variety ofopharmaeokinetie and pharmaco»
`dynamic phenomena. He defined effect equations applicable to several classical
`compartment models and extended the approach to accommodate the effect
`compartment concentration being driven from a peripheral compartment as
`shown in Figure 60:»). In the interest ofmodel identifiabiiity, he recommended
`that centtni and peripheral compartment models be fit to each data set and that
`
`InnoPharma Exhibit 1029.0015
`
`

`

`PHARMnCOKINETlC}l’l‘ll’lRl‘leCODYNr-‘llvliC MODELlNG
`
`29
`
`drug be administered by several routes of administration before extrapolating
`the concentration—effect relationship beyond observed data AddltionallyS a
`model selected from fitting to singledose data should be tested for adequacy by
`studying the transition from single to multiple doses} since predicted and
`observed effects will
`systematically diverge when multiple doses
`are
`administered if an incorrect model has been chosen (iii). Potential divergence
`due to inappropriate model selection is illustrated in Figure 8.
`The peripheral compartment effect model (Figure 64:17)} can be used to explain
`apparent changes in pharmacoltineticlpharmacodynamic relationships as a
`function of route of administration, or other phenomena not explained by the
`central compartment effect model {Figure as) (14). The peripheral compart~
`merit effect model provides an additional tool for explaining non—parallelism
`between concentration and effect modeled using the central compartment effect
`model. Modeling the effect compartment as driven by a peripheral compart-
`ment may be more physiologically relevant if the effector tissue is believed to
`he a phatmacoklnetically 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}r effector site
`can be isolated (14).
`
`is
`Further refinement of the pharmacoklnetic}pharmacodynamic model
`possible using specially designed studies to isolate and identify the rate-limiting
`components of the proposed model {12). By using a varying first-order rate of
`intravenous administration,
`rate-limiting and} or controlling steps such as
`receptor binding can be isolated from the model. f‘ilternativel1vj 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-l’nRAMETRIC AFPROACH
`
`the intrinsic
`thorough understanding of
`Parametric modeling requires
`pharmacoklnetic and pharmacodynamic 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 pharmacodynatnic data sets and the characteristics of the underlying
`models for a given drug. In an attempt to minimize these factors, Foseau and
`Sheiner (18} proposed that the phartnacodynainic component of the combined
`model could be modeled non-parametrically using the relationship between
`observed effect and the effect compartment drug concentration (Ce) predicted
`using a parametric pharmacokinetic model. To achieve this, it is necessary to
`assume that
`the relationship betvveen Cs and effect
`is instantaneous and
`invariant with time, i.e.,‘ tolerance and sensitization do not occur. As in the
`parametric approach, the effect compartment is modeled as receiving negligible
`
`InnoPharma Exhibit 1029.0016
`
`

`

`3i}
`
`....31...No
`
`.935).-_
`
`6OIOmcu...)
`
`O9w?
`
`xxnHmxuxm.
`uu“auauxxnunW,
`
`Nauru»?uxuxuW»
`
`dmmm‘0um...wm.wnwdu.aw86w
`
`
`
`
`
`«.NwwN“,wovw9NW0ovmmwN"m0cmwwmu,mofittifllqlxxij114154311}1114'le
`
`3mm.wg885%2E.
`
`uQnESCQmOQ
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`on?93338mm3“803%“me8Go1338wmgwmmmofimbwfigmwwowafifiafimafimwSwatHmumwomoonmbfim$8605.w83mg
`
`
`Eomwwnfiumfig"SBEES950%SufiummfionvMakmfifiomEH8@838magmammSofia
`
`InnoPharma Exhibit 10290017
`
`
`

`

`PHARMACOKINETIC; PHARMACODYNAMIC MODELING
`
`31
`
`
`
`amounts of drug with the concentration profile determined by Kim and the
`plasma drt g concentration (Cpl. In the non-paz‘ametric pharmacodynamic
`approach, ysteresis between effect and Ca is suppressed by choosing K50 such
`that the ascending and descending arms of the effect-Ce plot are sanct-
`imposable (18). The best estimate of K53 is determined using a univariate search
`method wh'ch minimises the average squared difference between observed and
`interpolated effect values from the hysteresis plot as shown in Figure 9.
`Fuseau and Sheinet (18) tested the nonparametric pharmacodynamic method
`using simu ations based on hyperbolic and sigmoid Emax models as well as
`models of the g3~function {convex Cc-E relationship), tolerance) sensitization
`and non-eouilibrlum between Ca 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 Ema): models and the #3 function model when adequate numbers
`of data having minimal error were used. itlowever> the non—parametric method
`could not provide accurate or precise estimates of Kgo when applied to data from
`the tolerance, sensitization or nonequilibrium simulation models. Additionally,
`performance was reduced for all simulation models omen too few data or noisy
`data were used.
`
`Subsequently, Unadkat at of. (19) extended the nonapatametric pharmaco—
`dynamic approach to include pharmacokinetic modeling such that pharmaco»
`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/pharmaco
`dynamic: modelling. The advantages of this approach are that fewer assumptions
`about either the underlying pharmaeokinetie or pharmacodynamic model are
`
`lIl
`
`
`
`
`
`l
`t
`I
`l
`i
`e
`I
`l
`I
`l
`l
`l
`i
`l
`l
`|
`lI
`g
`I
`I
`I
`g
`l
`I
`II
`t
`l
`l
`r
`I
`Mf“
`CaH Cai
`cog?
`
`Figure 9. Application of the non—parametric pharmacodynamie method of Fuseau and
`Sheiner {18) to estimate K59 by minimizing the average squared difference between
`observed {Eu and Egg) and interpolated (53mg) effect data. Ce values are corresponding
`effect compartment concentration values estimated using a parametric pharmacoltinetlc
`model {From reference 18, with permission)
`
`InnoPharma Exhibit 1029.0018
`
`

`

`32
`
`OVERVIEW OF PHARMACODYNAMZCS
`
`required. Unadltat at of. {19} described this as a two-stage process where
`observed Ce values are used to model pharmacodynamics and determine the
`linking KEG value. Simple linear interpolation is used to estimate Cit—time
`values if missing from the Clo—effect data set as shown in Figure 10. The
`resultant (Sp-time data set is used to estimate Ce as a function of time by
`numerically integrating the following equation for a given value of Kg; (19):
`
`aegis: = K] ~ op - Kay Ce
`
`(8}
`
`Where K1 is effect compartment input rate constant (assumed to be equal to
`K50} and all 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
`K50 value which minimizes the area within the hysteresis loop is found.
`This approach assumes that Cs and hence effect is a function of observed (and
`interpolated) C}? as determined by the value of K503 independent of intrinsic
`pharmacoklnetics. Based on a series of simulations, the authors (19) suggested
`that this approach is nearly as efficient as the parametric approach even when
`
`
`
`
`
`
`Plasmaconeentration
`
`Time
`
`Figure 19. Example of non-parametric ‘fit’of plasma concentration (Ce) versus time
`data {G}. If Co was not observed at a pharmacodynamic observation time, it is estimated
`using linear interpolation between the nearest bracketing observed values (8339-) and
`Cpiz +}) (From reference 19, with permission)
`
`InnoPharma Exhibit 1029.0019
`
`

`

`PHARMACOKlNETIC;PHARMACODYNAMIC MODELING
`
`33
`
`the underlying models were known, but considerably more robust when the
`underlying models were misspecitied.
`Shafcr at all. (20) reported a comparison of the above method with parametric
`pharmacokinetic/pharmacodvnamic modeling in the evaluation of the neuro-
`museular blocking drug metocurine in laboratorfi,t animals. The semi-parametric
`method was found to give results in close agreement with the parametric
`method. When results differe

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