throbber
See discussions, stats, and author profiles for this publication at: http://www.researchgate.net/publication/13877791
`
`Basic concepts of
`pharmacokinetic/pharmacodynamic (PK/PD)
`modelling
`
`ARTICLE in INTERNATIONAL JOURNAL OF CLINICAL PHARMACOLOGY AND THERAPEUTICS · NOVEMBER 1997
`
`Impact Factor: 1.22 · Source: PubMed
`
`CITATIONS
`83
`
`2 AUTHORS:
`
`READS
`2,113
`
`Bernd Meibohm
`The University of Tennessee Health Science …
`
`Hartmut Derendorf
`University of Florida
`
`125 PUBLICATIONS 2,811 CITATIONS
`
`432 PUBLICATIONS 8,778 CITATIONS
`
`SEE PROFILE
`
`SEE PROFILE
`
`Available from: Hartmut Derendorf
`Retrieved on: 28 September 2015
`
`Page 1 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`/11rernat/011af.lour'nal of Clinical Pharmacology and Therapeutics, Vol. 35, No. /0- 1997 (401-413)
`
`Basic concepts of pharmacokinetic/
`pharmacodynamic (PK/PD) modelling
`
`B. MEIBOHM and H. DERENDORF
`
`Department of Pharmaceutics, College of Pharmacy.University of Florida. Goinesvillr'. Fl., USA
`
`Abstract. Pharmacokinetic (PK) and pharmacodynamic (PD) information from the scientific basis of
`modern pharmacotherapy. Pharmacokinetics describes the drug concentration-time courses in body
`fluids resulting from administration of a certain drug dose, pharmacodynamics the observed effect
`resulting from a certain drug concentration. The rationale for PK/PD-modelling is to link pharmaco(cid:173)
`kinetics and pharmacodynamics in order to establish and evaluate dose-concentration-response
`relationships and subsequently describe and predict the effect-time courses resulting from a drug dose.
`Under phannacokinctic steady-state conditions, concentration-effect relationships can be described
`by several relatively simple pharmacodynamic models, which comprise the fixed effect model, the
`linear model, the log-linear model, the Emax-model and the sigmoid Emax-model. Under non steady(cid:173)
`state conditions, more complex integrated PK/PD-models are necessary to link and account for a
`possible temporal dissociation between the plasma concentration and the observed effect. Four basic
`attributes may be used to characterize PK/PD-models: First, the link between measured concentration
`and the pharmacologic response mechanism that mediates the observed effect, direct vs. indirect link;
`second, the respons.e mechanism that mediates the observed effect, direct vs. indirect response; third,
`the information used to establish the link between measured concentration and observed effect, hard
`vs. soft link; and fourth, the time dependency of the involved pharmacodynamic parameters, time(cid:173)
`variam vs. time-invariant. In general, PK/PD-modelling based on the underlying physiological process
`should be preferred whenever possible. The expanded use of PK/PD-modelling is assumed to be highly
`beneficial for drug development as well as applied phannacotherapy and will most likely improve the
`current state of applied therapeutics.
`
`Key words: pharmacology pharmacokinetics - phannacodynamics modelling
`
`Rationale for PK/PD-modelling
`
`The rational use of drugs and the design of effective
`dosage regimens is facilitated by the appreciation of the
`relatiunship5 between the administered dose of a drug, the
`resulting drug concentrations in body fluids accessible for
`measurements, and the intensity of pharmacologic effects
`caused by these concentrations l Gihaldi et al. 1971 J. These
`relationships and thus the dose of a drug required to achieve
`a certain effect is determined by its pharmacokinetic and
`pharmacodynamic properties.
`Pharmacokinetics describes the time course of the
`concentration of a drug in a body fluid, preferably plasma
`or blood, that results from the administration of a certain
`dose. In simple words, phannacokinetics is · wlwr rile hody
`does to the drug'. Pharmacodynamics describes the inten-
`
`Correspondence lo: Prof. Dr. H. Derendorf, 100494. College of
`Pharmacy. University of Florida. Gainesville. FL 32610, USA
`
`sity of a drug effect in relation to its concentration in the
`body tluid. It can be simplified to 'what the drug does to
`the body' [Holford and Sheiner 1982].
`Pharmacokinetic/pharmacodynamic (PK/PD)-mod(cid:173)
`elling combines both approaches and tries to estabiish
`models in order to describe the effect-time course directly
`resulting from the administration of a certain dose (Fig(cid:173)
`.ure I). Thus, a so-called integrated PK/PD-model consists
`of a pharmacokinetic model component that describes the
`time course of a drug in some body fluid and a pharma(cid:173)
`codynamic model component that relates the concentration
`in this fluid to the drug effect.
`
`Models in general are simplified descriptions of true
`biological process and can be used for data reduction and
`interpolation, but their major value is derived from their
`ability to extrapolate relationships beyond the ~xisting
`data. In the case of PK/PD-models, that means to extrapo(cid:173)
`late dose-effect relationships for example from single to
`multiple dosing situations or from intravenous to oral or
`other administration routes.
`
`Page 2 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`. 402
`
`MPihnhm and DPrPndnrf
`
`Pharmacokinetics
`Dose¢ Cone.vs.time
`
`Time
`
`Pharmacodynamics
`Conc.¢Effect
`
`PK/PD
`Dose¢ Effect vs.time
`
`TI mo
`
`Fig. I
`Interrelationship
`between phannacokinetics,
`pharmacodynamic' and
`PK/PD-modelling.
`
`Evolution of PK/PD-modelling
`
`Early in the evolution of pharmacokinetics, relation(cid:173)
`ships between drug disposition and pharmacologic effect
`have been described [Gibaldi and Levy 1972, Gibaldi et al.
`1971, Levy 1964, Wagner 1968]. It could be shown that
`the intensity and time course of action of numerous directly
`and reversibly acting drugs are related to and largely deter(cid:173)
`mined by the time course of drug concentrations in the
`body. When the concentration-time courses and the con(cid:173)
`centration-effect relationships ot these drugs were known,
`it was often possible to predict the temporal pattern of their
`pharmacological effects, including maximum intensity and
`duration of action.
`However. for many other drugs, it was believed that
`there is no relationship between the drug concentration in
`plasma and the time course of action, predominantly hased
`on the observation that the pharmacologic effect of many
`drugs lags behind their concentration in plasma. Pharma(cid:173)
`cologic effects often increase in their intensity despite
`decreasing drug concentrations and may persist well be(cid:173)
`yond the time, when drug concentrations in plasma are no
`longer determinable. As a consequence plasma concentra(cid:173)
`tion vs. effect plots show a more or less pronounced hys(cid:173)
`teresis.
`The apparent dissociation between drug concentra(cid:173)
`tion and effect was first overcome by Sheiner and co-work(cid:173)
`ers [Holford and Sheiner 1982, Sheineret al. 1979] based
`on the concepts of Segre f 1968 J who proposed to use a
`hypothetical effect compartment to account for the lag
`between concentration and response, which is described in
`detail in one of the following sections of this paper. This
`approach led to a collapse of the hysteresis loop for drugs
`with a temporal delay between effect and plasma concen(cid:173)
`tration by plotting the effect intensity versus the concentra(cid:173)
`tion in the effect compartment. All of a sudden, the delay
`
`between the time courses of drug concentration and effect
`made sense [Levy 1994].
`Since then, PK/PD-modelling has exponentially
`evolved as a research area with increasing importance and
`dedication in academia, industry and regulatory authori(cid:173)
`ties. The present paper aims to give a short overview over
`the basic concepts in pharmacokinetic/pharmacodynamic
`modelling and provide some structural information for
`classification of the applied approaches.
`
`Pharmacokinetic models
`
`The plasma concentration-time course of a drug is
`determined by the pharmacokinetic processes of distribu(cid:173)
`tion, metabolism and excretion as well as absorption in case
`of nonsystemic administration. The currently used pharma(cid:173)
`cokinetic models can basically be distinguished into com(cid:173)
`partmental, physiological and statistical models.
`Although nonparametric and physiologically based
`pharmacokinetic models have been used as a basis for
`PK/PD-approaches [Holford et al. 1994, Unadkat et al.
`1986, Veng-Pedersen and Gillespie 1988], compartmental
`models are the most frequently preferred, probably due to
`the fact that they provide a continuous concentration-time
`profile in a body fluid that can be related to a continuous
`effect-time profile and that the popular effect compartment
`concept can easily be implemented. Thus, the present paper
`will focus on PK/PD-modelling based on compartmental
`pharmacokinetic models.
`
`Pharmacodynamic models for steady-state situations
`
`Pharmacodynamic analysis involves quantifying
`drug concentration/effect relationships. Ideally, concentra(cid:173)
`tions should be measured at the effect site, the site-of-action
`or biophase, where the interaction with the respective bio-
`
`Page 3 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Basic concepts ofpharmacokineric!pharmacodynamic (PK/PD) modelling
`
`403
`
`logical receptor system takes place, but this is in most cases
`not possible. Thus, concentrations in easily accessible body
`fluids like plasma or blood are frequently used to establish
`these relationships under the assumption, that the pharma(cid:173)
`cologically active, unbound concentration at the effect site
`is directly related to the one in the respective body fluid
`and that they are, under phannacokinetic steady-state con(cid:173)
`ditions, in equilibrium. Since the same proportionality
`holds for different steady-state plasma levels, steady-state
`plasma concentrations may serve as the only determinant
`of the observed effect.
`A drug effect can be defined as any drug-induced
`change in a physiological parameter when compared to the
`rc:spective prt::dost:: or bast::line value. The ba:seli11e value is
`the value of the same physiological parameter in the ab(cid:173)
`sence of drug dosing. Baseline values do not necessarily
`have to be constant but can change, e.g. as a function of
`time of day or of food intake. Furthermore, the term effect
`has to be clearly separated from the term efficacy. Efficacy
`is the sum of all therapeutically beneficial drug effects and
`is the most relevant target parameter in PK/ PD-modelling.
`However, in many PK/PD-studies, efficacy is difficult to
`quantify and thus, easily accessible surrogate markers as
`effect parameters are used instead. In these cases, it is
`necessary to present evidence that the pharrnacodynamic
`effect parameter used correlates with the desired efficacy
`to provide valid results.
`For steady-state conditions, the most commonly·used
`phannacodynamic models are the
`
`-
`
`fixed effect model,
`linear model,
`log-linear model,
`-
`- Emax-model, and
`sigmoid Emax-rnodel.
`-
`
`Fixed effect model
`
`A fixed effect model, also known as quanta! effect
`model, is a statistical approach based on a logistic regres(cid:173)
`sion analysis. It relates a certain drug concentration with
`the statistical likelihood of a predefined. fixed effect to be
`present or absent.
`The simplest case of a fixed effect model is a threshold
`model, where the effect Efoed occurs after reaching a
`certain threshold concentration Cthrcshold, as for example
`described for the ototoxicity occurring during gentamicin
`therapy with trough levels exceeding 4 ~Lg/ml for longer
`than l 0 days of therapy ! Mawer et al. l 97 4 j:
`
`E
`
`Et1xed
`
`if
`
`C 2: Critreshold
`
`(Eq. I)
`
`certain concentration will be a function of the threshold
`concentration distribution in the population. For example,
`at a digoxin plasma concentration of 2.0 ng/ml there is a
`50% probability to observe digox.in toxicity, whereas at a
`concentration of 4.1 ng/ml the probability is 90% [Beiler
`et al. 197 l, Holford and Sheiner 1982]. This approach may
`be useful in the clinical setting as an approximation of
`dose-response relationships but has major limitations for
`the prediction of complete effect-time profiles.
`
`Linear model
`
`The linear model assumes a direct proportionality
`between drug concentration and drug effect, as shown by
`Weaver et al. [ 1992] for the correlation of salivary flow rate
`and plasma concentration after pilocarpine infusions:
`
`E=m xC+ Eo
`where Eo is the baseline effect in the absence of drug
`and ma proportionality factor, that characterizes the slope
`of a plot of effect E versus concentration C. Although the
`linear model is the one that intuitively is the most popular,
`it rarely applies.
`
`(Eq. 2)
`
`log-linear model
`
`A much more common situation than the linear model
`is the log-linear model with
`
`E""' m x log C + b
`
`(Eq. 3)
`
`where m and bare slope and intercept in a plot of effect
`E versus the logarithm of the concentration C. Although b
`should have the unit of the effect, it is an empiric constant
`that has no real physiologic meaning, especially not that of.
`a baseline value. The log-linear model is applicable in
`many situations and can be considered a special case of the
`Emax-model, regarding the range between 20% to 80% of
`Emax, where effect E and logarithm of the concentration C
`follow a linear relationship. Nagashima et al. [ 1969] for
`example used it to relate the synthesis rate of prothrombin
`complex activity to the plasma concentration of warfarin.
`
`Einax-model
`
`In the maximum effect Ema"-model, concentration C
`and effect E are related as
`
`E- Emax xC
`Eso-1 C
`
`(Eq.4)
`
`where E is the measured effect and C is the measured
`concentration.
`Since the threshold concentration will vary among
`patients, the probability of the effect to be present at a
`
`where Emax is the maximum effect possible and E5o
`is the concentration that causes 50% of Emax. Emax refers
`to the intrinsic activity of a drug, E5o to its potency. Lalonde
`et al.! 1987) gave an ex.ample for applying the Emax-model
`
`II
`
`Page 4 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`404
`
`100
`
`80
`
`60
`
`40
`
`20
`
`u Q)
`~
`E
`:J
`E
`·;;:
`!1l
`E
`0
`~ 0
`
`slope: Ema/4
`
`100
`
`80
`
`60
`
`40
`
`20
`
`b
`
`intercept: In E50 - 2
`
`!
`//
`
`0-4-...... ~~.,.;._~~~~~~~~
`-4 -3 -2 -1 0 1 2 3 4 5 6 7 6 9 10
`
`Concentration (ng/ml)
`
`In C (ng/ml)
`
`Meibohm and Derendorf
`
`Fig. 2 Concentration-effect
`relationship resulting from a
`phurmacodynamic Emax-model,
`where the effect is either plotted
`against the concentration (a) or
`the logarithm of the concentra(cid:173)
`tion (b), the Inst resulting in the
`classical sigmoidal shape. [Em ax
`maximum effect, Eso
`con(cid:173)
`centration at 50% ofEmaxl.
`
`by describing the relationship between propranolol plasma
`concentrations and the resulting decrease in heart rate.
`The equation of the Emax-model (Eq. 4) is based on
`the receptor theory relationship [Ariens and Simonis 1964]
`that can be derived for the equilibrium interaction of a drug
`(D) with its site of action (R), e.g. a receptor, enzyme or
`ion channel, producing the effect E:
`
`_
`[D]+[R]<=:>[DR]Hc:ffect =>
`
`[DR]-
`
`[nto,]x[D]
`[ ]
`Kd+ D
`
`(E 5)
`q.
`
`where Kct is the equilibrium constant and Riot the total
`number of interaction sites. Under the assumption, that the
`observed effect E is directly proportional to the number of
`occupied interaction sites DR, Eq. 4 and Eq. 5 are equiva(cid:173)
`lent indicating that maximum effect would be observed if
`all interaction sites are occupied. Kct is the concentration at
`which half of the interaction sites are occupied and, hence,
`equivalent to Eso.
`The Emax-model describes the concentration-effect
`relationship over a wide range of concentrations from zero
`effect in the absence of a drug to the maximum effect at
`concentrations much higher than Eso (C >> Eso). In the
`presence of a baseline effect Eo, this term can simply be
`added to Eq. 4 as shown in Eq. 6:
`xc
`£ 50 +C
`The clear non-proportional concentration-effect rela(cid:173)
`tionship of the Emax-model is presented in Figure 2 as linear
`and semilogarithmic plot. Whereas small increases in con(cid:173)
`centration may result in significant increases of the effect
`for low concentrations, this is much less pronounced for
`higher concentrations where only small changes in effect
`
`(Eq. 6)
`
`E=E0
`
`will result from changes in concentration. From the
`semilogarithmic presentation, it is apparent that in the
`range from 20% to 80% of the maximum effect, the rela(cid:173)
`tionship between effect and the logarithm of the concentra(cid:173)
`tion is linear. This is consistent with the log-linear model
`(Eq. 3). The slope of the linear phase can be calculated as
`EmaJ4, the respective x-intercept as In Eso - 2, and the
`y-intercept b as Emax x (2
`In Eso)/4, [Hochhaus and
`lJerendort 1995 J. At concentrations below 20% and above
`80% of the maximum effect the Emax-model clearly devi(cid:173)
`ates from the log-linear model. For concentrations much
`smaller than Eso (C << Eso), it reduces to a linear model
`with a slope m of Emax/Eso. Hence, both, the log-linear as
`well as the linear model may be interpreted as special case
`of the Emax-model.
`The Emax-model presented in Eq. 4 assumes an in(cid:173)
`crease of the effect with increasing concentrations, i. e. a
`stimulated effect. Opposite, inhibitory effects can be de(cid:173)
`scribed by Eq. 7:
`
`xC
`E10 +C
`where Eo is the baseline effect. If the maximum effect
`Emax is complete suppression of the baseline effect Eo, Eq.
`7 simplifies to Eq. 8:
`
`(Eq. 7)
`
`E=E0x(1--C-)
`
`(Eq. 8)
`
`E5o+C
`In accordance with the receptor theory the Emax(cid:173)
`model also allows to describe more complex concentra(cid:173)
`tion-effect relationships, e.g. competitive or noncompeti(cid:173)
`tive agonist and antagonist interactions at the response
`system, if appropriately modified Emax-equations are ap(cid:173)
`plied. This was shown by Braat et al. 11992] for the com-
`
`Page 5 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Basic concepts of pharmacokineticlpharmacodynamic (PK/PD) modelling
`
`405
`
`100
`
`80
`
`60
`
`40
`
`20
`
`Q)
`
`u
`~
`E
`:::>
`E ·x
`ro
`E
`0
`~ 0
`
`0
`
`0
`
`Fig. 3 Concentration-effect re(cid:173)
`lationship resulting from a sig(cid:173)
`moid Emax-model, presented a~
`a function of various shape fac(cid:173)
`tors in a normal (a) and
`semilogarithmic plot (b).
`
`shape factor
`
`40
`
`a
`
`shape factor
`
`b
`
`2
`3
`4
`Concentrationti;0
`
`5
`
`101
`
`1 ()l
`
`1 ()1
`
`Concentrationll::so
`
`petitive effect of dexamethasone and hydrocortisone on
`lymphocytes.
`
`Sigmoid Emax-model
`
`The sigmoid Emax-model is an expansion of the Emax(cid:173)
`model. Effect and concentration are related as
`
`(Eq.9)
`
`xC"
`E
`£- ma,\
`E;o +C"
`Theoretically, this relationship can be derived to de(cid:173)
`scribe the interaction between n drug molecules and one
`interaction site similar to Eq. 5. However, in most cases n
`has no molecular basis and is merely used as an operational
`shape factor that allows a better data fit. This also explains
`why in many studies noninteger values for n are reported
`which could not be possible based on the mentioned deri(cid:173)
`vation. Figure 3 shows the effect of different values of non
`the concentration-effect curves. The larger n, the steeper
`the linear phase of the log-concentration-effect curve. It
`could be shown that the slope is n x Emax/4, hence directly
`proportional to n. For this reason, n is also referred to as
`the slope factor. The respective x-intercept is then In Eso
`2/n fHochhaus and Derendorf 1995]. The Emax-model can
`be considered as a special case of the sigmoid Emax-model
`with a slope factor n = l.
`The sigmoid Emax-model is the most versatile model
`of those presented. Meffin et al. f 1977} fur exam pit: used
`it to characterize the relationship between plasma concen(cid:173)
`tration and antiarrhythmic activity of tocainide. The re(cid:173)
`ported slope factor of n 2.3 - 20.6 for individuals indi(cid:173)
`cates a very steep concentration-effect curve.
`In summary, several relatively simple pharmacody(cid:173)
`namic models are available to describe the relationship
`
`between drug concentration and effect under steady-state
`conditions. The preferred model in any given situation
`depends on many factors, including the drug used, the
`response to be measured, the effect seen after administra(cid:173)
`tion of drug and of placebo, the degree of linearity in the
`effect-concentration-curve, and the potential for achieving
`the maximum possible response.
`
`Pharmacodynamic models for non steady-state
`situations
`
`Under non steady-state conditions, drug concentra(cid:173)
`tions in plasma (or respective other sampled body fluids
`like blood. or serum, which will be summarized in the
`expression plasma from hen: on) undergo time-dependent
`changes due to the involved pharmacokinetic process and
`an equilibrium between plasma and effect site concentra(cid:173)
`tion does not necessarily exist. Furthermore, another time
`consuming process might be involved in mediating the
`observed effect after interaction of the drug with its physi(cid:173)
`ological response structure. As a result, time courses of
`plasma concentration and effect might dissociate. Thus, to
`characterize the time course of drug action under non
`steady-state conditions, pharmacokinetics and pharma(cid:173)
`codynamics have to be adequately linked to predict firstly
`the dose/concentration-relationship, and secondly the con(cid:173)
`centration/effect-relationship. This link is provided by the
`multiple concepts of integrated PK/PD-models.
`
`Four basic attributes of PK/PD-models
`
`At present, usually applied integrated PK/PD-models
`for reversible drug effects can be classified by four major
`attributes (Figure 4), characterizing the link between the
`
`-
`
`••
`
`Page 6 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`406
`
`Meibohm and Derendorf
`
`Dose-concentration-effect relationship
`to be modeled
`
`Dhec.• Link
`
`""·
`
`Indirect Link
`
`Olrv4:,;l fi'npvnH'
`vs.
`lndlroct Response
`
`.•.
`
`Hiird Link
`
`Soft Link
`
`Time~lnv111d•11t
`vs .
`Time~variant
`
`Selected PK/PD-approach
`
`•
`
`•
`
`.,
`lso
`Q. «> w
`.5 30
`
`10
`
`I J ~
`
`0 ......... --.;:::::;;;.,.~....,....--.~--~...-....... ~..--.
`0
`2
`4
`IS
`I
`10 12 14
`tlS 18 ~
`Cp (µ.g/m~
`
`Fig. 4 Four basic attributes of PK/PD-models to be considered during
`the selection of an appropriate modelling approach.
`
`Fig. 5 Counter-clockwise hysteresis loop for the relationship between
`plasma concentration (Cp) of $-ibuprofen and its analgesic effect quanti(cid:173)
`fied as decrease in evoked potential (EP) amplitudes [from Suri et al.
`1997).
`
`plasma concentration and the response mechanism respon(cid:173)
`sible for the observed effect, the response mechanism by
`which the effect is mediated, the information used to estab(cid:173)
`lish the link between plasma concentration and observed
`effect, and the time-dependency of the involved parameters
`of the pharmacodynamic model component. The resulting
`alternatives are
`Direct link vs. indirect link models
`- Direct response vs. indirect response models
`- Soft link vs. hard link models
`Time-invariant vs. time-variant models
`
`Direct link versus indirect link
`
`Plasma and effect site concentrations can be linked
`either directly or indirectly, dependent on their temporal
`arrangement. Due to the constant change in drug plasma
`levels during non steady-state situations, the equilibrium
`between the concentrations in plasma and at the site of
`action 1s permanently readjusted. Since this re-equilibra(cid:173)
`tion is dependent on the involved distribution process and
`more or less time consuming, the concentration at the site
`of action may lag behind that in plasma. This is reflected
`in a more or less pronounced counter-clockwise hysteresis
`between plasma concentration and effect, as shown in
`Figure 5 for the analgesic effect of S-ibuprofen quantified
`by the amplitude decrease of evoked potentials [Suri et al.
`1997]. The extent of hysteresis is then dependent on the
`degree of delay between the concentrations in plasma and
`at the effect site.
`
`and at the effect site occurs rapidly and the concentrations
`are directly proportional to each other at any time despite
`the pharmacokinetic non steady-state conditions. Thus, the
`time of maximum measured concentration is also the time
`of maximum effect, if the pharmacodynamic response is
`directly mediated (see following chapter). In such situ(cid:173)
`ations effect and plasma concentration lack any hysteresis
`and may be linked by simple substitution of the pharma(cid:173)
`cokinetic model for concentration C in one of the pharma(cid:173)
`codynamic models for steady-state situations. Schaefer et
`al. [ 1997] for example used plasma concentrations directly
`in an Emax-model to describe the effect-time course, quan(cid:173)
`tified as blood pressure reduction, after different doses and
`dosage forms of nisoldipine (Figure 6).
`If the pharmacokinetic properties of a drug are de(cid:173)
`scribed by a multicompartmental phannacokinetic model,
`the effect might not only be related to the central compart(cid:173)
`ment, but also to the concentration predicted for one of the
`peripheral compartments. This approach was used hy
`Hochhaus et al. [ l 992b] to characterize the pulmonary and
`cardiac effects of fenoterol (Figure 7), where the plasma
`concentration-time profile was described with a three com(cid:173)
`partment mammillary model and the concentration pre(cid:173)
`dicted for the shallow tissue compartment was used as an
`input function for a sigmoid Emax-model. Derendorf et al.
`[ 1991] used a similar approach with a two companmem
`model to relate fasting blood sugar levels to methylpred(cid:173)
`nisolone concentrations in the peripheral compartment. For
`most drugs, however, a specific compartment does not
`reflect the drug concentration at the site of action.
`
`Direct link models
`
`lndin:cl link rrwdels
`
`A direct link has to be applied in a situation where
`equilibration between the drug concentrations in plasma
`
`An indirect link is needed if the time courses of
`concentration and effect are dissociated and the observed
`
`Page 7 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Basic l'Oncepts of pharmacokineticlpharmacodynamic (PK/PD) modelling
`
`407
`
`a
`
`b
`
`c
`
`Dose
`Plasma concentration
`First-order rate constant
`
`a
`
`D
`
`D
`Dose
`C
`Plasma concentration
`c01 , cp2 Peripheral compartment
`concentration
`k10, k~ 1 , k,,, k.,, k 13
`First-order rate constants
`ko
`Zero-order rate constant
`
`I Effect I
`
`b 1600
`
`::::;
`e
`
`' Cl s-
`ti z
`0
`u
`
`1200
`
`800
`
`400
`
`tw-+1----1}
`t+h
`
`U
`
`H
`
`JO
`
`l4
`
`0
`
`0
`
`40
`
`80
`
`120
`TIME (mlnl
`
`160
`
`200
`
`240
`
`c
`
`•
`
`Time !hi
`
`•
`
`~r.::============;~~~~~~~~~-,
`
`_...... pndlacd ~· t_ mw.:1 I
`I -0- metl' \-al1,1e ~f ,wdy OIQ.4 \
`
`l 5
`
`'i
`;:: 2S
`~
`.! JO
`=
`= J5
`i 10
`
`•
`...
`-.-:r-;-AA-------"--------~
`....
`-~~.!
`~AA--..A..--x------~----­
`----0
`--'2..------
`,1-1{ 0
`10
`0 I
`........... ________________ /
`o/'
`I
`
`• . .,,,(cid:173)
`
`.,...
`./
`
`0
`
`10•& !'( N11t-<W'I'
`u .....
`
`5-s N .._...
`
`10.1 N MhtiM
`
`lO•t N tOhltiN
`
`•o+-~-+-~-+~~'--~f--~+-~~~-+~--1
`1.00
`D.00
`2.00
`3.00
`Time (hr)
`
`Fig. 6 Direct link model for the effect of oral nisoldipine (Nl on diastolic
`blood pressure (DRP): (a) PK/PD-model scheme: (b) plasma concentra(cid:173)
`tion vs. time and (c) effect vs. time profiles for different doses and dosing
`forms (study means) ffrom Schaefer et al.1997].
`
`counter-clockwise hysteresis in a plot of effect versus
`plasma concentration is most likely related to a distribu(cid:173)
`tional delay. The important conceptual advance in indi(cid:173)
`rectly linking pharmacokinetic and pharmacodynamic
`models was the realization that the time course of the effect
`itself can be used to define the rate of drug movement to
`the effect site as expressed in the effect compartment model
`of Holford and Sheiner [ 1981 a, 1982]
`
`Fig. 7 Direct link model for the effect of fenoterol on heart rate and
`airway resistance'. (a) PK/PO-model scheme; (b) plasma concentration vs.
`time profile after i. v. bolus (25 µg; solid line) or constant i. v. infusion
`incl. loading dose (200 µg/180 min+ 25 µg loading dose; dashed line;
`• ti) and airway resistance(•. 0)
`mean± SD); (c) effect on heart rate (
`vs. time pmfilf' ~ftl"r boil" (0. i\) resp. constant infusion(•.
`) (study
`means. lines are modeled) [from Hochhaus ct al. 1992a].
`
`This concept uses a hypothetical effect compartment
`that is modeled as an additional compartment of a pharma(cid:173)
`cokinetic compartment model and represents the active
`drug concentration at the effect site. It is linked to the
`kinetic model by a first-order process but receives negli(cid:173)
`gible mass of drug. Therefore, the first-order rate constant
`
`-
`
`...
`
`Page 8 of 14
`
`YEDA EXHIBIT NO. 2038
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`. 408
`
`Meibohm and Derendorf
`
`for the transfer from the respective pharmacokinetic com(cid:173)
`partment into the effect compartment is negligible. Thus,
`the time-dependent aspects of the equilibrium between
`plasma concentration and effect are only characterized by
`the first-order rate constant ke0, which describes the disap(cid:173)
`pearance of the drug from the effect compartment and is
`not directed to any of the pharmacokinetic compartments.
`The time course of the effect site concentration (Ce)
`for a one compartment body model with bolus input is
`
`a
`
`0
`
`D Dose
`C Plasma concentration
`c. Effect compartment
`concentration
`k10• k1 • k10, koa
`First-order rate constants
`
`ka0 ~::~-.~~) ~ I Effect I
`
`Effect compartment
`
`(Eq. 10)
`
`where Dis the dose, Vd the volume of distribution in
`the central compartment, and k the first order elimination
`rate constant of the drug. The respective equations for other
`compartment models and input functions are described
`elsewhere in detail [Colburn 1981, Holford and Sheiner
`198lb].
`Suri et al. [ 1997] used the effect compartment concept
`to model the analgesic effect of S-ibuprofen, Sheiner et al.
`[ 1979]
`to describe
`the muscle relaxant effect of
`d-tubocurarine. In the first example, the effect compart(cid:173)
`ment was attached to a pharmacokinetic one-compartment
`body model (Figure 8), in the last to the central compart(cid:173)
`ment of a three-compartment body model (Figure 9). For
`other drugs, it might theoretically also be possible to link
`the effect compartment to peripheral pharmacokinetic
`compartments as
`theoretically evaluated by Colburn
`[ 1981 ]. However, this seems only adequate, if the drug
`concentration is also measured in this pharmacokinetic
`compartment, for example by invasive techniques or by
`imaging technology. Otherwise, basing a hypothetical ef(cid:173)
`fect compartment concentration on a predicted peripheral
`compaitment concentration seems vague and obscures the
`elaborated PK/PD-relationship.
`The effect compartment concept achieved wide popu(cid:173)
`larity in PK/PD-modelling of various drugs. However, the
`link between measured plasma concentration and observed
`effect is based on an unk.11ow11 1m::cha11is111, a "black. bux".
`Therefore, the validity of the relationship has to be estab(cid:173)
`lished thoroughly by evaluating concentrations and effects
`under a variety of doses and input functions, before predic(cid:173)
`tions and extrapolations to other therapeutic situations may
`be performed.
`
`1~-1-. ....... ..,...,.. .................. .,.......,...,...,..,..,~ ................ .....-. ....... ..,...,...-j
`0
`
`Tme(hra)
`
`..
`
`c
`

`
`i•
`~
`i5., ID
`
`... Q c10
`i;.ll "' ·~.
`" £ ID
`
`40
`
`•
`s
`Time (hrs)
`
`Fig. 8
`Indirect link model for the analgesic effect of 400 mg oral
`ibuprofen quantified by subjective pain intensity rating: (a) PK/PD-model
`scheme; (b) plasma concentration vs. time courses of the enantiomcrs
`R-ibuprofen (OJ and S-ibuprofen (•), (c) effect vs. time profile modeled
`with the concentration of the active S-enantiomer (mean± SD. lines arc
`modeled) [from Suri et al. 1997].
`
`Direct response versus indirect response
`
`Direct response models
`
`Two general pathways can be distinguished for medi(cid:173)
`ating the effect of a drug, a direct response pathway, where
`the interaction of the drug with a response structure at its
`effect site directly results in the observed effect, and an
`indirect response pathway, where a physiological factor
`that governs the observed effect, is modulated.
`
`Direct response models

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