throbber

`
`
`
`
`
`
`
`1d!J93nU9V\lJ0l41nVVd'H|N1d!J93nU9V\|J0l—l1nVVd'H|N1d!J93nU9V\|J0ll1nVVd'H|N
`
`
`
`
`
`
`
`
`
`INS)‘
`NIH Public Access
`st‘I:
`/6
`gfié) 5‘ Author Manuscript
`HEP~\lSz‘
`
`01¢
`
`Published in final edited form as:
`
`D1ugMetab P11am1ac0k1'11et. 2009 ; 24(1): 16-24.
`
`Scaling Pharmacodynamics from In Vitro and Preclinical Animal
`
`Studies to Humans
`
`Donald E. Mager*, Sukyung Wool, and William J. Jusko
`Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, New
`York, USA
`
`Summary
`
`An important feature of mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) models is
`the identification of drug- and system-specific factors that determine the intensity and time-course
`of pharmacological effects. This provides an opportunity to integrate information obtained from 111
`Vitro bioassays and preclinical pharmacological studies in animals to anticipate the clinical and
`adverse responses to drugs in humans. The fact that contemporary PK/PD modeling continues to
`evolve and seeks to emulate systems level properties should provide enhanced capabilities to
`scale-up pharmacodynamic data. Critical steps in drug discovery and development, such as lead
`compound and first in human dose selection, may become more efficient with the implementation
`and further refinement of translational PK/PD modeling. In this review, we highlight fundamental
`principles in pharmacodynamics and the basic expectations for 111 Vitro bioassays and traditional
`allometric scaling in PK/PD modeling. Discussion of PK/PD modeling efforts for recombinant
`human erythropoietin is also included as a case study showing the potential for advanced systems
`analysis to facilitate extrapolations and improve understanding of inter-species differences in drug
`responses.
`
`Keywords
`
`allometric scaling; cell life span models; mechanism-based modeling; pharmacodynamics, PD;
`pharmacokinetics, PK; receptor occupancy; recombinant human erythropoietin, rHuEpo; target-
`mediated drug disposition, TMDD
`
`Introduction
`
`The extrapolation of 111 511100, 111 Vitro, and preclinical animal studies to predict the likely
`pharmacokinetic properties of drugs in humans now appears within reach, largely due to
`advancements in physiologically-based pharmacokinetic (PBPK) modeling. L2) Whereas
`traditional allometry continues to prove useful under certain conditions for inter-species
`scaling of PK properties, significant progress has been achieved by transitioning from
`models of data (e.g., classic compartmental models) to those of biological systems. The
`PBPK modeling approach provides a framework for integrating drug-specific calculated
`parameters (e.g., octanolzwater and bloodztissue partition coefficients) and 111 Vitro
`measurements (e.g., plasma protein binding and hepatocyte intrinsic clearance) with
`physiological system-specific parameters (e.g., tissue volumes and blood flows). Given the
`
`‘To whom correspondence should be addressed: Dr. Donald E. Mager, Department of Pharmaceutical Sciences, University at Buffalo,
`SUNY, Buffalo, New York 14260, USA. Tel. +1-716-645-2842 (ext. 277), Fax. +1-716-645-3693, dmager@buffalo.edu.
`lPresent address: Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, Maryland 20892, USA.
`Full text of this paper is available at http://www.jstage.j st. go.jp/browse/dmpk
`
`|nnoPharma Exhibit 1028.0001
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJOHWVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 2
`
`relative success of anticipating human exposures to drugs and toxicants, 3=4) there is
`considerable interest in the development of techniques for the scaling of pharmacodynamic
`systems. Although drug responses are considerably more complex than processes controlling
`pharmacokinetics, the shift from empirical to mechar1ism-based PK/PD modeling5=6) should
`provide the best means for translating 1'11 Vjtro and ammal data to human climcal
`pharmacology. 7)
`
`In this review, we discuss the basic tenets of pharmacodynarnics, namely 1)
`pharmacokinetics or drug exposure as the driving function, 2) capacity-lin1itation of drug-
`receptor interactions, and 3) physiological turnover processes and functional adaptation or
`homeostatic feedback mechanisms. As with PBPK models, these basic components identify
`drug and system specific properties that might be anticipated using 111 Vjtro assays,
`allometry, and/or preclimcal ar1imal experiments. A case study showing how human
`responses to recombinant human erythropoietin (rHuEpo) can be predicted from scaling a
`mathematical model developed in rats is provided as an example of utilizing mechar1ism-
`based PK/PD models to scale complex pharmacological systems.
`
`Basic Principles of Pharmacodynamics
`
`The basic tenets of pharmacokinetics (PK), pharmacology, and physiology continue to form
`the basis for contemporary pharmacodynamic systems analysis (Fig. 1). Pharrnacokinetics,
`or the processes controlling the time-course of drug concentrations in relevant biological
`fluids, tissues, and sites of action (biophase), is the driving force for subsequent
`pharmacological and most toxicological effects. Although mammillary plasma clearance
`models (simple linear compartmental models) and area/moment analysis are the most
`commonly applied techmques for characterizing the absorption and disposition (distribution
`and elimination) properties of drugs, PBPK models provide a comprehensive platform for
`describing the major processes influencing the concentration time-course and net exposure
`of drugs in various fluids and tissues (Fig. 1, left panel). Each tissue of interest is
`anatomically arranged and described by a series of mass balance differential equations.
`Fick’s law of perfusion/diffusion and drug partitiomng are featured along with a capacity-
`limited function for various drug binding, transport, and elimination processes. This
`approach provides insights into expected drug concentrations in important tissues, and
`potentially sites of action, and the intrinsic scalability of predictions across species and
`molecular drug properties is unparalleled. Whereas traditional PBPK model development
`has relied on destructive sampling in preclimcal studies, advances in nomnvasive imaging
`(such as positron emission tomography and magnetic resonance imaging) and microdialysis
`may eventually provide even finer details of 111 V1'Vo drug disposition.8=9)
`
`At the biophase, the law of mass action and the limited concentration of pharmacological
`targets often manifest as nonlinear, capacity-lin1ited systems”) The rate of change of a
`dmg-receptor complex (RC) can be defined as:
`
`dRC
`7:/can ' (Rfot _
`
`' C _ kolf '
`
`(1)
`
`where Rm is the maximum receptor concentration, C is the drug concentration at the site of
`action, and km and kogare the second-order association and first-order dissociation rate
`constants. Assuming equilibrium conditions, this equation can be rearranged to yield the
`general binding equation:
`
`RC:
`
`Rtot ‘ C
`KD+C (2)
`
`Drug Metab Pliazmacokinet. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0002
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0Ll1nVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 3
`
`where KD is the equilibrium dissociation constant (lfoff/[(011). Based on Clark’s theory of
`receptor occupancy, the stimulus or drug effect (15) can be directly proportional to the
`fraction of occupied receptors, such that E=cL ~ RC, thus deriving a classic form of the Hill
`equation or sigmoid Em,“ model:
`
`Ernax ' C7
`E: ECO’
`Cw
`‘ 50+ ’
`
`(3)
`
`where limax is the maximum effect, 7 (or Hill coefficient) is a slope term that reflects the
`steepness of the effect-concentration curve, and the EC50 is a sensitivity parameter
`representing the drug concentration producing 50% of Emax. The typical stimulus/effect-log
`concentration relationship is thus curvilinear, and typical profiles for varying values of y are
`shown in the center panel of Figure 1.
`
`In contrast to the linear transduction of receptor occupancy (Eq. 3), Black and Leff
`introduced the operational model of agomsm to provide a mechanistic interpretation of
`concentration-effect curves“) The stimulus or effect is assumed to be nonlinearly related to
`the dmg-receptor complex:
`
`Emax ‘RC
`' KE+RC
`
`(4)
`
`where KE is the RCvalue producing half-maximal effect. Combimng Equations 2 and 4
`yields:
`
`_ Ernax ‘T‘C
`_KL,+(T+l)-C (5)
`
`where EmaXis a system maximum and ( represents a transducer or efficacy function (R,0,/
`KE). This model can accommodate complex relationships, such as partial agomsm, where
`observed capacity and sensitivity properties are actually hybrid terms composed of drug
`specific (KD and t) and system specific (Emax) parameters. Regardless of whether linear or
`nonlinear transduction is operational, capacity-limitation is a hallmark property of
`pharmacology, and consequentially, a suitable range of dose-levels (or concentrations) are
`required to define the parameters of such systems. In addition, the implementation of
`Equation 5 requires pharmacodynamic data, or at least prior information, on the properties
`of a full agonist to identify the maximal system response.
`
`Physiological turnover processes and homeostatic feedback mechamsms represent the third
`major component of pharmacodynamics (Fig. 1, right panel). An open system for a
`biological substance, R, with zero-order production (k,~,,) and first-order removal (kout) can
`be defined by the following differential equation:
`
`dR/dt=/an - kout -R (6)
`
`Assuming a time-invariant baseline or steady-state, the imtial or baseline value (R0) can be
`defined as the ratio of the production and loss terms: R°=1(,~,/kout. A family of basic indirect
`response models apply to many drugs where interaction with the pharmacological target (Eq.
`3) serves to inhibit or stimulate either 19,, or kw, 12) A series of transit compartments can
`also be factored into such models to emulate time-dependent transduction processes that
`often exhibit significant onset delays and exposure-response hysteresis. 13) Knowledge of the
`turnover rates for physiological system components is important for the identification of the
`rate-lin1iting steps for specific pharmacological responses and might impact study design.
`Such information might also facilitate the characterization of feedback mechamsms that
`
`Drug Metab Pliazmacokinet. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0003
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0Ll1nVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 4
`
`might result in tolerance and/or rebound phenomena.6) As both drugs and diseases often
`interfere with normal physiological processes, the turnover aspect of both indirect response
`models”) and transduction models”) renders them well suited for the simultaneous
`
`consideration of these factors in the time-course of disease progression.
`
`Mechanism-based models seek to integrate these basic components to identify critical
`pharmacological and (patho)-physiological system properties as well as the rate-limiting
`steps in responses to drugs.6=16) Useful models with a potential for translational medicine
`also provide a structural framework for incorporating 111 517100, 111 Vitro, and preclimcal PK/
`PD measurements to predict the effects of new drugs in humans and across levels of
`biological organization (Fig. 2). A discussion of all these methods is beyond the scope of
`this review, which will focus on 111 Vitro assays and allometric principles in the context of
`mechamstic PK/PD models. The derivation of quantitative structure-PK/PD relationships (111
`517100 modeling) to predict the exposure-response profiles of new chemical entities has been
`recently reviewed?)
`
`Extrapolation of In Vitro Bioassays
`
`Pharmacodynamic modeling of several systems has revealed that properties of drug
`interactions with pharmacological targets measured 1'11 Vitro may be correlated with specific
`model parameters often reflective of drug potency. Shimada and colleagues developed an
`ion-channel binding model based on 111 Vitro binding data of calcium channel antagomsts,
`which demonstrate relatively slow rates of association and dissociation”) The
`pharmacologic effect was assumed to be proportional to the concentration of the drug-
`receptor complex and, as an extension of Equation 1, the rate of change of the effect was
`defined as:
`
`%:krJn ' (Emaix _ E) ' C _ kofl ' E (7)
`
`The inclusion of the binding parameters was sufficient to explain the hysteresis observed
`between the PK and antihypertensive effect of eight calcium channel antagomsts in Japanese
`patients. The calculated KD values based on estimates of km and kogwere shown to be
`sigmficantly correlated with those obtained from 111 Vitro experiments. These results suggest
`that PK and 111 Vitro binding data alone could be used to predict the pharmacodynamic
`profile of future drugs in this class. Kalvass and colleagueslg) performed extremely
`insightful PK/PD studies with seven opioids in mice showing the importance of time-course
`of brain distribution and binding in deterinimng their antinociceptive effects. The EC50 of
`unbound drugs in brain showed excellent correlation with 111 Vitro receptor binding affimties
`(KD). From a drug development perspective, these examples demonstrate how 111 Vitro
`assays may be coupled with useful PK/PD models to anticipate the outcomes of similar
`compounds and may guide lead compound selection.
`
`Relative receptor affimty has been shown to be correlated with 111 V1'Vo estimates of drug
`potency for several drugs, and 111 Vitro measurements could be used in scaling of EC50
`values across species. In a 5-way randomized placebo-controlled crossover study aimed at
`evaluating the dosing equivalency of four systemically admimstered corticosteroids,
`mechamsm-based PK/PD models were used to estimate EC50 values for several
`immunomodulatory effects, including cortisol suppression, lymphocyte and neutrophil
`trafficking, and ex V1'Vo inhibition of lymphocyte proliferation. 1930) The estimated potencies
`for all of these responses were highly correlated with relative receptor affimty (1'11 Vitro KD
`values normalized to dexamethasone). Differences in protein homology and other genetic
`sources of variability may result in altered drug binding affimty among species. Chien and
`colleagues corrected an EC50 value for a competitor drug measured in humans, using several
`
`Drug Metab P11am1ac0k111et. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0004
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0Ll1nVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 5
`
`factors including receptor binding, to predict the 111 V1'Vo human EC50 for a new chemical
`entity (NCE):21)
`
`EC
`
`5O.NCE,hu1ndu‘
`
`-EC
`
`50.CuIupeLiLo1',hu1uL11A '
`
`EC5U.NCE
`
`(EC50,con1pct1t01‘
`
`_
`
`)
`
`rat
`
`' ‘sfu ' 6K!)
`
`(8)
`
`where Sfu and SKD are correction factors for differences in the free fraction in plasma (fa)
`and binding affimty (KD). For example,
`
`6K _ KD.l1ux1ia11
`D" K
`D,rat
`
`KD.ra.t
`' K
`D.hiuu-an C0n1petjt01-
`
`NOE
`
`(9)
`
`The scaled EC50 from ammal and 111 V1'tro data (Eq. 8) was coupled with other projected
`parameters to simulate a dose-response curve (Eq. 3 with an added baseline) for a new
`antihypertensive agent, relative to a competitor, in the preclimcal phase of development.
`Monte Carlo simulations included a relatively large confidence interval about expected
`outcomes; however, data from climcal studies would eventually be used to confirm and
`update the model.
`
`Traditional Allometric Scaling in PK/PD
`
`Although the structural nature of physiologically-based models makes them uniquely suited
`for scaling and predicting human drug exposures, the extrapolation of PK-PD models from
`ammals to humans is primarily based on classic allometric principles.22) There is a general
`expectation that many physiological processes and organ sizes (9) tend to obey a power
`law:23)
`
`6=u- Wk
`
`(10)
`
`with Wrepresenting body weight and a and b as dmg/process coefficients. The exponent, 1),
`tends to be around 0.75 for clearance processes, 1.0 for organ sizes or physiological
`volumes, and 0.25 for physiological times or the duration of physiological events (e. g.,
`heart-beat and breath duration, cell lifespans, and turnover times of endogenous substances
`or processes).24) A theoretical basis for allometric scaling has been proposed by West and
`colleagues based on the fractal nature of biological systems and energy conservation
`principles.25) Empirical models have also been coupled with allometric relationships and 111
`V1'tro metabolism experiments using nonlinear mixed effects modeling to improve the
`scalability of such models.26:27)
`
`The basic expectations in pharmacodynamics are that physiological turnover rate constants
`of most general structures and functions should be predictable among species based on
`allometric principles, whereas capacity (Emax) and sensitivity (EC50) parameters tend to be
`similar across species. Brodie and colleagues were the first to examine some PK-PD
`properties across species, revealing inter-species differences in duration of action and
`biological half-life, but similarity in plasma concentrations on awakemng (i.e., concentration
`producing a standard response analogous to an EC50), following hexobarbital
`admimstration.28) There has long been a general belief that the plasma drug concentration
`required to elicit a certain (intensity of) action is often similar in experimental animals and
`humans”) While interspecies differences in relative receptor affimty and plasma protein
`binding occur (Eqs. 8 and 9),“) there are several examples that show reasonable agreement
`of such properties between rats and humans for chemically-related series of drugs. Ito and
`colleagues demonstrated a linear correlation between the logarithm of KD values of
`benzodiazepines in rat and human cerebral cortex tissue over several orders of magmtude.3°)
`
`Drug Metab P11a1mac0k111et. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0005
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0Ll1nVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 6
`
`Cox and coworkers also showed good agreement for the EC50 values of four synthetic
`opioids between these same species“) Mechanistic modeling was applied to PK/PD data
`for S(+)-ketoprofen obtained from several species, and estimated parameters further support
`these basic expectations. Pharmacokinetic parameters were shown to scale proportionally to
`body weight (albeit with unusual power coefficients), and anti-inflammatory PD parameters
`exhibited lin1ited ranges that were essentially independent of body weight”)
`
`Mechanism-Based PK/PD Modeling of rHuEpo
`
`To demonstrate the use of scaling principles within mechar1ism-based PK/PD models, we
`present here scaled phannacodynamic responses using a rat model of rHuEpo PK/PD to
`anticipate the time-course of several biomarkers in humans. This drug is climcally indicated
`for the treatment of specific types of anemia, and binding of this endogenous protein to its
`biological receptor (EPOR) expressed by progemtor cells in bone marrow elicits
`proliferation and differentiation of erythroid cells, thereby increasing reticulocytes, red
`blood cells, and hemoglobin concentrations in blood. Erythropoietin exhibits a high degree
`of homology among mammals, which explains the conserved biological activity of rHuEpo
`in various species.
`
`The disposition of rHuEpo in several species is polyexponential and nonlinear, and typical
`PK profiles have been described using a two-compartment model with a concentration-
`dependent Michaelis-Menten elimination function operating in parallel with a linear
`nonsaturable clearance pathway.33’35) Target-mediated drug disposition (TMDD) represents
`a likely explanation for the capacity-limited elimination of erythropoietin; a condition where
`a sigmficant proportion of the drug (relative to dose) is bound to its pharmacological target,
`such that this interaction influences the PK properties of the drug.36=37) Receptor-mediated
`endocytosis is a major clearance mechanism for many protein drugs,38) and this saturable
`process can result in nonlinear drug disposition.39) The binding of erythropoietin to EPOR is
`specific and results in saturable internalization of the dmg-receptor complex. 40) Chapel and
`colleagues demonstrated that bone marrow ablation in sheep produced a sigr1ificant decrease
`in erythropoietin clearance, providing experimental evidence that target binding and
`transport plays a major role in the 111 V1'Vo disposition of erythropoietinf”) Interestingly, the
`simultaneous modeling of PK profiles of rHuEpo from a wide-range of intravenous dose
`levels in rats, monkeys, and humans revealed that full and reduced TMDD models42:43) well
`characterized rHuEpo disposition and provided a basis for linking an established
`pharmacodynamic model.44)
`
`Woo and Jusko have provided a comparison of interspecies PK/PD properties of rHuEpo.45)
`Although the prospective use of allometric scaling can be limited,46) it is generally
`considered that peptide and protein drugs are more likely to exhibit allometric PK
`relationships than small molecules owing to the relative species conservation of mechamsms
`that control the biodistribution and elimination of such compounds.47’49) Despite the non-
`linear disposition of rHuEpo, total systemic clearance and the steady-state volume of
`distribution show good correlation with body weight. The exponent for clearance (0.708)
`was close to the expected value of 0.7 5; however, the exponent for the volume of
`distribution (0.853) was slightly lower than the expected value (1.0). Pharmacokinetic model
`specific parameters, such as Michaelis-Menten capacity or Vmax, the central volume of
`distribution, and a first-order rate constant of absorption, also scaled to body weight with
`exponents of 0.504, 0.983, and -0.349, respectively (based on rat, monkey, and human
`data). As anticipated, the pharmacological capacity and sensitivity parameters were
`essentially species-independent.
`
`Drug Metab Pliazmacokinet. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0006
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJOHWVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 7
`
`We sought to predict the time-course of reticulocytes, red blood cells, and hemoglobin
`concentrations in humans after rHuEpo administration from an established PK/PD model
`developed in rodents. Pharmacodynamic data were extracted from a climcal study in which
`healthy male volunteers were given 150 IU/kg subcutaneously (SC) three times weekly for 4
`weeks.34) The general structure of the PK/PD model for rHuEpo developed from rat
`preclimcal data is shown in Figure 3.35) The PK component of the model can be described
`by:
`
`dA”=in ut(t)=
`dt
`p ‘
`
`Km _ ‘/p+AP
`
`+k
`pt
`
`el
`
`-A +k -A
`p
`I]:
`
`I
`
`11
`
`)
`
`(
`
`LIA
`d\f=pr-Ap—/WA:
`
`<12)
`
`where Ap and At represent the amounts of rHuEpo in the central and tissue compartments,
`Vmax and Km are Michaelis-Menten parameters, Vp is the volume of the central rHuEpo
`compartment, [(61 is first-order elimination rate constant, and kpt and Iqp are first-order
`distribution rate constants. The imtial conditions of Equations 11 and 12 are zero, and the
`input function after SC drug admimstration is defined as:
`
`I
`
`t
`
`=
`
`npu (0 { ka - F - fr - Dose - e‘k""’"T)
`
`F~(l—fr)~Dose
`T
`
`T
`
`,0<t < T
`’
`_
`
`;I>T
`
`'
`
`(13)
`
`where F is bioavailability, firis the fraction of the dose undergoing first-order absorption
`(kg), and ‘I: is the time period of zero-order input. This input function is based on the
`complex absorption profile due in part to the sigmfrcant role of the lymphatics in the uptake
`of proteins admimstered subcutaneously.5°=51)
`
`The catenary PD model (Fig. 3) contains two precursor compartments (P1 and P2) linked to
`reticulocyte (REY), red blood cell (RBC), and hemoglobin (Hb) compartments. This model
`mimics the process of erythropoiesis from bone marrow to blood, and is based on cell life
`span concepts integrated into indirect response models for drugs that alter the generation of
`natural cells”) Cells are assumed to be produced at a constant rate, circulate for a specific
`duration of time (Ti), and are then eliminated from the system not by a first-order process,
`but at the same rate as the input, delayed by the cell life span (senescence or conversion to
`another cell type). The precursor compartments represent early progemtor cells and
`erythroblasts, and TP1 and TP2 are the average times taken for cells to differentiate. The
`rates of change of the reticulocyte (RE?) and mature RBC (RBCM) counts are described by:
`
`JRE T
`
`dt
`
`:kir1'S
`
`Tpl T}12)'S (I
`
`Tpl T112) kirL'S
`
`Tpl Tp2
`
`(t—Tp2—TRE'I‘)'I(t—T}1l—T}I2—TRE’1‘)
`
`(14)
`
`Drug Metab Pliazmacokinet. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.000?
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJOHWVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 8
`
`dRBCM
`dt
`
`=k,-,7 . 5 (z
`
`_ P1 _ TF3 _ TRET) ' SO
`
`— T1,; — TRET) - I(t
`— T,,1 — T,,2 — Tm) — k,-,, .50
`
`(15)
`
`_ Pl _ TP2 _ TRET _ TRBC) ' SO
`
`_ TF3 _ THET _ REC) ' [(1
`
`_ 1'1 _ T112 _ TRET — TRBC)
`
`with TRETand TR30 as the average life span of these cells, and 19,, is the zero-order
`production rate constant. The initial conditions for Equations 14 and 15 are RE7}) and
`RBC0—RET0, where RE1}) and RBC0 are baseline measurements. The stimulation function
`S(t) was defined as:
`
`5(1):]
`
`S C5()+AP(I)/VP
`
`(16)
`
`where Smax is the maximal stimulation factor and SC50 is the rHuEpo concentration
`resulting in 50% of Smax. Hemoglobin concentrations were calculated as the product of the
`mean corpuscular hemoglobin (MCH; measured) and sum of RET and RBCM (Eqs. 14, 15).
`A counter regulation feedback loop is also included, I(t), driven by the difference in Hb from
`baseline values, and was defined as:
`
`[max '
`=1 _T
`IC5o+AI-Ib(z)
`
`1(1)
`
`(17)
`
`where the maximal inhibition factor (lmax) was fixed to 1, and [C50 is the Hb difference
`from baseline producing 50% feedback inhibition.
`
`The parameter values, their sources, and scaled-up values in humans used for the PK/PD
`model simulations are listed in Table 1. Inter-individual variability (IIV) for each parameter
`used in the Monte Carlo simulations is also reported. Volume, clearance, and first-order rate
`constants were scaled with allometric exponents of 1, 0.75, and -0.25. The baseline values
`for RET, RBC, and MCH (and their respective IIV) were considered drug and species
`independent and were set to literature values for humans.53) Life span parameters were
`scaled using an allometric exponent of 0.124 which was previously estimated using RBC
`data obtained from over 20 species/*5) Only nominal variability was assigned to PK terms
`(10% CV%), whereas CV% values were set to 20% for Smax and 30% for sensitivity
`parameters (SC50 and IC50).
`
`Monte Carlo simulations were conducted using ADAPT II (Biomedical Simulation
`Resource, USC, Los Angeles), and mean observed data and model predicted profiles are
`shown in Figure 4. The predicted values of the three biomarkers are in good agreement with
`observed data, which fall well within the 90% prediction interval (gray areas). The
`successful scaling of the rat PK/PD model of rHuEpo to human responses demonstrates how
`basic allometric principles and preclimcal data may be integrated using mechar1ism-based
`models to make useful predictions. It is important to recognize that the biomarkers of drug
`activity and preclimcal PK/PD models must be meaningful across species. The likelihood of
`these appears to be greater for macromolecules as compared to small molecules;54) however,
`a similar interspecies scaling approach was shown to apply to two 5-HT1A receptor
`agomsts.55)
`
`Drug Metab Pliazmacokinet. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0008
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0Ll1nVVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 9
`
`In summary, the scaling of pharmacodynamic data relies heavily on the ability to predict and
`integrate the fundamental processes controlling drug exposure (pharmacokinetics), drug
`action (pharmacology), and interactions with physiological systems. Preclimcal data and 111
`V1'tro bioassays can provide important insights into these properties, especially as
`pharmacodynamic parameters tend to be species independent; however, it is important to
`verify whether measurements of drug effects are meaningful across species.
`Notwithstanding the linritations of prospective allometry, such power law relation-ships
`have proven useful in scaling-up physiological turnover processes and PK properties for
`many drugs. New techmques are needed to identify conditions under which allometric
`scaling may or may not be appropriate in PK/PD models. Physiologically-based PK models
`will likely become commonplace given their intrinsic potential for projecting human PK
`properties from 111 V1'tro and 111 517100 measurements and data obtained in other species.
`Ammal studies can provide preliminary data for the development of mechanism-based PK/
`PD models, which will continue to evolve toward efficient descriptions of pharmacological
`systems. Such models offer the best approach toward effectively combining and interpreting
`the major determinants of drug action across species.
`
`Acknowledgments
`
`This research was supported in part by Grant GM 57980 from the National Institutes of Health (to W.J. J.), and a
`New Investigator Grant from the American Association of Pharmaceutical Scientists (to D.E.M.).
`
`References
`
`1. Nestorov I. Whole-body physiologically based phannacokinetic models. Expert Opin. Drug. Metab.
`Toxicol. 2007', 3:235-249. [PubMed: 17428153]
`2. Mager DE. Quantitative structure-pharmacokinetic/pharmacodynamic relationships. Adv. Drug
`Deliv. Rev. 2006', 58:1326-1356. [PubMed: 17092600]
`3. Beliveau M, Lipscomb J, Tardif R, Krishnan K. Quantitative structure-property relationships for
`interspecies extrapolation of the inhalation phannacokinetics of organic chemicals. Chem. Res.
`Toxicol. 2005', 18:475-485. [PubMed: 15777087]
`4. Parrott N, Paquereau N, Coassolo P, Lave T. An evaluation of the utility of physiologically based
`models of pharmacokinetics in early drug discovery. J. Pharm. Sci. 2005', 94:2327—2343. [PubMed:
`161 36543]
`5. Danhof M, de Jongh J, De Lange ECM, Della Pasqua O, Ploeger BA, Voskuyl RA. Mechanism-
`based phannacokinetic-phannacodynamic modeling: Biophase distribution, receptor theory, and
`dynamical systems analysis. Annu. Rev. Pharmacol. Toxicol. 2007', 47:21.21-21.44.
`6. Mager DE, Wyska E, Jusko WJ. Diversity of mechanism-based phannacodynamic models. Drug
`Metab. Dispos. 2003', 31:510-518. [PubMed: 12695336]
`7. Mager DE, Jusko WJ. Development of translational phannacokinetic-pharmacodynamic models.
`Clin. Pharmacol. Ther. 2008', 83:909-912. [PubMed: 18388873]
`8. Brunner M, Langer O. Microdialysis versus other techniques for the clinical assessment of in vivo
`tissue drug distribution. AAPS J. 2006', 8:E263-E271. [PubMed: 16796376]
`9. Liefaard LC, Ploeger BA, Molthoff CF, Boellaard R, Lammertsma AA, Danhof M, Voskuyl RA.
`Population pharrnacokinetic analysis for simultaneous detennination of B (max) and K (D) in vivo
`by positron emission tomography. Mol. Imaging. Biol. 2005', 7:411-421. [PubMed: 16328648]
`10. Ariens EJ. Affinity and intrinsic activity in the theory of competitive inhibition. Arch. Int.
`Phannacodyn. Ther. 1954', 99:32-49. [PubMed: 13229418]
`11. Black JW, Leff P. Operational models of pharmacological agonist. Proc. R. Soc. Lond. B. Biol.
`Sci. 1983', 220:141-162. [PubMed: 6141562]
`12. Dayneka NL, Garg V, Jusko WJ. Comparison of four basic models of indirect pharmacodynamic
`responses. J. Phannacokinet. Biopharm. 1993', 21:457-478. [PubMed: 8133465]
`13. Mager DE, Jusko WJ. Pharmacodynamic modeling of tirne-dependent transduction systems. Clin.
`Phannacol. Ther. 2001', 70:210-216. [PubMed: 11557908]
`
`Drug Metab Plrazmacokirret. Author manuscript; available in PMC 2013 July 30.
`
`|nnoPharma Exhibit 1028.0009
`
`

`

`
`
`
`
`
`
`
`
`1d!J93nU9V\|JOHWVVd'H|N1d!J39nU9V\lJ0l-110VVd'H|N1d!J93nU9V\|J0l«|1hVVd'H|N
`
`
`
`
`
`
`
`
`
`Mager et al.
`
`Page 10
`
`14.
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`
`28.
`
`29.
`
`30.
`
`31.
`
`32.
`
`Post TM, Freijer JI, DeJongh J, Danhof M. Disease system analysis: basic disease progression
`models in degenerative disease. Pharm. Res. 2005', 22: 1038-1049. [PubMed: 16028004]
`Earp JC, Dubois DC, Molano DS, Pyszczynski NA, Keller CE, Almon RR, Jusko WJ. Modeling
`corticosteroid effects in a rat model of rheumatoid arthritis I: mechanistic disease progression
`model for the time course of collagen-induced arthritis in Lewis rats. J. Phannacol. Exp. Ther.
`2008', 326

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