throbber
Chapter 8
`Foundations of Pharmacodynamic
`Systems Analysis
`
`William J. Jusko
`
`Abstract The pillars of pharmacodynamic modeling are the pharmacokinetics of
`the drug, the nature of the pharmacology that underlies drug interactions with their
`targets, and the physiology of the system considering molecular to whole body
`levels of organization and functioning. This chapter provides a general assessment
`of the fundamental components and some interactions of each of these pillars
`indicating how they serve as building blocks for systems models. Key elements of
`pharmacokinetics include the operation of Fick's Laws for diffusion and perfusion
`along with the often nonlinear mechanisms of drug distribution and elimination.
`Target- binding relationships in pharmacology evolve from the law of mass action
`producing capacity -limitation in most operative control functions. Mammalian
`physiology and pathophysiology feature a wide breadth of turnover rates for bio-
`logical compounds, structures, and functions ranging from rapid electrical signals to
`lengthy human lifespans, which often determine the rate -limiting process and basic
`type of model to be applied. Appreciation of the diverse array, mechanisms, and
`interactions of individual components that comprise the pillars of pharmacody-
`namics can serve as the foundation for building more complex systems models.
`
`Keywords Fick's laws
`Target- binding Drug- biological interface Affinity
`Turnover Homeostasis
`Substrate control Operational efficacy
`Capacity
`Gaddum equation
`
`8.1
`
`Introduction
`
`The three pillars of pharmacodynamics (PD), as depicted in Fig. 8.1, are the
`pharmacokinetics (PK) of the drug, the pharmacology and mechanism of the
`drug- biological interface, and the physiology or pathophysiology of the system
`
`W.J. Jusko (2)
`Department of Pharmaceutical Sciences, University at Buffalo,
`404 Kapoor Hall, Buffalo, NY, USA
`e -mail: wjjusko @buffalo.edu
`
`© American Association of Pharmaceutical Scientists 2016
`D.E. Mager and H.H.C. Kimko (eds.), Systems Pharmacology and Pharmacodynamics,
`AAPS Advances in the Pharmaceutical Sciences Series 23,
`DOI 10.1007/978-3-319-44534-2_8
`
`161
`
`Page 1
`
`SHIRE EX. 2068
`KVK v. SHIRE
`IPR2018-00290
`
`

`

`162
`
`W.J. Jusko
`
`Computation
`
`Mathematics
`
`N
`
`:N=
`co
`
`a
`Lq
`O
`t?
`
`_L
`0i
`_c
`
`'
`
`.
`Statistics
`
`C?
`' o ,

`v
`co
`
`L
`a
`._
`
`Fig. 8.1 The palace of pharmacodynamics with its foundation, structural components, and three
`pillars
`
`being altered by the drug (Mager et al. 2003; Jusko 2013). Each can contribute to
`the extent and time -course of observed pharmacodynamic responses depending on
`their intrinsic properties and rate -limiting step(s). The foundations of systems
`analysis will be explored by delineating the basic "rules of biology" for the major
`components that govern each of the pillars of pharmacodynamics. Along with the
`determinants of PK, two general principles, namely capacity -limitation and turn-
`over, form the basis for a variety of commonly used PK/PD and systems models.
`Genomics is included in Fig. 8.1 as the presence, location, and functioning of
`determinants of PK/PD are governed by genomics and genetics. The quantitative
`skills of mathematics, statistics, and computation are needed to identify relation-
`integrate them into models, analyze experimental data, and perform
`ships,
`simulations.
`
`8.2 Pharmacokinetics
`
`Common approaches for analyzing pharmacokinetic data utilize noncompartmental,
`compartmental (mammalian), and physiological concepts and methods, with
`ascending degrees of complexity. Physiologically -based PK (PBPK) models pro-
`vide mechanistic and insightful separation of drug and systems properties as well as
`their interfaces and interactions. Three key relationships that underpin drug distri-
`butional processes in PBPK models are:
`
`Page 2
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`163
`
`Fick's Law of Diffusion:
`
`dA = PS(Ch - Cl)
`dt
`
`where the rate of drug movement (Amount/time, dA/dt) from higher (Ch) to lower
`(C1) concentrations is governed by the permeability -surface area (PS) coefficient
`(Fick 1855). Permeability (P) is governed by molecular size and lipid solubility of
`the compound along with the nature of the biological membrane and its surface area
`(S). This equation is often invoked to describe small molecule (drug) absorption
`rates, movement between interstitial fluids (ISF) and cell water spaces, and has been
`adapted to account for biophase distribution of drugs.
`
`Fick's Law of Perfusion: d = Q(Ca - C)
`
`where the rate of organ uptake (dA/dt) is governed by arterial (Ca) and venous (Cr)
`drug concentrations and organ blood flow (Q) (Teorell 1937). The ratio of
`(Ca - C,,) /Ca is also termed the Extraction Ratio (ER). This equation is commonly
`used in PBPK models to describe drug distribution to various organs and tissues via
`blood flow.
`
`Convection: d = L(1 - a) C = fL Q(1 - 6) C
`
`where organ uptake of molecules is determined by water movement equaling lymph
`flow (L) and the vascular reflection coefficient (a) associated with water flux across
`capillary membranes into ISF (Renkin 1979). Lymph flow is usually assumed as a
`small fraction (fL = 0.02 -4 %) of blood flow to each organ or tissue as determined
`by the Starling (1896) approximation, while the reflection coefficient varies with
`type of organ capillaries (some `leaky' such as liver, some `tight' such as muscle).
`This equation is used in PBPK models of monoclonal antibodies (mAbs) and other
`large molecules to describe their limited and slow movement from plasma to ISF
`(Cao et al. 2013). Glomerular filtration rate is primarily a convection process as
`well.
`The joint roles of blood flow and permeability for control of the distribution of
`molecules from blood to tissues is termed Distribution Clearance (CLd) in PK and
`quantified as:
`
`CLd=fdQ=Q(1 -e-PS/Q)
`
`where fd is the fraction of Q accounting for organ uptake of drug, PS is the
`rate -limiting factor when Q is small, and Q is the rate -limiting factor when PS is
`large (Stec and Atkinson 1981).
`The array of nonlinear protein binding, metabolism, transport, and clearance
`relationships commonly encountered in PK are listed in Table 8.1 (Jusko 1989).
`
`Page 3
`
`

`

`Wilkinson and Shand (1975)
`
`ConcentrationRowland et al. (1973) and
`
`CLbu: Intrinsic clearance
`
`Goldstein, (1949)
`
`KA: Equilibrium Association constantDf free drug
`
`Shannon (1939)
`Menten (1913)
`ConcentrationMichaelis and
`Substrate
`
`Concentration
`
`References
`
`Km
`
`Km
`
`Affinity
`
`Q: Blood Flow
`Pt: Protein Conc.
`n: No. Binding Sites
`
`Vtt
`
`Vt t,,
`
`Capacity
`
`Q +cL
`Q.c"
`
`=
`
`CL
`
`Clearance
`
`Organ clearance
`
`b = 1 /KA +Df
`
`nPeD1
`K + C
`v,,,x'c
`
`D
`
`dt
`dAT
`
`ar - m +c
`'c
`
`v
`
`a
`
`Equation
`
`Bound drug
`
`Protein binding
`
`Flux
`formation rate
`Metabolite (M)
`Function
`
`Transport
`
`Metabolism
`Process
`
`Table 8.1 Common capacity -limited functions in pharmacokinetics
`
`4,
`
`Page 4
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`165
`
`They all evolve from the law of mass action where the limited quantity of binding
`substances, metabolic enzymes, or transporters results in capacity -limited pro-
`cessing of drugs and other substrates. At low drug concentrations, the functions
`operate linearly, such as with the common relationship for intrinsic clearance,
`CLint = Vm/Km, pertaining to drug metabolism. Often the preferred or operative
`drug concentration is the free or unbound drug in either plasma or in tissues. These
`distributional and elimination relationships are components of full PBPK models
`and are presented here partly owing to their fundamental value in PK, but also
`because they are helpful in describing the kinetics of physiological substances or
`biomarkers when analyzed in PK/PD and systems models. For example, the PK/PD
`modeling of cortisol as an indicator of adrenal suppression and of nitrate as a
`biomarker of inflammation is best handled by considering their intrinsic kinetics
`(Krzyzanski and Jusko 2001; Sukumaran et al. 2012).
`
`8.3 Pharmacology
`
`The interaction of drugs (D) with their biophase targets (R) is the interface that
`controls the array of subsequent genomic, proteomic, biochemical, and physio-
`logical changes. These targets may be receptors, enzymes, transporters, ion chan-
`nels, and/or DNA. A common feature is that the concentration or quantity of such
`targets is limited and can be described with the law of mass action:
`
`as described by:
`
`D
`
`+R DR
`k-
`
`kopf
`
`dR
`dt
`
`k0riDR-k0ffDR
`
`where k0n is the association rate constant, keis the dissociation rate constant and, at
`equilibrium, the equilibrium dissociation constant is KD = k0yk0n. This type of
`interaction leads to a nonlinear relationship that the author calls "The Equation of
`Life ":
`
`Function =
`
`Capacity Substrate
`Affinity -I- Substrate
`
`In this fashion, Capacity, Affinity, and Substrate control numerous biological pro-
`cesses: those involved in PK as listed in Table 8.1 and those describing many
`pharmacological actions as listed in Table 8.2. These pharmacologic processes or
`
`Page 5
`
`

`

`and Ko (1994)
`Dayneka et al. (1993) and Jusko
`
`)
`
`(
`
`Concentration
`
`SC50
`
`Sma,
`
`=1 +^uu+C
`Numerical change
`
`Scso
`
`C
`
`Jusko and Ko (1994)
`Dayneka et al. (1993),
`Zhi et al. (1988)
`Jusko (1971),
`
`Black and Leff (1983)
`
`Hill (1910)
`
`Clark (1933)
`References
`
`Concentration
`n: Power Coefficient
`concentration
`[AR]: Agonist- receptor
`y = Hill Coefficient
`C or C v
`
`Di. free drug
`Substrate
`
`Concentration
`
`IC50
`
`Constant
`KE: Transducer
`
`EC50
`
`Ema,
`
`KD orkt
`13,,
`CapacityAffinity
`
`Imp
`
`K
`
`Em
`
`Fractional Change
`
`Ics0 + c
`'max 'C
`
`1
`
`=
`
`removal
`Altered input or
`
`removal
`Altered input or
`
`Stimulation
`
`Inhibition
`
`Kc50 +c
`Km,. c
`
`dr
`ax
`
`CytotoxicityKilling rate
`
`KÉ + [R]
`E - [ARln'Em
`
`TransductionEffect
`
`=Eqo + 0
`= Kp +Df
`
`Eÿ' CY
`
`Db
`
`Equation
`
`Bound drug
`Function
`
`PD effects
`binding
`Receptor
`Process
`
`E
`
`Direct effect
`
`Table 8.2 Common capacity -limited functions in pharmacology
`
`Page 6
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`167
`
`mechanisms include receptor binding, transduction, cytotoxicity, inhibitory and
`stimulatory changes, as well as simple directly observed drug effects.l
`It is important to appreciate the need for sufficiently high doses or drug con-
`centrations to attain an observed maximum response (Capacity) and the occurrence
`of the Affinity constant at concentrations when responses equal one -half of
`Capacity. Such conditions facilitate operation of all PK/PD models and assist in
`resolution of the parameter values (Dutta et al. 1996; Krzyzanski et al. 2006). Some
`of the relationships include a power coefficient (n, y) accompanying the concen-
`tration and Affinity terms. Although this power coefficient may not have a mecha-
`nistic basis, it sometimes adds flexibility in fitting pharmacological data. Of special
`note is that the Affinity term in the equations is of the nature that lower, rather than
`higher, concentration values reflect greater potency of the drug.
`The simple pharmacologic relationships listed in Table 8.2 have been applied for
`a vast array of drugs and response measures. However, pharmacology textbooks
`may offer additional more complex relationships that have usually evolved from
`in vitro systems (Kenakin 1997). One of note is the Adair equation (1925) relevant
`for biphasic or hormesis drug effects:
`
`E=
`
`EmCaC
`EC50 + C + K2 C2
`
`where K2 is a secondary binding coefficient. This equation produces a bell- shaped
`Effect (E) versus concentration (C) relationship. Cao et al. (2012) applied this
`equation to describe the effects of GLP -1 on stimulating insulin secretion for a wide
`range of doses examined in rats.
`Another complex relationship of considerable value is the Black and Leff (1983)
`equation for nonlinear transduction of the effects of an agonist:
`
`Em Tn Cn
`Effect = (KD + Cr + 2n Cn
`
`where i is Operational Efficacy defined as Bmax/KE (Other symbols are defined in
`Table 8.2). Resolution of all parameters in this equation may require assessment of
`both nonlinear drug- receptor binding (Bmax, KD) as well as nonlinear responses (Em,
`KE) in relation to receptor occupancy. This equation is of great value as the PK and
`drug- receptor interactions are usually specific for individual compounds, while the
`subsequent events yielding an effect are controlled by the biological transduction or
`signaling system. One applied example is where the plasma concentrations of
`methylprednisolone were found to control receptor binding while the receptor
`
`'The author tells his students that this equation will also predict their future success in pharma-
`cometrics: a function of the combination of brain capacity (IQ), affinity for mathematics, statistics,
`and computation, and the relevant assimilated information (coursework and studies).
`
`Page 7
`
`

`

`168
`
`W.J. Jusko
`
`binding of free and liposomal- incorporated drug served as the basis for the
`time -course of immunosuppression for lymphocytes in the spleen of rats (Mishina
`and Jusko 1994).
`
`8.4 Physiology
`
`Nearly all mechanistic PD models are based on the concepts of turnover and
`homeostasis (Mager et al. 2003; Jusko 2013). Biological compounds (biomarkers),
`structures, and functions are continually being produced and degraded. The starting
`condition or baseline of most PD models is thus the steady -state that exists in the
`organism. Numerous physiological controls can be invoked to maintain home-
`ostasis of the system and factor being measured.
`Figure 8.2 provides a listing of many biological entities that have served as PD
`measures. Their time -frames for turnover range from very fast (electrical signals) to
`very slow (human lifetimes). The factors in the upper part of the list are often
`biomarkers of body processes while the lower components may require clinical
`measures of major organ or system functioning (e.g., arthritis or depression
`symptom scores). Of course, patient survival is a key endpoint in cancer
`chemotherapy where measurements are made in a population sense.
`The turnover rate may determine which type of PK/PD model applies. For very
`rapid turnover processes, direct effect or biophase models are relevant as the PK of
`the drug will be rate - limiting in controlling observed responses. When the pro-
`duction (kin) and loss (kout) rates of the biological factors are slower and directly
`altered by drugs, indirect response models pertain. As systems become more
`complex with multiple controls, then transduction, multi- component, or systems
`models are needed. These time -frames also determine study designs as slow pro-
`cesses need lengthier monitoring of the response measures.
`
`SY STEM S M ODE l
`
`.
`S
`
`Fig. 8.2 Diversity of
`turnover rates and models
`(adapted from Mager and
`Jusko 2008)
`
`Biological Turnover Rates of Structures or Functions
`Fast Electrical Signals (msec) -,
`Neurotransmitters (msec)
`¡ á
`Chemical Signals (min)
`¡ o
`Mediators, Electrolytes (min) : n
`R
`Hormones (hr)
`1 K
`mRNA (hr)
`(
`)
`Proteins / Enzymes (hr)
`Cells (days)
`
`Direct
`Effect
`Models
`
`Turnover
`Models
`
`Tissues (mo)
`Organs (year)
`Person ( .8 century)
`
`V
`Slow
`
`1 E
`R
`I s
`I
`
`--J
`--%c
`1 Ì
`1 N
`1 1
`
`1 ñ
`-- L
`
`Transduction
`Models
`
`Page 8
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`169
`
`Table 8.3 Basic types of turnover models or components used in pharmacodynamics
`
`Model type
`
`Diagram
`
`Indirect response
`
`k
`
`i-r
`
`Precursor -indirect
`
`kp
`
`P
`
`R
`
`kout
`
`->
`
`Cytotoxiciry
`
`Irreversible
`
`Transit
`
`kg
`
`kin
`
`t
`
`Feedback
`Tolerance (Tol)
`
`kt
`--->
`
`k,
`
`R
`
`k°t
`k1
`
`ti
`
`kt
`
`x
`
`Receptor binding
`Target- mediated
`
`k5,.n 1
`
`C
`
`+
`
`R
`
`k,°ss
`
`kon
`
`k
`
`°"
`
`CR
`
`Input
`function
`kin (drug
`modified)
`Zero -order
`
`kp P (drug
`modified)
`First -order
`
`kg
`First -order
`
`kin
`Zero -order
`
`Loss
`function
`km (drug
`modified)
`First -order
`
`knot
`First -order
`
`k1 C (drug
`modified)
`Second -order
`k1 C (drug
`modified)
`Second -order
`
`First -order
`1/i
`kt
`First -order
`knn C R
`Second -order
`
`First -order
`1/r
`kt
`First -order
`
`ko ff CR
`First -order
`
`References
`
`Dayneka
`et al. (1993),
`Jusko and
`Ko (1994)
`Sharma et al.
`(1998)
`
`Jusko,
`(1971), Zhi
`et al. (1988)
`Yamamoto
`et al. (1996)
`
`Sun and
`Jusko (1998)
`Friberg et al.
`(2002)
`
`Shimada
`et al. (1996),
`Mager and
`Jusko (2001)
`
`R
`
`I Usually nonlinear inhibition: (1
`
`.150
`
`ic +cc
`Usually nonlinear stimulation: (1-I- SCSO+c)
`I Drug- induced loss may be simple 2nd -order k1 C R or nonlinear: (xmc
`
`)
`
`KC50 + C
`
`Turnover and homeostasis are part of most of the basic PD models or compo-
`nents currently known.2 Homeostasis reflects the baseline and final steady -state
`condition of body systems before and after drug administration and the diverse
`feedback and set -point mechanisms that help maintain normal functioning. The
`primary PD turnover models are depicted in Table 8.3 with indication of input and
`loss functions, types of rate processes, and commonly associated pharmacological
`functions. In the receptor and target- mediated models, turnover may be viewed as
`drug binding and dissociating from receptors particularly when kon is relatively
`slow (Swinney 2009). The synthesis (k,) and degradation (kdeg) rates of the
`receptors also contribute to the PD, particularly for longer studies. The Precursor
`model and Feedback component are often employed to account for tolerance and
`rebound phenomena.
`
`2Here turnover is generalized to include any process where the response or control factor is
`affected by production and loss. Some authors consider only basic indirect response models as
`turnover models.
`
`Page 9
`
`

`

`170
`
`W.J. Jusko
`
`The listings in Table 8.3 provide the most basic models or components. Various
`complexities can be added to any model such as transit steps, feedback, additional
`compartments, circadian baselines (Krzyzanski et al. 2000a), disease -altered base-
`lines (Lepist and Jusko 2004), life -span loss ( Krzyzanski et al. 2000b), and/or
`physiological limits in responses (Yao et al. 2006).
`
`8.5 Disease Progression
`
`Disease progression models often reflect the time- course of disturbance of PD
`baselines, turnover components, or subsystems with changes in homeostasis
`(Mould et al. 2007; Earp et al. 2008a, b). A classic disease model for cell prolif-
`eration and tumor growth is the `resistance to death' Gompertz function (1825) as
`shown in the modern convenient form:
`
`N = NSS e- No e
`
`]nNss
`
`-kg.t
`
`where kg is a first -order growth constant, N0 is the initial number, and NSS is the
`steady -state number of cells, tumor size, and/or body mass. This equation accounts
`for early exponential growth with an ultimate attainment of a plateau value.
`The simpler, preferred equation used for size measures or numbers of cells in
`many chemotherapy studies is the logistic function (Robertson 1923):
`
`Growth Rate = kg 1 - N° N
`
`NSS
`
`Cell proliferation rates have also been modeled with an adaptation of the
`Michaelis -Menten equation (Meagher et al. 2004). All of these growth or disease
`models are "Equations of Life" in a somewhat different format as they are nonlinear
`and exhibit capacity -limitation.
`
`8.6 Drug Interactions
`
`The wealth of existing drug -drug interactions and quantitative methods are
`well- appreciated in pharmacokinetics. When two or more drugs are administered,
`additional interactions can occur owing to either the nature of their pharmacological
`mechanisms or their alteration of the same or convergent turnover process or both.
`The isobolograph approach based on Loewe Additivity (1926) is often used in
`assessing pharmacologic interactions of two agents (Gessner 1974). However, this
`and most drug interaction methodology in pharmacology has involved measure-
`ment of a static endpoint and do not take into account the PK/PD time- course of
`
`Page 10
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`171
`
`drug action. Fortunately, time -honored mechanistic equations allow the PK to be
`incorporated into their interaction relationships:
`Gaddum (1937) Equation for Competitive Interactions:
`
`EA+B
`
`EmaxA ' ECCso + EmaxB ECScBOB
`+ 1
`CA +
`
`EC DA
`
`ECSOB
`
`where CA is the concentration of agonist and CB is the concentration of a drug
`competing for the same target site. This equation is applicable for two agents
`typically having similar molecular structures and targets. The Gaddum equation
`simplifies for an antagonist when EmaxB = 0 as:
`
`Effect =
`
`Em,axA CA
`
`CA+EC50Al1+ECo$I
`
`The ability to fully resolve antagonistic drug effects requires careful assessment of
`the actions of the agonist to obtain its EmaxA and EC50A values and further
`examining the offsetting effects of the antagonist in order to calculate EC50B. It is
`difficult to do this in most in vivo studies, but Mandema et al. (1992) accomplished
`this in quantifying the agonist effects of midazolam and antagonistic action of
`flumazenil in studies in human subjects.
`Ariens et al. (1957) provided basic equations for more complex drug interactions
`such as noncompetitive, uncompetitive, and irreversible drug combinations that
`require careful enactment for experimental data. The Kenakin (1997) book provides
`highly useful instructions regarding these diverse relationships.
`If one of these basic mechanistic equations does not suffice in accounting for the
`joint effects of two agents, then an empirical drug interaction parameter (fr) can be
`introduced by multiplying it times one of the EC50 terms. Chakraborty et al. (1999)
`used the inhibitory forms of the Gaddum equation and Ariens equations and
`demonstrated how adding the * term allowed assessment of possible immuno-
`suppressive interactions between IL -10 and prednisolone for inhibiting lymphocyte
`proliferation. In fitting joint drug data, * < 1 reflects synergy and * > 1 reflects
`r does not equal 1, is that a more complex
`antagonism. Another interpretation, if
`mechanism may exist than accounted for by these basic interaction equations.
`Turnover models often allow a mechanistic approach for discernment of natural
`synergy occurring for two or more drugs. Earp et al. (2004) provided equations for
`indirect response models and demonstrated how strong synergism can result when
`there is either joint inhibition of kin and stimulation of kou,. or, conversely, stimu-
`TheThe principle that synergy or augmented drug
`lation of kin and inhibition of
`
`Page 11
`
`

`

`172
`
`W.J. Jusko
`
`effects are produced by opposing drug effects on the two sides of a turnover process
`also applies in chemotherapy when inhibition of growth along with cytotoxicity on
`the loss side occur. This was nicely demonstrated for the effects of rituximab and
`rhApo2L on tumor xenografts in a small systems model enacted by Harrold et al.
`(2012).
`
`8.7 Summary and Prospectus
`
`This compilation of PK/PD models and components provides a `toolbox' of kinetic
`processes, pharmacological functions, and turnover features of major basic models.
`Enhanced PK/PD or small to large systems models can often be assembled by
`consideration of the sequence of events leading to an observed drug effect and
`utilization of the appropriate mechanistic pieces that capture major rate -limiting
`steps. For example, Earp et al. (2008a, b) described disease progression in arthritic
`rats and inhibitory effects of dexamethasone on pro -inflammatory cytokines and
`edema with model components that included PK, receptor binding, transduction,
`competitive interactions, end organ Turnover (paw and bone), and inhibitory
`pharmacological functions. Similarly, Fang et al. (2013) assembled a small systems
`model to account for the diabetogenic effects of methylprednisolone in a
`meta- analysis of receptor, genomic, and biomarker (glucose, insulin, FFA) data
`from several studies in rats. The similarity that exists for numerous capacity -limited
`pharmacologic and disease progression functions and the fundamental nature of
`diverse turnover processes greatly facilitates the meshing of these components in
`assembling complex models.
`
`Acknowledgments This work was supported by NIH Grants GM 24211 and GM 57980 and by
`the University at Buffalo Center of Excellence in Pharmacokinetics and Pharmacodynamics.
`Technical assistance was provided by Mrs. Suzette Mis. The author greatly appreciates the
`mentorship and friendship of Gerhard Levy, deemed the "Father of Pharmacodynamies ", for his
`seminal contributions in recognizing concepts and models for simple direct drug effects (Levy
`1966), indirect responses (Nagashima et al. 1969), target -mediated drug disposition (Levy 1994),
`and many other aspects of PK/PD.
`
`References
`
`Adair GS (1925) The hemoglobin system. VI. The oxygen dissociation curve of hemoglobin.
`J Biol Chem 63:529 -545
`Ariens EJ, Van Rossum JM, Simons AM (1957) Affinity, intrinsic activity and drug interactions.
`Pharmacol Rev 9:218 -236
`Black JW, Leff P (1983) Operational models of pharmacological agonism. Proc R Soc Lond B
`Biol Sei 220:141 -162
`Cao Y, Gao W, Jusko WJ (2012) Pharmacokinetic /pharmacodynamic modeling of GLP -1 in
`healthy rats. Pharm Res 29:1078 -1086
`
`Page 12
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`173
`
`Cao Y, Balthasar JP, Jusko WJ (2013) Second -generation minimal physiologically -based
`pharmacokinetic model for monoclonal antibodies. J Pharmacokin Pharmacodyn 40:597 -607
`Chakraborty A, Blum RA, Cutler DL, Jusko WJ (1999) Pharmacoimmunodynamic interactions of
`IL -10 and prednisone in healthy volunteers. Clin Pharmacol Ther 65:304 -318
`Clark AJ (1933) The mode of action of drugs on cells. Edward Arnold, London
`Dayneka N, Garg V, Jusko WJ (1993) Comparison of four basic models of indirect
`pharmacodynamic responses. J Pharmacokin Biopharm 21:457 -478
`Dutta A, Matsumoto Y, Ebling WF (1996) Is it possible to estimate the parameters of the sigmoid
`Emax model with truncated data typical of clinical studies? J Pharm Sci 85:232 -239
`Earp J, Krzyzanski W, Chakraborty A, Zamacona MK, Jusko WJ (2004) Assessment of drug
`to pharmacodynamic indirect
`response models.
`interactions
`relevant
`J Pharmacokin
`Pharmacodyn 31:345 -380
`Earp JC, DuBois DC, Molano DS, Pyszczynski NA, Keller CE, Almon RR, Jusko WJ (2008a)
`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 Pharmacol
`Exp Ther 326:532 -545
`Earp JC, DuBois DC, Molano DS, Pyszczynski NA, Almon RR, Jusko WJ (2008b) Modeling
`corticosteroid effects in a rat model of rheumatoid arthritis II: mechanistic pharmacodynamic
`model for dexamethasone effects in Lewis rats with collagen -induced arthritis. J Pharmacol
`Exp Ther 326:546 -554
`Fang J, Sukumaran S, DuBois DC, Almon RR, Jusko WJ (2013) Meta -modeling of
`methylprednisolone effects on glucose regulation in rats. PLoS ONE 8:e81679
`Fick A (1855) On liquid diffusion. Philos Mag 10:30 -39
`Friberg LE, Henningsson A, Maas H, Nguyen L, Karlsson MO (2002) Model of
`chemotherapy -induced myelosuppression with parameter consistency across drugs. J Clin
`Oncol 20:4713 -4721
`Gaddum JH (1937) The quantitative effects of antagonistic drugs. J Physiol 89:7P -9P
`Gessner PK (1974) In: Moselli PL, Garattini S, Cohen SN (eds) Drug interactions. Raven Press,
`New York
`Goldstein A (1949) The interaction of drugs and plasma proteins. Pharmacol Rev 1:102 -165
`Gompertz B (1825) On the nature of the function expressive of the law of human mortality, and on a
`new mode of determining the value of life contingencies. Philos Trans R Soc Lond 36:513 -585
`Harrold JM, Straubinger RM, Mager DE (2012) Combinatorial chemotherapeutic efficacy in
`non -Hodgkin lymphoma can be predicted by a signaling model of CD20 pharmacodynamics.
`Cancer Res 72:1632 -1641
`Hill AV (1910) The possible effects of the aggregation of the molecules of haemoglobin on its
`dissociation curves. J Physiol 40:ív -vii
`Jusko WJ (1971) Pharmacodynamics of chemotherapeutic effects: Dose -time - response relation-
`ships for phase- nonspecific agents. J Pharm Sci 60:892 -895
`Jusko WJ (1989) Pharmacokinetics of capacity -limited pharmacokinetic systems. J Clin Pharmacol
`29:488 -493
`Jusko WI (2013) Moving from basic toward systems pharmacodynamics models. J Pharm Sci
`102:2930 -2940
`Jusko WJ, Ko HC (1994) Physiologic indirect response models characterize diverse types of
`pharmacodynamic effects. Clin Pharmacol Ther 56:406-419
`Kenakin TP (1997) Pharmacologic analysis of drug- receptor interaction, 3rd edn. Raven Press,
`New York
`Krzyzanski W, Chakraborty A, Jusko WJ (2000a) Algorithm for application of Fourier analysis for
`biorhythmic baselines of pharmacodynamic indirect response models. Chronobiol Internat
`17:77 -93
`Krzyzanski W, Ramakrishnan R, Jusko WJ (2000b) Basic pharmacodynamic models for agents
`which alter production of natural cells. J Pharmacokin Biopharm 27:467 -489
`
`Page 13
`
`

`

`174
`
`W.J. Jusko
`
`Krzyzanski W, Jusko WJ (2001) Indirect pharmacodynamic models for responses with
`Pharmacokin
`polyexponential
`disposition.
`distribution
`or
`J
`multicompartmental
`Pharmacodyn 28:57 -78
`Krzyzanski W, Dmochowski J, Matsushima N, Jusko WJ (2006) Assessment of dosing impact on
`infra- individual variability in estimation of parameters for basic indirect response models.
`J Pharmacokin Pharmacodyn 33:635 -655
`Lepist E -I, Jusko WJ (2004) Modeling and allometric scaling of s( +)ketoprofen pharmacokinetics
`and pharmacodynamics: a retrospective analysis. J Vet Pharmacol Ther 27:211 -218
`Levy G (1966) Kinetics of pharmacologic effects. Clin Pharmacol Ther 7:362 -372
`Levy G (1994) Pharmacologic target -mediated drug disposition. Clin Pharmacol Ther 56:248 -252
`Loewe S, Muischnek H (1926) Effect of combinations: mathematical basis of the problem. Arch
`Exp Pathol Pharmakol 114:313 -326
`Mager DE, Jusko WJ (2001) General pharmacokinetic model for drugs exhibiting target -mediated
`drug disposition. J Pharmacokin Biopharm 28:507 -532
`Mager DE, Jusko WJ (2008) Development of translational pharmacokinetic -pharmacodynamic
`models. Clin Pharmacol Ther 83:909 -912
`Mager DE, Wyska E, Jusko WJ (2003) Diversity of mechanism -based pharmacodynamic models.
`Drug Met Disp 31:510 -518
`Mandema JW, Tukker E, Danhof M (1992) In vivo characterization of the pharmacodynamic
`interaction of a benzodiazepine agonist and antagonist: midazolam and flumazenil. J Pharmacol
`Exp Ther 260:36 -44
`Meagher AK, Forrest A, Dahhoff A, Stass H, Schentag JJ (2004) Novel pharmacokinetic-
`pharmacodynamic model for prediction of outcomes with an extended -release formulation of
`ciprofloxacin. Antimicrob Agents Chemother 48:2061 -2068
`Michaelis L, Menten ML (1913) Die Kinetik der Invertinwirkung. Biochem Z 49:333 -369
`Mishina EV, Jusko WJ (1994) Inhibition of rat splenocyte proliferation with methylprednisolone:
`in vivo effect of liposomal formulation. Pharm Res 11:848 -854
`Mould DR, Denman NG, Duffull S (2007) Using disease progression models as a tool to detect
`drug effect. Clin Pharmacol Ther 82:81 -86
`Nagashima R, O'Reilly RA, Levy G (1969) Kinetics of pharmacologic effects in man: the
`anticoagulant action of warfarin. Clin Pharmacol Ther 10:22 -35
`Renkin EM (1979) Relation of capillary morphology to transport of fluid and large molecules.
`Acta Physiol Scand Supp1463:81 -91
`Robertson TB (1923) The chemical basis of growth and senescence, Lippincott, Philidelphia
`Rowland M, Benet L, Graham C (1973) Clearance concepts in pharmacokinetics. J Pharmacokin
`Biopharm 1:123 -136
`Shannon JA (1939) Renal tubular excretion. Physiol Rev 19:63 -93
`Sharma A, Ebling WF, Jusko WJ (1998) Precursor- dependent indirect pharmacodynamic response
`model for tolerance and rebound phenomena. J Pharm Sci 87:1577 -1584
`Shimada S, Nakajima Y, Yamamoto K, Sawada Y, Iga T (1996) Comparative pharmacodynamics
`of eight calcium channel blocking agents in Japanese essential hypertensive patients. Biol
`Pharm Bull 19:430 -437
`Starling EH (1896) On the absorption of fluids from connective tissue spaces. J Physiol 19:312 -326
`Stec GP, Atkinson AJ (1981) Analysis of the contributions of permeability and flow to
`intercompartmental clearance. J Pharmacokin Biopharm 9:167 -180
`Sukumaran S, Lepist E -I, DuBois DC, Almon RR, Jusko WJ (2012) Pharmacokinetic /
`pharmacodynamic modeling of methylprednisolone effects on iNOS mRNA expression and
`nitric oxide during LPS- induced inflammation in rats. Pharm Res 29:2060 -2069
`Sun Y -N, Jusko WI (1998) Transit compartments versus gamma distribution function to model
`signal transduction processes in pharmacodynamics. J Pharm Sci 87:732 -737
`Swinney DC (2009) The role of binding kinetics in therapeutically useful drug action. Curr Opin
`Drug Discov Devel 12:31 -39
`Teorell T (1937) Kinetics of distribution of substances administered to the body. II. The
`intravascular modes of administration. Arch Int Pharmacodyn Ther 57:226 -240
`
`Page 14
`
`

`

`8 Foundations of Pharmacodynamic Systems Analysis
`
`175
`
`Wilkinson G, Shand D (1975) A physiologic approach to hepatic drug clearance. Clin Pharmacol
`Ther 18:377 -390
`Yamamoto K, Abe M, Katashima M, Yamada Y, Sawada Y, Iga T (1996) Pharmacodynamic
`analysis of antiplatelet effect of aspirin in the litera

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