throbber
Journal ofPharmacokinetics and Biopttarmuccutics. Vol. 27. No. 6, J99?
`
`Modeling Interactions between Adrenal Suppression
`and T-Helper Lymphocyte Trafficking during Multiple
`Dosing of Methylprednisolone
`
`Fung-Sing Chow,"2 Amaranth Shanna,L2 and William J. Juskom
`
`Received September [4, 1999—F‘inal March 22, 2000
`
`A physioiogic pharmacodynamic model was developed to jointly characterize the effects ofcortico-
`steroid treatment on adrenai suppression and T-heiper ceii traffictting during single and muitipie
`dosing in asthmatic patients. clothyiprcdnisoione (MP). cortisoi. and T-iteiper ceii concentrations
`obtained from a previousiy pubiisited study during singie day and 6 days of muitipie dosing MP
`treatment were examined. Tire formation and disposition kinetics of MP were described wttk a
`compartmentai model. The bioritytiimic profiie of basal cortisol secretion rate was analyzed using
`a recent Fourier approach based on circadian harmonics. A three—compartment toop model was
`proposed to represent titres major T-netfirer ceii poois: Mood, extravascuiar site. and iyrnpit nodes.
`T—iteiper ceii synthesis and degradation rate constants were obtained from the literature. The
`Suppressive effects ofcortisoi andr MP on T—iteiper ceii concentrations were described with a joint
`additive inhibition function aitcrt'ng the cell migration rate from iymph nodes to blood. The model
`adequately described both plasma cortisol profiles and T-iteiper cells in Moon" after single and
`muitipie doses of MP. The potency of MP for suppression of'eortisoi secretion was estimated as
`ICm = 0.8 rig/rat. Tire biorhytitmic nature of the basai T—iteiper ceiis in blood was well described
`as under the influence of basal circadian cortisol concentrations with [C,9 = 79 rig/mi. Tire model
`fitted potency of MPfor suppression of T-iteiper ceiis was lCm = 4.6 rig/mi. Tire observed rebound
`of T-iteiper ceiis in Mood can also be described by the proposed modei. The rhythm and suppression
`ofptastna cortisol and T—itetjoer ceiis before and during singie and rnuitipie dose MP treatment
`were adequately described ity these extended indirect response models.
`
`KEY WORDS: T-helper cells; trafficking; rebound; corticosteroids; circadian rhythm; methyl-
`prednisolone; drug interactions; pharmacokinetics; pharmacodynamics.
`
`Supported by Grant GM 24211 from the National Institute of General Medical Sciences,
`National Institutes of Health.
`
`‘Department of Pharmaceutics. School of Pharmacy. State University of New York at Buffalo.
`New York 1426!).
`2Present address: Drug Metabolism and Phannacokinctics, SmithKlinc Beecham Pharmaceut-
`icals, King of Prussia, Pennsylvania 19406.
`’To whom correspondence should be addressed at 565 Hochatctter Hall, Department of Phar-
`maceatics, School of Pharmacy, State University ofNew York at Buffalo, New York 14260;
`e-mail: wjjusko@acsu.buffalo.edu
`
`559
`
`lltflfl-dfibXNWI20ll-lliS'35lfiflU/{l © 199') Plenum Publishing Corporation
`
`Apotex v. Novartis
`lPR2017-00854
`
`NOVARTIS 2059
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2059
`
`

`

`560
`
`Chow, Shanna, and Jusko
`
`INTRODUCTION
`
`The trafficking of lymphocytes between blood and extravascular pools
`occurs throughout their life-span. Newly formed lymphocytes migrate from
`bone marow via the thymus to lymph nodes during which they undergo
`maturation. This migration process may take a period of weeks prior to
`their full differentiation into T lymphocytes. The trafficking of mature non-
`dividing lymphocytes involves migration from blood into lymphoid tissue
`and back to the blood. This trafficking process is relatively fast and can be
`measured in hours (1). The T lymphocytes in tissues move via the lymphatic
`system to blood. Therefore, the drainage of lymphocytes into blood from
`the thoracic duct lymph is the predominant process. The extent of lympho-
`cyte trafficking through the blood is significant; in a 24—hr period the total
`mass of lymphocytes in blood can be replenished several times (2,3). The
`ability of lymphocytes to exchange between the blood and tissues is essential
`to enable the immune system to react to antigens virtually anywhere in the
`body. This allows lymphocytes to be concentrated at sites where they can
`most effectively respond to and eliminate foreign antigens (4).
`Corticosteroids trigger multiple effects in the body. In addition to inhi-
`bition of cortisol secretion, one of the rapid effects of synthetic cortico-
`steroids is producing lymphocytopenia (5). Lymphocytes in blood exhibit a
`circadian rhythm (6). This rhythm is partly caused and regulated by cortisol
`concentrations, which has a similar but opposite circadian profile (7).
`Among the lymphocytes, the T-helper cells are the most sensitive subset to
`corticosteroid treatment (8) and reside in the heart of the immune cross-
`talking network. Therefore, characterizing the T-helper lymphocyte tem-
`poral profile in response to corticosteroids is of importance for understand—
`ing the action of this important class of drugs.
`The effort to model cortisol concentrations and lymphocyte trafficking
`under the influence of corticosteroids has been an evolutionary process.
`Dunn er of. (9) used a cosine function to quantitate the circadian rhythm of
`cortisol concentratious and the trafficking of T-helper cells from extra-
`vascular sites to blood. These two sets of pharrnacodynamic (PD) data were
`modeled separately with the assumption that trafficking of T-helper cells to
`blood followed a zero-order process. Explicit equations were employed to
`describe the change in T-helper cells as affected by the corticosteroid. Fisher
`er of. (10) applied the concepts of indirect response modeling (11) on T-
`helper cell dynamics. Corticosteroids were assumed to inhibit the circadian
`input of cells into blood. The combined action of dexamethasone and
`hydrocortisone effects on lymphocyte distribution in blood was suggested
`by Braat at at. (12) based on a competitive interaction model of Ariens
`and Simonis (13). A multiple-dosing study of methylprednisolone (MP) was
`
`

`

`Dynamics of T-Helper Cell Trafficking
`
`561
`
`Dose
`Harmonic
`
`
`Function
`
`
`
`Fig. 1. Schematic diagram of the joint cortisol and T-helper cell trafficking
`models.
`
`published by Milad et ai. (14) in which the T-helper lymphocyte time profile
`was described with a model similar to Fisher er a1. (10) and included the
`joint competitive effects of exogenous and endogenous corticosteroids.
`However, some important features of these data had not been explored.
`First, a higher degree of T-cell suppression occurred after multiple dosing.
`After seven doses of MP, the predose T—helper cell concentration showed a
`higher value than baseline with a pronounced rebound after the last dose.
`Moreover, differences in estimated [C50 values were found when separately
`fitting the single and multiple-dose data. In this paper, data from Milad e:
`at. (14) were reexamined to develop a physiological explanation and model
`for characterizing the effects of MP on cortisol suppression and their dual
`role in T-helper cell dynamics.
`
`THEORETICAL
`
`The proposed model is shown in Fig. 1. Blood (THBL), extravascular
`{THEV ), and lymph node (THLN) pools as well as natural production (1:5,...)
`and loss (kdcg) of T-cells along with the joint effects of exogenous and
`
`

`

`562
`
`Chow, Sharma, and Jusko
`
`endogenous corticosteroids are utilized to explain the natural baseline cir-
`cadian rhythms, acute suppression, and rebound phenomena of the time
`profiles
`of
`blood T-helper
`cells
`during multiple
`dosing with
`methylprednisolone.
`
`Pharmacokinefics
`
`The kinetics of MP after doses of its prodrug have been well charac-
`terized (14,15). The soluble prodrug, methylprednisolone sodium succinate
`(MP5) was rapidly metabolized with a first-order rate (lg) to its active form.
`The elimination of methylprednisolone was best described by a monoexpon—
`ential decline. The equations used were
`
`dMPs
`a: : «ram:
`
`dM—P2 k, - MP3 —9- MP
`
`a:
`
`V
`
`(1)
`
`(2)
`
`where CL and V are the systemic clearance and volume of distribution of
`MP. Model-estimated parameters were kr, CL, and V.
`
`Circadian Rhythmic Secretion Rate of Cortisol
`
`Over the past 5 years, various extensions of indirect response models
`have been proposed to describe the circadian nature of cortisol time profiles
`(15—19). Each of these models has its unique features, advantages, and dis—
`advantages in regard to characterizing the nature of cortisol secretion. A
`review and comparison of these models was done by Chakraborty er al.
`(20). Among these models, the multicomponent harmonic function with a
`24-hr rhythm was the most accurate model for the cortisol secretion rate. A
`new Fourier analysis method with a circadian (Le, 24 hr) constraint was
`used to describe the secretion rate of’cortisol (19). Briefly, the baseline eor~
`tisol concentrations (Cc‘mw) are represented as the Fourier series
`
`Comm : no + E [aI cos(2:tnt/24) + b,1 smarter/24)]
`n = I
`
`(3}
`
`where an, a“ and b;- are Fourier coefficients which can be obtained by fitting
`Eq. (3) to baseline or placebo data. The value of n represents the frequency
`of the harmonic function. For example, when n = 0, the harmonic function
`describes a steady baseline value of .520, when n = 1, the harmonic function
`has a period of 24 hr; when n = 2, the period is 12 hr; and so on. The L2-
`norm approximation method was used to derive the secretion rate function.
`Rama). The selection of the numbers of harmonics which are best to
`
`

`

`Dynamics of T-Helper Cell Trafficking
`
`563
`
`describe the baseline cortisol concentrations was determined by the percent-
`age contribution of each harmonic to the overall data fitting. To generate
`the cortisol secretion rate, the following equation was applied:
`
`
`
`RCort( I) =
`
`+ kC I CCort
`
`(4)
`
`where kc is the cortisol disposition rate constant. Endogenous cortisol syn-
`thesis and loss is then described by
`
`
`(16' on:
`C : RCorlU) I 1(3) _kC' CCorL
`
`.
`(5)
`
`where the inhibition function (1(1)) is added when MP is present. The phar-
`macodynarnics is related to MP and cortisol via
`
`If:
`
`I )1“? C t
`
`CM?
`_ o, =——
`
`{GIMP-Cort + CMp
`
`6
`
`(
`
`)
`
`Equation (6) depicts the effect of MP on the suppression of cortisol
`secretion rate. The circadian secretion rate of cortisol is inhibited decreasing
`hyperbolic function (I(I)MP.CM) with methylprednisolone concentration
`from Eq. (2) as the driving fierce.
`
`Circadian Rhythmic Trafficking of T-Helper Cells
`
`Movement of TH cells between the three pools is given by
`
`
`
`dig“ : km _ km", - THEV — kEL . THEV + kEE - THBL
`
`
`
`dTZLN = km. - THEV — I‘m; ‘ 1(0Cort-TH ‘ THLN
`
`
`
`‘17:”: km . Itasca-m - mm — knE - THBL
`
`The inhibition function related to cortisol concentrations is
`
`mam” = 1—#
`ICSO Cort-TH + CCort
`
`(7)
`
`(3)
`
`(9)
`
`“0)
`
`where ICsu Comm reflects the cortisol concentration producing a 50% change
`in the rate constant, km.
`The synthesis rate of T-helper cells follows a zero-order rate constant
`(ksyn) while the elimination of these cells from blood is by a first—order rate
`
`

`

`564
`
`Chow. Sharma, and Josh:
`
`constant (kdc31-»HJ. Intercompartmental rate constants are km; , It“, and kg.
`to describe the trafficking process. The circadian rhythm of T-helper cells
`in blood observed under basal conditions is controlled by the inhibitory
`effect of cortisol
`(ICSUCMTH), which is secreted in a circadian rhythm.
`Hence, T-helper cells in blood have a rhythmic profile similar but opposite
`to cortisol.
`
`The total recirculating lymphocyte pool in extravascular sites has been
`estimated to contain 30 to 45 times as many cells as found in blood (21,22).
`The lymphocyte subset composition is similar in these pools (23). In this
`model, we separated the lymph from the extravascular site. The T-helper
`cell pool sizes of the compartments in this model were assigned (initial con-
`ditions) based on estimates from Trepel (22). The T-helper cell pool
`in
`lymph was [9 times larger than the pool size in blood (TH3L)- The T-helper
`cell pool size in the rest of the lymphoid tissues (EV) was 26 times larger
`than that in the circulation. The T-helper cell production (km) and degra-
`dation (meg-rt.) rates were calculated based on the life-span estimated by
`Hellerstein et a1. (24). With an estimated km.“ : 0.00033 hr“, we related
`kg,“ to the initial condition of the EV site (THEV) by kwfl = THEY x kdcgm.
`Therefore,
`the production rate was specified for each subject.
`It was
`assumed that these parameters apply to asthmatic patients.
`The joint effects of cortisol and MP on T—helper cells arises from
`
`[UlcertaMP-TH '= 1 —
`
`CMP ' §+ CCorl
`— (1 la)
`Ing Cort-TH + CH? ' §+ CCort
`
`fixes—mm
`ICSD MF—TH
`
`(11b)
`
`which is inserted into Eq. (9). Equation (1 l) was adapted from Milad et at.
`(14), and has a similar structure as Eq. (10) with the inclusion of the additive
`effects from both corticosteroids. The g is an index of the potency ratio of
`cortisol and MP for inhibition of T—helper cells input from lymph to blood
`(km), It is assumed that short-term corticosteroid treatment will not affect
`
`the production rate of T-helper cells in bone marrow. Parameters estimated
`in modeling the dynamics were kc and ICE Mag,“ for adrenal suppression
`and kLE, kBE, kEt, [Quests-m, and a for T—helper cell trafficking.
`
`METHOD
`
`Data from Milad e: a]. (14) were used to test the applicability of this
`model. Six asthmatic male volunteers were studied. Five data sets were used
`
`for this modeling as the sixth subject had irregular responses. Analytical
`and other study details can be found in the original report. The study design
`
`

`

`Dynamics of T-Helper Cell Trellicking
`
`565
`
`consisted of three sessions. In Session I, the basal cortisol and T-helper cell
`concentrations were monitored for 24 hr. [11 Session 2, a single dose of 20 mg
`MP was administered as MP5. The MP, cortisol, and T—helper cell concen-
`trations were monitored for 32 hr, In Session 3, beginning 48 hr after the
`initiation of Session 2, a daily dose of 20 mg of MP was given for 6 days.
`On the last dosing day, monitoring the same scheme as in Session 2 was
`carried out for 32 hr.
`
`Data were analyzed using a piecewise approach. The pharmacokinetie
`(PK) parameters were first estimated and then were fixed as constants when
`fitting the cortisol suppression data. Then, parameter estimates from PK
`and cortisol fittings were subsequently fixed as constants when the T-helper
`cell data were fitted. The estimated potency factor (5) for each individual
`was fixed during the final fittings to obtain the precision of the remaining
`parameter estimates for the T-helper cell PD.
`All fittings were done with subroutines written for Adapt II release 4
`(27). Assuming that the additive error is normally distributed, the Maximum
`Likelihood Estimator along with a general variance model was used to
`obtain the system and variance parameter estimates. This error model
`described a nonconstant relationship of variance and predicted values with
`a slope and a power function based on predicted values: Variance of Y:
`(slope of 3m2 x (predicted WW
`
`RESULTS
`
`Pharmacokinetics of MP
`
`Equations (1') and (2), which describe the pharmacokinetics of MP,
`were fitted simultaneously to the first and multiple—dose MP concentrations
`for each subject. The profiles for one of the subjects is shown in Fig. 2. The
`estimated parameters are summarized in Table 1. These estimated param-
`eters were then fixed to generate the forcing functions for the later fitting of
`cortisol and T-helper cell concentrations. The kinetics of MP is well
`described with a first-order formation rate and monoexponential decay of
`MP. The formation rate (kr) of MP from MP3 was rapid with a mean value
`of 6.2 hr“. The mean half-life of MP was 2.6 hr, with an average V of 841
`and CL of 231/hr. These parameter estimates were similar to those pre-
`viously reported (9,10,25,26). With a half-life of 2.6 hr and a daily dosing
`scheme, there was no accumulation during 6 days of once-daily treatment.
`Therefore, differences in response to single and multiple doses were not
`caused by any changes in the pharmacokinetics of MP.
`
`

`

`566
`
`Chow, Shanna, and Jusko
`
`100
`
`
`
`
`
`MethylprednlcaohneConcentration(ngi'I'nL)
`
`i—_|—|—|—I—I—t—l
`0
`20
`4D
`30
`ED
`10!)
`mo
`ND
`
`Time (hr)
`
`time profile of one
`Fig. 2. Metltylprednisolone concentration vs.
`subject during multiple dosing. Symbols are the observed data with
`line fitted using Eqs. [1) and (2).
`
`Table 1. Summary of Methylprednisolme Phannacokinetic Parameters"
`
`Subject
`
`1
`2
`3
`4
`5
`
`M
`SD
`
`kr
`(hr' ')
`
`7.7 (39.4)
`3.2 (27.1)
`4.9 {12.6)
`4.0 (9.6)
`6.1 {24.3)
`
`6.2
`1.3
`
`V
`(L)
`
`100.7 (6.9}
`75.7 (5.0)
`79.5 (3.3)
`73.7 (2.4)
`34.3 (4.5)
`
`83.3
`9.9
`
`CL
`[L/hr)
`
`23.6 (8.1)
`21501.2)
`24.3 (3.5)
`19.4 (1.1)
`20.2 (2.9)
`
`22.3
`3.7
`
`[”2
`(hr)
`
`2.4 (12.3)
`2.4 (6.3)
`2.3
`(5.3)
`2.3 (2.5)
`2.9 (3.3]
`
`2.6
`0.3
`
`”Data presented as parameter estimate {% C V).
`
`Baseline Cortisol
`
`A baseline profile of cortisol for one subject is shown in Fig. 3. The
`insert of Fig. 3 further illustrates its circadian nature. The harmonic func—
`tions that contributed more than 1% to the overall function of cortisol
`
`secretion rate were selected. The baseline cortisol concentrations of these
`
`subjects from the first study session were well described with three to four
`harmonic functions plus an average cortisol secretion rate. (it 2 0). The har-
`monics selected for each subject are summarized in Table II. The contri-
`bution of each harmonic to the total function is listed in rank order. This
`
`cortisol function was stable and reproducible over the whole study period.
`Hence, the effect of MP on suppression of cortisol secretion can be assessed.
`
`

`

`567
`
`o
`
`Dynamics of T-Helper Cell Trafficking
`
`0
`
`m
`
`’
`
`‘
`
`120
`
`(nglmL} S8
`CortisolConcentration
`
`
`
`
`Time (hr)
`
`Fig. 3. The basal cirCadian profile of cortisol concentration versus time for one
`subject. Symbols are the observed data with the line fitted using the Fourier
`approach.
`
`Table ll. Summary of Cortisol Pharmacodynamic Parameters”
`
`Subject
`
`Harmonic functions
`(it)
`
`Initial CCall
`(mg/ml)
`
`0. l. 3, 2, 4
`0. l, 2, 4
`0. 1. 3. 2
`01 1.2.3
`0. 1.3, 4
`
`107.6
`26.2
`47.5
`104.?
`61.4
`
`l
`2
`3
`4
`5
`
`M
`SD
`
`kc
`[hr")
`
`(12)
`0.2]
`0.36 [20.2)
`0.30 {15.3)
`0.22 (10.4)
`0.16 (10.5)
`
`0.25
`0.03
`
`[C50 MP—Corl
`(Hg/ml)
`
`0.25 (21.3)
`0.36 (44.”
`1-62 (42.9)
`(17491.3)
`0.37 (“5.5)
`
`0.77
`0.54
`
`“Data presented as parameter estimate (%C V).
`
`Cortisol after MP “Treatment
`
`Cortisol concentrations of each subject from baseline, single dose, and
`multiple doses were arranged according to their sample collection times
`based on the study design. Therefore, a single differential equation [Eq. (5)]
`with a single initial condition of cortisol (at r= 0) was used. This avoided
`the need to assume initial conditions for each treatment session. The pharm-
`acokinetics of MP was used as a fixed function for suppression of the circad-
`ian cortisol secretion rate. Figure 4 shows the fitting of this model to the
`
`

`

`568
`
`Chow, Slant-me, and Jusko
`
`
`
`
`
`CortisolConcentration(nymL')
`
`120
`
`asD
`
`ha
`
`7
`
`ti 1
`
`1 W
`
`Time (hr)
`
`300
`
`Fig. 4. The cortisol concentration vs. time profile for one subject in all
`sessions. Symbols are the observed data with the line fitted using Eqs. (5)
`and (6}.
`
`cortisol data of one subject. The fitting well described all cortisol concen-
`trations From the study. Table II summarizes the cortisol initial condition,
`disposition rate constant (kc), and MP potency (ICSG MPCM) for each sub-
`ject. The mean kc is 0.25 hr‘I and {(350 Mpg,” is 0.77 ng/ml. These parameter
`estimates are similar to those previously reported using a single cosine cir-
`cadian cortisol secretion rate (26,28). The mean kc value obtained using the
`Fourier approach is similar to that (0.38 hr'l) reported by Lima 6! a]. (28)
`following short-term infusions of cortisol.
`
`Dynamics of T-Helper Cells
`
`The T-helper cell concentrations were also arranged in the same
`sequential manner as the cortisol data for each subject. The parameters for
`MP pharmacokinetics and cortisol pharmacodynamics were used according
`to the model as fixed joint effects on the suppression of the kw rate process.
`Figure 5 shows fitting of the model to T—helper cell concentrations for one
`subject. The T-helper cell concentrations of Session 1 (baseline) were placed
`on Day 22 to demonstrate the stability of model. The fitting was able to
`simultaneously describe data from all sessions. The nature of T-helper cell
`rebound after multiple doses was well captured. The rhythm of T—helper
`cells in blood was predicted to be restored about 5 days after the last treat-
`ment. The parameter estimates are summarized in Table III. The mean
`
`

`

`Dynamics of T-Helper Cell Trafficking
`
`569
`
`1600
`
`1200
`
`800
`
`400
`
`
`
`
`
`T-halperCells(cellsluL)
`
`
`0|
`I
`r
`T
`0
`
`300
`
`350
`
`400
`
`50l
`
`1““1 it fit
`
`2i”
`
`Time {hr}
`
`Fig. 5. The T-helper cell concentration in blood vs. time profile for one
`subject in all sessions. Symbols are the observed data with line fitted using
`Eqs. m—(n).
`
`1C5.) Comm and 15:35.3,“qu were 79.3 and 4.6 ng/ml with potency ratio (.5) of
`17.1. The mean blood T-helper cell trafficking rate constants were kw =
`0.0291hr" and kBE=0.379 hr". The EV to LN rate constant (km) was
`0.018hr“. The cell dynamic parameter estimates were similar among the
`subjects except for Subject 2. The mean basal cell level of Subject 2 was
`higher at about 1200 cells/n1. His degree of rebound was more pronounced
`(maximum of 2000 cells/p1) after multiple doses. Although the estimated
`parameters suggested Subject 2 was insensitive to cortisol-induced T-hclper
`cell suppression (high ICSOCD,,_CD4), the high basal T-helper cell
`level
`in
`blood indicated a higher production rate.
`Figure 6 shows the simulated profiles of cells in LN and EV of the
`indicator subject. The LN and EV pools in this model show an opposite
`profile to each other. The magnitude of change in these pools is similar.
`During MP treatment, the LN pool increased from about 15,500 to 20,000
`cells/m1 (—30% increase) and the EV pool decreased from about 20,000 to
`16,000 cells/ml (~20% decrease). With the assumption that MP does not
`affect the trafficking of T-helper cells from EV to LN, this simulation shows
`that T-helper cells were retained in the LN while the EV pool continuously
`depletes over the treatment period. Upon the cessation of MP treatment,
`both pools rapidly return to the steady-state cell counts. This swift equili-
`bration of these circulation pools agrees with indications that lymphocyte
`trafficking through the blood compartment occurs at a high rate (2).
`
`

`

`570
`
`Chow, Sharma, and Jusko
`
`
`
`“259$;geniuoanfifi:6E35.3Sufism.5”:5.
`
`
`
`
`
`
`EEEBV:1Eng:13.ro:1}:ro{a{a9-3.gums.M.“8.5%
`
`
`
`
`Ewan.—7W¥mrhn50mm.“meHdn—QWflbfimay.Elf—GU@mhuk
`
`
`
`
`
`M3anon80.:anm5582mm;tammom_
`
`a:83mm:85”32«:32.2V8:.S:M3:N9.52.82.3:23
`
`81mm3.3Ga:Ga:3Raw93.39353$3wasSnv.2mclawSA:Gd:
`
`”was
`
`3m33mm8.2”a2:3.83owed<293d.
`
`3.83m3:2mom:3$3#8.:2m:2ma.5adRm:
`
`:3:
`
`E...«RR98.939was2.2.$3.a?a:E3.»:839.2..3.5
`
`mm:58mma“Bed39.:83.EN.“an
`
`
`
`
`
`
`
`.Euuuax:meH.53.MT:—mmooodHFiona”:hdfifibtzheacfibmHm..gnuaxfi035:8gun—05min—mmuufluumflnEma...
`
`
`
`
`
`
`
`

`

`Dynamics of T-Helper Cell Trafficking
`
`571
`
`20000
`
`18005
`
`16000
`
`
`
`
`
`T-helperCells[CellaiuLl
`
`14000
`
`...°..
`
`t
`
`
`I
`I
`I
`I
`I
`I
`I
`50
`10
`150
`
`r ”H 1 1h 25"
`
`400
`
`Fig. 6. The simulated 'T—helper cell concentrations in the extravascular (solid
`line) and lymph node pools (dashed line) vs. time profile for one subject in all
`sessions.
`
`DISCUSSION
`
`The circadian rhythm of cortisol concentrations in humans is often
`asymmetric and variable. The patterns of‘ the basal cortisol secretion rate of
`each individual are different. A single cosine function may not be sufficient
`to describe such a complex pattern. This issue has been illustrated in studies
`that compared various modeling approaches for biorhythmic basal cortisol
`concentrations (16,20). In this model, we utilized the Fourier Lzflnorm ana—
`lytical approach (19,20) to generate the appropriate circadian harmonic
`function for the cortisol secretion rate profile of each subject. This approach
`is flexible, accurate, and stable over time. Therefore, it was used as a part of
`the model for driving the cortisol-induced changes in T-helper cell rhythms.
`The majority of lymphocytes in the human body reside in lymph nodes,
`spleen, thymus, and bone marrow. Only about 2% of the total lymphocytes
`are found in the blood (22). Among the lymphoid tissues, the lymph nodes
`and spleen are the main compartments in lymphocyte trafficking (1). Circul-
`ating blood disseminates lymphocytes to various regions of the body. The
`extravasation process of lymphocytes is rapid. It takes about 15 min from
`the initiation of contact of lymphocytes with endothelial cells to diapedesis
`of the lymphocytes into tissue (29). The lymphocytes accumulate in either
`the white pulp of spleen or the high endothelial venule (HEV) of the lymph
`nodes (30). Cells in lymph nodes complete their journey and return to the
`bloodstream via the thoracic duct. Our three-pool model was structured
`
`

`

`572
`
`Chow, Sharma, and Jusko
`
`accordingly with the kBE value found as expected to be much larger than
`the other rate constants.
`
`Different types of lymphocytes follow a specific trafficking pathway.
`Peripheral
`lymph nodes retain mostly the T lymphocytes (3|,32), while
`Spleen retains mostly the B lymphocytes (23) from the recirculating pool.
`Therefore, we considered only the lymph node pool as a model compart~
`merit apart from the extravascular site for T-helper cell
`trafficking. By
`including these two compartments along with a recirculation system, this
`model provides a more plausible and physiological mechanism or T lympho-
`cyte trafficking. However, with the increase of model complexity, the rela-
`tive sizes of the model compartments determine the rate process of the T-
`helpcr cell migration between each site. Therefore, published values of rela-
`tive lymphocyte distribution in healthy volunteers (22) were assigned for the
`sizes of the T-helpcr cell pools in lymph nodes and extravascular sites under
`steady-state conditions. The influence of disease status on T lymphocyte
`
`distribution should be considered when this model is used to estimate drug
`effects on cell trafficking in patients. Our asthmatic patients were otherwise
`relatively healthy and could be well characterized with this model.
`The T-cell production and degradation rates are important factors in
`our attempt to quantify the temporal changes in these model compartments
`over several days. In normal conditions, these rate processes occur at sites
`other than blood and lymph nodes. Hellerstein et al. (24) measured the
`kinetics of circulating T lymphocytes in normal humans. By intravenously
`infusing the stable isotope-labeled metabolite 2H—glucose to healthy volun—
`teers and following the rate of its disappearance, they Were able to estimate
`the normal life-span of T-helper cells by calculating the rate constant of
`replacement of unlabeled by labeled DNA strands in blood. Since the loss
`of labeled DNA strands would not be affected by cell proliferation during
`the postlabeling period, the estimated production rate and life-span from
`this study represent the true kinetics of T—helper cells in humans. Therefore,
`these synthesis and degradation rates of T—cells were used in our model.
`However, in the same study, when the life-span of T-helper cells were com-
`pared in healthy subjects with those in HIV patients, the degradation pro-
`cess was faster in HIV patients.
`Our model provides a physiologic approach to determine the potency
`of a therapeutic agent for suppressing T lymphocyte trafficking. However,
`it is not possible to obtain appropriate parameter estimates without the
`prior understanding of T lymphocyte distribution and production and
`degradation rates.
`This model considered the dynamics of recirculating T-‘nelper cells with
`a simple three-compartment loop system. Although it is a simplified version
`
`

`

`Dynamics of TuHelper Cell Trafficking
`
`573
`
`of the physiological system, our model contains the main components of T-
`helper cell dynamics in humans. With use of some information about T-
`helper cell pools and production from the literature, we were able to con-
`struct this model to describe (i) the biorhythmic nature of T-helper cells in
`blood under the influence of basal cortisol concentrations; (ii) the acute
`suppression of T-helper cells after a single MP dose; (iii) the suppression of
`T-helper cells after multiple MP doses; (iv) the increase of blood T-helper
`cells during multiple MP doses; and (v) the restoration of the normal T-
`helper cell biorhythm after cessation of therapy.
`In basic indirect response models (IRM) (ll), zero-order input and
`first-order output rate constants for a single compartment are used to
`describe the dynamics of a physiological response. Drugs are assumed to
`modulate one of these rate processes. A natural extension of [RM is a two-
`compartment catenary IRM. Sharma et a1. (33) explored precursor-depen-
`dent indirect response models (PIRM) with a zero-order rate in the precur-
`sor preceding the response compartment. A first-order
`rate constant
`connects the precursor to the response compartment while the response was
`dissipated via a first—order process. When drug suppresses or enhances the
`formation rate of response,
`the model predicts tolerance or
`rebound
`phenomena upon repeated dosing. Our current model is partly an extension
`of these PlRM allowing the phenomenon of rebound to be predicted with
`a suppressive drug effect. New developments presented in our models are
`the use of a third dynamic compartment, the description of the production
`and loss of response on the third compartment (THEV )3 and the recircu-
`lation system of these pools (i.e., THEV to THm to THBL and return to
`THE, ). This third compartment is needed when the natural production rate
`(km) of the dynamic response is not associated with the precursor compart-
`ment, and /or the dissipation of response from the response compartment
`(THBL) is not the natural degradation process (kdcgm). Furthermore, a
`threecompartment loop system provides a delay of cell movement in that
`a cell leaving the blood compartment requires time for trafficking through
`the EV site before it becomes available for the blood compartment. There-
`fore, this model is most suitable to describe physiological trafficking pro-
`cesses of T-helper cells where the site of main drug effect is not at the
`synthesis and degradation steps. Additional features of this model are the
`exploration of the intricacies of methylprednisolone and cortisol effects on
`T-cells.
`
`ACKNOWLEDGMENT
`
`The authors thank Dr. Wojciech Krzyzanski
`discussion.
`
`for his
`
`insightful
`
`

`

`574
`
`REFERENCES
`
`Chow, Sharma, and Jusko
`
`l. W. L. Ford and J. L. Gowans. The traffic of lymphocytes. Semin. Hematol. 6:67—83 (1969).
`2. A. J. Young, J. B. Hay, and C. R. Mackay. Lymphocyte recirculation and life span in
`viva. Curr. Topics Microbiol. Immunol. 184:161—173 (1993}.
`3.. A. J. Young and J. B. Hay. Rapid turnover of the recirculating lymphocyte pool in viva.
`frat. Immortal. 7:1607—1615 (1995).
`4. A. K. Abbas. A. H. Lichtman, and J. S. Pober. Cellular and Molecular Immunology,
`W. B. Saunders, Philadfelphia, PA, 1994.
`5. Y. Shoenfeld, Y. Gurewich, L. A. Gallant, and J. Pinkhas. Prednisone—induced leuko-
`cytosis. Influence of dosage, method and duration of administration on the degree of leuko~
`cytosis. Am. J. Med. 71:7?3—778 ([93]).
`6. T. Miyawaki. K. Taga, T. Nagaoki, H. Seki. Y. Suzuki, and N. Taniguchi. Circadian
`changes of T lymphocyte subsets in human peripheral blood. Clin. Exp. Immortal. 55:618—
`622 (1984).
`'2’. H. B . Tavadia, K. A. Fleming, P. D. Hume, and H. W. Simpson. Circadian rhythmicity of
`human plasma cortisol and FHA-induced lymphocyte transformation. Cfin. Exp. Immunol.
`22:190—193 (1975).
`B. B. Zweiman, P. C. Atkins. P. M. Bedard, S. L. Flaschcn. and R. P. Lisak. Corticosteroid
`effects on circulating lymphocyte subset levels in normal humans. J. Clin. Immunol. 4:15!~
`155 (1984).
`9. T. E. Dunn. E. A. Ludwig, R. L. Slaughter, D. 5. Camera. and W. J. Jusko. Pharmacoki-
`notice and pharmacodynamics of methylprednisolone in obesity. Clin. Pharmacol. Ther.
`49:536—549 (1991).
`10. L. E. Fisher, E. A. Ludwig, and W. J. Jusko. Phannacoimmunodynamics of methyl-
`prednisolone: trafficking oi" helper T lymphocytes. J. Pharmacokr’n. Biophorm. 20:319—331
`(1992).
`11. N. L. Dayneka, V. Garg, and W. J. Jusko. Comparison of four basic models of indirect
`pharmacodynamic responses. J. Phormccokr'o. Biopharm. 11:45‘i—478 (1993).
`12. M. C. Brant, B. Oosterhuis, R. P. Koopmans, J. M. Meewis, and C. J. Van Boxtel. Kinetic-
`dynamic modeling of lymphocytopcnia induced by the combined action of dexamethasone
`and hydrocortisone in humans, after inhalation and intravenous administration of dexame-
`thasone. J. Pharmacol. Exp. Ther. 262:5

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