Sie sind auf Seite 1von 12

PHARMACOKINETICS AND PHARMACODYNAMICS

ARTICLE
MAGER
10.1177/0091270003258651
DOSE
PHARMACOKINETICS
EQUIVALENCY
ET AL EVALUATION
AND PHARMACODYNAMICS
OF CORTICOSTEROIDS

Dose Equivalency Evaluation of Major


Corticosteroids: Pharmacokinetics and
Cell Trafficking and Cortisol Dynamics
Donald E. Mager, PharmD, PhD, Sheren X. Lin, PharmD,
Robert A. Blum, PharmD, Christian D. Lates, MD, and William J. Jusko, PhD

The integrity of current corticosteroid dose equivalency ta- of all PD responses was calculated using a linear-trapezoidal
bles, as assessed by mechanistic models for cell trafficking method. Although T helper cell trafficking and adrenal
and cortisol dynamics, was investigated in this study. Single, suppression achieved significant differences by repeated-
presumably equivalent, doses of intravenous hydrocortisone, measures ANOVA (p = 0.014 and 0.022), post hoc analysis
methylprednisolone, dexamethasone, and oral prednisolone using the Bonferroni method revealed no difference between
were given to 5 white men, according to total body weight, in a treatments. Although limited by the use of single doses and a
5-way crossover, placebo-controlled study. Pharma- relatively small sample size, this study applies mechanistic
codynamic (PD) response-time profiles for T helper cells, T models for several biomarkers showing that currently used
suppressor cells, neutrophils, and adrenal suppression were dosing tables reflect reasonable dose equivalency relation-
evaluated by extended indirect response models. For adrenal ships for four corticosteroids.
suppression, prednisolone appears to be less potent than
methylprednisolone or dexamethasone. A good correlation Keywords: Glucocorticosteroids; dose equivalency tables;
was found between the estimated in vivo EC50 values and rel- cell trafficking; pharmacokinetics
ative receptor affinity (equilibrium dissociation constants Journal of Clinical Pharmacology, 2003;43:1216-1227
normalized to dexamethasone). Area under the effect curves ©2003 the American College of Clinical Pharmacology

S ystemic glucocorticosteroids (GC) currently play a


major role as immunosuppressive agents for
diseases in which inflammation or immunologically me-
other. One of the problems encountered in the usage of
systemic GC is the integrity of dose equivalency tables.
Most equivalency tables used today have been derived
diated phenomena play a predominant pathophysiologic from clinical experience and older literature. Currently
role.1-5 To optimize therapy, it is not uncommon for cli- acceptable equivalent doses for the major systemic GC
nicians to switch from one agent of this group to an- are hydrocortisone (20 mg), prednisolone (5 mg),
methylprednisolone (4 mg), and dexamethasone
(0.75 mg).5 Although traditional pharmacokinetic/
From the Buffalo Clinical Research Center, Buffalo, New York (Dr. Lin, Dr.
pharmacodynamic (PK/PD) models have been applied
Blum, Dr. Lates) and the Department of Pharmaceutical Sciences, School
of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State
to characterize the major effects of steroids in the con-
University of New York, Buffalo, New York (Dr. Mager, Dr. Jusko). Sup- text of dose equivalency,6 such assessments using more
ported by grant GM24211 to Dr. Jusko from the National Institutes of contemporary mechanistic PK/PD models to follow the
General Medical Sciences (National Institutes of Health); fellowship sup- potency of several immunosuppressive effects have yet
port for Dr. Lin from GlaxoSmithKline Pharmaceuticals, Philadelphia; and to be performed.
a predoctoral fellowship for Dr. Mager from the American Foundation for The precise mechanisms of action of GC still remain
Pharmaceutical Education. Submitted for publication March 3, 2003; re-
elusive.7-9 Suggested mechanisms include the inhibi-
vised version accepted August 10, 2003. Address for reprints: William J.
Jusko, PhD, Department of Pharmaceutical Sciences, School of Pharmacy tion of specific cellular functions and cellular redistri-
and Pharmaceutical Sciences, University at Buffalo, State University of bution of lymphocytes and neutrophils.7 Lymphocytes
New York, Buffalo, NY 14260. normally migrate between blood and tissue, which al-
DOI: 10.1177/0091270003258651 lows the immune system to react to antigens through-

1216 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

out the body.10,11 The GC transiently deplete lympho- known to alter the metabolism of corticosteroids. All
cytes from the circulation, and T helper and T volunteers were prohibited from using any prescrip-
suppressor cells, in particular, are more sensitive to tion or over-the-counter medications (with exception
this effect.12,13 Lymphocytes kept out of circulation for acetaminophen ≤ 2 g/day and vitamins) within 14
have less access to the target organs in which they exert days prior to each of the study periods. Alcohol and
their effects.14 The characterization of lymphocyte dy- beverages containing caffeine or xanthine derivatives
namics is therefore important for understanding the were not permitted for 24 hours prior to the start of each
effect of GC on the immune system. period until the end of each 32-hour blood collection
The rise in neutrophil count following GC exposure period. This study was approved by the Kaleida Health
is thought to be due to accelerated release from the Millard Fillmore Hospital Institutional Review Board
bone marrow and reduced movement out of the blood (Buffalo, NY) and performed in accordance with the
into inflammatory sites.14 In the presence of inflamma- Declaration of Helsinki. All subjects were informed of
tion, the migration of neutrophils from blood to areas of the nature, purposes, and risks of the study and
inflamed tissues is an essential step in the inflamma- provided informed consent.
tory process. Evidence suggests that, in patients with
chronic inflammatory conditions, the accumulation of Procedure
neutrophils in the blood is due to the effect of GC on the
adhesion properties of neutrophils.15 It is uncertain This was a single-dose, randomized, five-period cross-
whether this process plays a significant role in the over study with placebo control. It was single blinded,
neutrophilia seen in subjects not experiencing an in- with all subjects confined in the Buffalo Clinical Re-
flammatory event; we therefore have assessed search Center during all five periods of the study to
neutrophil dynamics in blood. control for identical eating and sleeping patterns. The
The primary purpose of this study is to further char- confinement period was approximately 34 hours dur-
acterize and compare the immunosuppressive and ing each period, beginning with dosing at 8 a.m. and
anti-inflammatory properties of four systemic GC— ending with the 32-hour blood draw. Each period was
hydrocortisone, prednisolone, methylprednisolone, separated by a 2-week washout. Dosing was according
and dexamethasone—at currently accepted equivalent to total body weight (TBW) of each subject based on Ta-
doses. This was approached by assessing T helper, T ble I. During each period, subjects received a single
suppressor, and neutrophil cell trafficking using mech- dose of intravenous hydrocortisone (hydrocortisone
anistic dynamic models. One of the major limitations sodium succinate, Solu-Cortef ® , Pharmacia),
of using systemic GC is their ability to suppress the re- methylprednisolone (methylprednisolone sodium
lease of corticotropin from the pituitary, thus decreas- succinate, Solu-Medrol®, Pharmacia), dexamethasone
ing the secretion of cortisol by the adrenal cortex.7,16 A (dexamethasone phosphate, Decadron®, Merck), oral
secondary goal of this study was to compare the rela- prednisolone (generic prednisolone tablets, Schein), or
tive potency of adrenal suppression among the four saline injection for placebo control.
systemic GC. On day 1 of each study period, two 18-gauge
angiocatheters were inserted into veins of both arms to
METHODS facilitate drug dosing and blood sample collections.
The catheter for drug administration was promptly re-
Subjects moved after dosing, and the remaining catheter was
kept patent with the use of a dilute heparin solution (10
Five healthy male volunteers were recruited to partici- units/mL). Samples of approximately 7 mL of blood
pate in this study. All subjects were 23 to 40 years of age were drawn into heparin-containing collection tubes
(mean ± SD, 32.2 ± 7.6 years), were within 25% of ideal for drug analysis. The blood samples were centrifuged
body weight (73.6 ± 10.7 kg), and had a normal circa- at 2000 rpm for 15 minutes, and plasma was harvested
dian rhythm (nightshift workers were excluded). All and frozen at –20°C until assayed. In addition, 3-mL
subjects were determined to be healthy by assessment blood samples were drawn into EDTA-containing
of medical history, physical examination, EKG, blood tubes for the determination of cell counts. These sam-
chemistry, and hematologic profile. Creatinine clear- ples were kept at room temperature and the assays per-
ances averaged 98.9 ± 18.4 mL/min. None of the sub- formed within 12 hours. All blood samples were col-
jects had a documented allergy to corticosteroids, and lected at 0, 0.5, 1, 1.5, 2, 4, 8, 12, 16, 24, 28, and 32 hours
none were receiving any concurrent medications after dosing.

PHARMACOKINETICS AND PHARMACODYNAMICS 1217


MAGER ET AL

Table I Dosages (mg) of Study Drugs


a
Weight Range Hydrocortisone Prednisolone Methylprednisolone Dexamethasone

50-59 kg 110 28 22 4
60-69 kg 130 32 25 5
70-79 kg 150 38 30 6
80-89 kg 170 42 34 6.5
90-100 kg 185 46 36 7
a. Based on total body weight.

Bioanalysis Pharmacokinetics

Plasma Glucocorticoid Concentrations Prednisolone


Corticosteroid concentrations were determined using The PK of free prednisolone were described by a one-
the normal-phase high-performance liquid chromatog- compartment model with first-order absorption and
raphy (HPLC) procedure of Rose and Jusko,17 as revised elimination rate constants (ka and kel) according to the
by Jusko et al.18 The lower limit of detection was 5.0 ng/ following:
mL for all steroids, and both the interday coefficients of D • ka
variation (CV) and percent error were ≤ 13%. Samples CP =
(Vd / F)(ka − kel )
[
• e− k el • t − e− k a • t ,] (1)
falling below the limit of detection were excluded from
data analysis, except for cortisol, which was set equal where Cp represents the mean plasma concentration of
to half the limit of quantification owing to expecta- free prednisolone, D is the dose administered, and Vd/F
tions of its continual presence. Plasma protein is the apparent volume of distribution.
binding of prednisolone was determined by
ultrafiltration using the Amicon Centrifree Device, as Methylprednisolone
previously reported.19 The concentrations of methylprednisolone following
Cell Responses an IV bolus dose were described by
Complete leukocyte counts and differentials were ob- D
Cp = • e− k el • t . (2)
tained using an automated hemocytometer (CELL- Vd
DYN 1700, Abbott Laboratories, Abbott Park, IL). Total
lymphocyte and segmented neutrophil counts were Dexamethasone
obtained. The T helper and T suppressor cell counts
were obtained by measuring fluorescence after per- Dexamethasone concentration-time data were de-
forming immunoreaction of lymphocytes with anti- scribed using the following biexponential equation:
CD3, anti-CD4, and anti-CD8 antibodies. Whole-blood C p = C1 • e− λ 1 • t + C 2 • e− λ 2 • t . (3)
samples (50 µL) with mouse IgG were reacted with
mouse anti-CD4 and anti-CD8 monoclonal antibody The intercept (C1, C2) and exponential coefficients (λ1,
conjugated with fluorescein isothiocyanate and λ2) were used to calculate kel, Vd, and the first-order dis-
phycoerythrin for 15 minutes. After lysing and wash- tribution rate constants between the plasma and tissue
ing, samples were analyzed for T helper (CD4+:CD8–) compartments (k12, k21).
and T cytotoxic (CD4+:CD8+) cells using a FACScan
flow cytometer (Becton-Dickinson, Mountain View, Hydrocortisone
CA). The T helper cells were CD3+ and CD4+, whereas T Since exogenous hydrocortisone (Chc) and endogenous
suppressor cells were CD3+ and CD8+. The total num- cortisol (Cen) are indistinguishable, plasma hydrocorti-
ber of T helper and T suppressor cells was obtained by sone concentrations were estimated using a joint PK/
multiplying the respective proportion of fluorescent PD model, as shown in Figure 1. Under normal physio-
cells generated and the total number of lymphocytes. logical conditions, cortisol concentrations in the blood

1218 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

follow a circadian-episodic profile. The rate of change


of cortisol concentration (dCen/dt) can be described by a
periodic time-dependent production function, kin(t),
and the first-order elimination rate constant, kout,
according to
dC en (4)
= kin ( t ) − kout • C en .
dt

The mean baseline cortisol profile (placebo data) was


used to characterize kin(t) using Fourier analysis with
two harmonics. Mean baseline cortisol concentrations
were approximated by 24- and 12-hour period harmon-
ics. The number of harmonics was chosen based on the
percent contribution of each harmonic to the baseline
function, which was described by
 2π • nt   2π • nt 
2
C en = a 0 + ∑a
n =1
n • cos 
 24 
 + bn • sin 
 24 
, (5) Figure 1. PK/PD model of cortisol following intravenous (IV) hydro-
cortisone. Cen and Chc represent the concentrations of endogenous
and exogenous cortisol. The secretion of Cen is represented by a circa-
dian input function (kin), which is suppressed by Chc (black rectan-
where a0, a1, a2, b1, and b2 are Fourier coefficients obtained gle). The same kout value, the first-order cortisol elimination rate con-
by L2 norm approximation using FOURPHARM.20 stant, is used for Cen and Chc.
Hence, the cortisol input function, kin(t), was derived
from equations (4) and (5):
kin (t ) = kout • a 0 where C 0hc is the concentration of exogenous hydrocor-
tisone at time zero (D/Vd), and kout is the first-order
2
 b •2 π • n • t   2π • n • t 
+ ∑  kout • a n + n  • cos  + elimination rate constant. Total cortisol concentrations
n =1 
 24   24 
(Cm) can thus be defined as
(6)
 a •2 π • n • t   2π • n • t  
 kout • bn − n  • sin   .
 24   24   Cm = Chc + Cen. (10)

The PK and PD parameters were determined simulta-


The suppressive effect of GC on cortisol secretion can neously by fitting the cortisol data following hydrocor-
be characterized by an indirect response model,21 de- tisone dosing to equations (7) through (10), yielding Vd,
fined as kout, and IC50.
dC en
= kin ( t ) • I C ( t ) − kout • C en , (7)
dt Pharmacodynamics

CP Cortisol
IC (t) = 1 − , (8)
IC 50 + C p The dynamics of cortisol after methylprednisolone and
dexamethasone doses were modeled in a similar fash-
where IC(t) is the inhibition function that accounts for ion to hydrocortisone by fixing the PK parameters to
the suppression of kin by exogenous GC, Cp is the define Cp in equation (8) for each drug. The IC50 values
plasma concentrations of exogenous GC, and IC50 is the were estimated using equations (7) and (8) by fixing
concentration of exogenous GC achieving 50% inhibi- both kout and kin (equation (7)) to be the same value and
tion of cortisol secretion. function as during hydrocortisone treatment. Cortisol
The concentrations of exogenous hydrocortisone suppression by prednisolone was modeled similarly,
were represented by except kout in equation (7) was estimated. Prednisolone
is believed to displace cortisol from its binding sites in
dC hc (9)
= − kout • C hc , a concentration-dependent fashion,22 thus potentially
dt altering cortisol elimination (kout). Therefore, cortisol

PHARMACOKINETICS AND PHARMACODYNAMICS 1219


MAGER ET AL

data after prednisolone treatment were fitted by equa-


tions (7) and (8) to obtain both kout and IC50.
T Helper and T Suppressor Cell Dynamics
Under normal physiological conditions, T lymphocyte
trafficking in the blood exhibits a circadian rhythm,
with migration of cells between blood and
extravascular tissue compartments.23 Endogenous
cortisol and exogenous GC inhibit the movement of
lymphocytes from the peripheral tissues to the blood.24
Therefore, the lymphocytopenia observed after GC
doses is the net result of a direct effect of the dosed GC
and influence of the endogenous GC, as shown in Fig-
ure 2. Movement of lymphocytes between the two
pools can be represented by Figure 2. Pharmacodynamic model for lymphocyte trafficking.
Glucocorticoids (Cp) inhibit the endogenous cortisol (Cen) secretion
dEV rate (kin). Cp and Cen competitively inhibit movement of lymphocytes
= − keb • I T ( t ) • EV + k be • BL, (11)
dt from the extravascular (EV) to the blood (BL) pool.

dBL
= keb • I T ( t ) • EV − k be • BL, (12)
dt

where EV and BL represent the extravascular and only Cen is relevant according to equation (5). When hy-
blood pools of lymphocytes, while keb and kbe are drocortisone is dosed, Cp reflects Che based on equation
intercompartmental rate constants. Since the total lym- (10). Parameters estimated from modeling were kin, kbe,
phocyte pool in the EV compartment has been esti- and IC50, C. Parameters of lymphocyte dynamics for
mated to be 30 to 45 times that of the BL pool,25,26 equa- prednisolone, methylprednisolone, and dexametha-
tion (12) simplifies to sone treatment were also estimated using equations
(13) and (14). Plasma concentrations of the dosed GC
dBL and their resultant cortisol concentrations were fixed
= kin • I T ( t ) − k be • BL, (13)
dt functions derived from previous sections. The IC50, C
that was estimated from the placebo and hydrocorti-
where kin = keb • EV, an apparent zero-order rate con-
sone data was also retained as a fixed value. The esti-
stant, and IT(t) is the inhibition function related to the
mated parameters were kin, kbe, and IC50, GC.
GC effect on lymphocytes. The inhibitory function was
adapted from Milad et al24 and describes the joint ef- Neutrophil Dynamics
fects of exogenous GC and endogenous cortisol:
The elevation in blood neutrophils can be described
  IC   using indirect response model III21:
 C p •  50, C  + C en 
  IC 50, GC   dR  S max • C p 
I T (t) = 1 −  , (14) = kin • 1 +  − kout • R, (15)
IC  IC 50, C   dt  SC 50 + C p 
50, C + C p •   + C en
  IC 50, GC  
 
where R represents blood neutrophil counts, Smax is the
where IC50, C and IC50, GC reflect the cortisol and exoge- maximum stimulation factor, SC50 is the GC concentra-
nous GC concentrations that produce a 50% change in tion producing 50% of Smax, kout is the first-order re-
keb. For the analysis of data collected during placebo moval rate of neutrophils, and the apparent zero-order
administration, Cp is fixed at 0. appearance of neutrophils is defined as kin = kout • R(0).
The pharmacodynamics of lymphocyte trafficking The Smax was fixed to 3.0 according to earlier observa-
during hydrocortisone treatment were characterized tions.15,19
first by fitting placebo and hydrocortisone lymphocyte Since baseline neutrophils appear to be constant, it
data with equations (13) and (14). For the placebo data, was assumed that baseline cortisol concentrations

1220 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

have little or no effect on neutrophil profiles. The Cp for Var(Y) = (σ1 + σ2 • Y)2, (17)
hydrocortisone treatment is therefore the difference be-
tween the plasma concentration after treatment and where the σi values are the variance model parameters,
baseline cortisol concentrations (Chc in equation (9)). and Y represents the model-predicted values. The
By fixing the PK parameters derived for Cp, the goodness of fit was assessed by convergence of the re-
neutrophil cell counts from each treatment group were gression analysis, the Akaike Information Criterion
fitted to estimate kout and SC50. and Schwarz Criterion (AIC and SC), the estimator cri-
terion value in ADAPT II, the correlation coefficients
Dose Equivalency Determination (r 2), examination of residuals, and visual inspection.
The AUEC values of the biomarkers were compared
To determine the validity of currently accepted dose across drugs using repeated-measures ANOVA. If the
equivalency tables, area under the effect-time curve ANOVA was significant, then the Bonferroni correc-
(AUEC) values were used as indicators of net tion test was performed. Statistical significance was
pharmacodynamic effects. The AUECs were generated defined as p < 0.05. All statistical analyses were done
for each biomarker and were calculated as using SAS version 8.1 for Windows.

AUEC = |AUECplacebo – AUECtreatment|, (16) RESULTS


with areas obtained using the linear trapezoidal rule.
Due to their rebound patterns, the AUEC of T helper Pharmacokinetics
and T suppressor cells only includes the 0- to 20-hour
time frames. Figure 3 shows the mean plasma concentrations (± SD)
versus time, with the fitted curves generated from mod-
Data Analysis eling for each drug. Hydrocortisone, prednisolone, and
methylprednisolone PK profiles were best fitted with
All PK/PD model parameters were estimated by non- monoexponential disposition functions. The disposi-
linear regression analysis. The above models were fit- tion of dexamethasone was best described by a
ted to mean data (naive averaged data technique) using biexponential equation. The estimated parameters re-
WinNonlin (Pharsight Corp., Apex, NC), and data were sulting from the PK analyses are listed in Table II. The
weighted by the inverse of the variance (1/y2). A stan- relatively low CV% values for the naive averaged data
dard two-stage analysis was also conducted in which were indicative of good model fitting. There was gener-
individual PK/PD profiles were fitted using the maxi- ally good agreement between parameter estimates ob-
mum likelihood estimator in ADAPT II (Biomedical tained by fitting mean or individual data. Note that the
Simulations Resource, Los Angeles, CA). The variance CV% values for the standard two-stage method reflect
model in ADAPT was defined as interindividual variability.

Table II Estimated Pharmacokinetic Parameters


a
Hydrocortisone Prednisolone Methylprednisolone Dexamethasone
NAD STS NAD STS NAD STS NAD STS
–1
k12 (h ) NA NA NA NA NA NA 1.11 (39) 1.73 (89)
k21 (h–1) NA NA NA NA NA NA 0.919 (13) 0.895 (26)
ka (h–1) NA NA 1.25 (21) 2.84 (110) NA NA NA NA
V/F (L)b 119 (15) 115 (14) 168 (9.9) 156 (20) 83.4 (8.1) 77.1 (14) 42.8 (22) 37.6 (44)
kel (h–1) 0.405 (9.4) 0.420 (18) 0.272 (4.8) 0.296 (13) 0.27 (4.2) 0.296 (13) 0.416 (20) 0.572 (45)
CL/F (L/h)b,c 48.2 48.6 (25) 45.7 45.3 (8.1) 22.5 22.7 (13) 17.8 18.2 (27)
Data reported as estimate and CV% (naive averaged data [NAD]) or mean and CV% of individual estimates (standard two-stage [STS]). NA, not applicable; k12,
k21, first-order rate constants of drug transport from central to peripheral compartment; ka, first-order absorption rate constant; V/F, volume of distribution cor-
rected for bioavailability; kel, first-order drug elimination rate constant; CL/F, total systemic clearance corrected for bioavailability.
a. Free drug.
b. F = 1 for intravenous hydrocortisone, methylprednisolone, and dexamethasone.
c. Calculated as kel • V/F.

PHARMACOKINETICS AND PHARMACODYNAMICS 1221


MAGER ET AL

Placebo Prednisolone
Hydrocortisone Methylprednisolone
10000 160

1000 120

80
100
Plasma Glucocorticoid Concentration (ng/mL)

Cortisol Plasma Concentration (ng/mL)


40
10

0
1

Methylprednisolone Dexamethasone
Free Prednisolone Dexamethasone
1000 160

120
100

80

10 40

0
0 8 16 24 32 0 8 16 24 32
1
0 8 16 24 32 0 4 8 12 16 20 Time (hour)

Time (hour) Figure 4. Time course of mean (± SD) and fitted plasma cortisol
concentrations. The symbols represent experimental data, and the
Figure 3. Time course of mean (± SD) and fitted plasma glucocor- solid lines show the fittings of the model (equations (7) and (8)). The
ticoid concentrations. solid line for exogenous hydrocortisone is simulated based on Figure 1.

Pharmacodynamics kout, min value was calculated (0.32 h–1) and is the mini-
mum acceptable value of kout that ensures positive
values of Cen for the function represented by equation
Endogenous Cortisol
(7). The Fourier coefficients were substituted into
The mean cortisol concentrations (± SD) versus time equation (6) to characterize baseline cortisol secretion.
with the model fittings for each GC are shown in Fig- The estimated PD parameters of cortisol suppression
ure 4. The normal circadian variation in cortisol con- for each drug are shown in Table III. The estimated IC50
centrations is shown in Figure 4 as the baseline plot. of prednisolone of 1.25 ng/mL was comparable to that
The episodic nature of cortisol secretion is evident, of previous studies. 1 9 Although the methyl-
with the peak occurring at 8 a.m. and the nadir at about prednisolone IC50 was threefold lower than that of pre-
midnight. Figure 4 also shows the cortisol suppression vious studies,27 the variability for both studies was
after administration of GC. Cortisol concentrations rap- high, as suggested from individual fittings. Such infor-
idly decline and reach a nadir at approximately 5 mation is unavailable for hydrocortisone and
hours. After hydrocortisone and prednisolone doses, dexamethasone.
the circadian rhythm resumes around 15 hours later.
T Helper and T Suppressor Lymphocytes
After methylprednisolone, the circadian rhythm re-
sumes closer to 28 hours, whereas for dexamethasone, During baseline conditions, T helper and T suppressor
cortisol suppression continues past 32 hours. lymphocytes exhibit circadian rhythms reversed to
The Fourier coefficients obtained for the character- that of cortisol secretion, with the nadir occurring at 8
ization of mean baseline cortisol dynamics were a0 = a.m. and the peak at midnight, as shown in Figures 5
49.66, a1 = 28.23, b1 = 9.955, a2 = 10.39, and b2 = –8.606 and 6. After GC administration, a rapid decline of cell
(number of harmonics and calculated Fourier coeffi- counts occurs, with the nadir at about 5 hours. The T
cients for individual cortisol profiles is not shown). A helper cell counts returned to baseline levels at 12, 20,

1222 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

Table III Pharmacodynamic Parameters of Cortisol Suppression and Cell Trafficking


Placebo and
Hydrocortisone Prednisolonea Methylprednisolone Dexamethasone
Treatment NAD STS NAD STS NAD STS NAD STS

Cortisol suppression
IC50 (ng/mL) 8.01 (70) 4.61 (114) 1.25 (14) 1.87 (34) 0.52 (37) 1.87 (89) 0.172 (30) 0.105 (62)
kout (h–1) 0.405 (9.4) 0.420 (18) 1.54 (26) 1.73 (62) 0.405b —c 0.405b —c
T helper cell
trafficking
IC50, C (ng/mL) 52.4 (30) 56.4 (31) 52.4b —c 52.4b —c 52.4b —c
IC50, GC (ng/mL) NA NA 5.76 (14) 5.35 (41) 9.93 (12) 11.1 (138) 3.36 (14) 4.00 (118)
kin (cell/µL/h) 695 (16) 724 (23) 947(8.5) 1120 (16) 862 (7.2) 1090 (33) 960 (9.2) 989 (51)
kbe (h–1) 0.324 (16) 0.329 (30) 0.349 (7.4) 0.361 (24) 0.350 (6.1) 0.415 (35) 0.441 (7.8) 0.415 (34)
T suppressor
cell trafficking
IC50, C (ng/mL) 99.5 (40) 103 (17) 99.5b —c 99.5b —c 99.5b —c
IC50, GC (ng/mL) NA NA 21.1 (10) 19.2 (11) 58.0 (11) 41.7 (37) 18.7 (18) 18.0 (26)
kin (cell/µL/h) 197 (19) 328 (34) 328 (8.1) 375 (20) 338 (9.7) 494 (113) 568 (20) 596 (102)
kbe (h–1) 0.197 (24) 0.327 (39) 0.282 (8.4) 0.281 (32) 0.308 (10) 0.389 (82) 0.492 (21) 0.427 (54)
Neutrophil trafficking
SC50 (ng/mL) 155 (25) 179 (37) 20.5 (21) 41.0 (102) 26.4 (32) 36.0 (90) 7.93 (27) 6.07 (88)
kout (h–1) 0.0732 (13) 0.0562 (22) 0.0451 (11) 0.0657 (13) 0.0621 (16) 0.0883 (49) 0.0646 (17) 0.0715 (35)
Data reported as estimate and CV% (naive averaged data [NAD]) or mean and CV% of individual estimates (standard two-stage [STS]). IC50, drug concentration
producing 50% of cortisol secretion; IC50, C, cortisol concentration producing 50% inhibition of T cell trafficking into circulation; IC50, GC, drug concentration
producing 50% inhibition of T cell trafficking into circulation; kbe, first-order rate constant of removal of T cells from circulation; kin, apparent zero-order rate
constant of appearance of T cells in circulation; kout, first-order rate constant of cortisol elimination or systemic removal of neutrophils; NA, not applicable; SC50,
drug concentration producing 50% maximum stimulation of neutrophil cell production.
a. Free drug.
b. Fixed values.
c. Fixed to individual values estimated for placebo and hydrocortisone.

18, and 24 hours after dosing with hydrocortisone, Dose Equivalency Determination
prednisolone, methylprednisolone, and dexametha-
sone, respectively. The T suppressor cell counts re- Comparisons of relative responses were made by exam-
turned to baseline at 16 hours after dosing. A rebound ining calculated AUEC values. The mean AUEC (± SD)
phenomenon is observed that continues past the 32- values of the evaluated biomarkers are listed in Table IV.
hour time point. The PD parameters obtained from For T helper cells, there was a significant difference be-
modeling are presented in Table III. Parameters esti- tween the groups (p = 0.014), with dexamethasone
mated for the prednisolone treatment were comparable showing almost a twofold higher activity than hydro-
to those of previous studies.19 The IC50, GC value of cortisone. Subsequent post hoc analysis using the
methylprednisolone was found to be only half of the Bonferroni method showed no difference. For cortisol
previous study but again may reflect interindividual suppression, prednisolone and dexamethasone
variability.27 Such PD parameters of the other GC for showed a greater effect than hydrocortisone and
cell trafficking were not found. methylprednisolone. Similar to the T helper cell com-
Neutrophils parison, there was a significant difference between
groups (p = 0.022) with the initial analysis, while sub-
Figure 7 shows the mean plasma neutrophil cell counts sequent post hoc analysis using the Bonferroni method
(± SD) versus time with the fitted curves. Baseline showed no differences.
neutrophil levels were constant at approximately 3.58 ×
106 cells/mL (data not shown). The estimated PD pa- DISCUSSION
rameters also are listed in Table III. The IC50 estimated
for prednisolone was similar to that found by Magee We have reevaluated the validity of the dose equiva-
et al.19 Similar data were unavailable for the other GC. lency table of GC by following cell trafficking dynam-

PHARMACOKINETICS AND PHARMACODYNAMICS 1223


MAGER ET AL

Placebo and Hydrocortisone Prednisolone Hydrocortisone Prednisolone

2500 10

2000 8

1500 6

1000 4

Neutrophil Cell Count (x106 cells/mL)


T-helper Cell Count (x103 cells/mL)

500 2

0 0

Methylprednisolone Dexamethasone Methylprednisolone Dexamethasone


2500 10

2000
8

1500
6

1000
4

500
2

0
0 8 16 24 32 0 8 16 24 32 0
0 8 16 24 32 0 8 16 24 32
Time (hour)
Time (hour)

Figure 5. Time course of mean (± SD) and fitted T helper lympho- Figure 7. Time course of mean (± SD) and fitted neutrophil cell
cyte cell counts. The placebo baseline phase is represented by the counts. Symbols and lines are as in Figure 5.
open circles. The treatment data (䊉) and model-fitted profiles (solid
and broken lines) are shown.

ics and adrenal suppression effects using moderate


Placebo and Hydrocortisone Prednisolone
doses of the drugs. Although studies have shown no
1600
differences in the qualitative anti-inflammatory prop-
erties with equivalent doses, these were reported in the
1200 late 1960s and early 1970s.28,29 With the increase in use
of GC, the integrity of the dose equivalency table has
800
become an important clinical consideration. The ad-
T-supressor Cell Count (x 103 cells/mL)

400
vent of sophisticated assays and mechanistic modeling
approaches can provide for a scientifically based
0 evaluation of the dose equivalency table.
The PK parameter values estimated for this study
were comparable to those of previous investiga-
Methylprednisolone Dexamethasone tions.19,30 The free prednisolone concentrations were
2000 used in this study due to nonlinear protein-binding
1600
characteristics.30 The estimated PK parameters were
fixed and used in indirect response models that were
1200 applied to estimate PD parameters. The results of fit-
800
ting mean and individual PK/PD data are reported, and
final estimates were similar between methods (Tables
400 II and III).
0
The cortisol dynamic model presented in this study
0 8 16 24 32 0 8 16 24 32 is an extension of the indirect response model I,21 in
Time (hour)
which the baseline input followed a circadian-episodic
profile. The input function for the mean cortisol base-
Figure 6. Time course of mean (± SD) and fitted T suppressor lym-
phocyte cell counts. Symbols and lines are as in Figure 5. line was modeled using a two-harmonic Fourier analy-

1224 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

Table IV AUEC Values of Measured Pharmacodynamic Markers


Marker Hydrocortisone Prednisolone Methylprednisolone Dexamethasone p-Value

T helper cells (103 cells•h/mL)a


Mean 9730 13,100 12,500 16,800 0.014
Standard deviation 4630 8700 7910 8000
T suppressor cells (103 cells•h/mL)a,b
Mean 4870 6070 5490 6900 0.43
Standard deviation 2270 6050 4120 3660
Neutrophils (103 cells•h/mL)
Mean 42.7 65.3 111 129 0.14
Standard deviation 26.8 30.1 57.9 40.6
Cortisol (ng•h/mL)
Mean 1070 1510 1150 1430 0.022
Standard deviation 230 452 394 237
a. Area under the effect-time curve (AUEC) value calculated from 0 to 20 hours only.
b. AUEC value of 1 subject was excluded due to abnormally low T suppressor cells during the placebo period.

sis, as presented by Chakraborty et al.31 This technique


provided a very good fitting to the data, as shown in the
cortisol baseline graph of Figure 4. The inhibition func- 2

tion of equation (8) then accounts for the suppression


of cortisol secretion by the administered drugs. Due to
the inability of our assay to distinguish between endog- 1

enous and administered hydrocortisone, the measured


log ( 1 / EC50 )

cortisol concentrations reflect both values. The PK pa-


rameters and cortisol PD parameters of hydrocortisone 0
treatment were therefore estimated simultaneously by
linking the two models together, as shown in Figure 1.
For all other treatments, the PK parameters were first -1
determined and then fixed in the cortisol dynamic
model.
The lymphocyte PD model used in this study was -2
1 2
modified from previous studies,32,33 in which the lym-
phocyte cell trafficking compartments were simplified log RRA
to the extravascular and blood pools. This model Figure 8. Correlation between log-reciprocal EC50 values (IC50 and
jointly accounts for the direct effect of GC and the GC- SC50 values by naive averaged data [NAD] in Table III) and relative
30
suppressed cortisol effect on lymphocyte trafficking. receptor affinity (RRA) from Jusko and Ludwig (hydrocortisone, 9;
prednisolone, 16; methylprednisolone, 42; dexamethasone, 100).
The neutrophil PD model used in this study was the Symbols and regression lines are shown for cortisol suppression (䊉,
simple indirect response model III.21 The baseline 2
r 2 = 0.95, p < 0.05), T helper cells (䉮, r = 0.999, p < 0.001), and T sup-
2
neutrophil counts, under the influence of baseline pressor cells and neutrophils (䊏, 䉫, r = 0.829, p < 0.005).
cortisol concentrations, were considered to be con-
stant. Therefore, the drug concentrations, Cp, that drive
the response during hydrocortisone treatment were the
difference between total cortisol measured and the fact well correlated with reported relative receptor af-
baseline concentrations. For all other treatments, finity values (equilibrium dissociation constants
cortisol concentrations were less than baseline and normalized to dexamethasone),30 as shown in Figure 8.
thus have little or no contribution to neutrophil To compare values with prednisolone, which was
dynamics. based on free drug concentrations, we corrected the
The major initiating event in corticosteroid pharma- EC50 values for hydrocortisone, methylprednisolone,
codynamics is receptor binding. The EC50 values re- and dexamethasone for protein binding (respective
ported in Table III (IC50 and SC50 parameters) were in free fractions = 0.20, 0.23, and 0.32).34 Whereas the EC50

PHARMACOKINETICS AND PHARMACODYNAMICS 1225


MAGER ET AL

values for each GC were biomarker dependent, the 7. Chrousos GP: The hypothalamic-pituitary-adrenal axis and im-
good correlation to receptor affinity was found across mune-mediated inflammation. N Engl J Med 1995;332:1351-1362.
all GC for each tested biomarker and is in agreement 8. Cronstein BN, Kimmel SC, Levin RI, Martiniuk F, Weissmann G: A
mechanism for the antiinflammatory effects of corticosteroids: the
with previous reports of this relationship.34 glucocorticoid receptor regulates leukocyte adhesion to endothelial
Although significant differences were found with T cells and expression of endothelial-leukocyte adhesion molecule 1
helper cells and cortisol suppression effects in the and intracellular adhesion molecule 1. Proc Natl Acad Sci USA 1992;
comparison of AUEC to assess dose equivalency, 89:9991-9995.
post hoc Bonferroni analysis showed no difference be- 9. Nakano T, Ohara O, Teraoka H, Arita H: Glucocorticoids suppress
tween treatments. Post hoc analysis using the Fisher’s group II phospholipase A2 production by blocking mRNA synthesis
least significant difference test (LSD) resulted in a dif- and post transcriptional expression. J Bio Chem 1990;265:12745-
12748.
ference between hydrocortisone and dexamethasone
10. Steer JH, Vuong Q, Joyce DA: Suppression of human monocyte tu-
effects on T helper cells. For the cortisol suppression mor necrosis factor-release by glucocorticoid therapy: relationship to
comparison, LSD also found hydrocortisone to differ systemic monocytopenia and cortisol suppression. Br J Clin
from dexamethasone and prednisolone, while Pharmacol 1997;43:383-389.
methylprednisolone differs from prednisolone. The 11. Fauci AS: Mechanisms of corticosteroid action on lymphocyte
Bonferroni method is stricter than LSD and is the better subpopulation. Immunology 1975;78:71-100.
of the two methods for this type of comparison. Al- 12. Yu DT, Clements PJ, Paulus HE, Peter JB, Levy J, Barnett EV: Hu-
though limited by the study of single doses and a small man lymphocyte subpopulations: effect of corticosteroids. J Clin In-
sample size, these results suggest that there are no sig- vest 1974;53:565-571.
nificant differences between the treatments based on 13. Zweiman B, Atkins PC, Bedard PM, Flaschen SL, Lisak RP:
AUEC analysis. In consideration of the small sample Corticosteroid effects on circulating lymphocyte subset levels in nor-
mal humans. J Clin Immunol 1984;4:151-155.
size, the number of subjects may have been inadequate,
14. Fauci AS, Dale DA, Balow JE: Glucocorticosteroid therapy: mech-
and thus there is a possibility that true differences exist
anism of action and clinical considerations. Ann Int Med 1976;
among the drugs, which may or may not be clinically 84:304-315.
relevant. Study of a larger population size using more 15. Burton JL, Kehrli ME Jr, Kapil S, Horst RL: Regulation of L-selectin
than one GC dosage level may be needed to ascertain and CD18 on bovine neutrophils by glucocorticoids: effects of
possible modest differences between these drugs. The cortisol and dexamethasone. J Leukoc Biol 1995;57:317-325.
dosage levels are important as AUEC values change 16. Lipworth BJ: Systemic adverse effects of inhaled corticosteroid
nonlinearly with dose, although they are proportional therapy: a systemic review and meta-analysis. Arch Int Med
to log dose at higher dosage levels, making compari- 1999;159:941-955.
sons such as these somewhat complicated.35 17. Rose JQ, Jusko WJ: Corticosteroid analysis in biological fluids by
high performance liquid chromatography. J Chromatogr 1979;
We thank the clinical staff of the Buffalo Clinical Research Labora- 162:273-280.
tory and Ms. Nancy Pyszczynski and Ms. Suzette Mis for providing 18. Jusko WJ, Pyszczynski NA, Bushway MS, D’Ambrosio R, Mis SM:
valuable technical support. Special thanks also go to Mr. David Collins Fifteen years of operation of a high performance liquid chromato-
for his assistance with the statistical analyses. graphic assay for prednisolone, cortisol and prednisone in plasma. J
Chromatogr B Biomed Appl 1994;658:47-54.
REFERENCES 19. Magee MH, Blum RA, Lates CD, Jusko WJ: Prednisolone
pharmacokinetics and pharmacodynamics in relation to sex and
1. Dipiro JT, Talbert RL, Yee GC, Matzke GR, Wells GR, Posey LM: race. J Clin Pharmacol 2001;41:1180-1194.
Pharmacotherapy. 3rd ed. Stamford, CT: Appleton & Lang, 1996. 20. Krzyzanski W, Chakraborty A, Jusko WJ: Algorithm for applica-
2. Liddle GW: Clinical pharmacology of the anti-inflammatory ste- tion of Fourier analysis for biorhythmic baselines of pharma-
roids. Clin Pharmacol Ther 1961;2:615-635. codynamic indirect response models. Chronobiol Int 2000;17:77-93.
3. Thorn GW: Clinical considerations in the use of corticosteroids. N 21. Dayneka NL, Garg V, Jusko WJ: Comparison of four basic models
Engl J Med 1966;274:775-781. of indirect pharmacodynamic responses. J Pharmacokin Biopharm
4. David DS, Grieco MH, Cushman P Jr: Adrenal glucocorticoids after 1993;21:457-478.
twenty years: a review of their clinically relevant consequences. J 22. Rocci ML, D’Ambrosio R, Jusko WJ: Prednisolone binding to albu-
Chronic Dis 1970;22:637-711. min and transcortin in the presence of cortisol. Biochem Pharmacol
5. Hardman JG, Limbird LE, Molinoff PB, Ruddon RW, Gilman AG: 1982;31:289-292.
Goodman & Gilman’s The Pharmacological Basis of Therapeutics. 23. Miyawaki T, Taga K, Nagaoki T, Seki H, Suzuki Y, Taniguchi N:
9th ed. New York: McGraw-Hill, 1995. Circadian changes of T lymphocyte subsets in human peripheral
6. Derendorf H, Hochhaus G, Mollmann H, Barth J, Krieg M, Tunn S, blood. Clin Exp Immunol 1984;55:618-622.
et al: Receptor-based pharmacokinetic-pharmacodynamic analysis of 24. Milad MA, Ludwig EA, Ann S, Middleton E Jr, Jusko WJ:
corticosteroids. J Clin Pharmacol 1993;33:115-123. Pharmacodynamic model for joint exogenous and endogenous

1226 • J Clin Pharmacol 2003;43:1216-1227


DOSE EQUIVALENCY EVALUATION OF CORTICOSTEROIDS

corticosteroid suppression of lymphocyte trafficking. J Pharmacokin 30. Jusko WJ, Ludwig EA: Corticosteroids, in: Evans WE, Schentag JJ,
Biopharm 1994;22:469-480. Jusko WJ (eds.), Applied Pharmacokinetics. 3rd ed. Vancouver: Ap-
25. Field EO, Sharpe HB, Dawson KB, Anderson V, Killmann SA, plied Therapeutics, Inc., 1992.
Weeke E: Turnover rate of normal blood lymphocytes and exchange- 31. Chakraborty A, Krzyzanski W, Jusko WJ: Mathematical modeling
able pool size in man, calculated from analysis of chromosomal aber- of circadian cortisol concentrations using indirect response models:
rations sustained during extracorporeal irradiation of the blood. comparison of seven methods. J Pharmacokin Biopharm 1999;
Blood 1972;39:39-56. 27:23-43.
26. Trepel F: Number and distribution of lymphocytes in man: a criti- 32. Chow F, Sharma A, Jusko WJ: Modeling interactions between ad-
cal analysis. Klin Wocheuschr 1974;52:511-515. renal suppression and T-helper lymphocyte trafficking during multi-
27. Lew KH, Ludwig EA, Milad MA, Donovan K, Middleton E Jr, ple dosing of methylprednisolone. J Pharmacokinet Biopharm
Ferry JJ, et al: Gender based effects on methylprednisolone 1999;27:559-575.
pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 33. Meibohm B, Derendorf H, Mollmann H, Frohlich P, Tromm A,
1993;54:402-414. Wagner M, et al: Mechanism-based PK/PD model for the
28. Peters WP, Holland JF, Senn H, Rhomberg W, Banerjee T: lymphocytopenia induced by endogenous and exogenous
Corticosteroid administration and localized leukocyte mobilization corticosteroids. Int J Clin Pharmacol Ther 1999;37:367-376.
in man. N Engl J Med 1972;286:342-345. 34. Derendorf H, Mollmann H, Hochhaus G, Meibohm B, Barth J:
29. Fauve RM, Pierce-Chase CH: Comparative effects of corti- Clinical PK/PD modeling as a tool in drug development of
costeroids on host resistance to infection in relation to chemical corticosteroids. Int J Clin Pharmacol Ther 1997;35:481-488.
structure. J Exp Med 1967;125:807-821. 35. Krzyzanski W, Jusko WJ: Integrated functions for four basic
models of indirect pharmacodynamic response. J Pharm Sci 1998;
87:67-72.

PHARMACOKINETICS AND PHARMACODYNAMICS 1227

Das könnte Ihnen auch gefallen