Influence of Everolimus On Steady-State Pharmacokinetics of Cyclosporine

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

The Journal of Clinical

Pharmacology
http://www.jclinpharm.org

Influence of Everolimus on Steady-State Pharmacokinetics of Cyclosporine in Maintenance Renal


Transplant Patients
Klemens Budde, Gustav Lehne, Michael Winkler, Lutz Renders, Arno Lison, Lutz Fritsche, Jean-Paul Soulillou, Per
Fauchald, Hans-Hellmut Neumayer, Jaques Dantal and RADW 102 Renal Transplant Study Group
J. Clin. Pharmacol. 2005; 45; 781
DOI: 10.1177/0091270005277196

The online version of this article can be found at:


http://www.jclinpharm.org/cgi/content/abstract/45/7/781

Published by:

http://www.sagepublications.com

On behalf of:
American College of Clinical Pharmacology

Additional services and information for The Journal of Clinical Pharmacology can be found at:

Email Alerts: http://www.jclinpharm.org/cgi/alerts

Subscriptions: http://www.jclinpharm.org/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

Downloaded from http://www.jclinpharm.org by Luciana Oliveira on April 21, 2009


BUDDEINTERACTIONS
10.1177/0091270005277196
EVEROLIMUS
DRUG ET AL AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

Influence of Everolimus on Steady-State


Pharmacokinetics of Cyclosporine
in Maintenance Renal Transplant Patients
Klemens Budde, Gustav Lehne, Michael Winkler, Lutz Renders, Arno Lison, Lutz Fritsche,
Jean-Paul Soulillou, Per Fauchald, Hans-Hellmut Neumayer, and Jaques Dantal,
on Behalf of the RADW 102 Renal Transplant Study Group

To investigate possible interactions of the novel 43%, which were not significantly different across all dosing
immunosuppressant everolimus with cyclosporine, a cohorts including placebo. Linear regression of everolimus
multicenter, randomized, double-blind, placebo-controlled, AUC on day 21 versus the increase in cyclosporine AUC0-12
dose-escalating phase I study was performed. Everolimus yielded a slope not significantly different from a horizontal
regimens (0.75-10 mg/d) were administered for 28 days to sta- line (P = ns). In conclusion, these results suggest that steady-
ble renal allograft recipients receiving the microemulsion state everolimus exposure over the wide range assessed
form of cyclosporine. Steady-state cyclosporine profiles were in this study did not affect steady-state cyclosporine
assessed at baseline on day 0 (cyclosporine alone) and on day pharmacokinetics.
21 with everolimus on steady state. By day 21, mean dose-
normalized cyclosporine AUC0-12 increased by 15% in pa- Keywords: Cyclosporine; everolimus; pharmacokinetics; re-
tients receiving placebo. In everolimus-treated patients, nal transplantation
mean increases in cyclosporine AUC0-12 ranged from 7% to Journal of Clinical Pharmacology, 2005;45:781-791
©2005 the American College of Clinical Pharmacology

T he novel macrocyclic immunosuppressant


everolimus (40-O-(hydroxy)ethyl-rapamycin) is a
promising candidate for potent adjunctive
tation.1,2 In experimental models, everolimus was syn-
ergistic with cyclosporine (CsA) in the prevention of
acute and chronic rejection and has been approved in
immunosuppressive therapy in solid organ transplan- several countries in combination therapy with CsA af-
ter kidney and heart transplantation.1-3 Everolimus be-
longs to the same drug class as sirolimus, but relatively
From the University Clinic Charité, Berlin, Germany (Dr Budde, Dr Fritsche, minor structural differences between the 2 agents con-
Dr Neumayer); Nephrology Service, CHU Hotel Dieu, Nantes, France (Dr fer different pharmacokinetic characteristics. 4-8
Soulillou, Dr Dantal); Rikshospitalet, Oslo, Norway (Dr Lehne, Dr
Everolimus has a shorter half-life (approximately 24
Fauchald); Medizinische Hochschule, Hannover, Germany (Dr Winkler);
Universitätsklinikum Kiel, Germany (Dr Renders); and Zentral-
hours vs 60 hours), exhibits more stable/consistent ab-
krankenhaus, Bremen, Germany (Dr Lison). The study group contains the sorption characteristics than oral sirolimus does,4-8 and
following additional members: L. Lerat (Nantes, France), K. Nordal (Oslo, may exert different effects on microsomal and mito-
Norway), L. Müller, R. Brunkhost (Hannover, Germany), I. A. Hauser chondrial metabolism.9,10
(Frankfurt, Germany), B. Charpentier (Safety Board member, Le Kremlin Sirolimus and everolimus have potent immunosup-
Bicêntre, France), P. McMaster (Safety Board member, Birmingham, United pressive activity and have a mode of action different
Kingdom), Albrecht-Georg Schmidt, Annette Jappe, Nathalie Cambon,
from that of CsA and other classes of immuno-
Janet Von Fellenberg, Khazal Paradis, Silke Appel-Dingemanse, Francois
Legay (Basel, Switzerland), and Christophe Gerbeau (Paris, France). Ad- suppressants.1,11 They target the signal transduction
dress for reprints: Klemens Budde, University Clinic Charité, Schumannstr. pathway involved in cell-cycle progression by inhibit-
20/21, Berlin 10098, Germany. ing the mammalian target of rapamycin (mTOR), thus
DOI: 10.1177/0091270005277196 leading to subsequent inhibition of interleukin-2 (IL-

J Clin Pharmacol 2005;45:781-791 781


BUDDE ET AL

2)–induced T-cell proliferation as well as inhibition of MATERIAL AND METHODS


general growth-factor–dependent proliferation of
hematopoietic and nonhematopoietic cells, including Patients
vascular smooth muscle cells.11,12 Chronic rejection has
been attributed to proliferation of vascular smooth Stable recipients of a primary renal transplant
muscle cells.12 Therefore, improved immunosup- (cadaveric or living donor) were eligible for inclusion
pression and a direct effect of these drugs on smooth in the study. All subjects gave written informed con-
muscle cell proliferation may have an impact on long- sent prior to inclusion. The study was conducted in ac-
term outcomes.12 cordance with good clinical practice guidelines and in
Sirolimus, the first drug of the class of mTOR inhibi- line with the Declaration of Helsinki. The protocol was
tors, is primarily metabolized through the hepatic approved by the Ethics Committee at each center (see
cytochrome P450 (CYP) 3A4 enzyme system, which is the appendix). For at least 3 months before enrollment
shared with CsA mutual microsomal metabolism.7-10 In into the study, all patients were receiving the
addition, both drugs are substrates for p-glycoprotein microemulsion formulation of CsA (Neoral; Novartis
countertransport.7-10 Not surprising, several studies Pharma, Basel, Switzerland) sufficient to produce
have demonstrated a strong pharmacokinetic interac- morning trough levels of 80 to 200 ng/mL. For inclu-
tion between sirolimus and CsA.13-20 Interestingly, CsA sion in the study, women had to be practicing an ac-
pharmacokinetics is unaffected by sirolimus in renal cepted method of birth control. Individuals were ex-
transplant recipients.13,14 In contrast, CsA administra- cluded from the study if they had received any other
tion resulted in markedly (1.8- to 2.3-fold) elevated immunosuppressive medication within 4 months of
sirolimus exposure20; even the timing of CsA dosing af- baseline. Any treatment interfering with CsA
fected sirolimus pharmacokinetics. Whole-blood pharmacokinetics was not allowed for 4 weeks prior to
trough levels increased by about 30% with concomi- enrollment. The detailed study protocol and patient
tant dosing; the time to peak levels was also shorter (1.8 characteristics were published recently.21 In brief, a to-
vs 2.5 hours) compared to timely separated administra- tal of 54 patients were randomized into 1 of the 7 differ-
tion.20 As a consequence, sirolimus should be given 4 ent treatment groups, 44 subjects received everolimus
hours after CsA according to the dosing recommenda- (Novartis Pharma, Basel, Switzerland), and 10 patients
tions. No such effect of timing of drug administration, received placebo.
however, could be found on CsA pharmacokinetic
parameters.20 Study Design
Since everolimus is intended to be used with CsA
and both compounds are metabolized in part by This was a randomized, double-blind, placebo-
CYP3A4,9,10 the potential influence of everolimus on controlled, dose-escalation study conducted at 6 cen-
CsA pharmacokinetics needs to be investigated. This is ters. Subjects fulfilling the entry criteria were ran-
the first study to investigate the tablet formulation of domly assigned in a 3:1 ratio to either 1 of 4 everolimus
everolimus across a wide range of doses, unlike the dose groups or placebo. This comprised 7 treatment
previously reported phase I single-dose study5 and a groups to examine the effect of dose level, dosing fre-
multiple-dose study that used in development a ser- quency (once daily [QD] or twice daily [BID]), and for-
vice capsule formulation6 that will not be commer- mulation (capsule and tablet at 0.75 mg QD). Dose
cially available. This is of particular importance as the groups (0.75, 5, and 10 mg/d) were initiated sequen-
tablet has a 2.6-fold higher bioavailability compared to tially at weekly intervals so that a preliminary assess-
the service capsule.21 The aim of the present study was ment of safety and tolerability could be made at each
to assess the effect of steady-state everolimus dose level prior to escalation to the next dose level.
coadministration over a wide dose range on detailed Safety and tolerability were monitored in an ongoing
steady-state pharmacokinetics of CsA in stable re- manner throughout the study by an independent, un-
nal allograft recipients. Unlike previously reported blinded Safety Monitoring Board constituted specifi-
studies, 22-26 complete 12-hour CsA pharmaco- cally for this study. An additional dose group (2.5 mg
kinetics in stable renal allograft recipients receiv- QD, open label) was added toward the end of the study.
ing the microemulsion formulation of CsA was as- In addition, 4 patients were added in an open-label
sessed, as it is well known that CsA exposure varies fashion to the 2.5-mg-BID group to further assess the
over time22 and trough levels are a poor indicator of safety and tolerability of the 5-mg daily dose, given
CsA exposure.27 concomitantly with cyclosporine in a BID dosing regi-

782 • J Clin Pharmacol 2005;45:781-791


EVEROLIMUS AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

men. Treatment at each dose level was administered for curve (25-2500 ng/mL) and 5 quality control concen-
4 weeks. At the baseline visit (before administration of trations (30-1600 ng/mL). The precision of quality
study drug, while the patients were hospitalized for the control samples (coefficients of variation) ranged from
baseline CsA pharmacokinetic profile), creatinine 5.1% to 13.5%, and the assay quantification limit was
clearance was determined from a 24-hour urine sam- 25 ng/mL.
pling period using the local laboratory at each center.
Pneumocystis carinii prophylaxis with cotrimoxazole/ Statistical Evaluation
sulfamethoxazole was mandated during the course of
the study. This study was of an exploratory design; that Standard noncompartmental pharmacokinetic param-
is, the study was not powered to address a specific sta- eters were derived including the peak concentration
tistical hypothesis. The results of the safety and (Cmax) and the time of its occurrence (tmax), the area un-
pharmacokinetic exploration of everolimus were der the concentration-time curve over the dosing inter-
published separately.21 val (AUC), and the percentage peak-trough fluctuation
(PTF). Attainment of steady state for everolimus and
Pharmacokinetic Assessment CsA was assessed by linear regression analysis of the
serial trough concentrations over time. A slope not sig-
Morning trough whole-blood levels (Cmin) of CsA were nificantly different from zero (a horizontal line) was
measured at 1- to 3-day intervals throughout the study. taken as evidence for steady-state conditions. Serial
Steady-state CsA pharmacokinetic profiles over the 12- CsA trough concentrations were plotted for each
hour dosing interval were obtained at baseline in the everolimus dosing group and inspected for trends over
absence of everolimus and on day 21 during the study duration. Linear regression analysis was per-
everolimus coadministration when steady-state formed for each patient’s troughs over time. A signifi-
everolimus blood concentrations would have been at- cant positive slope was interpreted as a rise in CsA
tained. For CsA profiling, blood samples were drawn exposure over the course of the study.
predose and after 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 8, The influence of everolimus on CsA pharmaco-
10, and 12 hours. Full pharmacokinetic profiles of kinetics was explored by an analysis of variance
everolimus over a 12- or 24-hour dosing interval were (ANOVA) on dose-normalized CsA parameters from
obtained on days 1 and 21. For everolimus profiles, baseline (without everolimus) and day 21 (under
blood samples were obtained from a forearm vein via steady-state everolimus). The ANOVA included terms
an indwelling cannula predose and then 0.25, 0.5, 0.75, for everolimus dose level (including placebo), patient
1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 8, 10, 12, 16, and 24 nested within everolimus dose level, study day (days 0
hours thereafter. On profiling occasions, patients had and 21), and day by everolimus dose-level interaction.
an overnight fasting period of at least 12 hours, and af- This model explored whether differences in CsA
ter administration of the medication, they remained pharmacokinetic parameters between baseline in the
fasting for an additional 4 hours; only water was al- absence of everolimus and day 21 during everolimus
lowed in this period. All blood samples were drawn coadministration were different among the everolimus
into EDTA-coated collection tubes, gently inverted sev- dose levels including placebo. Pharmacokinetic pa-
eral times, frozen at –20°C or below, and analyzed in a rameter ratios (coadministration on day 21/baseline
central laboratory. on day 0) were also derived and compared among
Whole-blood everolimus concentrations were deter- everolimus dosing levels in a 1-way ANOVA. Linear
mined in duplicate by a validated enzyme-linked regression analysis was performed on everolimus
immunosorbent assay method.5,6 Assay performance AUC on day 21 versus the CsA AUC ratio. A slope
was based on a 7-point calibration curve (1.6 to 100 ng/ not significantly different from zero was taken as evi-
mL) and 5 quality control concentrations (2 to 80 ng/ dence that everolimus had no influence on CsA
mL) determined with each assay run. Assay precision pharmacokinetics.
(coefficients of variation) ranged from 11.2% to 26.3%,
and accuracy (deviation from nominal value) ranged RESULTS
from –1.6% to –8.8%. The limit of quantification was 2
ng/mL. CsA was quantified in whole blood by a com- Subjects
mercially available radioimmunoassay using a
monoclonal antibody specific for the parent com- A total of 54 subjects were randomized for treatment
pound (INCSTAR Cyclo-Trac SP, Stillwater, Minn). As- with everolimus (n = 44) or placebo (n = 10). There
say performance was based on a 7-point calibration were no significant demographic differences between

DRUG INTERACTIONS 783


BUDDE ET AL

different treatment groups and placebo (Table I). The Table I Baseline Demographics and Medical
median age was 47 years (range, 25-66), the majority of Characteristics
patients were Caucasian (93%) and male (69%), and no
black patients participated. They were at least 6 Everolimus Placebo
months posttransplant (mean = 5.3 years) and had sta- Variable (n = 44) (n = 10)
ble serum creatinine levels (mean = 147 ± 46 µmol/L;
Age, y
everolimus 149 ± 49 µmol/L; placebo 139 ± 42 µmol/L).
Median (range) 47 (25-66) 46 (25-57)
The average glomerular filtration rate (using 24-hour
Gender, %
urine collection) was 68 ± 22 mL/min (range, 28-128) at
Male:female 68:32 70:30
baseline. As shown in Table I, the immunosuppressive
Race, % Caucasian 93 90
therapy mainly consisted of CsA and prednisone; the
Weight, kg 69 ±14 75 ± 6
mean prednisone dose was 6.7 mg/d (everolimus 6.9 ±
Height, cm 170 ± 10 172 ± 9
1.7 mg/d; placebo 6.0 ± 1.8 mg/d). Eighty percent of the
Years since transplantation 5.3 ± 4.5 5.1 ± 4.9
patients were hypertensive, and only 2 diabetic pa- Baseline creatinine clearance, 66 ± 20 80 ± 28
tients (4%) were included. Thus, the most frequent mL/min
concomitant medications were antihypertensive drugs Medical disorders, %
(Table I). In general, the patients’ usual medication re- Hypertension 79.5 80
mained unchanged over the study period. Pre- Diabetes 4.5 0
transplant and transplant history as well as past or co- Concomitant medication, %
existing medical conditions revealed no specific Glucocorticoids 93.2 100
clustering in certain treatment groups and reflected the Calcium channel blockers 47.7 30
intended target population. Treatment groups were (dihydropyridines)
matched for demographic data, prior nonimmunosup- β-Blockers 45.5 50
pressant medications, kidney transplant history, and Diuretics 22.7 20
background medical characteristics, as reported Angiotensin-converting 18.2 10
elsewhere in greater detail.21 enzyme inhibitors
Eight (80%) of the 10 patients in the placebo group α-Blockers 13.6 10
and 29 (80.5%) of the 36 patients in the everolimus Statins 27.3 10
dose group up to 5 mg/d received study drug for the full
duration of treatment, while only 1 patient (12.5%) of
the 8 enrolled in the 10-mg dose group completed the treatment discontinuations nor serious AEs were
study. If samples for pharmacokinetic analysis were reported for the placebo group. One (10%) subject in
obtained, these were included in the results, even if the the placebo group withdrew because of a protocol
patients discontinued the study. A total of 9 serious ad- violation.
verse events (AEs) were reported in 8 everolimus-
treated patients, consisting of 2 viral infections and 1 Cyclosporine Dosing
case each of gastroenteritis, pneumonia, intestinal ob-
struction, myocardial infarction, stomatitis, increased Initial CsA doses were comparable in each of the
creatinine, and thrombopenia, leading to 4 treatment everolimus dosing cohorts: in the placebo group, the
discontinuations. Treatment discontinuations in the mean dose was 107 ± 36 mg BID and among the patients
everolimus group were due to AEs in 11 subjects, and receiving everolimus, the mean cohort doses ranged
in a further 3 subjects, discontinuation was due to the from 100 ± 50 (0.75 mg QD), 133 ± 44 (2.5 mg QD), 133 ±
decision of the Safety Monitoring Board to discontinue 17 (5 mg QD), 135 ± 45 (2.5 mg BID), to 137 ± 40 (0.75
treatment at the 10-mg/d dose level. AEs leading to dis- mg QD capsule mg BID). At the end of the study, the
continuation were thrombocytopenia (n = 4), infection mean CsA dose differed not significantly from starting
(n = 2), stomatitis and thrombocytopenia, hypertension doses (placebo: 107 ± 36; 0.75-mg capsule: 133 ± 33;
and headache, increased serum creatinine, intestinal 0.75 mg QD: 95 ± 48; 2.5 mg QD: 129 ± 51; 5 mg QD: 133
obstruction (each n = 1), and a patient with gastritis, ± 17; 2.5 mg BID: 129 ± 47; 10 mg QD (n = 1): unchanged
atrial fibrillation, edema, and elevated lipids. Most AEs 150 mg BID).
leading to discontinuation (especially thrombo- Of the 52 patients providing CsA concentration-
cytopenia) occurred in the highest dosage groups.21 time data (profiles and/or troughs), 38 (73%) main-
The highest dosage group was therefore prematurely tained the same dosage schedule throughout the trial.
terminated by the Safety Monitoring Board. Neither The remaining 14 patients had dose reductions of 20

784 • J Clin Pharmacol 2005;45:781-791


EVEROLIMUS AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

250
ng/ml

200
CsA trough level

150

100

0.75mg capsule 5.0mg QD placebo


0.75mg tablet 10mg QD
2.5mg QD 2.5mg BID
50
0 7 14 21 28

day

Figure 1. Synoptic view of mean cyclosporine trough concentrations in patients receiving different doses of everolimus or placebo.

mg (n = 1), 25 mg (n = 4), 50 mg (n = 8), and 75 mg (n = prior to day 28 or who had a CsA dose reduction, con-
1). These dose reductions were started roughly equally centrations up to the day of these events were used.
in each study week: 3 in week 1, 5 in week 2, 2 in week Linear regression was performed on through trough
3, and 4 in week 4. While no patient randomized to re- levels over time to test for a significantly positive slope.
ceive placebo had a CsA dose alteration, the 14 dose re- This was taken as an indication for a rise in CsA expo-
ductions were equally spread among the rising sure over the course of the trial in the absence of CsA
everolimus dose levels. dose changes. Interestingly, there was a general rise in
troughs over time; indeed, the majority of patients re-
Serial Trough Concentrations ceiving placebo (6 of 9, or 67%) had a significantly pos-
itive regression slope. The proportion of patients in
Serial trough concentrations were available from 52 everolimus dosing cohorts for which regression slopes
patients. Not surprisingly, CsA trough levels exhibited were significantly positive was similar or lower com-
inter- and intrapatient variability. For patients without pared with the placebo group: 0/6 (0.75-mg QD cap-
a dose change, the intrapatient coefficient of variation sule), 3/6 (0.75 mg QD), 2/6 (2.5 mg QD), 5/6 (5 mg QD),
was calculated. Placebo-treated patients had a coeffi- 1/2 (10 mg QD), 7/11 (2.5 mg BID), and 2/6 (5 mg BID).
cient of variation of CsA trough levels of 16.8% ± 6.1%, Across all dose levels including placebo, ANOVA did
similar to everolimus-treated patients (16.0% ± 7.5%). not detect any differences in the magnitude of the
Synoptic views of mean CsA trough levels are shown in trough increases among everolimus dose levels or
Figure 1. For patients who withdrew from the study placebo (P = .11).

DRUG INTERACTIONS 785


BUDDE ET AL

Table II Cyclosporine Pharmacokinetics


RAD-Placebo (n = 9) 0.75 mg QD (n = 5) 2.5 mg QD (n = 6) 5 mg QD (n = 5)
Parameter Day 0 Day 21 Day 0 Day 21 Day 0 Day 21 Day 0 Day 21

tmax, h 1.0 1.5 1.0 1.0 1.5 1.5 1.0 1.5


(1.0-2.0) (1.0-1.5) (1.0-1.5) (1.0-1.5) (1.0-1.5) (1.5-2.5) (1.0-1.5) (1.0-2.0)
Cmin, ng/mL 119 ± 62 133 ± 46 149 ± 56 163 ± 93 112 ± 26 117 ± 39 89 ± 26 140 ± 38
Cmin/dosea 1.12 ± 0.47 1.29 ± 0.37 1.56 ± 0.30 1.70 ± 0.62 0.95 ± 0.54 1.05 ± 0.54 0.67 ± 0.15 1.06 ± 0.28
Cmax, ng/mL 978 ± 284 1061 ± 267 1162 ± 560 1278 ± 725 1106 ± 444 843 ± 451 1176 ± 187 1621 ± 366
Cmax/dosea 9.9 ± 3.4 10.6 ± 3.4 11.8 ± 1.5 13.0 ± 2.1 8.5 ± 3.2 7.2 ± 3.8 8.9 ± 0.9 12.3 ± 2.9
C2h, ng/mL 681 ± 146 834 ± 339 829 ± 397 945 ± 599 794 ± 339 730 ± 328 717 ± 194 1080 ± 275
AUC12, 3470 ± 974 3942 ± 1064 4078 ± 1554 4774 ± 2448 3917 ± 1148 3664 ± 1102 3684 ± 775 5215 ± 824
ng•h/mL
AUC12/dosea 34.0 ± 8.8 38.9 ± 10.8 42.5 ± 5.2 49.9 ± 12.6 31.0 ± 11.5 32.1 ± 14.2 27.7 ± 4.0 39.5 ± 6.4
Peak-trough 309 ± 87 287 ± 43 293 ± 62 281 ± 62 290 ± 81 226 ± 74 362 ± 70 344 ± 88
fluctuation, %
Values are mean ± SD except for tmax, which is median (range).
a. Dose-normalized parameters are per milligram cyclosporine.

Cyclosporine Profiles average in patients receiving placebo and between


10% and 19% for the lower dose everolimus regimens
Paired CsA profiles from day 0 (CsA alone) and day 21 (0.75-2.5 mg QD). Of the 2 regimens delivering the
(CsA + everolimus) were evaluable from 9 patients re- same total daily dose of 5 mg, there was a 58% increase
ceiving placebo and 32 patients receiving everolimus. for 5 mg QD but a 28% increase for 2.5 mg BID; this lack
None of the patients randomized to receive 5 mg of an apparent dose relationship was due to individual
everolimus BID remained in the study to day 21; hence, variability. At the highest dose level of 10 mg QD (n =
paired CsA profiles were not available at this dose 1), no change in CsA trough was noted. Similar pat-
level. Pharmacokinetic parameters are summarized in terns were noted for dose-normalized Cmax and AUC. In
Table II for 3 representative everolimus dose levels us- the placebo arm, these parameters rose on average by
ing the tablet formulation. The administration of the 11% and 15%, respectively, with similar increases at
capsule formulation yielded similar results (not all other everolimus dose levels with the apparent ex-
shown). The global ANOVA did not detect any group- ception of 5 mg QD (38% and 43%). Nonetheless,
by-day interactions; hence, the main effects compari- ANOVA did not detect any differences in the magni-
son was used to assess differences in parameters be- tude of the Cmax or AUC increases (P = .17 and .13, re-
tween days. No differences in tmax or PTF were noted spectively) across all dose levels including placebo.
when CsA was coadministered with everolimus com- The plot of dose-normalized AUC ratios is reproduced
pared with administration alone (P = .60 and .17, re- in Figure 3. In agreement with the global ANOVA, CsA
spectively). Dose-normalized Cmin and AUC were sig- peak-trough fluctuation was not altered upon
nificantly increased (P < .01 for both) across all dose coadministration with everolimus (P = .17).
groups (including placebo), while dose-normalized An additional analysis of the impact of everolimus
Cmax showed a similar trend (P = .06). Representative exposure on CsA disposition was based on linear re-
plots to examine the mean dose-normalized parame- gression of everolimus AUC versus the
ters across 3 dosing groups and the placebo group on coadministration/baseline CsA AUC ratios (day 21/0).
the 2 profiling occasions are shown in Figure 2. As shown in Figure 4, the slope of the relationship was
not significantly different from that of a horizontal line
Cyclosporine Parameter Ratios (P = .24).

An alternative approach to examine CsA changes DISCUSSION


across everolimus dose groups was based on the CsA
parameter ratios (ratio coadministration/baseline). This article reports the results of the first multiple oral
These data are summarized in Table III. Over the course dose clinical study of everolimus tablets. Everolimus
of the study, CsA trough concentrations rose by 24% on or placebo was administered as add-on therapy to

786 • J Clin Pharmacol 2005;45:781-791


EVEROLIMUS AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

2000

RAD-placebo
CsA Conc (ng/mL)

1500

1000

500

0
0 2 4 6 8 10 12
Time (h)

2000 2000
CsA Conc (ng/mL)

CsA Conc (ng/mL)


0.75mg qd 2.5mg qd
1500 1500

1000 1000

500 500

0 0
0 2 4 6 8 10 12 0 2 4 6 8 10 12
Time (h) Time (h)

2000
CsA Conc (ng/mL)

5mg qd
1500

1000

500

0
0 2 4 6 8 10 12
Time (h)

Figure 2. Mean (SD) cyclosporine plots comparing administration alone ( ) and with various everolimus regimens ( ).

Table III Cyclosporine Parameter Ratios


(Ratio Coadministration/Baseline)
RAD Placebo 0.75 mg QD Tablet 2.5 mg QD Tablet 5 mg QD Tablet
Parameter (n = 9) (n = 5) (n = 6) (n = 5)

Cmin/dose, ng/mL/mg 1.24 ± 0.30 1.10 ± 0.34 1.12 ± 0.32 1.58 ± 0.18
Cmax/dose, ng/mL/mg 1.11 ± 0.24 1.12 ± 0.22 0.93 ± 0.47 1.38 ± 0.26
AUC/dose, ng•h/mL/mg 1.15 ± 0.16 1.18 ± 0.28 1.07 ± 0.38 1.43 ± 0.16
Peak-trough fluctuation, % 0.98 ± 0.24 0.96 ± 0.10 0.80 ± 0.21 0.95 ± 0.13
A ratio of 1.00 implies no change in the respective cyclosporine parameter with everolimus or placebo coadministration. Values are mean ± SD.

maintenance renal transplant patients on a double teristics of subjects enrolled in the study matched those
immunosuppressive regimen consisting of CsA of the intended target population; however, it is impor-
microemulsion formulation and steroids. The charac- tant to note that no black patients were included in this

DRUG INTERACTIONS 787


BUDDE ET AL

2.00 2.00

CsA AUC/Dose Ratio (Day 21/Day 0)


1.75 1.75
AUC/Dose Ratio (Day 21/Day 0)

1.50 1.50

1.25
1.25
1.00
1.00
0.75
0.75
0.50
0.50
0.25
0.25 0.00
0.00 0 200 400 600 800 1000 1200
Placebo 0.75qd* 0.75qd 2.5qd 5qd 2.5bid 10qd RAD AUC(0-24) (ng.h/mL)
RAD regimen (mg)

Figure 4. Linear regression to explore the relationship of


Figure 3. Cyclosporine AUC ratios compared across everolimus everolimus exposure and the change in cyclosporine AUC during QD
dosage levels and placebo cohorts. Shown are individual values ( ) steady-state coadministration. Regression equation: Y = 0.0001 • X +
and group means ( ). A ratio of 1.00 implies no change in 1.13; slope not significantly different from zero (P = .24).
cyclosporine exposure with everolimus or placebo coadministration.
All everolimus regimens are with the tablet formulation except 0.75
mg QD (capsule).
pact on CsA pharmacokinetics under steady-state con-
ditions. However, the fact that all patients who re-
ceived CsA dosage adjustments were on active
trial. It is noteworthy that patients randomized to re- everolimus and none on placebo and that all were CsA
ceive placebo had a significant increase in CsA expo- dosage reductions might suggest that there was indeed
sure over the course of the study on average by 15%. It a minor interaction occurring. Unfortunately, even the
is likely that this reflects improved compliance while analysis of 2 large prospective trials with more than
participating in a controlled clinical trial. A similar 580 patients each could not solve this question as the
15% to 20% increase of CsA pharmacokinetic parame- mean CsA dose was about 9% lower in everolimus-
ters was noted in placebo-treated patients of a parallel treated patients, but this reached statistical signifi-
North American trial.6 As a consequence, changes in cance only in the global trial, not in the American
CsA pharmacokinetics in patients receiving trial.22 In this regard, it is important to note that smaller
everolimus needed to be assessed in comparison to the drug interactions cannot be detected based on CsA-
placebo arm of the study. This was addressed at 3 levels dosing histories alone, mainly because CsA dose
of increasing refinement: (1) CsA dose histories, (2) changes depend on the fluctuating CsA trough levels
comparison of CsA pharmacokinetic parameters across (with a coefficient of variation in this study of 14%-
everolimus dose groups, and (3) regression of 20%), which precludes the detection of smaller
everolimus exposure versus the change in CsA pharmacokinetic interactions.
exposure. We next analyzed CsA trough concentrations in
No CsA dose reductions were performed in the pla- greater detail. To avoid any additional interassay vari-
cebo arm, and 28 of 44 patients (64%) randomized to ability, all CsA concentrations were determined in a
receive everolimus maintained the same CsA dose central laboratory. CsA trough concentration trajecto-
throughout the study. The remaining 14 patients had ries demonstrated a general rise in troughs over the
generally small dose reductions of ≤50 mg, probably re- study duration in the majority of patients receiving pla-
flecting the improved compliance during the course of cebo, suggesting better patient compliance during the
the study. Only 1 patient had a reduction of 75 mg. course of the clinical trial. In cohorts of patients receiv-
Since these patients were evenly distributed across all ing everolimus, a similar or even lower proportion of
everolimus dose levels, increasing everolimus expo- patients had significant increases in trough levels.
sure did not appear to have a strong influence on CsA Again, this observation is in line with the previously
exposure based on CsA-dosing histories. This observa- reported North American study, in which 29% of pa-
tion extends the previous report on 24 patients receiv- tients had increasing CsA trough levels over the course
ing the service capsule formulation of everolimus.6 No of the study.6 Furthermore, even large studies in de
dose changes under everolimus treatment were re- novo renal allograft recipients failed to demonstrate a
ported in this smaller study, suggesting no major im- clear effect of low everolimus doses (1.5-3 mg/d) on

788 • J Clin Pharmacol 2005;45:781-791


EVEROLIMUS AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

CsA trough level.22 Although dose-normalized Cmin was between everolimus-treated patients and patients re-
approximately 10% higher in everolimus-treated pa- ceiving mycophenolate mofetil with regard to CsA
tients, this difference reached statistical significance AUC and Cmax. In 953 paired observations, they ob-
only in the global trial and failed to reach statistical sig- served only a weak correlation (r = 0.38) between
nificance in the American trial. Using a population everolimus exposure and CsA exposure and only a
pharmacokinetic approach, it was found that modest rise in dose-adjusted Cmin. The effect of
everolimus-treated patients had similar CsA trough everolimus on CsA exposure was clearly much less
levels compared to mycophenolate mofetil–treated pa- pronounced than the intra- and interindividual
tients, and there was only a weak correlation between variability of CsA.22
everolimus exposure and CsA trough levels, suggesting Within the limitations of the study, our comprehen-
congregation of high absorber status and/or poor clear- sive and thorough evaluations over a broad range of
ance for both drugs.25 In addition, 3 de novo trials could everolimus exposures suggest that steady-state CsA
not demonstrate a significant impact of everolimus pharmacokinetics was not influenced by steady-state
dosing (0.5-4 mg) on CsA levels24,28,29; however, these coadministration of everolimus to a significant degree
trials had no control group of patients not receiving that would be expected to have a clinical effect, based
everolimus. The detailed analysis in our European co- on what is known about CsA pharmacokinetics. This
hort of stable maintenance patients provided further conclusion is in agreement with preclinical pharma-
evidence that everolimus, assessed over a wide dosing cokinetic studies in rats and monkeys9,30 and with pre-
range, has no major impact on CsA trough levels, vious clinical studies6,22,24,26 in which steady-state
which would be expected to have a clinical meaning- coadministration of 0.5 to 7.5 mg everolimus with CsA
ful effect. Important to note is the rather large coeffi- did not affect the pharmacokinetics of the latter. In
cient of variation (14%-20%) in consecutive CsA addition, single-dose administration of everolimus to
trough level determinations, even in this closely moni- renal transplant patients5 or healthy volunteers23 re-
tored, selected, and compliant study population. This sulted in no interaction with CsA pharmacokin-
probably reflects the highly variable CsA pharma- etics. Kirchner and colleagues31,32 performed a detailed
cokinetics and makes the detection of smaller changes analysis of CsA metabolites in a small number of pa-
of CsA exposure based on trough level monitoring tients. They could not detect an impact of everolimus
difficult.27 on CsA metabolites after single-dose and multiple-dose
Because CsA trough levels do not adequately reflect administration,31,32 which excluded a significant influ-
CsA drug exposure,27 individual CsA pharmacokinetic ence of everolimus on the time-concentration relation-
parameters from the full profiles were compared be- ship and the metabolism of CsA under steady-state
tween baseline and coadministration with everolimus conditions. Finally, the single and multiple oral ad-
across different everolimus dose groups. ANOVA de- ministration of sirolimus did not reveal any pharmaco-
tected higher CsA exposure during coadministration kinetic interaction on CsA blood concentrations13-15,20
with everolimus; however, this was also detected in the in renal transplant recipients. Because both drugs are
placebo arm. To more specifically address potential structurally closely related and share most metabolic
differences among everolimus dose levels, the CsA pathways, this observation provides further evidence
AUC ratios (coadministration/baseline) were com- that there is no significant drug interaction of evero-
pared across groups. The mean ratio was 1.15 for pla- limus on CsA pharmacokinetics, but this cannot rule
cebo and did not significantly differ in the everolimus out pharmacodynamic interactions of both drugs.
dose groups in which the ratios ranged from 1.08 to Similar to tacrolimus and CsA, sirolimus and
1.43. Finally, the above approaches categorize each pa- everolimus are metabolized to some extent in the small
tient’s everolimus exposure into the nominal intestine and extensively in the liver via cytochrome
everolimus regimen they received, which may down- P450 3A4 enzymes and are countertransported in
play interindividual variation in the systemic the gut lumen by the multidrug efflux pump, p-
everolimus exposure. By contrast, linear regression glycoprotein; these processes account for low
analysis incorporates everolimus exposure (AUC on bioavailability and high pharmacokinetic variabil-
day 21) as a continuous variable. When regressed ity.2,4,13 As a consequence of this wide interpatient vari-
against the change in CsA exposure, the relationship ability, it is not possible to rule out that some patients
was not different from a horizontal line over the full have an interaction to everolimus. This has been de-
range of everolimus AUCs achieved in this study. Us- scribed for other drugs interacting with CsA, in which
ing abbreviated pharmacokinetic profiles from 256 de some patients are more sensitive to the interaction than
novo patients, Kovarik et al22 could not find differences are others (eg, serial metabolic inhibition studies with

DRUG INTERACTIONS 789


BUDDE ET AL

diltiazem33). Larger, specifically designed studies are edge on this interesting new immunosuppressive com-
needed to further explore this issue. pound, suggesting that everolimus could be used con-
As immunosuppressive doses and concentrations of comitantly with the microemulsion formulation of
CsA in humans are usually 100-fold higher than those CsA using either QD or the standard BID dosage. How-
of everolimus, the competition between both drugs for ever, the present study did not address the question of
elimination pathways would favor an influence of CsA pharmacodynamic interactions, which might be re-
on everolimus compared with the reverse situation. sponsible for the increased incidence of calcineurin-
This indeed has been shown in animal models, in related toxicity, seen in clinical studies, when
which CsA increased the systemic exposure to everolimus or sirolimus were coadministered with
everolimus. Similarly, CsA seems to exert rather strong CsA.1,3,10,16,28,29 Because of limitations of the present
effects on everolimus exposure in humans, as demon- study, which do not allow us to detect smaller changes
strated in a single-dose study.23 The effect of CsA seems or changes in subpopulations (eg, black patients; pa-
to be dependent on the CsA formulation, as Neoral ex- tients who differ in their expression of CYP 3A or p-
hibited much stronger effects on everolimus exposure glycoprotein), this finding will, however, need to be
(168% increase) compared with the previous CsA for- confirmed over longer periods of coadministration in
mulation (Sandimmun; 74% increase). Because broader patient populations.
everolimus was added to patients already receiving
This study was supported by Novartis Pharma AG, Basel, Swit-
CsA, there was no opportunity to examine the possible zerland. The authors are grateful for the help of J. Kovarik in prepar-
effect of CsA on everolimus pharmacokinetics in this ing the article.
study. Similar to everolimus, in preclinical models,
sirolimus increased the bioavailability of CsA by 2- to
3-fold,19 with an approximately 2-fold increase in APPENDIX
the CsA concentrations in rat tissues.9,16,18 From these List of Institutional Review Boards
studies, it was suggested that the pharmacokinetic in-
teraction between both drugs might contribute to the Ethik-Komitee der Medizinischen Hochschule
observed in vivo synergism. However, this pharmaco- Hannover; 30623 Hannover, Germany
kinetic interaction with increased drug concentrations Ethikkommission, Universitätsklinikum Charité;
in renal tissue might be responsible for the aggravation Schumannstrasse 20/21, 10177 Berlin, Germany
of CsA-induced renal dysfunction, as has been sug-
Der Dekan der Medizinischen Fakultät (Ethik-
gested by Podder et al.16 Whether these data derived
kommission); Universitätsstrasse 40; 91054 Erlangen,
from experimental models transfer to humans is yet Germany
unclear.
Ethikkommision des Landes Bremen, ZKH St.-
Despite their structural similarity and identical Jürgenstr.; 28205 Bremen, Germany
mode of action, sirolimus and everolimus clearly differ
Comite consultatif de protection des persones; CHU
in their pharmacological metabolism.17 Although CYP
de Nantes, Immeuble Deurbroucq, 44093 Nantes,
3A4 is mainly responsible for the metabolism of both
France
sirolimus and everolimus, the total intrinsic clearance
Regional Komite for Medicinsk Forskningsetikk
of the CYP-dependent formation of everolimus metab-
Forskningsparken, Gaustadalleen 21 0371 Oslo,
olites is 3-fold lower than for sirolimus, and some of the
Norway
main metabolic rate constants are drastically (up to 15-
fold) reduced.17 In contrast to sirolimus, everolimus de-
creased CsA concentration in rat brain mitochondria.9
Furthermore, sirolimus enhances CsA toxicity on rat REFERENCES
brain; everolimus, however, had no effect on CsA toxic-
ity and even improved some deleterious effects of CsA 1. Nashan B. Review of the proliferation inhibitor everolimus. Expert
on high-energy phosphate metabolism.9 Opin Investig Drugs. 2002;11:1845-1857.
2. Kovarik JM. Everolimus: a proliferation signal inhibitor targeting
Taken together, our data provide further evidence
primary causes of allograft dysfunction. Drugs Today. 2004;40:101-
that everolimus coadministration exhibits no relevant 109.
effect on pharmacokinetics and CsA exposure in Cau- 3. Nashan B. Early clinical experience with the novel rapamycin de-
casians that would be expected to have a clinical effect, rivative. Ther Drug Monit. 2002;24:53-58.
based on what we know today about CsA pharmaco- 4. Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharma-
kinetics. This observation clearly extends our knowl- cokinetics of everolimus. Clin Pharmacokinet. 2004;43:83-95.

790 • J Clin Pharmacol 2005;45:781-791


EVEROLIMUS AND CYCLOSPORINE IN RENAL TRANSPLANT PATIENTS

5. Neumayer H-H, Paradis K, Korn A, et al. Entry-into-human study 20. Kaplan B, Meier-Kriesche HU, Napoli KL, Kahan BD. The effect of
with the novel immunosuppressant SDZ RAD in stable renal trans- relative timing of sirolimus and cyclosporine microemulsion formu-
plant recipients. Br J Clin Pharmacol. 1999;48:694-703. lation coadministration on the pharmacokinetics of each agent. Clin
6. Kahan BD, Wong R, Carter C, et al. A phase I study of a 4-week Pharmacol Ther. 1998;63:48-53.
course of SDZ-RAD (RAD) in quiescent cyclosporine-prednisone- 21. Budde K, Neumayer HH, Lehne G, et al. Tolerability and steady
treated renal transplant recipients. Transplantation. 1999;67:1100- state pharmacokinetics of everolimus in maintenance renal trans-
1106. plant patients. Nephrol Dial Transplant. 2004;19:2606-2614.
7. MacDonald A, Scarola J, Burke JT, Zimmermann JJ. Clinical phar- 22. Kovarik JM, Kaplan B, Silva HT, et al. Pharmacokinetics of an
macology and therapeutic drug monitoring of sirolimus. Clin Ther. everolimus-cyclosporine immunosuppressive regimen over the first
2000;22:B101-B121. 6 months after kidney transplantation. Am J Transplant. 2003;3:606-
8. Gallant-Haidner HL, Trepanier DJ, Freitag DG, Yatskoff RW. 613.
Pharmacokinetics and metabolism of sirolimus. Ther Drug Monit. 23. Kovarik JM, Kalbag J, Figueiredo J, Rouilly M, Frazier OL, Rordorf
2000;22:31-35. C. Differential influence of two cyclosporine formulations on
9. Serkova N, Jacobsen W, Niemann CU, et al. Sirolimus, but not the everolimus pharmacokinetics: a clinically relevant pharmacokinetic
structurally related RAD (everolimus), enhances the negative effects interaction. J Clin Pharmacol. 2002;42:95-99.
of cyclosporine on mitochondrial metabolism in the rat brain. Br J 24. Kovarik JM, Kahan BD, Kaplan B, et al. Longitudinal assessment
Pharmacol. 2001;133:875-885. of everolimus in de novo renal transplant recipients over the first
10. Jacobsen W, Serkova N, Hausen B, et al. Comparison of the in vitro year: pharmacokinetics, exposure-response relationships and influ-
metabolism of the macrolide immunosuppressants sirolimus and ence on cyclosporine. Clin Pharmacol Ther. 2001;69:48-56.
RAD. Transplant Proc. 2001;33:514-515. 25. Kovarik JM, Kaplan B, Tedesco Silva H, et al. Exposure-response
11. Kahan BD, Chang JY, Sehgal SN. Preclinical evaluation of a new relationships for everolimus in de novo kidney transplantation: de-
potent immunosuppressive agent, rapamycin. Transplantation. fining a therapeutic range. Transplantation. 2002;73:920-925.
1991;52:185-191. 26. Kovarik JM, Hsu CH, McMahon L, Berthier S, Rordorf C. Popula-
12. Nashan B. The role of Certican (everolimus, Rad) in the many tion pharmacokinetics of everolimus in de novo renal transplant pa-
pathways of chronic rejection. Transplant Proc. 2001;33:3215-3220. tients: impact of ethnicity and comedications. Clin Pharmacol Ther.
13. Ferron GM, Mishina EV, Zimmerman JJ, Jusko WJ. Population 2001;70:247-254.
pharmacokinetics of sirolimus in kidney transplant patients. Clin 27. Jorga A, Holt DW, Johnston A. Therapeutic drug monitoring of
Pharmacol Ther. 1997;61:416-428. CsA. Transplant Proc. 2004;36:396S-403S.
14. Zimmerman JJ, Kahan BD. Pharmacokinetics of sirolimus in sta- 28. Vitko S, Tedesco H, Eris J, et al. Everolimus with optimized
ble renal transplant patients after multiple oral dose administration. J cyclosporine dosing in renal transplant recipients: 6-month safety
Clin Pharmacol. 1997;37:405-415. and efficacy results of two randomized studies. Am J Transplant.
15. Brattstrom C, Sawe J, Tyden G, et al. Kinetics and dynamics of sin- 2004;4:626-635.
gle oral doses of sirolimus in sixteen renal transplant recipients. Ther 29. Kahan BD, Kaplan B, Lorber MI, Winkler M, Cambon N, Boger RS.
Drug Monit. 1997;19:397-406. RAD in de novo renal transplantation: comparison of three doses on
16. Podder H, Stepkowski SM, Napoli KL, et al. Pharmacokinetic in- the incidence and severity of acute rejection. Transplantation.
teractions augment toxicities of sirolimus/cyclosporine combina- 2001;71:1400-1406.
tions. J Am Soc Nephrol. 2001;12:1059-1071. 30. Serkova N, Hausen B, Berry GJ, et al. Tissue distribution and clini-
17. Kuhn B, Jacobsen W, Christians U, Benet LZ, Kollman PA. Metab- cal monitoring of the novel macrolide immunosuppressant SDZ-
olism of sirolimus and its derivative everolimus by cyctochrome RAD and its metabolites in monkey lung transplant recipients: inter-
P450 3A4: insights from docking, molecular dynamics, and quantum action with cyclosporine. J Pharmacol Exp Ther. 2000;294:323-332.
chemical calculation. J Mol Chem. 2001;44:2027-2034. 31. Kirchner GI, Mueller L, Winkler M, et al. Long-term
18. Napoli KL, Wang ME, Stepkowski SM, Kahan BD. Relative tissue pharmacokinetics of the metabolites of everolimus and cyclosporine
distributions of cyclosporine and sirolimus after concomitant peroral in renal transplant recipients. Transplant Proc. 2002;34:2233-2234.
administration to the rat: evidence for pharmacokinetic interactions. 32. Kirchner G, Winkler M, Mueller L, et al. Pharmacokinetics of SDZ
Ther Drug Monit. 1998;20:123-133. RAD and cyclosporine including their metabolites in seven kidney
19. Stepkowski SM, Napoli KL, Wang ME, Qu X, Chou TC, Kahan BD. graft patients after the first dose of SDZ RAD. Br J Clin Pharmacol.
Effects of the pharmacokinetic interaction between orally adminis- 2000;50:449-454.
tered sirolimus and cyclosporine on the synergistic prolongation of 33. Jones TE, Morris RG, Mathew TH. Diltiazem-cyclosporine
heart allograft survival. Transplantation. 1996;62:986-994. pharmacokinetic interaction: dose-response relationship. Br J Clin
Pharmacol. 1997;44:499-504.

DRUG INTERACTIONS 791

You might also like