throbber
Longitudinal assessment of everolimus in
`de novo renal transplant recipients over the
`first post-transplant year: Pharmacokinetics,
`exposure-response relationships, and
`influence on cyclosporine
`
`Objective: Our objective was to characterize the steady-state pharmacokinetics of everolimus and
`cyclosporine (INN, ciclosporin) when coadministered in de novo kidney allograft recipients during the
`first year after transplantation.
`Methods: This study was a multicenter randomized double-blind study of 101 patients who were randomly
`assigned 1:1:1 to receive everolimus tablets at doses of 0.5 mg, 1 mg, or 2 mg twice daily with cyclosporine
`and prednisone. Blood sampling for the pharmacokinetics of everolimus and cyclosporine was performed
`on day 1, on weeks 1, 2, 3, and 4, and on months 2, 3, 6, 9, and 12. Everolimus dose-proportionality
`and stability over time were assessed in the context of linear regression and ANOVA models. Everolimus
`exposure-response relationships between area under the blood concentration-time curve (AUC) and
`changes in platelets, leukocytes, and lipids were explored with the median-effect model. Potential differ-
`ences in cyclosporine dosing and pharmacokinetics at different levels of everolimus exposure were assessed
`in the context of ANOVA.
`Results: Everolimus steady state was reached on or before day 7, with a median 3-fold accumulation of drug
`exposure compared with that after the first postoperative dose. Both steady-state maximum concentration
`and AUC were dose proportional over the full dose range when assessed on day 1, as well as for the full
`duration of the study at steady state. There was evidence for longitudinal stability in AUC of everolimus
`during the course of the study. The interindividual pharmacokinetic variability for AUC was 85.4% and
`intraindividual, interoccasion variability was 40.8%. Age (range, 17-69 years), weight (range, 49-106 kg),
`and sex (65 men and 36 women) were not significant contributors to variability. There was an increasing
`incidence of transient thrombocytopenia (≤100 × 109/L) with increasing everolimus AUC (P = .03).
`Cyclosporine doses, trough concentrations, and AUC exhibited similar temporal patterns during the course
`of the study regardless of the co-administered everolimus dose level (P = .13, .82, and .76, respectively).
`Conclusions: Everolimus exhibited dose-proportional, stable exposure during the first post-transplant year.
`For a 4-fold range of everolimus doses there were no differential effects on cyclosporine dosing or phar-
`macokinetics. (Clin Pharmacol Ther 2001;69:48-56.)
`
`John M. Kovarik, PhD, Barry D. Kahan, MD, Bruce Kaplan, MD, Marc Lorber, MD,
`Michael Winkler, MD, Marisel Rouilly, PhD, Christophe Gerbeau, PhD,
`Natalie Cambon, MD, Robert Boger, MD, and Christiane Rordorf, MD,
`on behalf of the Everolimus Phase 2 Study Group* Basel, Switzerland, Houston, Texas,
`Livingston and East Hanover, NJ, New Haven, Conn, and Hannover, Germany
`
`From Novartis Pharmaceuticals, Basel and East Hanover; University
`of Texas Medical School-Houston; St Barnabas Medical Center,
`Livingston; Yale University School of Medicine, New Haven; and
`Medizinische Hochschule, Hannover.
`*Study group members: S. Cockfield, University of Alberta Hospi-
`tals, Edmonton; D. Hricik, University Hospitals of Cleveland,
`Ohio; K. Rigg and M. Shehata, Nottingham City Hospital, Eng-
`land; W. Land, Klinikum Grosshadern, Munich, Germany; and E.
`Mueller, A. G. Schmidt, S. Wehr, M. M. Wilkie, and R. L. Wong,
`Novartis Pharmaceuticals, East Hanover, NJ.
`
`Supported by research grants from the National Institute of Diabetes,
`Digestive and Kidney Diseases (NIDDK#38016-12) and Novartis
`Pharmaceuticals.
`Received for publication Aug 9, 2000; accepted Nov 14, 2000.
`Reprint requests: John Kovarik, PhD, Novartis Pharma AG, Build-
`ing WSJ 27.4093, 4002 Basel, Switzerland.
`Copyright © 2001 by Mosby, Inc.
`0009-9236/2001/$35.00 + 0 13/1/112969
`doi:10.1067/mcp.2001.112969
`
`48
`
`Ex. 1050-0001
`
`

`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 69, NUMBER 1
`
`Kovarik et al 49
`
`Temporal changes in the pharmacokinetics of
`immunosuppressants used in acute rejection prophylaxis
`after organ transplantation have been reported.1-4 Two
`examples are cyclosporine (INN, ciclosporin) and
`mycophenolate mofetil. For the non-microemulsion for-
`mulation of cyclosporine the relationship between dose
`and area under the blood concentration-time curve
`(AUC) changes in the first 3 to 4 months after a trans-
`plant and stabilizes thereafter.1,2 Although this pattern is
`also evident for the microemulsion formulation, the
`period of dose-AUC stability is achieved earlier (by the
`end of the first month) compared with the non-
`microemulsion formulation.3 This suggests that the oral
`absorption and bioavailability of cyclosporine in the
`early post-transplant period (which is improved with the
`microemulsion formulation) is probably contributing to
`this phenomenon. For mycophenolate mofetil, dose-
`normalized AUC is lower in the early post-transplant
`period and increases slowly over several months by an
`average 50% to reach late post-transplant values. Because
`bioavailability is nearly complete throughout this period,
`poor absorption cannot explain these observations. Spec-
`ulation has centered on changes in protein binding and
`enterohepatic recirculation.4 These examples underscore
`the importance of characterizing the longitudinal phar-
`macokinetics of new immunosuppressants, especially in
`the early post-transplant period so that time-dependent
`changes in the disposition of the immunosuppressants
`may be taken into account in the dosing regimen to yield
`more stable systemic exposure over time.
`Everolimus (development name, RAD) is a macro-
`cyclic lactone immunosuppressant that is primarily
`metabolized and eliminated in the bile. At therapeutic
`concentrations, more than 75% of everolimus is parti-
`tioned into red blood cells and approximately 75% of
`the plasma fraction is protein bound. Everolimus blocks
`growth factor–driven transduction signals in the T-cell
`response to alloantigen5 and thus acts at a later stage
`than the calcineurin inhibitors cyclosporine and
`tacrolimus. The complimentary modes of action of
`everolimus and cyclosporine suggest a synergistic inter-
`action that has indeed been shown in vitro and in vivo
`in preclinical models.5 This provided a rationale for the
`addition of everolimus to cyclosporine-based immuno-
`suppression. Subsequent phase 1 studies showed good
`tolerability and an acceptable side-effect profile when
`everolimus was administered for 1 month to stable
`patients with renal transplants who were receiving a
`cyclosporine-prednisone regimen.6,7 A recently com-
`pleted phase 2 study constituted the first experience
`with everolimus in patients immediately after kidney
`transplantation in the de novo setting.8 In that trial,
`
`extensive pharmacokinetic evaluations were performed
`during a 1-year period after transplantation for longitu-
`dinal characterization of the disposition of everolimus.
`The clinical results will be reported elsewhere; this arti-
`cle focuses on the pharmacokinetic results of the study
`in which dose proportionality, longitudinal stability in
`exposure, and pharmacokinetic variability with poten-
`tial covariates were addressed.
`The inhibitory effects of everolimus are not restricted
`to T cells. Everolumus also inhibits the signals provided
`by some hematopoietic and nonhematopoietic cell
`growth factors.5 In this context, phase 1 experience with
`this compound in patients indicated that treatment with
`everolimus may be associated with decreases in
`platelets and leukocytes and increases in lipids that gen-
`erally occur in the first 2 months after initiation of treat-
`ment.6,7 This has also been the experience with
`sirolimus.9,10 Because we used a broader range of
`everolimus doses in this phase 2 study than are cur-
`rently under investigation in phase 3 efficacy trials, it
`provided the opportunity for us to screen for exposure-
`response relationships between AUC and changes in
`laboratory parameters.
`Both everolimus and cyclosporine are extensively
`biotransformed by means of CYP3A and are substrates
`for P-glycoprotein. Therefore there is a potential for a
`drug-drug interaction when these two agents are co-
`administered. Although everolimus was the primary
`focus of this study,
`the longitudinal influence of
`everolimus on cyclosporine pharmacokinetics was also
`explored as a secondary objective.
`
`METHODS
`Study design and dose regimens. This was a 1-year,
`randomized, double-blind trial performed at 8 study cen-
`ters. The study protocol was approved by local medical
`ethics committees for each center. One hundred three
`de novo kidney allograft recipients gave written
`informed consent to participate in the study and were
`randomly assigned to receive everolimus (Certican,
`Novartis Pharmaceuticals, East Hanover, NJ) at oral
`doses of 0.5 mg (n = 34), 1 mg (n = 34), or 2 mg (n =
`35) twice daily in addition to cyclosporine and pred-
`nisone. The first dose of everolimus was given once it
`had been ascertained that the allograft was functional
`(maximum, 48 hours after transplantation). This was
`defined as study day 1. Everolimus was supplied as a
`tablet formulation in strengths of 0.25 mg and 1 mg with
`matching placebos. Patients were instructed to take the
`prescribed number of tablets every 12 hours simultane-
`ously with cyclosporine. Cyclosporine (Neoral, Novar-
`tis Pharmaceuticals) was initiated orally at 6 to 12
`
`Ex. 1050-0002
`
`

`
`50 Kovarik et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`JANUARY 2001
`
`mg/kg/d in two divided doses. Thereafter, the doses
`were adjusted to maintain whole blood predose concen-
`trations in the range of 150 to 400 ng/mL in the first
`post-transplant month and in the range of 5 to 300
`ng/mL from month 2 to month 12. Prednisone was
`dosed according to a protocol-specified taper.
`Pharmacokinetic assessments. Blood sampling for
`the pharmacokinetics of everolimus and cyclosporine
`was performed at protocol-scheduled visits throughout
`the study duration: day 1, weeks 1, 2, 3, and 4, and
`months 2, 3, 6, 9, and 12. At each visit, either a full
`pharmacokinetic profile that consisted of 10 blood sam-
`ples (predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours)
`or an abbreviated profile (predose, and 1, 2, 5, and 8
`hours or predose, and 1, 2, and 5 hours) was performed
`during the morning dosing interval. At the week 3 and
`month 2 and 12 visits, only a predose trough sample
`was obtained. For the 5-point abbreviated profiles, the
`12-hour concentration was estimated by log-linear
`decrease from the 5-hour and 8-hour measured concen-
`trations; for the 4-point abbreviated profiles, the 12-
`hour concentration was assumed to be the same as the
`predose measured concentration under the assumption
`of steady state. When maximum concentration (Cmax)
`and AUC from the full profiles in this study were re-
`evaluated on the basis of the 4- or 5-sample abbrevi-
`ated approach described previously, they were highly
`correlated (r2 = 0.91 for Cmax and r2 ≥ 0.96 for AUC);
`this indicated that parameters from full and abbreviated
`sampling could be pooled for analysis.
`Bioanalysis of everolimus. Everolimus whole-blood
`concentrations were determined with a validated
`enzyme-linked immunosorbent assay. Performance was
`assessed on the basis of a 5-point quality control con-
`centration range from 2 to 80 ng/mL of everolimus.
`Coefficients of variation ranged from 13.3% to 16.1%
`and bias ranged from –7.0% to –1.8%. The assay quan-
`tification limit was 2 ng/mL.
`Sample preparation consisted of the mixing of 250 µL
`of blood sample with 1 mL of extraction buffer (Tris
`buffer, 0.05 mol/L, pH 9) and 1 mL of extraction solvent
`(diethyl ether with 0.04% Tween 20). The samples were
`shaken for 10 minutes and centrifuged for 10 minutes at
`4000 rpm and 4°C. They were subsequently frozen in
`dry ice, and the organic phase was decanted and evapo-
`rated at 40°C for 20 to 30 minutes. The dry residues were
`reconstituted with 120 µL of a reconstitution solution
`that consisted of 200 ng/mL mouse antirapamycin anti-
`body in phosphate-buffered saline solution (PBS) buffer
`(1:10 dilution) with 1% bovine serum albumin. For the
`determination of nonspecific binding, the dry residue
`was dissolved in dilution buffer without antibody. The
`
`samples were shaken for 10 minutes and then incubated
`for at least 15 minutes at room temperature.
`Plastic 96-well assay plates were coated overnight
`with 120 µL per well of 10 µg/mL goat anti-mouse anti-
`body in PBS buffer (Goat Anti-Mouse IgG Fc,
`Immunopure, Rockford, Ill) at a temperature of approx-
`imately 4°C. The plates were then washed with wash-
`ing buffer (PBS buffer with 0.05% Tween 20) and
`blocked with 300 µL per well of blocking solution by
`incubation for 5 minutes at room temperature. The
`plates were washed and 100 µL per well of the
`processed blood samples was added. After 1 hour of
`incubation at room temperature with gentle shaking, 20
`µL of tracer solution (19 ng/mL of rapamycin
`biotinyled in PBS buffer with 1% bovine serum albu-
`min and 0.04% Tween 20) were added to each well. The
`plates were incubated overnight at approximately 4°C
`and then washed with PBS buffer with 0.05% Tween 20
`and incubated with 120 µL per well of streptavidin per-
`oxidase solution (diluted 1:10,000 in assay buffer) for
`15 minutes in the dark at room temperature with gentle
`shaking. Plates were washed with assay buffer and 120
`µL per well of o-phenylenediamine dihydrochloride
`solution (10 mg in 50 mL of 0.05 mol/L citrate/phos-
`phate buffer and 20 µL of hydrogen peroxide 30%) was
`added, and then the plates were shaken gently for 5 to
`6 minutes at room temperature. The reaction was
`stopped with 50 µL per well of 2 N sulfuric acid, and
`the optical density was measured at 492/620 nm.
`Bioanalysis of cyclosporine. Cyclosporine whole
`blood concentrations were determined with the use of
`the Incstar CYCLO-Trac radioimmunoassay kit (Still-
`water, Minn) according to the manufacturer’s directions.
`Performance was assessed on the basis of a 5-point qual-
`ity control concentration range from 30 to 1250 ng/mL.
`Coefficients of variation ranged from 5.1% to 10.6%
`and bias ranged from –12.0% to 0.4%. The assay quan-
`tification limit was 30 ng/mL.
`Pharmacokinetic and statistical evaluation. Con-
`ventional noncompartmental steady-state pharmaco-
`kinetic parameters were derived for both everolimus
`and cyclosporine, including the predose trough concen-
`tration (Cmin); the Cmax; the time to reach maximum
`concentration (tmax); the AUC during a dosing interval,
`by trapezoidal summation; the average concentration
`(Cavg), calculated as the quotient of AUC/12; and the
`peak-trough fluctuation, calculated as (Cmax – Cmin)/
`Cavg. The accumulation ratio was derived from the ratio
`of AUCs from day 7 and day 1.
`Dose proportionality for the Cmax and AUC of
`everolimus was assessed on day 1 (first dose) and day
`7 (steady state) with linear regression analysis of the
`
`Ex. 1050-0003
`
`

`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 69, NUMBER 1
`
`Kovarik et al 51
`
`Table I. Everolimus pharmacokinetic parameters*
`0.5 mg bid
`
`First dose
`tmax (h)
`Cmax (ng/mL)
`AUC (ng · h/mL)
`Steady state†
`Cmin (ng/mL)
`tmax (h)
`Cmax (ng/mL)
`Cmax/dose (ng/mL/mg)
`AUC (ng · h/mL)
`AUC/dose (ng · h/mL/mg)
`Cavg (ng/mL)
`PTF (%)
`
`3 (1-12)
`2.0 ± 2.1
`8 ± 12
`
`1.5 ± 1.8
`2 (1-5)
`5.0 ± 2.9
`10.0 ± 5.8
`34 ± 23
`68 ± 46
`2.8 ± 1.9
`84 ± 38
`
`bid, Twice a day; PTF, peak-trough fluctuation.
`*Values are median (range) for tmax and mean ± standard deviation for all other parameters.
`†Steady-state data are from week 1.
`
`1 mg bid
`
`3 (2-9)
`5.6 ± 3.7
`28 ± 23
`
`4.7 ± 2.6
`2 (1-5)
`11.6 ± 4.4
`11.6 ± 4.4
`81 ± 34
`81 ± 34
`6.7 ± 2.8
`97 ± 38
`
`2 mg bid
`
`3 (2-12)
`9.8 ± 7.0
`56 ± 37
`
`9.5 ± 5.2
`2 (1-8)
`21.9 ± 10.5
`11.0 ± 5.3
`164 ± 78
`82 ± 39
`13.6 ± 6.5
`90 ± 47
`
`parameter versus dose. Steady-state, dose-normalized
`parameters from day 7 to the end of the study were also
`evaluated in an ANOVA model with dose, subject-
`within-dose, visit, and dose-by-visit interaction terms.
`In the absence of a dose-by-visit interaction, lack of a
`dose-effect was taken as support for dose proportional-
`ity during the full study course; lack of a visit effect
`was evidence for longitudinal stability in exposure.
`Steady-state AUC per dose from the replicate pharma-
`cokinetic profiles was assessed with a two-way ANOVA
`with subject and visit as sources of variation. The mean
`square from the subject term was taken as a measure of
`interindividual variance, and the mean square from the
`error term was taken as a measure of intraindividual,
`interoccasion variance. The corresponding coefficients
`of variation were calculated as the standard deviation
`(square root of the mean square) divided by the grand
`mean of the parameter. The contribution of conven-
`tional demographic covariates to the pharmacokinetic
`variability was explored with graphical and regression
`techniques for continuous variables (age and weight)
`and with unpaired two-sided t tests for categorical vari-
`ables (sex and ethnicity).
`Dose-normalized cyclosporine pharmacokinetic param-
`eters were evaluated with a global ANOVA model identi-
`cal to that for everolimus in which the dose term referred
`to the everolimus dose level. The absence of a dose-by-
`visit interaction and of a dose-effect was taken as support
`that the influence of everolimus on cyclosporine was not
`different among the everolimus dose levels.
`Exposure-response relationships. Relationships
`between everolimus steady-state AUC and the incidence
`of thrombocytopenia (≤100 × 109/L), leukopenia (≤4.0
`
`× 109/L), hypertriglyceridemia (>2.9 mmol/L), and
`hypercholesterolemia (>6.5 mmol/L) were explored
`with the median-effect principle.11 This model relates
`the fraction of the population affected (fa) and unaf-
`fected (fu = 1 – fa) with respect to a given laboratory
`parameter change on the one hand and to the drug expo-
`sure (AUC) and the exposure at which half the popula-
`tion is affected (median effect [AUCm]) on the other
`hand: fa/fu = (AUC/AUCm)m. The relationship is lin-
`earized on the logarithmic scale as follows: log(fa/fu)
`= m log(AUC) – m log(AUCm). In this relationship, m
`is a Hill-type coefficient that describes the sigmoidic-
`ity in the exposure-response relationship. The average
`AUC for each patient was determined from the week 1,
`2, and 4 profiles and the overall distribution divided
`into quartiles. The fraction of patients whose labora-
`tory parameter did (fa) and did not (fu) exceed the
`defined cutoff value listed previously in the first two
`post-transplant months was determined in each of the
`quartiles. The resulting log(fa/fu) versus log(AUC) rela-
`tionship was assessed by linear regression analysis.
`Goodness of fit of the model to the data was indicated
`by a regression coefficient (r value) of more than 0.8;
`a regression P value of less than .05 indicated a signif-
`icant relationship between exposure and the occurrence
`of a laboratory parameter change.
`
`RESULTS
`Demographics and patient disposition. Of the 103
`patients enrolled in the study, 101 provided at least one
`pharmacokinetic profile and were included in this analy-
`sis. The two unevaluable patients withdrew from the trial
`in the first week and did not undergo pharmacokinetic
`
`Ex. 1050-0004
`
`

`
`52 Kovarik et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`JANUARY 2001
`
`A
`
`B
`
`Fig 1. Everolimus mean AUC trajectories during 9 months after transplants (A) and everolimus
`trough concentrations (Cmin) to 12 months after transplants (B) at dose levels of 0.5 mg (solid
`squares), 1 mg (solid circles), and 2 mg twice a day (solid triangles). Bars designate 95% confi-
`dence intervals.
`
`blood sampling. There were 65 men and 36 women who
`were 44.4 ± 11.6 years old and who weighed 76.7 ± 15.4
`kg. The majority of the patients were white (n = 82);
`there were 9 black patients, 1 Asian patient, and 9
`patients of other ethnicities.
`Everolimus pharmacokinetic profiles were evaluable
`at each visit if the patient remained on the dose to which
`he or she was randomly assigned and had not missed
`doses within 5 days before the visit. Over the 1-year
`duration of the study, 18 patients had dose reductions for
`safety reasons, 31 patients had transient dose interrup-
`tions, and 16 patients were removed from the study (8
`unsatisfactory therapeutic response, 3 died, 3 withdrew
`
`consent, and 2 violated the study protocol). Conse-
`quently, the number of evaluable profiles at each of the
`steady-state visits were: 94 (week 1), 88 (week 2), 83
`(week 4), 71 (month 3), 58 (month 6), and 55 (month 9).
`First-dose everolimus pharmacokinetics. A total of
`95 profiles were obtained after the first dose of
`everolimus on day 1: 33, 32, and 30 profiles, respec-
`tively, were obtained from patients who were receiving
`0.5, 1, and 2 mg twice daily. First-dose pharmacokinetic
`parameters are summarized in Table I. In postoperative
`conditions, there was generally a 1-hour lag time until
`everolimus was quantifiable in blood, and the peak con-
`centration occurred at 3 hours after the dose. Both Cmax
`
`Ex. 1050-0005
`
`

`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 69, NUMBER 1
`
`Kovarik et al 53
`
`Fig 2. Median-effect plots of everolimus steady-state AUC and incidence of hypercholesterolemia
`(solid diamonds), hypertriglyceridemia (solid triangles), and thrombocytopenia (solid circles) in
`patients with renal transplants during first 2 post-transplant months. For these laboratory parame-
`ters, the slopes were 1.27, 0.66, and 0.70, and the correlation coefficients were 0.94, 0.83, and 0.97,
`respectively. Incidence is expressed as the fraction of the population affected and unaffected (fa/fu).
`
`and AUC rose proportionally with increasing dose (r =
`0.57, P < .001 for Cmax and r = 0.61, P < .001 for AUC).
`Steady-state everolimus pharmacokinetics. Steady
`state was reached on or before day 7 on the basis of the
`ANOVA. Between the first dose and steady state there
`was a median accumulation ratio of 2.9. A total of 449
`steady-state profiles were evaluable between week 1
`and month 9 (100 with full 12-hour sampling and 349
`with abbreviated sampling). Steady-state parameters
`from week 1 are summarized in Table I. Cmin increased
`in a dose-proportional manner during week 1 at steady
`state (r = 0.47, P < .001). The Cmax was subsequently
`reached by 2 hours after the dose and also increased in
`a dose-proportional manner (r = 0.51; P < .001). The
`peak trough fluctuation was relatively narrow and
`approximately 90% at all dose levels. Overall steady-
`state exposure (AUC) exhibited dose proportionality
`during week 1 (r = 0.53, P < .001). In addition, the
`absence of dose-effects in the global ANOVA on dose-
`normalized Cmax (P = .65) and AUC (P = .10) from the
`week 1 to month 9 profiles indicated that steady-state
`dose proportionality was maintained during the full
`study course.
`Everolimus longitudinal stability and pharmacoki-
`netic variability. Everolimus mean AUC trajectories are
`shown in Fig 1, A for all three dose levels. There was
`no dose-by-visit interaction (P = .85); this indicated
`that the pattern in AUCs over time was similar for all
`dose levels. A significant visit effect was noted (P =
`
`.004); subsequent pairwise contrasts attributed this to a
`lower than average pooled AUC per dose at week 2 (68
`ng · h/mL) and a higher than average pooled value at
`month 9 (91 ng · h/mL) relative to the surrounding vis-
`its (70-80 ng · h/mL). Therefore the temporal pattern
`was generally stable, with two relatively small devia-
`tions at isolated visits, during the full study course.
`The global longitudinal estimate was 85.4% for
`interindividual variability for AUC per dose and was
`40.8% for intraindividual,
`interoccasion variability.
`Explorations for covariates that contributed to interindi-
`vidual variability in exposure indicated that age, which
`ranged from 17 to 69 years, explained only 2.6% of the
`variability and weight, which ranged from 49 to 106 kg,
`contributed less than 1.0% on the basis of the coefficients
`of determination (r2) from linear regression analysis.
`Likewise, AUC per dose was not different between sexes
`(P = .39) or ethnicities (white versus nonwhite, P = .30).
`Everolimus serial trough concentrations. A total of
`651 steady-state trough concentrations were available
`between week 1 and month 12. There was no dose-by-
`visit interaction (P = .98); this confirmed the impres-
`sion that the Cmin trajectories were similar among the
`everolimus dose levels over time (Fig 1, B). At the low-
`est dose level, several troughs were below the assay
`quantification limit and were set to 0 ng/mL by conven-
`tion. This biased the mean values downward and
`resulted in a significant dose-effect (P = .03). Specifi-
`cally, Cmin per dose pooled across visits at the upper two
`
`Ex. 1050-0006
`
`

`
`54 Kovarik et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`JANUARY 2001
`
`A
`
`B
`
`Fig 3. Cyclosporine (CsA) morning doses (A) and cyclosporine mean AUC per dose trajectories to
`month 9 (B) in the everolimus 0.5-mg (solid squares), 1-mg (solid circles), and 2-mg twice a day (solid
`triangles) dose cohorts during 1 year after transplants. Bars designate 95% confidence intervals.
`
`dose levels was consistent with dose proportionality (4.5
`± 3.0 ng/mL/mg at 1 mg and 4.8 ± 2.4 ng/mL/mg at 2
`mg) but was less than proportional at the 0.5-mg dose
`level (3.2 ± 3.7 ng/mL/mg). A significant visit effect (P
`< .001) was detected; subsequent pairwise comparisons
`attributed the visit effect to an isolated occasion of low
`values at week 3 relative to the surrounding visits dur-
`ing the study course. Steady-state Cmin was well corre-
`lated with AUC during the year-long study; this yielded
`a correlation coefficient (r value) of 0.88.
`Everolimus exposure-response relationships. The
`average minimum platelet count over the first 2 post-
`transplant months was 160 ± 51 × 109/L; on average, the
`minimum count occurred by day 16. As shown in Fig 2,
`there was a significant relationship between everolimus
`
`AUC and the incidence of thrombocytopenia (P = .03).
`The average maximum triglyceride level was 3.8 ± 1.8
`mmol/L (337 ± 159 mg/dL) and the maximum choles-
`terol level was 7.8 ± 1.4 mmol/L (302 ± 54 mg/dL),
`which occurred on average by days 29 and 35, respec-
`tively. There were trends toward an increased incidence
`of both hypertriglyceridemia (P = .14) and hypercholes-
`terolemia (P = .06) with increasing everolimus AUC. The
`average minimum leukocyte count was 5.7 ± 1.7 × 109/L,
`and it occurred by day 29. The incidence of leukopenia
`was not related to everolimus exposure (P = .35).
`Cyclosporine dosing. Dosing began at 240 ± 74 mg,
`252 ± 50 mg, and 245 ± 60 mg twice a day in the
`ascending everolimus dose cohorts. As shown in Fig 3,
`A, cyclosporine doses were reduced over time (P <
`
`Ex. 1050-0007
`
`

`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 69, NUMBER 1
`
`Kovarik et al 55
`
`.001) as is conventional in the first post-transplant year.
`Cyclosporine doses did not differ among the everolimus
`dose groups (P = .13) nor did the temporal patterns dif-
`fer over the study course as shown by lack of a dose-
`by-visit interaction (P = .99).
`Cyclosporine pharmacokinetics. A total of 586
`cyclosporine trough concentrations were evaluable during
`the year-long study course. Trough concentrations were
`significantly reduced during the first post-transplant year
`(P < .001). The temporal patterns did not differ among
`everolimus cohorts (P = .58 for dose-by-visit interaction)
`nor were cyclosporine troughs different among the
`everolimus dose levels (P = .82). This was also true for
`dose-normalized troughs (P = .70 and .25, respectively).
`There was a total of 417 cyclosporine profiles (109
`from full 12-hour sampling and 308 from abbreviated
`sampling). Because cyclosporine doses were reduced
`over time, the corresponding AUCs differed signifi-
`cantly across study visits (P < .001). The temporal pat-
`tern for AUC per dose shown in Fig 3, B exhibited an
`increase during the first month after the transplant with
`stability thereafter despite the fact that the cyclosporine
`dose was being continuously decreased. This was the
`result of an alteration in the dose-exposure relationship
`during the first month as has been described in previ-
`ously published series with cyclosporine (Neoral) in the
`absence of everolimus.3 There was no significant dose-
`by-visit interaction (P = .94) or everolimus dose-effect
`(P = .76) for AUC or the corresponding dose-normal-
`ized values; this confirmed that the temporal patterns
`in cyclosporine AUC were similar regardless of
`everolimus dose cohort.
`
`DISCUSSION
`This study provided the first pharmacokinetic and
`exposure-response data for everolimus in patients with
`renal transplants longitudinally followed up from the
`de novo period in the first 3 post-transplant months
`through the metastable phase from months 3 to 6 and
`into the stable phase from months 6 to 12. The impor-
`tance of careful assessment of the disposition of an
`immunosuppressant during this time frame stems from
`the experience with cyclosporine and mycophenolate
`mofetil. For both agents there are changes in the dose-
`AUC relationship in the first 3 months after a trans-
`plant; these changes may need to be taken into account
`in dosing.3,4 The cyclosporine data from this study
`clearly show this phenomenon, as illustrated in Fig 3.
`The pharmacokinetics of everolimus after the first
`dose indicated some delays in absorption (median tmax
`of 3 hours) that were probably related to postsurgical
`gastrointestinal ileus rather than to the dosage formu-
`
`lation because there was rapid absorption (tmax of 1-2
`hours) by the next assessment (day 7). A good absorp-
`tion profile was maintained during the trial period and
`was similar to that characterized in stable patients with
`renal transplants in a previous study.6 Although absorp-
`tion was delayed after the first dose, both Cmax and
`AUC were dose proportional; this was also the case lon-
`gitudinally at steady state over the full study course.
`The range of doses used in this trial extended below
`and above those currently being assessed in multicen-
`ter efficacy studies. Therefore the dose proportionality
`observed in this study implies that safety- or efficacy-
`related dose titrations within the potential therapeutic
`dose range should yield proportional changes in AUC
`in the clinical setting. Although it is currently unknown
`whether dose titration will need to be guided by thera-
`peutic drug monitoring as is used for some other
`immunosuppressants, it was encouraging to observe the
`good correlation between predose trough concentra-
`tions and AUC, which suggests that monitoring of
`trough concentrations may serve as a marker for over-
`all exposure if needed.
`With regard to temporal stability in exposure, visual
`inspection of the AUC and Cmin trajectories as shown
`in Fig 1 did not show any specific longitudinal trends.
`Although there were significant visit effects in ANOVA,
`further pairwise comparisons of the parameters across
`visits indicated that values at a few isolated nonsequen-
`tial visits were either higher or lower than at the other
`visits. These transient fluctuations may have been
`caused by the different number of evaluable profiles at
`each visit because of patient withdrawals, dose inter-
`ruptions, and samples not being obtained. Nonetheless,
`the magnitudes of these isolated deviations in exposure
`are unlikely to be of clinical relevance, and a generally
`stable pattern in everolimus exposure was observed.
`Pharmacokinetic variability has conventionally been
`assessed in a replicate single-dose study with a rela-
`tively short washout period between assessments so that
`the only source of variability is theoretically intraindi-
`vidual, interoccasion effects. In this study, pharmaco-
`kinetic profiling was performed under outpatient dos-
`ing conditions during an extended period of 9 months
`(that is, from day 7 to month 9) during which the
`patient’s clinical condition changed. Both the long
`period of assessment and the change in clinical status
`must be kept in mind when one consideres the intrain-
`dividual, interoccasion coefficient of variation of 40.8%
`for dose-normalized AUC. This contrasts with the esti-
`mate from a previous study of everolimus in clinically
`stable patients with renal transplants who underwent
`pharmacokinetic profiling on two occasions 1 week
`
`Ex. 1050-0008
`
`

`
`56 Kovarik et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`JANUARY 2001
`
`apart and in whom the coefficient of intraindividual,
`interoccasion variability was 10% to 19%.7 Post-trans-
`plant changes in renal function in our study population
`is unlikely to have contributed to variability because
`renal clearance of everolimus is minimal. The coeffi-
`cient of interindividual variation in the present study
`was 85.4%

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket