• No results found

Prediction of Area under the Cyclosporine Concentration Versus Time Curve in Children Undergoing Hematopoietic Stem Cell Transplantation

N/A
N/A
Protected

Academic year: 2021

Share "Prediction of Area under the Cyclosporine Concentration Versus Time Curve in Children Undergoing Hematopoietic Stem Cell Transplantation"

Copied!
6
0
0

Loading.... (view fulltext now)

Full text

(1)

Prediction of Area under the Cyclosporine Concentration

Versus Time Curve in Children Undergoing Hematopoietic

Stem Cell Transplantation

L. Lee Dupuis

1

,

2

,

3

,

4

,

*

, Winnie Seto

1

,

4

, Oliver Teuffel

2

, Paul Gibson

7

,

Kirk R. Schultz

8

, John D. Doyle

2

,

5

, Adam Gassas

2

,

5

, R. Maarten Egeler

2

,

3

,

5

,

Lillian Sung

2

,

3

,

6

, Tal Schechter

2

,

3

,

6

1Department of Pharmacy, SickKids, Toronto, Ontario, Canada 2Division of Haematology/Oncology, SickKids, Toronto, Ontario, Canada 3Research Institute, SickKids, Toronto, Ontario, Canada

4Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada 5Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

6Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada

7Pediatric Haematology/Oncology, Childrens Hospital, London Health Sciences Centre, London, Ontario, Canada 8Division of Pediatric Hematology/Oncology/BMT, BC Childrens Hospital, University of British Columbia, Vancouver, British

Columbia, Canada Article history: Received 31 August 2012 Accepted 28 October 2012 Key Words: Cyclosporine Pediatrics Pharmacokinetics Area under the curve Acute graft versus host disease

a b s t r a c t

This prospective study aimed to validate a previously developedfirst-dose limited sampling strategy (LSS) to predict the area under the cyclosporine concentration-versus-time curve (AUC) and to develop and then validate an LSS to predict cyclosporine AUC at steady state. This two-center Canadian study included children (ages .4 to 17.2 years) undergoing myeloablative allogeneic hematopoietic stem cell transplantation receiving cyclosporine for acute graft-versus-host disease prophylaxis. There were three cohorts, each incorporating 24 AUC determinations:first-dose LSS validation, steady-state LSS development, and steady-state LSS vali-dation. Patients contributing data to either of the development cohorts were excluded from the corre-sponding validation group. Cyclosporine was given every 12 hours as a 2-hour infusion. Cyclosporine AUC was determined after administration of thefirst cyclosporine dose (8 samples) and then once weekly (9 samples) until engraftment. Steady-state LSSs were developed using stepwise multiple linear regression. An LSS was considered to provide an acceptable estimate of AUC if the lower limit of the 95% confidence limit (CL) of the intraclass coefficient was .8 or higher and both bias and precision were 15% or less. Fifty-three children age .4 to 18 years participated. Cyclosporine concentrations drawn up to 4 hours from the start of the infusion correlated most strongly with AUC. The previously developedfirst-dose LSSs and three steady-state LSSs met criteria for acceptability. The intraclass coefficients of the three-pointfirst-dose LSS validation cohort, three-point steady-state LSS development cohort, and three-three-point steady-state LSS validation cohort were .974 (95% CL: .941 to .988), .984 (95% CL: .965 to .993), and .993 (95% CL: .984 to .997), respectively. The three-point first-dose (2, 6, and 8 hours) and steady-state (2, 2.5, and 8 hours) LSSs are valid measures of cyclosporine AUC after intravenous administration over 2 hours. Their use in a prospective evaluation of the relationship between cyclosporine AUC and hematopoietic stem cell transplantation clinical outcomes in children is suggested.

Ó2013 American Society for Blood and Marrow Transplantation.

INTRODUCTION

Acute graft-versus-host-disease (aGVHD) is the primary

cause of treatment-related mortality after hematopoietic

stem cell transplantation (HSCT) and has a great impact on

morbidity and quality of life

[1]

. Calcineurin inhibitors

(cyclosporine or tacrolimus) in combination with

short-course methotrexate or corticosteroids are most commonly

used for aGVHD prevention. Because inter- and

intra-individual variability in cyclosporine disposition is high,

the whole blood trough concentration (C

0

) is routinely used

to individualize cyclosporine doses in HSCT patients.

However, evidence to support a relationship between C

0

values and aGVHD prevention is limited and highly variable.

For example, reported target C

0

values range from 85 to

500 ng/mL

[2-6]

. As a result, there is no consensus regarding

the optimal target C

0

range, and targets vary widely among

institutions

[7]

.

Prevention of acute graft rejection correlates more

strongly with area under the cyclosporine

concentration-versus-time curve (AUC) than C

0

in both adult and pediatric

solid organ transplant patients

[8,9]

. Cyclosporine dose

adjustment based on AUC rather than C

0

has consequently

become the standard of care. This approach has only just

begun to be evaluated in HSCT

[10-19]

. Classical

determina-tion of AUC entails a large total blood sample volume, high

workload to obtain the samples, high assay costs, and

potentially a high level of patient inconvenience or distress.

Blood volume is especially a concern in pediatrics. A valid

limited sampling strategy (LSS) would facilitate large-scale

evaluations of the relationship between cyclosporine AUC

and clinical outcomes such as aGVHD prophylaxis in

pediatrics.

We described the relationship between AUC and whole

blood cyclosporine concentrations at various single time

American Society for Blood

ASBMT

and Marrow Transplantation

Financial disclosure:See Acknowledgments on page 422.

*Correspondence and reprint requests: L. Lee Dupuis, MScPhm, Department of Pharmacy, SickKids, 555 University Ave., Toronto, Ontario, Canada, M5M 1X3.

E-mail address:lee.dupuis@sickkids.ca(L.L. Dupuis).

1083-8791/$esee front matterÓ2013 American Society for Blood and Marrow Transplantation. http://dx.doi.org/10.1016/j.bbmt.2012.10.031

(2)

points within a dosing interval after the

rst intravenous

(i.v.) cyclosporine dose given as a 2-hour infusion in 24

children undergoing HSCT

[20]

. Strong correlations between

AUC and each of the concentrations obtained within the

rst

4 hours after the start of the infusion (2, 2.5, 3, and 4 hours)

(Spearman rho coef

cient

¼

.717 to .868) were observed

but only a moderate relationship (Spearman rho

coef-fi

cient

¼

.442 to .553) between AUC and the concentrations

obtained at the remaining time points. The correlation

between C

0

and AUC was moderate (Spearman rho

coef

cient

¼

.457). Using these data, we developed an LSS for

the prediction of AUC achieved in children after

adminis-tration of the

rst i.v. cyclosporine dose. An LSS for prediction

of cyclosporine AUC after steady-state cyclosporine i.v.

administration has not yet been developed.

The primary purpose of this prospective study was to

validate the LSS previously developed to estimate

cyclo-sporine AUC after the

rst i.v. cyclosporine dose and to

develop and validate an LSS for use once steady state was

achieved in children undergoing HSCT. These LSSs will be

used in future studies of the relationship between HSCT

clinical outcomes and cyclosporine AUC and cyclosporine

concentrations at time points other than C

0

.

METHODS

This study was approved by the Research Ethics Boards of both SickKids and BC Children’s Hospital. Patients or guardians provided informed consent and/or assent.

Study Population

Patients less than 18 years of age who were undergoing allogeneic myeloablative HSCT at either SickKids or Women’s and Children’s Hospital of British Columbia and who were scheduled to receive cyclosporine for aGVHD prophylaxis were eligible to participate. Children who received voriconazole or posaconazole within 14 days of thefirst cyclosporine dose, or at any time until engraftment, were excluded. Patients receivingfl uco-nazole were included in this study becausefluconazole does not alter the disposition of i.v. cyclosporine[21,22].

Cyclosporine Administration, Blood Sampling, and Dose Adjustment

Cyclosporine was administered as a 120-minute (15 minutes) i.v. infusion (including the lineflush) every 12 hours starting on day3,2, or1 (where the day of HSCT is day 0). One lumen of a central venous access device was used consistently for cyclosporine dose administration. Cyclo-sporine dose adjustments were made by the clinical team caring for the patient using the blood samples obtained for clinical care in accordance with the institutional target C0concentrations (100 to 200 ng/mL).

All samples for cyclosporine concentration determination were ob-tained from a lumen of a central venous access device not used for cyclo-sporine administration. After thefirst i.v. cyclosporine dose, blood samples were obtained for determination of cyclosporine concentration at 2 (end of the infusion), 2.5, 3, 4, 6, 8, 10, and 12 hours after the start of the 2-hour infusion. Samples drawn within 10 minutes of the ideal sampling time were acceptable.

After day 0, blood samples were obtained for determination of cyclo-sporine concentration after the morning dose weekly on a standard day of the week (eg, every Monday) until engraftment (absolute neutrophil count>.5109/L on 2 consecutive measurements separated by more than 1

day) occurred. In total, 9 blood samples per week were obtained: just before the morning cyclosporine dose and at 2 (end of the infusion), 2.5, 3, 4, 6, 8, 10, and 12 hours after the start of the 2-hour infusion. Blood samples were obtained weekly for no longer than 7 weeks after transplantation.

Cyclosporine Assay

Whole blood cyclosporine concentrations were determined in a single laboratory using tandem mass spectrometry. The interday coefficient of variation of this assay is 3.6% to 4.2% (Warren Walsh, Resource Technologist, Department of Laboratory Medicine, SickKids, personal communication, October 27, 2009) Study samples were frozen at20C until assayed.

Data Analysis

Statistical analysis was performed using SAS Enterprise Guide (version 4.3, SAS Institute, Cary, NC) or SPSS (version 17, SPSS Inc., Chicago, IL). APvalue of .05 or less was considered to indicate statistical significance.

Pharmacokinetics

The cyclosporine AUC achieved over the 12-hour dosing interval was calculated using all data points (AUC-12) for all patients using non-compartmental methods (Pharsight Phoenix WinNonLin 6.0.0.1648, St Louis, MO). The relationship between AUC and whole blood cyclosporine concentrations at individual time points was assessed by the Spearman rho correlation coefficient.

First-Dose LSS Validation

The previously developedfirst-dose LSS[20]was validated using the cyclosporine concentrations obtained from thefirst 24 patients enrolled for whom 8 properly timed cyclosporine concentrations were available. Each patient’s AUC was calculated using the previously developed LSSs. Agree-ment between the AUC-12 and the AUC calculated using each LSS was assessed using a single-measure, one-way, random effect, intraclass coeffi -cient (ICC)[23,24]. Agreement was considered to be acceptable if the lower limit of the 95% confidence limit of the ICC was .8 or higher. The extent of agreement between AUC-12 and predicted AUC was also assessed using Bland-Altman analysis. The limits of agreement (mean % difference2 standard deviations, where % difference¼100[(AUC-12eAUC-LSS)/(mean of

AUC-12 and AUC-LSS)]) were calculated. It was not possible to determine the clinically significant limits of agreement a priori for cyclosporine because the clinical significance of AUC-12 with respect to HSCT outcomes is not yet established. Bias (mean prediction error) and precision (root mean squared error) of 15% or less were considered to be acceptable[25]. In summary, an LSS was considered to provide an acceptable estimate of AUC-12 if the lower limit of the 95% confidence limit of the ICC was .8 or higher and both bias and precision were 15% or less. The association between patient age and percent bias of the AUC estimated by the 3-point LSS was undertaken using linear regression analysis.

Steady-State LSS Development and Validation

Steady state was defined as having been achieved once the patient had received at least 4 consecutive, identical cyclosporine doses. Thefirst 24 patients enrolled for whom all 9 cyclosporine concentrations were available at steady state constituted the development group; the second group of 24 patients enrolled for whom 9 properly timed cyclosporine concentrations were available at steady state constituted the validation group. None of the patients in the validation group was included in the development group.

LSSs were developed at steady state using stepwise multiple linear regression analysis [25]. LSSs involving 5 or fewer data points were preferred for model parsimony and clinical utility. Adjusted r2values greater

than .9 were considered acceptable. Agreement between AUC-12 and the AUC calculated using each steady-state LSS was assessed using ICC and Bland-Altman analysis as described above, as were the criteria for accep-tance of a steady-state LSS as an estimate of AUC-12. The association between patient age and percent bias of the AUC estimated by the 3-point LSSs was undertaken using linear regression analysis.

RESULTS

From June 2008 to March 2012, 104 patients were

screened for study eligibility. Thirty did not meet study entry

criteria, 12 declined to participate, the attending physician of

5 patients asked that they not be approached, and 3 were

missed due to logistical problems. Fifty-four patients were

enrolled; 1 later withdrew before sampling at steady state

occurred. Of the patients who enrolled, blood sampling

deviated from protocol after administration of the

rst dose

in 14 patients. Furthermore, blood sampling deviated from

protocol or cyclosporine dosing never achieved steady state

in 3 and 2 patients beyond the

rst cyclosporine dose,

respectively. Data from 53 children are reported here.

Demographic data relating to the study patients are

pre-sented in

Table 1

. It is notable that mycophenolate mofetil

was more commonly used in the steady-state validation

cohort (7 of 24 patients) compared with the other cohorts

(0 or 1 of 24 patients). Fourteen patients contributed data to

both the

rst-dose LSS validation group and the steady-state

LSS development group, whereas 7 contributed data to both

(3)

the

rst-dose LSS validation group and the steady-state LSS

validation group.

First-Dose LSS Validation

Table 2

illustrates that whole blood cyclosporine

concentrations drawn within the

rst 4 hours from the start

of the infusion correlated strongly with AUC-12 (Spearman

rho coef

cient

¼

.66 to .91) Correlations with other time

points were moderate.

The four previously developed

rst-dose LSS met the

criteria for acceptability (

Table 3

). That is, adjusted r

2

was

>

.9, ICC indicated good agreement, and mean bias and

precision were

<

15%. In addition, Bland-Altman analysis

indicated good agreement between the AUC-12 and

pre-dicted AUC values (

Table 3

,

Figure 1

A). No signi

cant

asso-ciation between patient age and percent bias of the AUC

estimates was observed (adjusted r

2

¼

.0332). AUC-12 and

AUC values calculated using the LSSs are presented in

Table 4

.

Steady-State LSS Development and Validation

The mean day of sampling relative to day 0 was

day

þ

18.0

8.7 and day

þ

17.6

7.4 in the steady-state LSS

development and validation cohorts, respectively. Whole

blood cyclosporine concentrations drawn from 2 to 4 hours

from the start of the 2-hour infusion correlated most strongly

with AUC-12 (

Table 2

; Spearman rho coef

cient

¼

.80609 to

.87913 [validation cohort] and .85409 to .95238

[develop-ment cohort]). Correlations with other time points were also

signi

cant in both cohorts. However, although the

correla-tions were weak to moderate at time points beyond 4 hours

from the start of the 2-hour infusion in the development

cohort, correlations with AUC-12 were strong at all time

points in the validation cohort.

Three steady-state LSSs were developed that met criteria

for acceptability: the three-, four-, and

ve-point LSSs

(

Table 3

,

Figure 1

B). That is, ICC indicated good agreement

between the AUC-12 value and that predicted by the three-,

four-, and

ve-point LSSs, adjusted r

2

was

>

.9, and both

mean bias and precision were

<

15%. The Bland-Altman

analysis also indicated good agreement. When evaluated in

the validation cohort, the 3 acceptable steady-state LSSs also

met criteria for acceptability and demonstrated good

agree-ment with AUC-12 values (

Table 3

,

Figure 1

C). No signi

cant

association between patient age and percent bias of the AUC

estimates

was

observed

in

either

cohort

(adjusted

r

2

¼

.0187 [development cohort] and

.0249 [validation

cohort]). AUC-12 and AUC values calculated using the LSSs

are presented in

Table 4

.

DISCUSSION

Using

complete

pharmacokinetic

pro

les

collected

prospectively, we developed and validated LSSs for

predict-ing AUC of cyclosporine after administration as a 2-hour i.v.

infusion to children undergoing HSCT both after the

rst

dose and at steady state. Speci

cally, we found that a 3-point

LSS is a valid method for estimating cyclosporine AUC after

both the initial i.v. cyclosporine dose and at steady state. This

information will facilitate assessment of cyclosporine AUC by

clinicians and the assessment of the relationship, if any,

between cyclosporine AUC and HSCT outcomes in future

studies.

Cyclosporine dose adjustment using the trough whole

blood cyclosporine concentration (C

0

) remains the standard

in HSCT, although the target concentration range is highly

variable among HSCT centers as stated previously. Several

investigators have reported cyclosporine AUC in HSCT

patients after i.v. administration

[3,10-14,16-19,26,27]

. The

method used to calculate or estimate cyclosporine AUC was

not described in some of these reports

[13]

, and serum rather

than whole blood concentrations (the accepted standard

matrix) were reported in another

[10]

. LSSs have been

developed and validated for estimation of AUC-12 in HSCT

patients older than 15 years receiving oral cyclosporine

[17]

.

The proposed LSSs had minimal bias. Agreement between

AUC-12 and the AUC estimates generated by the LSSs was not

evaluated. Most pediatric patients receive i.v. cyclosporine

Table 1

Description of patients in First Dose Development (N¼24) and Validation (N¼24) Cohorts and Steady-State Development (n¼24) and Validation (n¼24) Cohorts

Parameter First-Dose Development Cohort First-Dose Validation Cohort Steady-State Development Cohort Steady-State Validation Cohort Mean age, yr,SD (range) 8.84.81 (.5-16.9) 8.84.89 (.4-17.2) 10.25.06 (.4-17.2) 9.33.54 (3.3-17.2) Day of sampling (no. days after HSCT) d d 18.08.70 (10-39) 17.67.39 (4-32)

Mean cyclosporine dose

(mg/kg;SD) 1.5.05 2.1.57 2.31.28 2.51.57 (mg/m2;SD) NA 58.49.64 69.234.04 69.635.79 Number of patients Sex (male:female) 16:8 15:9 16:8 15:9 Underlying diagnosis ALL 8 5 4 7 AML 10 9 10 7 Other malignancies 2 2 3 3 Nonmalignancies 4 8 7 7 HSCT conditioning Cyclophosphamide-containing 21 24 24 22 Other 3 0 0 2 GVHD prophylaxis CSA alone 0 2 0 2 CSAþ Methotrexate 24 13 17 11 Mycophenolate mofetil 0 1 0 7 Corticosteroid 0 6 6 4 Methotrexateþcorticosteroid 0 2 1 0

(4)

immediately after HSCT and convert to oral dosing several

weeks later, usually after engraftment. Because achievement

of a minimum C

0

has been reported to be most important

just before engraftment

[2]

, it may be most clinically relevant

to evaluate cyclosporine AUC when patients are receiving i.v.

cyclosporine.

The relationship between cyclosporine AUC after i.v.

administration and HSCT outcomes such as aGVHD is just

beginning to be explored

[11,13,17-19,26,27]

. Among these

are 2 pediatric studies. In the

rst, Willemze et al.

retro-spectively estimated cyclosporine pharmacokinetic

param-eters including AUC in 91 children receiving i.v. cyclosporine

over 2 hours

[19]

. Cyclosporine concentrations were assayed

by either radioimmunoassay or

uorescence polarization

immunoassay. No comment regarding the comparability of

the results of these assays was provided. AUC-12 was

esti-mated using Bayesian methods based primarily on C

0

values

that the authors stated led to a potential error in AUC

esti-mations of approximately 20%. During the second week after

HSCT, AUC values greater than 3000

m

g/L/h were associated

with lower overall survival rates and higher risk of relapse,

most especially in children with acute myelogenous

leukemia. No association between AUC and the incidence of

acute GVHD was observed.

Gerard et al. reported the second evaluation of

cyclo-sporine AUC in children undergoing HSCT

[26]

. In this

retro-spective analysis, cyclosporine AUC in blood and other tissues

were estimated using Bayesian models. The sampling

schedule and the number of cyclosporine blood

concentra-tions available to inform the construction of the

pharmaco-kinetic model were not described. Children received

cyclosporine either as a continuous infusion (30 patients) or

as a 2-hour infusion every 12 hours (31 patients). Thirty-four

patients (56%) developed aGVHD. It was not clear if the

relationship between C

0

and aGVHD occurrence was

evalu-ated. However, there was a signi

cant relationship between

cyclosporine AUC at steady state (de

ned as the day before

the occurrence of aGVHD or day 13 after HSCT) and aGVHD.

For patients with underlying malignancy, the authors

concluded that cyclosporine doses be adjusted to achieve an

AUC of 10 h

mg/L over 24 hours (10,000

m

g/L/h over 24 hours;

5000

m

g/L/h over 12 hours), which corresponds to a C

2

and C

12

of 1000 and 80

m

g/L, respectively. It was furthermore

sug-gested that an AUC of 16 h

mg/L over 24 hours (16,000

m

g/L/h

over 24 hours; 8000

m

g/L/h over 12 hours) corresponding

to a C

2

and C

12

of 1600 and 120

m

g/L, respectively, be targeted

for patients without underlying malignancy.

The

ndings of these retrospective studies require

prospective validation before speci

c AUC values can be

adopted as the standard of care in clinical practice. The

availability of LSSs for estimation of cyclosporine AUC, such

as presented here, that can be performed without the use of

computer software will facilitate this process.

We found that the correlation between all data points and

AUC-12 was remarkably stronger in the steady-state LSS

validation cohort than in the other 3 study cohorts. Another

Table 3

Bias and Precision of Estimates of AUC Using Limited Sampling Strategies for i.v. Cyclosporine Involving Fewer Than Five Data Points and Extent of Agreement between Values of AUC Calculated Using All Data Points and Using the LSS

Sample Time (hours from start of 2-h infusion)

LSS Equation Adjusted r2 Mean Bias,

% (95% CI)

Precision (%) ICC (95% CI) Limits of Agreement (%) Validation offirst-dose LSS[20] 2, 6, and 8 1.429C2þ5.722C8þ2.741C6þ380.862 .965 2.1 (.1-4.3) 5.0 .974 (.941 to .988) 13.4-8.9 2, 2.5, 6, and 8 1.372C2þ5.449C8þ2.870C6þ.470C2.5þ 165.709 .985 2.5 (3.9-1.2) 3.2 .979 (.952-.991) 4.5-9.8 2, 2.5, 4, 6, and 8 1.229C2þ5.242C8þ.740C6þ.514C2.5þ 2.7C4þ23.740 .993 2.2 (2.9-1.4) 2.3 .994 (.985-.997) 1.7-6.1 Development of steady-state LSS 2, 2.5, and 8 1.22C2þ1.54C2.5þ8.26C8þ292 .9851 .34 (2-1) 3.5 .984 (.965-.993) 7.2-7.3 2, 2.5, 4, and 8 1.22C2þ.78C2.5þ2.55C4þ5.91C8þ331 .9920 .19 (1-1) 4.9 .994 (.986-.997) 4.6-4.3 2, 2.5, 3, 4, and 8 1.26C2þ1.65C3þ2.43C4þ4.80C8þ261 .9967 .13 (0-1) 1.2 .995 (.988-.998) 2.9-2.9 Validation of steady-state LSS 2, 2.5, and 8 1.22C2þ1.54C2.5þ8.26C8þ292 .9875 3.5 (2-5) 3.3 .993 (.984-.997) 9.3-3.3 2, 2.5, 4, and 8 1.22C2þ.78C2.5þ2.55C4þ5.91C8þ331 .9937 2.3 (1-3) 3.5 .996 (.991-.998) 7.3-2.7 2, 2.5, 3, 4, and 8 1.26C2þ1.65C3þ2.43C4þ4.80C8þ261 .9935 1.7 (1-3) 1.4 .998 (.996-.999) 7.1-3.9

CI indicates confidence interval.

An LSS was considered to provide an acceptable estimate of AUC-12 if the r2value was>.9, lower limit of the 95% condence limit of the ICC was .8 or higher, and

both bias and precision were 15% or less.

Table 2

Spearman Rho Correlation Coefficients for AUC-12 and Whole Blood Cyclosporine Concentrations at Time Points after the Start of a 2-Hour Infusion Sampling

Time (h)

First-Dose LSSValidation Cohort Steady-State LSS Development Cohort Steady-State LSS Validation Cohort Spearman

Rho Coefficient

Significance (Two-Tailed)

Spearman Rho Coefficient

Significance (Two-Tailed)

Spearman Rho Coefficient

Significance (Two-Tailed) 0 NA NA .43845 .0321 .80409 <.0001 2 .91324 <.0001 .87913 <.0001 .85478 <.0001 2.5 .66014 <.0004 .83652 <.0001 .95238 <.0001 3 .71739 <.0001 .80609 <.0001 .91933 <.0001 4 .90039 <.0001 .83826 <.0001 .94609 <.0001 6 .82775 <.0001 .54502 .0059 .92345 <.0001 8 .74543 <.0001 .47097 .0202 .90522 <.0001 10 .65810* <.0006 .45605 .0251 .89841 <.0001 12 .68099* <.0007 .34181* .1021 .87671 <.0001 *n¼23.

(5)

notable distinction in this cohort was the increased use of

mycophenolate mofetil compared with the other 3 cohorts.

The potential in

uence of mycophenolate mofetil on

cyclo-sporine pharmacokinetic disposition has been identi

ed

previously

[28]

and deserves further exploration.

For the purpose of this study, steady state was de

ned as

having been achieved when patients had received at least 4

identical cyclosporine doses. Judging by the discrepant

correlations between C

0

and the concentration 12 hours after

the start of the infusion (C

12

), true steady state likely was not

achieved in all patients. Indeed, the C

0

and C

12

differed

by

>

20% in 6 (25%) and 4 patients (17%) in the development

and validation

steady-state LSS

cohorts,

respectively.

Fluctuations in dose times, infusion duration and/or rate,

liver function, and extent of enterohepatic circulation may

preclude achievement of steady state in HSCT patients in the

rst weeks after HSCT. Interpretation of our study

ndings

may also be limited by differences between cohorts such as

differences in aGVHD prophylaxis, pharmacogenetic

poly-morphism

[29]

, and underlying diagnosis. Despite these

possible limitations, the proposed LSSs performed well in

estimating AUC-12 within each patient.

The LSSs developed and validated here permit con

dent

estimation of AUC-12 by clinicians. It must be noted that

their use is limited to the population in which they were

developed. That is, they may be used to estimate

cyclo-sporine AUC in children undergoing HSCT age .4 to

<

18 years

of age and receiving cyclosporine as a 2-hour i.v. infusion.

Use of the 3-point

rst-dose (2, 6, and 8 hours) and

steady-state (2, 2.5, and 8 hours) LSSs to prospectively evaluate

the relationship between cyclosporine AUC and HSCT

outcomes in children is suggested.

ACKNOWLEDGMENTS

We are thankful to the children and their families who

participated in this study; Warren Walsh and Daksha Jadav,

Department of Pediatric Laboratory Medicine, SickKids; the

Clinical Research Assistants at SickKids and Children &

Women

s Health Centre of British Columbia (Entela Zhuleku,

Kim Pagel, Alexandra Airhart, Calvin Liu, Sheliza Moledina,

and Colleen Fitzgerald); and the nursing and pharmacy staff

of the Blood and Bone Marrow Transplant units, SickKids,

and Children & Women

s Health Centre of British Columbia.

Financial disclosure:

This study was funded by the

Pedi-atric Oncology Group of Ontario and C

17

Research Network.

L.S. is supported by a New Investigator Award from the

Canadian Institutes of Health Research.

Con

ict of Interest Statement

: There are no con

icts of

interest to report.

REFERENCES

1. Jacobsohn DA. Acute graft-versus-host disease in children. Bone Marrow Transplant. 2008;41:215-221.

2. Punnett A, Sung L, Price V, et al. Achievement of target cyclosporine concentrations as a predictor of severe acute GvHD in children receiving cyclosporine and methotrexate prophylaxis.Ther Drug Monit. 2007;29:750-757.

3. Martin P, Bleyzac N, Souillet G, et al. Clinical and pharmacological risk factors for acute graft-versus-host disease after paediatric bone marrow transplantation from matched-sibling or unrelated donors.

Bone Marrow Transplant. 2003;32:881-887.

Table 4

Mean and Predicted AUC-12 Value Estimates Using the LSSs for Estimation of Cyclosporine AUC Estimation after i.v. Administration over 2 Hours

Mean AUC-12 (SD)mg/L/h) 95% CI Validation offirst-dose LSS AUC-12 2509672 2226-2793 Three point 2546588 2297-2794 Four point 2431595 2180-2682 Five point* 2480671 2190-2770 Development of steady-state LSS AUC-12 48881316 4279-5496 Three point 48651305 4262-5468 Four point 49401289 4424-5455 Five point 49351290 4419-5451 Validation of steady-state LSS AUC-12 42001142 3746-4657 Three point 43241168 3856-4791 Four point 42821110 3837-4726 Five point 42551105 3813-4697 *n¼23.

Figure 1.Bland-Altman plots. Percent difference against mean for actual (AUC-12) and predicted area under the whole blood cyclosporine concentra-tion versus time curve during 12-hour dosing interval generated by LSS. Percent difference¼100[(AUC-12eAUC predicted by LSS)]/mean of AUC-12

and AUC predicted by LSS]. (A) First-dose 4-point LSS validation: % difference against mean AUC. (B) Steady-state 3-point LSS development: % difference against mean AUC. (C) Steady-state three-point LSS validation: % difference against mean AUC.

(6)

4. Kanda Y, Hyo R, Yamashita T, et al. Effect of blood cyclosporine concentration on the outcome of hematopoietic stem cell trans-plantation from an HLA-matched sibling donor.Am J Hematol. 2006;81: 838-844.

5. Ram R, Storer B, Mielcarek M, et al. Association between calcineurin inhibitor blood concentration and outcomes after allogeneic hemato-poietic cell transplantation. Biol Blood Marrow Transplant. 2012;18: 414-422.

6. Malard F, Szydlo R, Brissot E, et al. Impact of cyclosporine-A concen-tration on the incidence of severe acute graft-versus-host-disease after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2010;16:28-34.

7. Ruutu T, Niederwieser D, Gratwohl A, Apperley JF. A survey of the prophylaxis and treatment of acute GVHD in Europe: a report of the European Group for Blood and Marrow, Transplantation (EBMT). Chronic Leukaemia Working Party of the EBMT.Bone Marrow Trans-plant. 1997;19:759-764.

8. Levy G, Thervet E, Lake J, Uchida K. Patient management by Neoral C2 monitoring: an international consensus statement. Transplantation. 2002;73:S12-S18.

9. Nashan B, Cole E, Levy G, Thervet E. Clinical validation studies of Neoral C2 monitoring: A review.Transplantation. 2002;73:S3-S11.

10. Schrauder A, Saleh S, Sykora KW, et al. Pharmacokinetic monitoring of intravenous cyclosporine A in pediatric stem-cell transplant recipients. The trough level is not enough.Pediatr Transplant. 2009;13:444-450. 11. Barkholt L, Remberger M, Bodegard H, et al. Cyclosporine A (CSA) 2-h

concentrations vary between patients without correlation to graft-versus-host disease after allogeneic haematopoietic stem cell trans-plantation.Bone Marrow Transplant. 2007;40:683-689.

12. Willemze AJ, Cremers SC, Schoemaker RC, et al. Ciclosporin kinetics in children after stem cell transplantation.Br J Clin Pharmacol. 2008;66: 539-545.

13. Hendriks MP, Blijlevens NM, Schattenberg AV, et al. Cyclosporine short infusion and C2 monitoring in haematopoietic stem cell transplant recipients.Bone Marrow Transplant. 2006;38:521-525.

14. Kong D, Shuttleworth P, Bailey M, Grigg A. CsA 2-h concentration correlates best with area under the concentration-time curve after allo-SCT compared with trough CsA.Bone Marrow Transplant. 2012;47:54-59. 15. Balci Y, Tavil B, Karabulut E, et al. Cyclosporine level at the second hour in pediatric hematopoietic stem cell transplant patients. Exp Clin Transplant. 2011;5:329-335.

16. Furukawa T, Kurasaki-Ida T, Masuko M, et al. Pharmacokinetic and pharmacodynamic analysis of cyclosporine A (CsA) tofind the best single time point for the monitoring and adjusting of CsA dose using twice-daily 3-h intravenous infusions in allogeneic hematopoietic stem cell transplantation.Int J Hematol. 2010;92:144-151.

17. Hadjibabaie M, Vazirian I, Iravani M, et al. Development and validation of limited sampling strategies for estimation of cyclosporine area under the concentration-time curve in hematopoietic stem cell transplant patients.Ther Drug Monit. 2011;33:673-680.

18. Inoue Y, Saito T, Ogawa K, et al. Pharmacokinetics of cyclosporine A at a high-peak concentration of twice-daily infusion and oral adminis-tration in allogeneic haematopoietic stem cell transplantation.J Clin Pharm Ther. 2011;36:518-524.

19. Willemze AJ, Press RR, Lankester AC, et al. CsA exposure is associated with acute GVHD and relapse in children after SCT. Bone Marrow Transplant. 2010;45:1056-1061.

20. Sibbald C, Seto W, Taylor T, et al. Determination of area under the whole blood concentration versus time curve afterfirst intravenous cyclosporine dose in children undergoing hematopoietic stem cell transplant: limited sampling strategies. Ther Drug Monit. 2008;30: 434-438.

21. Osowski CL, Dix SP, Lin LS, et al. Evaluation of the drug interaction between intravenous high-dose fluconazole and cyclosporine or tacrolimus in bone marrow transplant patients.Tranplantation. 1996; 61:1268-1272.

22. Canafax DM, Graves NM, Hilligoss DM, et al. Interaction between cyclosporine and fluconazole in renal allograft recipients. Trans-plantation. 1991;51:1014-1018.

23. Muller R, Buttner P. A critical discussion of intraclass correlation coefficients.Stat Med. 1994;13:2465-2476.

24. Lee J, Ong CN. Statistical evaluation of agreement between two methods for measuring a quantitative variable.Comput Biol Med. 1989; 19:61-70.

25. Ting LS, Partovi N, Levy RD, et al. Limited sampling strategy for pre-dicting area under the concentration-time curve of mycophenolic acid in adult lung transplant recipients. Pharmacotherapy. 2006;26: 1232-1240.

26. Gerard C, Bleyzac N, Girard P, et al. Links between cyclosporine exposure in tissues and graft-versus-host disease in pediatric bone marrow transplantation: analysis by a PBPK model.Pharm Res. 2011; 28:531-539.

27. Wilhelm A, de Graaf P, Veldkamp A, et al. Population pharmacokinetics of cyclosporin in haematopoietic allogeneic stem cell transplantation with emphasis on limited sampling strategy.Br J Clin Pharmacol. 2012; 73:553-563.

28. Pape L, Froede K, Strehlau J, et al. Alterations of cyclosporin A metab-olism induced by mycophenolate mofetil.Pediatr Transplant. 2003;7: 302-304.

29. Qui F, He X-J, Sun Y-X, et al. Influence of ABCB1, CYP3A4*18B and CYP3A5*3 polymorphisms on cyclosporine A pharmacokinetics in bone marrow transplant recipients.Pharmacol Rep. 2011;63:815-825.

http://dx.doi.org/10.1016/j.bbmt.2012.10.031Biol Blood Marrow Transplant 19 (2013) 418

References

Related documents

The SPI and GDI indices values showed good or high ecological status in all sampling seasons, corresponding with a II or I water quality class.. The TDI trophic index had the

A random sampling was utilized to determine the predictive relationship between independent variables leadership style (autocratic, laissez faire and democratic leadership

social justice illiteracy, Harry advocated and shared his ideas and perspectives on education, 460. especially the need to involve a wider community

In this paper, the effect of radius variation of beam light on output efficacy of SFD Yttrium aluminium borate laser doped with Neodymium ion, which is simultaneously a non-linear

Students who have successfully completed the Graduate Diploma of Research Studies in Science with a minimum GPA of 5 and a minimum grade of 5 in the dissertation component

We will discuss a credit union’s accounting for: (i) external and internal costs to modify existing internal-use computer software for the year 2000; (ii) purchases or development