DIAB-5944; No. of Pages 9 diabetes research and clinical practice xxx (2014) xxx–xxx
Contents available at ScienceDirect
Diabetes Research and Clinical Practice journ al h ome pa ge : www .elsevier.co m/lo cate/diabres
Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin Ling He a,*, Prachi Wickremasingha a, James Lee a, Ben Tao a, Jeanne Mendell-Harary a, Joseph Walker a, Douglas Wight b a b
Daiichi Sankyo Pharma Development, Edison, NJ, USA Milestone Pharmaceuticals, Montreal, Que´bec, Canada
article info
abstract
Article history:
Aims: Drug interactions with bile acid sequestrants are primarily due to the potential of
Received 29 August 2013
these agents to bind to concomitant drugs. Six clinical studies were performed to determine
Received in revised form
the effects of colesevelam on the pharmacokinetics of aspirin, atenolol, enalapril, phenyto-
5 November 2013
in, rosiglitazone, and sitagliptin.
Accepted 21 December 2013
Methods: All six studies enrolled healthy subjects aged 18–45 years. The phenytoin study
Available online xxx
used a single-dose, three-period crossover design (phenytoin alone, phenytoin simultaneously with colesevelam, and phenytoin 4 h before colesevelam). The other studies used a
Keywords:
two-period crossover design (test drug alone and test drug simultaneously with coleseve-
Drug–drug interactions
lam). Colesevelam (3750 mg once daily) was dosed throughout the pharmacokinetic sam-
Colesevelam
pling period. After each single dose of the test drug, serial blood samples were collected for
Antihypertensive agents
determination of plasma drug concentrations and calculation of pharmacokinetic param-
Hypoglycemic agents
eters.
Anticonvulsants
Results: For all six test drugs, 90% CIs for geometric least-squares mean ratios of AUC and Cmax for the measured analytes were within specified limits, indicating no interaction between the test drug and colesevelam. Conclusions: Aspirin, atenolol, enalapril, rosiglitazone, and sitagliptin may be taken with colesevelam. Although the phenytoin study indicated no pharmacokinetic interaction, phenytoin should continue to be taken 4 h before colesevelam in accordance with current prescribing information. # 2014 Elsevier Ireland Ltd. All rights reserved.
1.
Introduction
Bile acid sequestrants have been used for a number of years for the treatment of adults with primary hyperlipidemia [1], while the bile acid sequestrant colesevelam is additionally indicated to improve glycemic control in adults with type 2 diabetes
mellitus (T2DM) [2]. Bile acid sequestrants are not systemically absorbed and therefore do not interact with drugs systemically, but there is the potential for medications taken concomitantly to be bound by the bile acid sequestrants in the gastrointestinal tract, thereby reducing absorption and decreasing therapeutic efficacy [3]. Colesevelam has greater bile acid-binding capacity than the first-generation bile acid
* Corresponding author at: Daiichi Sankyo Pharma Development, 339 Thornall Street, Edison, NJ 08837, USA. Tel.: +1 732 590 5193; fax: +1 732 906 5690. E-mail address:
[email protected] (L. He). 0168-8227/$ – see front matter # 2014 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.diabres.2013.12.033 Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9
2
diabetes research and clinical practice xxx (2014) xxx–xxx
sequestrants colestipol and cholestyramine [4], which hypothetically could influence its propensity for binding to other drugs and causing drug–drug interactions [5]. Data on potential drug–drug interactions with colesevelam are available for 28 drugs based on testing of in vitro binding, in vivo drug interaction studies with colesevelam, and/or postmarketing reports consistent with potential drug–drug interactions; a complete listing is included in the colesevelam United States prescribing information [2]. Drug–drug interaction profiles are of particular importance when making treatment decisions for patients with T2DM, who may take multiple medications to manage their many comorbidities [6–8]. The American Diabetes Association recommends that patients with T2DM receive pharmacotherapy for the management of glycemic control, hypertension, and dyslipidemia, and for primary prevention of cardiovascular disease [9]. As the number of prescribed medications in a patient’s regimen increases, the risk of drug–drug interactions correspondingly increases [10,11]. The purpose of this paper is to report on a series of recent drug–drug interaction studies that showed no pharmacokinetic interactions. This series of studies was performed to determine the effects of colesevelam on the single-dose pharmacokinetics of aspirin, atenolol, enalapril (a prodrug that is metabolized in vivo to enalaprilat, the pharmacologically active chemical form, by various esterases), rosiglitazone, and sitagliptin, medications that may be commonly prescribed in patients with T2DM. In addition, a study was performed to determine the effects of colesevelam on the single-dose pharmacokinetics of phenytoin. This was based on postmarketing reports consistent with potential drug–drug interactions (increased seizure activity or decreased phenytoin levels) in patients concomitantly receiving colesevelam with phenytoin [2], a compound with a narrow therapeutic index (the ratio between the upper and lower limits of the therapeutic range) [12].
2.
Subjects, materials and methods
2.1.
Clinical drug interaction studies
The aspirin and atenolol studies were conducted at MDS Pharma Services, Neptune, New Jersey, USA; the enalapril and sitagliptin studies were conducted at MDS Pharma Services, Lincoln, Nebraska, USA; the phenytoin study was conducted at the Clinical Pharmacology Unit, Manipal AcuNova Kasturba Hospital Clinical Research Centre, Manipal, India; and the rosiglitazone study was conducted at Celerion [formerly MDS Pharma Services], Lincoln, Nebraska, USA. Each study enrolled healthy men and women aged 18–45 years with a body mass index of 18.0–30.0 kg/m2. Exclusion criteria varied between the studies, but the following were generally excluded: a history and/or presence of bowel obstruction (or being at risk for bowel obstruction), pancreatitis, gastrointestinal disorders within 4 weeks prior to the first dose; use of any drugs or substances known to be strong inducers or inhibitors of cytochrome P450 enzymes within 30 days prior to the first dose; and consuming or not agreeing to abstain from grapefruit/grapefruit juice from 10 days before the first dose and throughout confinement. Study details are summarized in Table 1. The dose of colesevelam for coadministration with the test drug in each study was 3750 mg (6 625 mg tablets) once daily until the plasma sampling for the test drug was completed. A quantitative structure property relationship (QSPR) model [13] based on the lipophilicity of the coadministered drug was used to predict the in vivo binding potential of colesevelam with the test drugs of interest. A two-period crossover study design was utilized in studies for the test drugs predicted by QSPR modeling to have a low potential for binding to colesevelam (aspirin [predicted binding 1.50%], atenolol [1.50%], enalapril [6.45%], rosiglitazone [16.8%], and sitagliptin
Table 1 – Study details. Test drug
Dose (mg)
Aspirin
650
Atenolol
100
Enalapril
40
Rosiglitazone
8
Sitagliptin
100
Phenytoin
300
Treatments Aspirin alone (reference) Aspirin simultaneously with colesevelam Atenolol alone (reference) Atenolol simultaneously with colesevelam Enalapril alone (reference) Enalapril simultaneously with colesevelam Rosiglitazone alone (reference) Rosiglitazone simultaneously with colesevelam Sitagliptin alone (reference) Sitagliptin simultaneously with colesevelam Phenytoin alone (reference) Phenytoin simultaneously with colesevelam Phenytoin 4 h before colesevelam
Participants enrolled, n
Participants completed, n
Pharmacokinetic sampling times
28
28
32
31
40
35
30
30
Predose and 0.08, 0.17, 0.33, 0.5, 0.75, 1.0, 1.25, 1.5, 2, 3, 4, 6, 8, 10, 12, and 24 h postdose Predose and 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 12, 24, 30, and 36 h postdose Predose and 0.167, 0.333, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 12, 24, 36, 48, 72, 96, 120, and 144 h postdose Predose and 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 9, 12, 16, and 24 h postdose
28
27
Predose and 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 24, 48, and 72 h postdose
24
24
Predose and 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 18, 24, 36, 48, 60, 72, 96, and 120 h postdose
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9 diabetes research and clinical practice xxx (2014) xxx–xxx
[1.50%]), with test drug given alone and test drug coadministered with daily colesevelam. If the QSPR model suggested that a drug had moderate or high potential to bind to colesevelam, an additional treatment period was added to evaluate the effects of the test drug given 4 h prior to colesevelam. This was the case for phenytoin (predicted binding 82.8%) [13], and therefore a three-period randomized crossover study design was utilized (i.e., phenytoin alone, phenytoin simultaneously with colesevelam, and phenytoin 4 h before colesevelam). Plasma samples for pharmacokinetic analysis were collected just prior to the first dose of medication and up to 144 h post-dose, depending on the reported half-life of the test drug. Since colesevelam is not absorbed systemically, plasma concentrations of colesevelam were not measured. For each study, the study protocol, informed consent form, and other study documents were approved by the relevant Institutional Review Board or Institutional Ethics Committee. Each study was conducted in compliance with the ethical principles that have their origin in the Declaration of Helsinki, International Conference on Harmonization Guideline E6 for Good Clinical Practice, and applicable regulatory requirement(s). Written informed consent was obtained from all participants before the start of any studyspecific procedures.
2.2. Analytical methods and concentration determinations Plasma samples were analyzed at PPD, LP, Richmond, Virginia, USA (aspirin [measured analytes were salicylic acid and acetylsalicylic acid]); MDS Pharma Services, Lincoln, Nebraska, USA (atenolol, enalapril [measured analytes enalapril and enalaprilat], and sitagliptin); Celerion [formerly MDS Pharma Services], Lincoln, Nebraska, USA (rosiglitazone); and Manipal AcuNova Kasturba Hospital Clinical Research Centre, Manipal, India (phenytoin). Plasma drug concentrations for all study drugs were determined by liquid chromatography– tandem mass spectrometry (LC–MS/MS) assays, which were validated in accordance with Guidance For Industry – Bioanalytical Method Validation (Food and Drug Administration, Center for Drug Evaluation and Research, May 2001). For all assay methods, a 1/concentration2 weighted linear regression model was used to calculate slope, intercept, and correlation coefficient of the calibration curve. Back-calculated results for study samples and quality control samples were then obtained by fitting the peak area ratio (analyte vs internal standard) to the regression equation constructed from calibration standards.
2.2.1.
Aspirin concentration determination
Reference standards were the analytes acetylsalicylic acid and salicylic acid (USP grade) and the internal standards 2acetoxybenzoic-3,4,5,6-d4 acid and 2-hydroxybenzoic-3,4,5,6d4 acid (C/D/N Isotopes, Pointe-Claire, Que´bec, Canada). A 50.0 mL human plasma (potassium oxalate/sodium fluoride) sample aliquot was fortified with 250 mL of internal standard working solution. Analytes were isolated through protein precipitation using acetonitrile. The final extract was analyzed via high-performance liquid chromatography (HPLC) with MS/ MS detection. The calibration curves ranged from 0.0200 to
3
10.0 mg/mL for acetylsalicylic acid and from 0.100 to 50.0 mg/ mL for salicylic acid.
2.2.2.
Atenolol concentration determination
Reference standards were the analyte atenolol (Cerilliant Corporation, Round Rock, TX, USA) and the internal standard d7-atenolol (C/D/N Isotopes, Pointe-Claire, Que´bec, Canada). A 0.100 mL aliquot of human plasma (K3EDTA) containing the analyte and internal standard was extracted using a liquid– liquid extraction procedure. The extracted samples were analyzed by an HPLC machine equipped with an ABjMDS Sciex API 4000 mass spectrometer. Positive ions were monitored in the multiple reaction monitoring (MRM) mode. The calibration curve ranged from 5.00 to 1500 ng/mL for atenolol.
2.2.3.
Enalapril concentration determination
Reference standards were enalapril maleate and enalaprilat (USP grade), and the corresponding internal standards, d5enalapril maleate (phenyl-d5) and d5-enalaprilat (phenyl-d5) (C/D/N isotopes), respectively. The analytes measured were enalapril and enalaprilat. A 0.200 mL aliquot of human plasma (K3EDTA) containing each analyte and internal standard was extracted using a solid phase extraction procedure. The extracted samples were analyzed by an HPLC equipped with an ABjMDS Sciex API 4000 mass spectrometer. Positive ions were monitored in the MRM mode. The calibration curves ranged from 0.500 to 100 ng/mL for enalapril and from 0.250 to 50.0 ng/mL for enalaprilat.
2.2.4.
Rosiglitazone concentration determination
Reference standards were the analyte rosiglitazone and the internal standard d4-rosiglitazone (Celerion). A 0.050 mL aliquot of human plasma (K3EDTA) containing the analyte and internal standard was extracted using a liquid–liquid extraction procedure. The extracted samples were analyzed by an HPLC equipped with an ABjMDS Sciex API 4000 mass spectrometer. Positive ions were monitored in the MRM mode. The calibration curve ranged from 2.5 to 500 ng/mL for rosiglitazone.
2.2.5.
Sitagliptin concentration determination
Reference standards were sitagliptin phosphate and the internal standard d4-sitagliptin phosphate (Toronto Research Chemicals, Inc., North York, Ontario, Canada). Sitagliptin was the analyte measured. A 0.100 mL aliquot of human plasma (K3EDTA) containing the analyte with added internal standard was extracted using a liquid–liquid extraction procedure. The extracted samples were analyzed by an HPLC equipped with an ABjMDS Sciex API 4000 mass spectrometer. Positive ions were monitored in the MRM mode. The calibration curve ranged from 2.5 to 500 ng/mL for sitagliptin.
2.2.6.
Phenytoin concentration determination
Reference standards were the analyte phenytoin (USP grade) and the internal standard phenytoin-D10 (Toronto Research Chemicals, Inc.). A 0.400 mL aliquot of human plasma (K2EDTA) containing the analyte and internal standard was extracted using a liquid–liquid extraction procedure. The samples were injected into a liquid chromatograph equipped
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9
4
diabetes research and clinical practice xxx (2014) xxx–xxx
with a tandem mass spectrometry detector. The calibration curve ranged from 20.03 to 10,015.00 ng/mL for phenytoin.
2.3.
Pharmacokinetic methods and endpoints
Pharmacokinetic parameters calculated in each study included maximum plasma concentration (Cmax), area under the concentration–time curve (AUC) from the time of dosing to the last measurable concentration (AUClast), AUC from the time of dosing extrapolated to infinity (AUC01), AUCextr (the percentage of AUC01 that is due to extrapolation from time to last measureable plasma drug concentration [tlast] to infinity), time of maximum plasma concentration (tmax), terminal halflife (t½), apparent terminal elimination rate constant (lZ), apparent total body clearance (CL/F), and apparent volume of distribution (V/F). The last measurable concentration in plasma (Clast) was calculated for atenolol, enalapril, rosiglitazone, and sitagliptin only. Parameters were calculated from plasma concentration using actual sampling times by noncompartmental methods. Calculations were performed using WinNonlin1 Professional (Pharsight, St Louis, Missouri, USA) version 5.0.1 for the phenytoin study and version 5.2 for the other studies. The primary endpoints were AUClast and AUC01 of salicylic acid for the aspirin study, and AUClast, AUC01, and Cmax for the other studies. Secondary endpoints included Clast, V/F, CL/F, tmax, and t½.
2.4.
Statistical methods
Pharmacokinetic parameters for each study were summarized using descriptive statistics. For the parameters of AUClast, AUC01, and Cmax, a mixed effect analysis of variance (ANOVA) model was used on the natural log-transformed data for statistical comparison between treatments, with the fixed effects of period, treatment, sequence, and the random effect of subject nested within sequence. The geometric mean ratio for the Test versus Reference (monotherapy) and corresponding 90% confidence interval (CI) were calculated by exponentiation of the difference in the least-square means (LSMs; on natural log-transformed data) between the treatments and the corresponding 90% CI for the difference between the LSMs calculated for the natural log-transformed values. Nonparametric analysis of the secondary endpoints of tmax and t½ was performed using the Hodges Lehmann estimator of the median of the differences between treatments with 90% CIs generated by the Moses method. For the primary endpoints, the ratio of geometric means derived from the ANOVA models of AUClast, AUC01, and Cmax were determined with two-sided 90% CIs to compare between treatments. For the aspirin study, no clinically significant interaction was concluded if the 90% CIs for the ratios of geometric LSM for AUClast and AUC01 values for salicylic acid were entirely contained within the range of 70–143%, based on the 32% decrease in bioavailability required to detect a clinically meaningful 10% decrease in serum thromboxane inhibition (from the correlation of serum thromboxane inhibition to an 81 mg dose of aspirin, the recommended dose for cardioprotection [14]). For the other studies, no statistically significant drug interaction was concluded if the 90% CIs for the ratios of geometric LSM for AUClast, AUC01,
and Cmax values for the measured analytes were entirely contained within the range of 80–125%. Sample sizes were calculated using the 80–125% bioequivalence limits for the 90% CIs for geometric mean ratios of AUC and Cmax, assuming an expected geometric mean ratio of 0.95 for the aspirin, atenolol, enalapril, and rosiglitazone studies, 0.94 for the phenytoin study, and 0.90 for the sitagliptin study. For the aspirin study, based on the estimated intra-subject coefficient of variation (CV) of 0.16 for salicylic acid AUC [15], 24 completed subjects would have provided more than 90% power to conclude bioequivalence for salicylic acid exposure; to ensure completion of 24 subjects, 28 subjects (14 per sequence) were enrolled. For the atenolol study, based on estimated intra-subject CVs of 21% and 20% for atenolol Cmax and AUC, respectively [16], 28 completed subjects would have provided at least 90% power to conclude no significant drug interaction; to ensure completion of 28 subjects, 32 subjects (16 per sequence) were enrolled. For the enalapril study, based on estimated intra-subject CVs of 25% and 23% for enalaprilat Cmax and AUC, respectively [17], 34 completed subjects would have provided at least 85% power to conclude no significant drug interaction; to ensure completion of 34 subjects, 40 subjects (20 per sequence) were enrolled. For the rosiglitazone study, based on estimated intra-subject CVs of 22% and 14% for rosiglitazone Cmax and AUC, respectively [18], 26 completed subjects would have provided at least 85% power to conclude no significant drug interaction; to ensure completion of 26 subjects, 30 subjects (15 subjects per sequence) were enrolled. For the sitagliptin study, based on estimated intra-subject CVs of 13% and 8% for sitagliptin Cmax and AUC, respectively [19], 24 completed subjects would have provided at least 90% power to conclude no significant drug interaction; to ensure completion of 24 subjects, 28 subjects (14 subjects per sequence) were enrolled. For the phenytoin study, based on the estimated intra-subject CV of 0.16 for phenytoin Cmax, 20 completed subjects would provide 90% power to conclude no significant drug interaction; to ensure completion of 20 subjects, 24 subjects (4 subjects per sequence for 6 sequences) were enrolled.
3.
Results
The majority of subjects in each study were male, and mean age ranged from 27.4 to 34.2 years (Supplemental Table S1). Supplementary material related to this article can be found, in the online version, at http://dx.doi.org/10.1016/ j.diabres.2013.12.033.
3.1.
Aspirin
The salicylic acid and acetylsalicylic acid concentration–time curves following administration of aspirin alone and simultaneously with colesevelam are shown in Fig. 1A. The ratios of geometric LSMs and their 90% CIs for salicylic acid AUClast (ratio 98.51%; 90% CI 94.86–102.30) and AUC01 (ratio 97.15%; 90% CI 93.88–100.52) values were entirely contained within the prespecified 70–143% limits (Table 2), indicating no interaction. The corresponding values for acetylsalicylic acid also fell within this range. Median tmax of salicylic acid and
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9 diabetes research and clinical practice xxx (2014) xxx–xxx
5
Fig. 1 – Concentration–time curves for salicylic acid and acetylsalicylic acid (following administration of aspirin) (A), atenolol (B), enalapril and enalaprilat (following administration of enalapril) (C), rosiglitazone (D), sitagliptin (E), and phenytoin (F).
acetylsalicylic acid were shortened by simultaneous administration of colesevelam with aspirin (Table 3).
3.2.
Atenolol
The atenolol concentration–time curves following administration of atenolol alone and simultaneously with colesevelam are shown in Fig. 1B. The ratios of geometric LSMs and their
90% CIs for atenolol AUClast (ratio 95.47%; 90% CI 88.76–102.68), AUC01 (ratio 101.14%; 90% CI 94.91–107.78), and Cmax (ratio 93.57%; 90% CI 85.28–102.67) values were entirely contained within the prespecified 80–125% limits (Table 2), indicating no interaction. Median t½ was longer following simultaneous administration of atenolol and colesevelam than with administration of atenolol alone (14.606 vs 10.089 h; Table 3). Although apparent t½ was prolonged, the accumulation
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9
6
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 2 – Primary pharmacokinetic results of drug–drug interaction studies. Drug/Analyte
Parameter
Aspirin Salicylic acida
AUClast (mg.h/mL)
240.3
236.7
AUC01 (mg.h/mL)
245.7
238.7
Reference LSM
Cmax (mg/mL) Acetylsalicylic acid
Atenolol
Enalapril Enalaprila
Enalaprilat
7.313
7.237
AUC01 (mg.h/mL)
7.853
7.576
Cmax (mg/mL)
3.268
3.829
AUClast (ng.h/mL)
4211
4020
AUC01 (ng.h/mL)
4721
4775
Cmax (ng/mL)
433.1
405.2
AUClast (ng.h/mL)
297.2
279.3
AUC01 (ng.h/mL)
302.1
282.4
Cmax (ng/mL)
147.1
139.6
AUClast (ng.h/mL)
1036
AUC01 (ng.h/mL)
1068
101.9
AUC01 (ng.h/mL)
2994
2978
501.6
500.2
AUClast (ng.h/mL)
2911
2907
AUC01 (ng.h/mL)
3000
2991
325.8
307.5
AUClast (ng.h/mL)
181,630.8
168,374.4
AUC01 (ng.h/mL)
183,045.5
170,583.1
5739.5
5147.9
AUClast (ng.h/mL)
181,630.8
177,010.3
AUC01 (ng.h/mL)
183,045.5
179,660.0
5739.5
5224.9
Cmax (ng/mL) a
104.1
2888
Cmax (ng/mL) 4 h before colesevelam
1017
2929
Cmax (ng/mL) Phenytoin Simultaneous with colesevelam
977.5
AUClast (ng.h/mL)
Cmax (ng/mL) Sitagliptin
26.68
AUClast (mg.h/mL)
Cmax (ng/mL) Rosiglitazone
28.24
Test LSM
Ratio of test/ reference, % (90% CI) 98.51 (94.86, 102.30) 97.15 (93.88, 100.52) 94.48 (88.19, 101.21) 98.95 (94.88, 103.19) 96.47 (91.47, 101.74) 117.18 (100.84, 136.16) 95.47 (88.76, 102.68) 101.14 (94.91, 107.78) 93.57 (85.28, 102.67) 93.97 (89.89, 93.50 (89.37, 94.88 (86.83, 94.39 (89.87, 95.24 (90.47, 97.90 (91.82, 98.61 (95.59, 99.47 (96.61, 99.72 (91.05, 99.85 (98.30, 99.70 (98.20, 94.38 (88.45, 92.7 (87.8, 93.2 (88.1, 89.7 (84.6, 97.5 (92.3, 98.1 (92.8, 91.0 (85.8,
98.23) 97.83) 103.67) 99.13) 100.27) 104.39) 101.74) 102.42) 109.21) 101.42) 101.23) 100.70)
97.8) 98.6) 95.1) 102.9) 103.8) 96.6)
Primary analyte.
factors (based on a 24-h dosing interval) were similar with administration of atenolol alone and simultaneously with colesevelam (1.233 and 1.293, respectively), indicating only a small change in the effective t½.
3.3.
Enalapril
The enalapril and enalaprilat concentration–time curves following administration of enalapril alone and simultaneously
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9
7
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 3 – Secondary pharmacokinetic results of drug–drug interaction studies. Drug/analyte Aspirin Salicylic acida Acetylsalicylic acid Atenolol Enalapril Enalaprila Enalaprilat Rosiglitazone Sitagliptin Phenytoin Simultaneous with colesevelam 4 h before colesevelam a
Parameter
Median reference value
Median test value
tmax (h) t½ (h) tmax (h) t½ (h) tmax (h) t½ (h)
4.000 2.744 2.000 0.542 1.500 10.089
3.000 2.677 1.134 0.893 1.500 14.606
tmax (h) t½ (h) tmax (h) t½ (h) tmax (h) t½ (h) tmax (h) t½ (h)
1.500 1.969 3.983 59.907 0.500 4.138 1.525 12.391
1.500 1.584 4.000 58.090 0.500 4.682 1.000 12.071
tmax (h) t½ (h) tmax (h) t½ (h)
8.0 13.1 8.0 13.1
8.0 12.9 7.0 13.2
Primary analyte.
with colesevelam are shown in Fig. 1C. The ratios of geometric LSMs and their 90% CIs for enalapril AUClast (ratio 93.97%; 90% CI 89.89–98.23), AUC01 (ratio 93.50%; 90% CI 89.37–97.83), and Cmax (ratio 94.88%; 90% CI 86.83–103.67) values were entirely contained within the prespecified 80–125% limits (Table 2), indicating no interaction. The corresponding values for enalaprilat also fell within this range. Following simultaneous administration of colesevelam with enalapril, median t½ of the prodrug enalapril was shorter (Table 3); however, primary and secondary pharmacokinetic parameters of the active moiety enalaprilat showed little change.
3.4.
Rosiglitazone
The rosiglitazone concentration–time curves following administration of rosiglitazone alone and simultaneously with colesevelam are shown in Fig. 1D. The ratios of geometric LSMs and their 90% CIs for rosiglitazone AUClast (ratio 98.61%; 90% CI 95.59–101.74), AUC01 (ratio 99.47%; 90% CI 96.61– 102.42), and Cmax (ratio 99.72%; 90% CI 91.05–109.21) values were entirely contained within the prespecified 80–125% limits (Table 2), indicating no interaction. Median t½ was longer with simultaneous administration of colesevelam with rosiglitazone (Table 3). Although apparent t½ was prolonged, the accumulation factors (based on a 24-h dosing interval) were similar with administration of rosiglitazone alone and simultaneously with colesevelam (1.039 and 1.047, respectively), indicating only a small change in the effective t½.
3.5.
Sitagliptin
The sitagliptin concentration–time curves following administration of sitagliptin alone and simultaneously with colesevelam are shown in Fig. 1E. The ratios of geometric LSMs and their 90% CIs for sitagliptin AUClast (ratio 99.85%; 90% CI 98.30– 101.42), AUC01 (ratio 99.70%; 90% CI 98.20–101.23), and Cmax (ratio 94.38%; 90% CI 88.45–100.70) values were entirely
contained within the prespecified 80–125% limits (Table 2), indicating no interaction.
3.6.
Phenytoin
The phenytoin concentration–time curves following administration of phenytoin alone, simultaneously with colesevelam, and 4 h before colesevelam are shown in Fig. 1F. When phenytoin was administered simultaneously with colesevelam, the ratios of geometric LSMs and their 90% CIs for phenytoin AUClast (ratio 92.7%; 90% CI 87.8–97.8), AUC01 (ratio 93.2%; 90% CI 88.1–98.6), and Cmax (ratio 89.7%; 90% CI 84.6–95.1) values were entirely contained within the prespecified 80–125% limits, indicating no interaction (Table 2). The corresponding values when phenytoin was administered 4 h before colesevelam also fell within this range (Table 2).
4.
Discussion
In this series of single-dose clinical studies performed to determine the effects of colesevelam on the pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin, no pharmacokinetic changes meeting the prespecified criteria for a drug–drug interaction were observed. The results of the current studies add to the existing body of study data supporting the favorable drug–drug interaction profile of colesevelam [20]. Fewer medications have demonstrated potential drug–drug interactions with colesevelam than with cholestyramine (Table 4) [2,21–37]. Previous studies have demonstrated an absence of clinically significant drug–drug interactions with concomitant administration of colesevelam with fenofibrate [38], pioglitazone [37], lovastatin [39], digoxin, metoprolol, quinidine, valproate, warfarin, and verapamil [40]. The current study utilized the results of QSPR modeling to predict the likelihood of observing drug–drug interactions, and to modify the design of the studies accordingly. Predicted
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9
8
diabetes research and clinical practice xxx (2014) xxx–xxx
Table 4 – Medications associated with potential drug– drug interactions with the bile acid sequestrants cholestyramine [21–36] and colesevelam [2,37], based on in vitro binding or in vivo interaction studies. Cholestyramine Digitalis/digoxin Estrogens and progestins Glipizide Hydrocortisone Penicillin G Phenobarbital Phenylbutazone Propranolol Rosiglitazone Simvastatin Tetracycline Thiazide diuretics Thyroxine Tricyclic antidepressants Valproic acid Warfarin Other drugs that undergo enterohepatic circulation (e.g., naproxen, phenprocoumon, sulindac) a
Colesevelam Cyclosporine Ethinyl estradiol and norethindrone Glimepiride Glipizide Glyburide Levothyroxine Metformin ERa (increases exposure) Olmesartan medoxomil
Extended-release.
binding was 82.8% for phenytoin and 1.50–16.8% for the other tested drugs [13]. In general, our observed data were consistent with the predictions of the QSPR modeling, suggesting that this may be a useful tool to screen for the potential for drug– drug interactions. The QSPR model is based on in vitro and physicochemical properties of the medications and does not include in vivo factors. Therefore, this model suggests which compounds show low risk for interaction with colesevelam and which may be at higher risk for interaction. Although our pharmacokinetic study has shown that the peak and total exposure of phenytoin is not altered when administered with colesevelam, concomitant use of phenytoin and colesevelam should remain in accordance with United States prescribing information for colesevelam. At present, the prescribing information indicates that phenytoin should be taken at least 4 h before colesevelam, based on the narrow therapeutic index of phenytoin and postmarketing reports consistent with potential interactions [2,12].
5.
Conclusions
Aspirin, atenolol, enalapril, rosiglitazone, and sitagliptin may be taken together with colesevelam. Although no pharmacokinetic-based drug–drug interaction between phenytoin and colesevelam is apparent, concomitant use of phenytoin and colesevelam should remain in accordance with colesevelam United States prescribing information, with phenytoin administered at least 4 h before colesevelam.
Author contributions P.W., L.H., J.M-H., J.L., B.T., D.W., and J.W. designed the studies, analyzed the data and contributed to discussions. L.H., D.W. and
P.W. conducted the studies. L.H. provided the assay support for the studies. L.H., J.L., B.T., J.M-H., and J.W. are employees of Daiichi Sankyo Pharma Development, the sponsor of these DDI studies, and P.W. was an employee of Daiichi Sankyo Pharma Development at the time that the studies were conducted. D.W. was an employee of MDS Pharma Services or Celerion Inc. at the time that these studies were conducted.
Conflicts of interest None.
Acknowledgments The studies were supported by Daiichi Sankyo, Inc. Robert Schupp, PharmD, and Sushma Soni of inScience Communications, Springer Healthcare, provided medical writing support funded by Daiichi Sankyo, Inc. The authors would like to acknowledge Richard Scheyer, MD, and Giorgio Sendali, MD, of Daiichi Sankyo Pharma Development for their participation in the studies as medical monitors; Mukesh Kumar, MBBS, MD, of Daiichi Sankyo India Development; Robert Noveck, MD, PhD, FCP, Frank Lee, MD, Srinivas Shenoy B, MBBS, MD, Satish Kumar, MBBS, MD, Daniel Weiss, MD, and Scott Rasmussen, MD, for their participation in the studies as principal investigators; Hamim Zahir, PhD, of Daiichi Sankyo Pharma Development for contributing to the study design; and SaeHeum Song, PhD, of Daiichi Sankyo Pharma Development for assistance with pharmacokinetic calculations and helpful discussion regarding interpretation of the data.
references
[1] Bays HE, Goldberg RB. The ‘forgotten’ bile acid sequestrants: is now a good time to remember? Am J Ther 2007;14:567–80. [2] Welchol [package insert]. Parsippany, New Jersey: Daiichi Sankyo, Inc.; 2012. [3] Bays HE, Dujovne CA. Drug interactions of lipid-altering drugs. Drug Saf 1998;19:355–71. [4] Braunlin W, Zhorov E, Smisek D, Guo A, Appruzese W, Xu Q, et al. In vitro comparison of bile acid binding to colesevelam HCl and other bile acid sequestrants. Polymer Prepr 2000;41:708–9. [5] Jacobson TA, Armani A, McKenney JM, Guyton JR. Safety considerations with gastrointestinally active lipid-lowering drugs. Am J Cardiol 2007;99(6A):47C–55C. [6] Corsonello A, Pedone C, Corica F, Incalzi RA, on behalf of the Gruppo Italiano di Farmacovigilanza nell’Anziano (GIFA) investigators. Polypharmacy in elderly patients at discharge from the acute care hospital. Ther Clin Risk Manag 2007;3:197–203. [7] Grant RW, Devita NG, Singer DE, Meigs JB. Polypharmacy and medication adherence in patients with type 2 diabetes. Diabetes Care 2003;26:1408–12. [8] Huang ES, Basu A, Finch M, Frytak J, Manning W. The complexity of medication regimens and test ordering for patients with diabetes from 1995 to 2003. Curr Med Res Opin 2007;23:1423–30.
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
DIAB-5944; No. of Pages 9 diabetes research and clinical practice xxx (2014) xxx–xxx
[9] American Diabetes Association. Standards of medical care in diabetes—2013. Diabetes Care 2013;36(Suppl 1):S11–66. [10] Goldberg RM, Mabee J, Chan L, Wong S. Drug–drug and drug–disease interactions in the ED: analysis of a high-risk population. Am J Emerg Med 1996;14:447–50. [11] Johnell K, Klarin I. The relationship between number of drugs and potential drug–drug interactions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed Drug Register. Drug Saf 2007;30:911–8. [12] Richens A, Dunlop A. Serum-phenytoin levels in management of epilepsy. Lancet 1975;306:247–8. [13] Walker JR, Brown K, Rohatagi S, Bathala MS, Xu C, Wickremasingha PK, et al. Quantitative structure-property relationships modeling to predict in vitro and in vivo binding of drugs to the bile sequestrant, colesevelam (Welchol). J Clin Pharmacol 2009;49:1185–95. [14] Patrignani P, Filabozzi P, Patrono C. Selective cumulative inhibition of platelet thromboxane production by low-dose aspirin in healthy subjects. J Clin Invest 1982;69:1366–72. [15] Benedek IH, Joshi AS, Pieniaszek HJ, King S-YP, Kornhauser DM. Variability in the pharmacokinetics and pharmacodynamics of low dose aspirin in healthy male volunteers. J Clin Pharmacol 1995;35:1181–6. [16] Lilja JJ, Juntti-Patinen L, Neuvonen PJ. Effect of rifampicin on the pharmacokinetics of atenolol. Basic Clin Pharmacol Toxicol 2006;98:555–8. [17] Howes LG, Conway EL, Phillips PA, Broadbear J, Drummer OH, Louis WJ. Pharmacokinetic comparison of a combination tablet of enalapril and hydrochlorothiazide with enalapril and hydrochlorothiazide tablets administered together and separately. Biopharm Drug Dispos 1991;12:447–55. [18] Freed MI, Allen A, Jorkasky DK, DiCicco RA. Systemic exposure to rosiglitazone is unaltered by food. Eur J Clin Pharmacol 1999;55:53–6. [19] Herman GA, Bergman A, Liu F, Stevens C, Wang AQ, Zeng W, et al. Pharmacokinetics and pharmacodynamic effects of the oral DPP-4 inhibitor sitagliptin in middle-aged obese subjects. J Clin Pharmacol 2006;46:876–86. [20] Florentin M, Liberopoulos EN, Mikhailidis DP, Elisaf MS. Colesevelam hydrochloride in clinical practice: a new approach in the treatment of hypercholesterolaemia. Curr Med Res Opin 2008;24:995–1009. [21] Cholestyramine for oral suspension USP [package insert]. Spring . Valley, NY: Par Pharmaceutical Companies Inc.; 2012 [22] Gallo DG, Bailey KR, Sheffner AL. The interaction between cholestyramine and drugs. Proc Soc Exp Biol Med 1965;120:60–5. [23] Ja¨hnchen E, Meinertz T, Gilfrich HJ, Kersting F, Groth U. Enhanced elimination of warfarin during treatment with cholestyramine. Br J Clin Pharmacol 1978;5:437–40. [24] Hahn KJ, Eiden W, Schettle M, Hahn M, Walter E, Weber E. Effect of cholestyramine on the gastrointestinal absorption of phenprocoumon and acetylosalicylic acid in man. Eur J Clin Pharmacol 1972;4:142–5. [25] Hunninghake DB, Hibbard DM. Influence of time intervals for cholestyramine dosing on the absorption
[26]
[27]
[28]
[29]
[30]
[31]
[32] [33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
of hydrochlorothiazide. Clin Pharmacol Ther 1986;39: 329–34. Hibbard DM, Peters JR, Hunninghake DB. Effects of cholestyramine and colestipol on the plasma concentrations of propranolol. Br J Clin Pharmacol 1984;18:337–42. Northcutt RC, Stiel JN, Hollifield JW, Stant Jr EG. The influence of cholestyramine on thyroxine absorption. JAMA 1969;208:1857–61. Brown DD, Juhl RP, Warner SL. Decreased bioavailability of digoxin due to hypocholesterolemic interventions. Circulation 1978;58:164–72. Nakai A, Nishikata M, Matsuyama K, Ichikawa M. Drug interaction between simvastatin and cholestyramine in vitro and in vivo. Biol Pharm Bull 1996;19:1231–3. Malloy MJ, Ravis WR, Pennell AT, Diskin CJ. Effect of cholestyramine resin on single dose valproate pharmacokinetics. Int J Clin Pharmacol Ther 1996;34: 208–11. Malloy MJ, Ravis WR, Pennell AT, Hagan DR, Betagari S, Doshi DH. Influence of cholestyramine resin administration on single dose sulindac pharmacokinetics. Int J Clin Pharmacol Ther 1994;32:286–9. Calvo MV, Dominguez-Gil A. Interaction of naproxen with cholestyramine. Biopharm Drug Dispos 1984;5:33–42. Johansson C, Adamsson U, Stierner U, Lindsten T. Interaction by cholestyramine on the uptake of hydrocortisone in the gastrointestinal tract. Acta Med Scand 1978;204:509–12. Muzeeb S, Venkatesh P, Mullangi R, Srinivas NR. Influence of cholestyramine on the pharmacokinetics of rosiglitazone and its metabolite, desmethylrosiglitazone, after oral and intravenous dosing of rosiglitazone: impact on oral bioavailability, absorption, and metabolic disposition in rats. Xenobiotica 2006;36:838–56. Kivisto¨ KT, Neuvonen PJ. The effect of cholestyramine and activated charcoal on glipizide absorption. Br J Clin Pharmacol 1990;30:733–6. Bailey DN, Coffee JJ, Anderson B, Manoguerra AS. Interaction of tricyclic antidepressants with cholestyramine in vitro. Ther Drug Monit 1992;14:339–42. Brown KS, Armstrong IC, Wang A, Walker JR, Noveck RJ, Swearingen D, et al. Effect of the bile acid sequestrant colesevelam on the pharmacokinetics of pioglitazone, repaglinide, estrogen estradiol, norethindrone, levothyroxine, and glyburide. J Clin Pharmacol 2010;50: 554–65. Jones MR, Baker BA, Mathew P. Effect of colesevelam HCl on single-dose fenofibrate pharmacokinetics. Clin Pharmacokinet 2004;43:943–50. Donovan JM, Kisicki JC, Stiles MR, Tracewell WG, Burke SK. Effect of colesevelam on lovastatin pharmacokinetics. Ann Pharmacother 2002;36:392–7. Donovan JM, Stypinski D, Stiles MR, Olson TA, Burke SK. Drug interactions with colesevelam hydrochloride, a novel, potent lipid-lowering agent. Cardiovasc Drugs Ther 2000;14:681–90.
Please cite this article in press as: He L, et al. Lack of effect of colesevelam HCl on the single-dose pharmacokinetics of aspirin, atenolol, enalapril, phenytoin, rosiglitazone, and sitagliptin. Diabetes Res Clin Pract (2014), http://dx.doi.org/10.1016/j.diabres.2013.12.033
9