ArticlePDF Available

Effect of drug transporter genotypes on pravastatin disposition in European- and African-American participants

Authors:

Abstract and Figures

Our aims were to evaluate the effects of polymorphisms in the hepatic drug uptake transporter organic anion transporting polypeptide 1B1 (OATP1B1, SLCO1B1) and efflux transporters multidrug resistance-associated protein 2 (MRP2, ABCC2), bile salt export pump (BSEP, ABCB11), and breast cancer-related protein (BCRP, ABCG2) on single-dose pravastatin pharmacokinetics in healthy European- and African-American participants. The pharmacokinetics of a single oral 40 mg dose of pravastatin was determined in 107 participants (69 European-Americans and 38 African-Americans). Participants were genotyped for known OATP1B1, MRP2, BSEP, and BCRP polymorphisms. Baseline serum total and unconjugated plasma bilirubin concentrations were also determined. OATP1B1 genotypes were ethnicity-dependent with a 521C allele frequency of approximately 15% in European-Americans and approximately 1% in African-Americans. SLCO1B1 521TC genotype was associated with significantly higher pravastatin area under the curve [AUC(0-5)] (P=0.01) and Cmax values (P<0.05). When analyzed by diplotype, SLCO1B1*1a/*15 (N=8) participants exhibited 45 and 80% higher AUC values than SLCO1B1*1a/*1a (N=29) (P=0.013) and SLCO1B1*1b/*1b (N=34) (P=0.001) carriers, respectively. SLCO1B1*15/*15 (N=2) participants exhibited 92 and 149% higher AUC values than SLCO1B1*1a/*1a (P=0.017) and SLCO1B1*1b/*1b (P=0.011) carriers, respectively. European-Americans had significantly higher plasma pravastatin AUC(0-5) (P=0.01) and Cmax values (P=0.009) than African-Americans. Neither ABCC2, ABCB11, nor ABCG2 genotypes were associated with differences in pravastatin pharmacokinetics. We did not observe an effect of SLCO1B1 genotype on baseline total or unconjugated bilirubin levels. SLCO1B1 genotype, in particular the 521C allele, had a significant effect on the pharmacokinetics of pravastatin. Even when adjusted for the presence of the SLCO1B1 521C or 388G variant allele, European-Americans demonstrated significantly higher pravastatin AUC and Cmax values than African-Americans.
Content may be subject to copyright.
Effect of drug transporter genotypes on pravastatin disposition
in European- and African-American participants
Richard H. Hoa, Leena Choib, Wooin Leec, Gail Mayoc, Ute I. Schwarzc,e, Rommel G.
Tironac,d,e, David G. Baileyd,e, C. Michael Steinc, and Richard B. Kimc,e
aDepartment of Pediatrics and Pharmacology, Vanderbilt University School of Medicine,
Nashville, Tennessee, USA
bDepartment of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee,
USA
cDivision of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville,
Tennessee, USA
dDepartment of Physiology and Pharmacology, Schulich School of Medicine and Dentistry,
University of Western Ontario, London, Ontario, Canada
eDivision of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and
Dentistry, University of Western Ontario, London, Ontario, Canada
Abstract
Objective—Our aims were to evaluate the effects of polymorphisms in the hepatic drug uptake
transporter organic anion transporting polypeptide 1B1 (OATP1B1, SLCO1B1) and efflux
transporters multidrug resistance-associated protein 2 (MRP2, ABCC2), bile salt export pump
(BSEP, ABCB11), and breast cancer-related protein (BCRP, ABCG2) on single-dose pravastatin
pharmacokinetics in healthy European- and African-American participants.
Methods—The pharmacokinetics of a single oral 40mg dose of pravastatin was determined in
107 participants (69 European-Americans and 38 African-Americans). Participants were
genotyped for known OATP1B1, MRP2, BSEP, and BCRP polymorphisms. Baseline serum total
and unconjugated plasma bilirubin concentrations were also determined.
Results—OATP1B1 genotypes were ethnicity-dependent with a 521C allele frequency of ~15%
in European-Americans and ~1% in African-Americans. SLCO1B1 521TC genotype was
associated with significantly higher pravastatin area under the curve [AUC(0–5)] (P =0.01) and
Cmax values (P< 0.05). When analyzed by diplotype, SLCO1B1*1a/*15 (N =8) participants
exhibited 45 and 80% higher AUC values than SLCO1B1*1a/*1a (N=29) (P=0.013) and
SLCO1B1*1b/*1b (N=34) (P=0.001) carriers, respectively. SLCO1B1*15/*15 (N=2) participants
exhibited 92 and 149% higher AUC values than SLCO1B1*1a/*1a (P=0.017) and
© 2007 Lippincott Williams & Wilkins.
Correspondence to Richard B. Kim, MD, Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine
& Dentistry, The University of Western Ontario, Room A-LL-152, LHSC-University Hospital, 339 Windermere Road, London, ON
N6A 5A5, Canada Tel: +519 663 3553; fax: +519 663 8619; Richard.Kim@LHSC.on.ca.
The authors have no conflicts of interest to disclose.
NIH Public Access
Author Manuscript
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
Published in final edited form as:
Pharmacogenet Genomics. 2007 August ; 17(8): 647–656. doi:10.1097/FPC.0b013e3280ef698f.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
SLCO1B1*1b/*1b (P= 0.011) carriers, respectively. European-Americans had significantly higher
plasma pravastatin AUC(0–5) (P =0.01) and Cmax values (P=0.009) than African-Americans.
Neither ABCC2, ABCB11, nor ABCG2 genotypes were associated with differences in pravastatin
pharmacokinetics. We did not observe an effect of SLCO1B1 genotype on baseline total or
unconjugated bilirubin levels.
Conclusion—SLCO1B1 genotype, in particular the 521C allele, had a significant effect on the
pharmacokinetics of pravastatin. Even when adjusted for the presence of the SLCO1B1 521C or
388G variant allele, European-Americans demonstrated significantly higher pravastatin AUC and
Cmax values than African-Americans.
Keywords
ABCB11; ABCC2; ABCG2; BCRP; bilirubin; BSEP; MRP2; OATP1B1; pharmacokinetics;
pravastatin; SLCO1B1; transporter
Introduction
In the liver, efficient extraction of drugs from the portal blood into hepatocytes is often
mediated by uptake transporters expressed on the sinusoidal (basolateral) membrane. For
many drugs, carrier-mediated uptake into hepatocytes may represent a critical rate-limiting
step to the overall disposition of such drugs and thus be an important component of hepatic
first-pass elimination. The organic anion transporting polypeptides (OATPs) represent an
important class of drug uptake transporters that mediate the sodium-independent transport of
a diverse range of amphipathic organic compounds including bile salts, steroid conjugates,
thyroid hormones, anionic peptides, numerous drugs and other xenobiotics [1]. Recently, the
HUGO Gene Nomenclature Committee approved a new species-independent classification
and nomenclature system based on divergent evolution that defines the SLCO-gene
superfamily through stratification into families, subfamilies and individual genes according
to evolutionary relationships and the degree of amino acid sequence identities [2] (http://
www.bioparadigms.org/slc/intro.asp).
Many OATP family members exhibit widespread tissue expression, but certain OATPs, such
as OATP1B1 (also known as OATP-C, LST1, OATP2) and OATP1B3 (also known as
OATP8, LST2), appear to exhibit an organ-selective pattern of expression, most notably in
the liver [3]. OATP1B1 is primarily expressed at the basolateral membrane of hepatocytes
and plays a key role in the hepatic disposition of structurally diverse substrates, including
bile salts, conjugated and unconjugated bilirubin, BSP, steroid conjugates, the thyroid
hormones T4 and T3, eicosanoids, cyclic peptides, and drugs such as benzylpenicillin,
methotrexate, pravastatin and rifampin [1,4].
We now know that there are a number of synonymous and nonsynonymous single
nucleotide polymorphisms (SNPs) in the coding region of the SLCO1B1 gene [5–8].
Moreover, the presence or frequency of these SNPs appears to be ethnicity-dependent. In-
vitro experiments in a number of cell-based assays demonstrated that certain SLCO1B1
variants exhibit significantly impaired transport activity for prototypical substrates such as
Ho et al. Page 2
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
estrone sulfate and estradiol-17β-glucuronide [5]. Decreased activity, in part, could be
attributed to decreased cell surface expression.
Pravastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase
(statins), is a drug widely used to treat hypercholesterolemia [9]. Owing to its hydrophilicity,
active transport mechanisms are thought to be important for its hepatic disposition.
Subsequently, pravastatin was shown to be a substrate for OATP1B1, and transport by this
mechanism is thought to be the rate-limiting step in pravastatin hepatic clearance [10,11].
Previous studies have demonstrated substantial interindividual variation (6-fold) in
pravastatin pharmacokinetic parameters, including area under the curve (AUC) and the
maximum concentration (Cmax) but not half-life, in healthy participants [9, 12]. More
recently, clinical studies have confirmed that variation in pravastatin pharmacokinetics can
be attributed in part to the presence of SLCO1B1 polymorphisms [7, 13, 14]. Specifically,
studies have demonstrated that individuals carrying the 521C allele either in combination
with the 388A allele (SLCO1B1*5) or the 388G allele (SLCO1B1*15) exhibited
significantly higher mean pravastatin AUC values compared with individuals carrying
reference alleles SLCO1B1*1a or *1b, consistent with in-vitro data indicating markedly
decreased transport function for the 521C variant.
In addition to OATP1B1, statins are substrates for efflux transporters including multidrug
resistance-associated protein 2 (MRP2; ABCC2) [15], breast cancer resistance protein
(BCRP; ABCG2) [16] and the bile salt export pump (BSEP; ABCB11) [17], all of which
have been localized to the canalicular membrane of hepatocytes [18]. Furthermore, MRP2
and BCRP expression has also been localized to the apical membrane of intestinal
enterocytes [18]. Therefore genetic variation in efflux transporters may be important
determinants of the oral bioavailability of statins. Polymorphisms have been identified in the
ABCC2, ABCG2 and ABCB11 genes, but there is a paucity of data regarding their
functional significance [19–23] (http://www.pharmgkb.org). Therefore, in addition to the
contribution of polymorphisms in SLCO1B1, it is plausible to hypothesize that functionally
relevant polymorphisms in these efflux transporters are also important determinants of
pravastatin disposition.
In this study, we assessed the relationship between genotype and phenotype of pravastatin
pharmacokinetics in a large number of European- and African-American participants to
effectively evaluate the functional significance of drug transporter polymorphisms on the in-
vivo disposition of pravastatin. We also describe the influence of ethnicity on pravastatin
pharmacokinetics among European- and African-Americans. Our findings suggest that
genetic variation in SLCO1B1 results in higher systemic exposure to pravastatin, whereas
known SNPs in the efflux transporters studied do not have a major influence. Importantly,
we report that plasma concentrations of pravastatin differ significantly between European-
versus African-American participants, suggesting that as yet undefined environmental or
genetic factors play additional roles in determining the interethnic variation in the
disposition of pravastatin.
Ho et al. Page 3
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Methods
Participants
One hundred and seven healthy, unrelated nonsmoking adult participants (69 European-
Americans and 38 African-Americans; age, 19–55 years; weight, 47.7–109 kg) were
enrolled in the study. Mean body surface area (BSA), weight and height was 1.96 m2, 77.3
kg and 1.81m for European-American men (n = 37) and 2.05 m2, 84.6 kg and 1.81m for
African-American men (n = 19), respectively. Mean BSA, weight and height was 1.70 m2,
63.3 kg and 1.64m for European-American women (n = 32) and 1.75 m2, 67 kg and 1.65m
for African-American women (n = 19), respectively. The health status of each individual
was confirmed by history and physical examination, including heart rate and blood pressure
measurements, and laboratory tests including a complete blood count and blood chemistry
screening including direct (conjugated) and total bilirubin values. After confirmation of
medical history and normal physical examination, participants were instructed to ingest no
drugs for at least 1 week before their assigned study day. The study protocol was approved
by the Institutional Review Board and General Clinical Research Center at Vanderbilt
University Medical Center, and participants provided written informed consent before study
enrollment.
Study design
The study was performed in the General Clinical Research Center at Vanderbilt University
Medical Center. After an overnight fast, each study participant received a single oral dose of
40 mg pravastatin (Bristol-Myers-Squibb, New York, USA) with water. An indwelling
venous cannula was placed and serial heparinized blood samples (10 ml) were obtained
immediately before and at 1, 2, 3, 4, 5, 6, 8, and 12 h after pravastatin administration.
Plasma was separated by centrifugation and immediately stored at −20°C until quantitative
analysis was performed.
Genotyping
During the screening visit, ~30 ml venous blood was obtained for pharmacogenetic
analyses. Genomic DNA was obtained from peripheral blood mononuclear cells via a
standard DNA extraction protocol (QIAmp DNA Mini Kit, Qiagen, Valencia, California,
USA) and samples were stored at − 20°C until genotyping was performed. Samples were
genotyped for previously reported SLCO1B1 polymorphisms (*1b – 388G, *5 – 521C, and
*9 – 1463C) as well as commonly occurring nonsynonymous polymorphisms in the efflux
transporters ABCC2 (1249A and 3563A), ABCG2 (34A and 421A) and ABCB11 (1331C
and 2029G). Genotyping was performed using a validated 5nuclease PCR-based assay with
allele specific fluorescent probes (TaqMan SNP Genotyping Assays, Applied Biosystems,
Foster City, California, USA) [24]. Fluorescent probes for each SNP were designed through
Applied Biosystems from reference sequences (Gen-Bank) for SLCO1B1, ABCC2,
ABCG2, and ABCB11 and are as follows: SLCO1B1 388G, SLCO1B1 521C, SLCO1B1
1463C, ABCC2 1249A, ABCC2 3563A, ABCG2 34A, ABCG2 421A, ABCB11 1331C, and
ABCB11 2029G. PCRs were performed on an Applied Biosystems Gene-Amp 9700 PCR
System (ABI, Foster City, California). Each 50μl reaction contained 100 ng genomic DNA,
10mmol/l Tris-HCl (pH 8.3), 50mmol/l KCl, 4mmol/l MgCl2, 200 μmol/l each dNTP, 200
Ho et al. Page 4
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
nmol/l each primer, 50 nmol/l each fluorescent-tagged probe, 0.5 U AmpErase uracil N-
glycosylase (Perkin-Elmer, Waltham, Massachusetts), and 1.25U AmpliTaq DNA
polymerase (Perkin-Elmer). Cycling conditions were 50°C, 2 min; 95°C, 10 min; 40 cycles
of 95°C, 15 s; 64°C, 1 min. Each individual TaqMan probe was validated by assaying a
sequenced-verified DNA standard for each wild-type and variant allele tested. Following
amplification, 40 μl of each reaction was transferred to a microtiter plate (Perkin-Elmer) and
fluorescence measured using the ABI Prism 7900HT Sequence Detection System. SNPs
were noted and grouped into cluster plots with software package SDS version 2.2 Enterprise
Edition Software Suite. OATP1B1 haplotypes were inferred using an algorithm based on
Bayesian inference with the statistical software package PHASE (http://
www.stat.washington.edu/stephens/software.html) [25,26] and named according to current
nomenclature conventions for SLCO1B1 haplotype [5,7].
Drug analysis and pharmacokinetics
A 100 mg C18 1ml capacity preparatory solid-phase extraction column (Bakerbond, JT
Baker, Phillipsburg, New Jersey, USA) was initially conditioned with 2 ml double-distilled
water. Aliquots of 1000 μl of plasma and 50 μl of internal standard 5-methoxypsoralen
(Indofine Chemical Company Inc., Somerville, New Jersey, USA) at 1000 ng/ml in
methanol/water (50: 50, v/v) were added and the column was washed with 3 ml of methanol/
water (10: 90, v/v) containing 0.05% acetic acid and then 3 ml of methanol/water (10: 90,
v/v) containing 0.05% ammonium hydroxide. The sample was eluted with 1 ml methanol
and was evaporated to dryness at 40°C under a gentle stream of nitrogen. The recovery of
pravastatin and 5-methoxypsoralen was complete. The residue was reconstituted with an
aliquot (50 μl) of methanol/water (50: 50, v/v) and centrifuged at 8000g for 3 min. An
aliquot (20 μl) of the supernatant was injected onto a Zorbax SB C18, 5μm particle size, 3.0
× 150 mm2 column (Agilent, VWR International Ltd, Mississauga, Ontario, Canada) that
was attached to a Hewlett Packard 1090 HPLC (high-pressure liquid chromatography)
system equipped with a Hewlett Packard 1050 variable wavelength ultraviolet detector. The
effluent was monitored at an absorbance wavelength of 238 nm. The HPLC mobile phase in
Pump A was acetonitrile/water (28: 72, v/v) containing 0.08% triethylamine at pH 3.0 with
10 mmol/l potassium phosphate buffer and in Pump B was acetonitrile/water (70: 30, v/v)
containing 0.08% triethylamine at pH 3.0 with 10mmol/l potassium phosphate buffer at a
flow rate of 0.5 ml/min. The timetable was 100% of mobile phase in Pump A from 0 to 18
min and 100% mobile phase in Pump B from 18 to 21 min. The retention times of
pravastatin and 5-methoxypsoralin were 10.4 and 17.4 min, respectively. The standard curve
of pravastatin was linear over the range tested. The intra-assay coefficient of variation (CV)
was 5% at 5 ng/ml (n = 5) and the limit of detection was 1 ng/ml. The inter-assay CV was
18% at 50 ng/ml (n = 27) and 11% at 100 ng/ml (n = 19). The sensitivity of the assay was
improved from a limit of detection 5–1 ng/ml after 73 participants had been studied. Assay
sensitivity accounted for 8% of variability in pravastatin AUC and was accounted for in the
statistical analysis but did not affect the overall results of the study.
Pharmacokinetic parameters of pravastatin were calculated using the time points 0 h before
pravastatin administration and 1, 2, 3, 4, and 5 h after pravastatin intake as the majority of
pravastatin concentrations at 6, 8, and 12 h were below the detection limits of our assay.
Ho et al. Page 5
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Pharmacokinetic analysis of pravastatin included peak concentration in plasma (Cmax) and
area under the plasma concentration vs. time curve from 0 to 5 h (AUC0–5). AUC0–5 values
were calculated by linear trapezoidal method.
Statistical analysis
All data are presented as mean ± SD for continuous variables and the number of participants
or proportion for categorical variables. The associations between each gene and ethnicity
were examined using Pearson χ2 test. Multiple regression models were used to examine the
association between outcomes and genotypes for each gene after adjustment for potential
confounding factors. The major outcomes of interest were AUC and the Cmax as
pharmacokinetic (PK) parameters, and total bilirubin and unconjugated bilirubin. Although
total bilirubin concentrations were measured for all participants, the conjugated bilirubin
concentrations were available for only 23 participants while 72 participants had partial
information only (conjugated bilirubin concentration < 0.1 owing to lower limits of assay
sensitivity). As a supplementary measure, the conjugated bilirubin concentrations were
imputed for the 12 missing values and the 72 values having partial information.
Theoretically, the fraction of the conjugated bilirubin is known to be 5% of the total
bilirubin [27]. To take into account uncertainty and measurement errors, the imputed values
were generated from log normal distribution with mean of 5% of the total bilirubin and SD
of 0.3 (assuming that the coefficient of variance for measurements is approximately 30%).
Then, the unconjugated bilirubin concentrations were calculated as the total bilirubin
concentration subtracted by either the measured or the imputed conjugated bilirubin
concentrations.
The potential confounding factors adjusted for included gender, [BSA = √height (cm) ×
weight (kg)/3600], ethnicity and assay sensitivity indicator in the models for pharmokinetic
parameters as outcomes, and sex, BSA, interaction between sex and BSA, and ethnicity in
the models for the total and unconjugated bilirubin as outcomes. These covariates were
selected based on potentially important factors affecting pravastatin pharmacokinetics in
humans as well as the considerations of the effect sample size rule where the number of
covariates should not exceed more than 1/10th–1/15th of the sample size [28]. The overall
gene effects on the outcomes and the interaction between covariates were tested using the
likelihood-ratio test. As the distributions of AUC, Cmax and the total and unconjugated
bilirubin were skewed to the right, they were transformed to square root of each value to
satisfy the normality assumption for regression models. All significant findings were
reanalyzed using the bootstrap method (1000 bootstraps) to reduce a chance of false-positive
finding and the bootstrapped P-values and 95% confidence intervals (CI) were reported, as
appropriate. All tests were two-tailed, and a P-value of < 0.05 was considered significant.
Hardy–Weinberg equilibrium was tested for each gene stratified by race using a STATA
user-written program, Hardy–Weinberg equilibrium tests and allele frequency estimation
[29]. All other analyses were performed with the software STATA 9.1 (StataCorp, College
Station, Texas, USA) and R (www.r-project.org).
Ho et al. Page 6
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Results
Comparison of transporter polymorphism allele frequencies between European-Americans
and African-Americans
The allele frequencies of SLCO1B1, ABCC2, ABCG2, and ABCB11 variants are
summarized in Table 1. The SLCO1B1 388G (*1b) variant was more common in African-
Americans than in European-American participants (77 vs. 38% allele frequency, P <
0.001). On the other hand, the SLCO1B1 521C (*5) variant had a relatively high allele
frequency in European-American participants than African-American participants (15 vs.
1%, P = 0.008), in line with our previous work [5]. Haplotype analysis revealed the 521C
variant was most likely to occur in conjunction with the 388G allele to form SLCO1B1*15
(combination of *1b and *5), consistent with several recently published observations in
Japanese and European Caucasian populations (Table 3) [7, 13]. The 1463C (*9) variant,
initially identified with a relatively high allele frequency in African-Americans [5], was only
identified with a 1% allele frequency in the current study, indicating this variant may be less
common in African-Americans than previously thought.
Participants were also genotyped for commonly occurring polymorphisms in the efflux
transporters ABCC2, ABCG2 and ABCB11. The frequency of ABCC2 4544A and ABCB11
1331C and 2029G variants differed significantly among European-American and African-
American participants, whereas there was a lack of difference in the prevalence of ABCG2
polymorphisms between ethnic groups (Table 1). Overall, these results reveal that allele
frequencies of commonly occurring SLCO1B1, ABCC2 and ABCB11 variants appear to be
dependent on race, consistent with previously published reports [5] (www.pharmgkb.org).
All genes were in Hardy–Weinberg equilibrium for both European-Americans and African-
Americans.
Pravastatin pharmacokinetics in relation to SLCO1B1 genotypes and ethnicity
There was wide interindividual and interethnic variability in pravastatin disposition. In our
model, the combination of gender, BSA, assay sensitivity, race and SLCO1B1 521TC
genotype accounted for 31% of the interindividual variability in pravastatin AUC, whereas
SLCO1B1 521TC genotype and race individually explained 6.5 and 6.2% of variability,
respectively (Table 2).
SLCO1B1 521TC genotype was significantly associated with higher AUC (P=0.01) and
Cmax values (P<0.05) after adjusting for gender, BSA and assay sensitivity (Fig. 1a).
SLCO1B1 wild-type T/T carriers had significantly lower AUC values compared with T/C
carriers (adjusted mean difference 31.3 ng · h/ml, 95% CI 6.2–56.4ng · h/ml, P= 0.015) and
C/C carriers (adjusted mean difference 58.2ng · h/ml, 95% CI 14.6–101.9ng · h/ ml, P =
0.009). T/T carriers also demonstrated significantly lower Cmax values compared with T/C
carriers (adjusted mean difference 14.6ng · h/ml, 95% CI 0.5–28.7 ng · h/ml, P = 0.042) and
tended toward lower Cmax values compared with homozygous C/C carriers (adjusted mean
difference 17.5 ng · h/ml, 95% CI −4.3 to 39.4 ng · h/ ml, P = 0.116).
When considering overall SLCO1B1 gene effects, SLCO1B1 diplotype (521C and 388G
alleles) was significantly associated with increase in AUC (P = 0.048), but not Cmax (P=
Ho et al. Page 7
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
0.153) (Table 3), after adjusting for gender, BSA, and assay sensitivity. SLCO1B1*1a/*15
participants had 45 and 80% higher AUC values than SLCO1B1*1a/*1a (adjusted mean
difference 42.9 ng · h/ml, 95% CI 8.9–76.8 ng · h/ml; P = 0.013) and SLCO1B1*1b/*1b
(adjusted mean difference 59.7 ng · h/ml, 95% CI 24.9–94.4ng · h/ml; P = 0.001) carriers,
respectively (Fig. 2). AUC was 92 and 149% higher in SLCO1B1*15/* 15 than
SLCO1B1*1a/*1a (adjusted mean difference 55.1 ng · h/ml, 95% CI 9.7–100.6ng · h/ml; P =
0.017) and SLCO1B1*1b/*1b (adjusted mean difference 71.9ng · h/ml, 95% CI 16.7–127.1
ng · h/ml; P = 0.011) carriers, respectively. Cmax was 46% and 78% higher in
SLCO1B1*1a/* 15 than *1a/*1a (adjusted mean difference 24.7 ng/ml, 95% CI 3.1–46.3
ng/ml; P = 0.025) or *1b/*1b (adjusted mean difference 29.8 ng/ml, 95% CI 8.0–51.6 ng/ml;
P = 0.007) carriers.
Overall, pravastatin AUC (98.3 ± 60.7 vs. 69.5 ± 56.3 ng · h/ml; P = 0.010) and Cmax values
(53.3 ± 33.3 vs. 37.4 ± 29.2 ng/ml; P = 0.009) were significantly higher in European-
Americans compared with African-Americans (Fig. 1b). After adjustment for SLCO1B1
521TC genotype, gender, BSA and assay sensitivity, European-Americans still
demonstrated significantly higher AUC (P = 0.028) and Cmax values (P = 0.036) than
African-Americans. Likewise, after adjustment for SLCO1B1 388AG genotype, gender,
BSA and assay sensitivity, European-Americans consistently had higher AUC (P = 0.004)
and Cmax values (P = 0.006). A subgroup analysis for European-Americans demonstrated
pravastatin AUC was significantly greater for those participants who carried the 521C allele
vs. wild-type individuals (adjusted mean difference 28.7 ng · h/ml, 95% CI 3.4–54.1 ng ·
h/ml; P = 0.026). Furthermore, SLCO1B1*1a/* 15 participants demonstrated significantly
higher pravastatin AUC values than *1a/*1a participants (adjusted mean difference 46.0ng ·
h/ml, 95% CI 11.2–80.7ng · h/ml; P = 0.011), but not *1b/*1b participants (adjusted mean
difference 33.9ng · h/ml, 95% CI 37.5–105.4ng · h/ml; P = 0.35). After adjusting for all
covariates, sex was not associated with significant differences in pravastatin
pharmacokinetics.
Pravastatin pharmacokinetics in relation to ABCC2, ABCG2, and ABCB11 genotypes
Commonly occurring ABCC2, ABCG2, or ABCB11 genotypes did not associate with
differences in pravastatin AUC or Cmax (Table 4) when adjusted for covariates. Although
some of these variants have demonstrated altered function for other substrates such as
estradiol-17β-glucuronide and methotrexate [19,20], it appears that they are not relevant to
the observed interindividual variability in pravastatin disposition.
Bilirubin concentrations and SLCO1B1 genotypes
Recent studies in Asian participants have noted a correlation between SLCO1B1 genotype
and unconjugated bilirubin concentrations and risk for hyperbilirubinemia [30–32]. Total
and direct bilirubin concentrations were measured in each subject as part of routine
laboratory screening for entry into the study. Unconjugated bilirubin was calculated by
subtracting the direct fraction from the total bilirubin concentration. Regarding SLCO1B1
genotype, there were no significant differences in total or unconjugated bilirubin
concentrations for individuals when grouped by the 521TC (data not shown) or 388AG (Fig.
Ho et al. Page 8
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
3) genotype. Moreover, there were no significant differences in total or unconjugated
bilirubin concentrations amongst SLCO1B1*1a/*1a, *1a/*1b, or *1b/*1b carriers (Table 3).
Discussion
There is an increasing appreciation of the important contribution of the OATP superfamily
of uptake transporters to the disposition of numerous endogenous and xenobiotic substrates.
Recently, polymorphisms have been identified in the SLCO1B1 gene and both in-vitro and
in-vivo studies have demonstrated that a number of these variants result in impaired function
compared with wild-type SLCO1B1. Previous studies using pravastatin as an in-vivo probe
for SLCO1B1 activity have demonstrated altered disposition profiles among Japanese or
European Caucasian participants who possess variant SLCO1B1 alleles *1b, *5 and *15
[7,13,14]. This report extends these findings by confirming higher AUC of the SLCO1B1
substrate pravastatin in individuals carrying the 521C allele (*5 or *15). Furthermore, we
observed significant ethnic differences in plasma exposure between European-Americans
and African-Americans after adjustment for SLCO1B1 genotype, suggesting additional
genetic factors may be of importance to the interethnic and interindividual variability in
pravastatin disposition.
Studies from our laboratory and others have demonstrated a number of SLCO1B1 variants,
most notably 521C, are associated with impaired transporter activity in vitro. Reduced
function of this variant appears to be due, in part, to mistrafficking of the protein to the cell
surface [5]. Importantly the 521C variant has a relatively high allele frequency of ~15–18%
in Asian and Caucasian populations [5,6]. Studies in healthy Asian and European Caucasian
participants utilizing single-dose pravastatin as a probe for SLCO1B1 activity demonstrated
that carriers of 521C polymorphism (*5), regardless of whether this was in linkage
equilibrium with 388A (*15), had significantly higher pravastatin AUC values [7,13,14],
consistent with in-vitro functional data.
In this report, we also used pravastatin as an in-vivo probe of transporter activity in a large
population of healthy European-American and African-American individuals. We genotyped
for commonly occurring polymorphisms in transporter genes of potential importance to
pravastatin disposition, including the uptake transporter OATP1B1 and the efflux
transporters MRP2, BSEP, and BCRP. Overall, the SLCO1B1 521C variant was
significantly associated with higher AUC and Cmax values, consistent with previous reports.
Individuals homozygous for the variant allele (C/C) had the highest pravastatin AUC,
whereas individuals heterozygous for the variant allele (T/C) or wild-type (T/T) exhibited
intermediate, and lowest AUC values respectively, suggesting a gene–dose effect. Similar
gene–dose effects were noted for Cmax values.
Previous reports have suggested there may be phenotypic differences between the two
reference wild-type alleles 388A (*1a) and 388G (*1b), as pravastatin AUC values for
individuals carrying the *1b allele tended to be lower than for the *1a allele, although this
has not been consistently statistically significant [7,14,33]. Likewise, in our study, we found
that individuals homozygous for the *1b allele (388GG) did have a tendency, albeit not
Ho et al. Page 9
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
statistically significant, to have lower AUC values than those homozygous for the *1a allele
(388AA).
When analyzed by haplotype, individuals carrying the SLCO1B1*1a/*15 or *15/*15
diplotype had significantly higher AUC values than carriers of either of the wild-type
diplotypes SLCO1B1*1a/*1a or *1b/*1b. Individuals carrying the SLCO1B1*1b/*15
diplotype tended to have higher AUC values than *1a/*1a or *1b/*1b carriers, but this did
not reach statistical significance, possibly related to the inverse effects of the *1b and *15
haplotypes. Individuals carrying the SLCO1B1*1a/*15 diplotype exhibited significantly
higher Cmax values than either *1a/*1a or *1b/*1b carriers. Participants carrying the
SLCO1B1*1b/*15 and *15/*15 diplotypes also tended to have higher Cmax values than
*1a/*1a or *1b/*1b carriers. Overall, these results are consistent with previously published
studies and confirm that individuals carrying the SLCO1B1 521C variant attain significantly
higher plasma pravastatin AUC and Cmax values than those carrying the wild-type reference
allele after single-dose administration of pravastatin.
The clinical implications of this observed genotypemediated variation in pravastatin
disposition are unclear. A recent prospective clinical study evaluating the effect of
SLCO1B1 genotype on pharmacokinetics and lipidlowering efficacy of multiple-dose
pravastatin confirmed significantly higher plasma AUC and Cmax values in individuals
carrying the 521C allele but did not observe a significant genotype dependence on lipid-
lowering effect [34]. As the number of participants in that study was, however, small,
potential subtle differences in lipidlowering efficacy among individuals with various
genotypes may not have been detected. Furthermore, SLCO1B1 genotype only explained
~7% of pravastatin AUC variability in our study. Niemi et al. [13] noted in their study that
characteristics such as body weight did not explain variability in pravastatin AUC. In
contrast, we noted that gender and BSA contributed 11 and 12% of pravastatin variability,
respectively. Therefore, consideration of other factors such as gender, BSA and race, in
addition to genotype may provide a more accurate prediction of the interindividual
variability in pravastatin pharmacokinetic and pharmacodynamic parameters.
Recently, interethnic differences in statin disposition have been noted. Rosuvastatin, with
similar physicochemical properties to pravastatin, had ~2-fold higher systemic exposure in
Asian participants compared with Caucasian participants in Western Europe or the United
States [35,36]. A recent study of rosuvastatin disposition in Asian and Caucasian
participants living in the same geographic area, Singapore, confirmed ~2-fold higher plasma
exposure in the Asian group [37]. Interestingly, neither the SLCO1B1 388G nor 521C allele
accounted for the observed pharmacokinetic differences between Asian and Caucasian
participants. In this study, we report ethnic differences in pravastatin pharmacokinetics
between European-Americans and African-Americans. Overall, European-American
individuals demonstrated significantly higher plasma pravastatin AUC and Cmax values than
African-American participants. As SLCO1B1 521C genotype had the most significant effect
on pravastatin pharmacokinetics and the allele frequency of this variant is very low (~1%) in
African-Americans, we adjusted the analysis for SLCO1B1 521TC genotype. Even after
adjustment for genotype, European-Americans had significantly higher AUC and Cmax
values than African-Americans, suggesting interethnic differences exist in pravastatin
Ho et al. Page 10
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
disposition between these two groups and that other genetic factors, possibly polymorphisms
in other transporter genes, may contribute to the observed interethnic variation in
disposition.
Recent studies have indicated that statins are substrates for efflux transporters, including
MRP2, BCRP and BSEP [15–17]. Polymorphisms have been identified in these transporters
and some have been shown to have functional consequences in vitro or in vivo [19,20,38–
40]. We genotyped individuals for commonly occurring polymorphisms in these
transporters. Although we noted significant interethnic differences in the frequencies of
some of these variants, significant associations between polymorphisms in these efflux
transporters and pravastatin AUC or Cmax were not found. The likely explanation is that
hepatic uptake mediated by OATP1B1 is thought to be the rate-limiting step in pravastatin
hepatic clearance [10]. Therefore, although efflux transporter polymorphisms influence the
pharmacokinetics of drugs such as diflomotecan [39], these do not clearly impact pravastatin
disposition.
Recent in-vitro studies have suggested that unconjugated bilirubin may be a substrate for
OATP1B1, although controversy exists as to whether OATP1B1 alone is sufficient to
mediate hepatic extraction of bilirubin from the systemic circulation [41,42]. Several clinical
studies have suggested that SLCO1B1 genotype, in particular the 521C and 388G variant
alleles, may predict risk for severe hyperbilirubinemia in Asian infants, influence serum
bilirubin concentrations, and be a risk for unconjugated hyperbilirubinemia in healthy Asian
adults [30–32]. Our clinical study enrolled healthy adults with normal complete blood
counts and comprehensive metabolic panels, including hepatic function panels. Therefore,
individuals with mild hyperbilirubinemia would not have been eligible for this study. Our
analysis, nevertheless, indicates that SLCO1B1 genotype alone, in particular the 521C and
388G variant alleles, was not associated with significant differences in total or unconjugated
bilirubin concentrations. Moreover, when analyzed by diplotype, there were no significant
differences in total or unconjugated bilirubin concentrations among SLCO1B1*1a/*1a,
*1a/*1b, *1b/*1b, *1a/*15, *1b/*15, or *15/*15 carriers.
In conclusion, our findings confirm SLCO1B1 variants, in particular SLCO1B1*15, are
important contributing factor to the observed interindividual variability in pravastatin
pharmacokinetics after single-dose oral administration. In addition, there appear to be
significant interethnic differences in pravastatin disposition between European-Americans
and African-American participants which are not attributable to SLCO1B1 521TC or
388AG genotype, suggesting other genetic factors may be of importance when considering
differences in pravastatin disposition between these two groups. The consequences of
SLCO1B1 polymorphisms on the pharmacokinetic variation of other statins and the
pharmacodynamic effects of SLCO1B1 polymorphisms to lipid response in patients with
dyslipidemia remain unclear and warrant further study.
Acknowledgments
We thank Katherine Lee, Rachel McCumsey, Santiago Vilanova and Scott Bailey for their technical assistance on
this project. We also thank Dr David Freeman for thoughtful advice relating to pravastatin HPLC analysis. This
work was funded in part by United States Public Health Service grants GM54724 (R.B.K.), M01 RR-00095 from
Ho et al. Page 11
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the National Center for Research Resources, and a Vanderbilt University Physician Scientist Development Award
(R.H.H.).
References
1. Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim
Biophys Acta. 2003; 1609:1–18. [PubMed: 12507753]
2. Hagenbuch B, Meier PJ. Organic anion transporting polypeptides of the OATP/ SLC21 family:
phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/
functional properties. Pflugers Arch. 2004; 447:653–665. [PubMed: 14579113]
3. Ho RH, Kim RB. Transporters and drug therapy: implications for drug disposition and disease. Clin
Pharmacol Ther. 2005; 78:260–277. [PubMed: 16153397]
4. Marzolini C, Tirona RG, Kim RB. Pharmacogenomics of the OATP and OAT families.
Pharmacogenomics. 2004; 5:273–282. [PubMed: 15102542]
5. Tirona RG, Leake BF, Merino G, Kim RB. Polymorphisms in OATP-C: identification of multiple
allelic variants associated with altered transport activity among European- and African-Americans. J
Biol Chem. 2001; 276:35669–35675. [PubMed: 11477075]
6. Nozawa T, Nakajima M, Tamai I, Noda K, Nezu J, Sai Y, Tsuji A, Yokoi T. Genetic
polymorphisms of human organic anion transporters OATP-C (SLC21A6) and OATP-B
(SLC21A9): allele frequencies in the Japanese population and functional analysis. J Pharmacol Exp
Ther. 2002; 302:804–813. [PubMed: 12130747]
7. Nishizato Y, Ieiri I, Suzuki H, Kimura M, Kawabata K, et al. Polymorphisms of OATP-C
(SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin
Pharmacol Ther. 2003; 73:554–565. [PubMed: 12811365]
8. Michalski C, Cui Y, Nies AT, Nuessler AK, Neuhaus P, Zanger UM, et al. A naturally occurring
mutation in the SLC21A6 gene causing impaired membrane localization of the hepatocyte uptake
transporter. J Biol Chem. 2002; 277:43058–43063. [PubMed: 12196548]
9. Hatanaka T. Clinical pharmacokinetics of pravastatin: mechanisms of pharmacokinetic events. Clin
Pharmacokinet. 2000; 39:397–412. [PubMed: 11192473]
10. Nakai D, Nakagomi R, Furuta Y, Tokui T, Abe T, Ikeda T, Nishimura K. Human liver-specific
organic anion transporter, LST-1, mediates uptake of pravastatin by human hepatocytes. J
Pharmacol Exp Ther. 2001; 297:861–867. [PubMed: 11356905]
11. Hsiang B, Zhu Y, Wang Z, Wu Y, Sasseville V, Yang WP, Kirchgessner TG. A novel human
hepatic organic anion transporting polypeptide (OATP2). Identification of a liver-specific human
organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-
CoA reductase inhibitor transporters. J Biol Chem. 1999; 274:37161–37168. [PubMed: 10601278]
12. Siekmeier R, Gross W, Marz W. Determination of pravastatin by high performance liquid
chromatography. Int J Clin Pharmacol Ther. 2000; 38:419–425. [PubMed: 11020028]
13. Niemi M, Schaeffeler E, Lang T, Fromm MF, Neuvonen M, Kyrklund C, et al. High plasma
pravastatin concentrations are associated with single nucleotide polymorphisms and haplotypes of
organic anion transporting polypeptide-C (OATP-C, SLCO1B1). Pharmacogenetics. 2004;
14:429–440. [PubMed: 15226675]
14. Mwinyi J, Johne A, Bauer S, Roots I, Gerloff T. Evidence for inverse effects of OATP-C
(SLC21A6) 5 and 1b haplotypes on pravastatin kinetics. Clin Pharmacol Ther. 2004; 75:415–421.
[PubMed: 15116054]
15. Kivisto KT, Grisk O, Hofmann U, Meissner K, Moritz KU, Ritter C, et al. Disposition of oral and
intravenous pravastatin in MRP2-deficient TR- rats. Drug Metab Dispos. 2005; 33:1593–1596.
[PubMed: 16107564]
16. Hirano M, Maeda K, Matsushima S, Nozaki Y, Kusuhara H, Sugiyama Y. Involvement of BCRP
(ABCG2) in the biliary excretion of pitavastatin. Mol Pharmacol. 2005; 68:800–807. [PubMed:
15955871]
17. Hirano M, Maeda K, Hayashi H, Kusuhara H, Sugiyama Y. Bile salt export pump (BSEP/
ABCB11) can transport a nonbile acid substrate, pravastatin. J Pharmacol Exp Ther. 2005;
314:876–882. [PubMed: 15901796]
Ho et al. Page 12
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
18. Chan LM, Lowes S, Hirst BH. The ABCs of drug transport in intestine and liver: efflux proteins
limiting drug absorption and bioavailability. Eur J Pharm Sci. 2004; 21:25–51. [PubMed:
14706810]
19. Hirouchi M, Suzuki H, Itoda M, Ozawa S, Sawada J, Ieiri I, et al. Characterization of the Cellular
Localization, Expression Level, and Function of SNP Variants of MRP2/ABCC2. Pharm Res.
2004; 21:742–748. [PubMed: 15180328]
20. Kondo C, Suzuki H, Itoda M, Ozawa S, Sawada J, Kobayashi D, et al. Functional analysis of SNPs
variants of BCRP/ABCG2. Pharm Res. 2004; 21:1895–1903. [PubMed: 15553238]
21. Zamber CP, Lamba JK, Yasuda K, Farnum J, Thummel K, Schuetz JD, Schuetz EG. Natural allelic
variants of breast cancer resistance protein (BCRP) and their relationship to BCRP expression in
human intestine. Pharmacogenetics. 2003; 13:19–28. [PubMed: 12544509]
22. Eloranta ML, Hakli T, Hiltunen M, Helisalmi S, Punnonen K, Heinonen S. Association of single
nucleotide polymorphisms of the bile salt export pump gene with intrahepatic cholestasis of
pregnancy. Scand J Gastroenterol. 2003; 38:648–652. [PubMed: 12825874]
23. Meier Y, Pauli-Magnus C, Zanger UM, Klein K, Schaeffeler E, Nussler AK, et al. Interindividual
variability of canalicular ATP-binding-cassette (ABC)-transporter expression in human liver.
Hepatology. 2006; 44:62–74. [PubMed: 16799996]
24. Livak KJ. SNP genotyping by the 5-nuclease reaction. Methods Mol Biol. 2003; 212:129–147.
[PubMed: 12491907]
25. Stephens M, Smith NJ, Donnelly P. A new statistical method for haplotype reconstruction from
population data. Am J Hum Genet. 2001; 68:978–989. [PubMed: 11254454]
26. Stephens M, Scheet P. Accounting for decay of linkage disequilibrium in haplotype inference and
missing-data imputation. Am J Hum Genet. 2005; 76:449–462. [PubMed: 15700229]
27. Lidofsky Steven, D. Jaundice. In: Feldman, M.; Friedman, LS.; Brandt, LJ., editors. Sleisender &
Fordtran’s gastrointestinal and liver disease. 7. St. Louis, MO: W.B. Saunders; 2002. p. 249-261.
28. Harrell, FE. Regression modeling strategies with applications to linear models, logistic regression,
and survival analysis. New York City: Springer; 2001.
29. Cleves MA. Hardy-Weinberg equilibrium tests and allele frequency estimation. STATA Tech Bull.
1999; 48:34–37.
30. Ieiri I, Suzuki H, Kimura M, Takane H, Nishizato Y, Irie S, et al. Influence of common variants in
the pharmacokinetic genes (OATP-C, UGT1A1, and MRP2) on serum bilirubin levels in healthy
subjects. Hepatol Res. 2004; 30:91–95. [PubMed: 15519273]
31. Huang MJ, Kua KE, Teng HC, Tang KS, Weng HW, Huang CS. Risk factors for severe
hyperbilirubinemia in neonates. Pediatr Res. 2004; 56:682–689. [PubMed: 15319464]
32. Huang CS, Huang MJ, Lin MS, Yang SS, Teng HC, Tang KS. Genetic factors related to
unconjugated hyperbilirubinemia amongst adults. Pharmacogenet Genomics. 2005; 15:43–50.
[PubMed: 15864125]
33. Maeda K, Ieiri I, Yasuda K, Fujino A, Fujiwara H, Otsubo K, et al. Effects of organic anion
transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and
temocapril. Clin Pharmacol Ther. 2006; 79:427–439. [PubMed: 16678545]
34. Igel M, Arnold KA, Niemi M, Hofmann U, Schwab M, Lutjohann D, et al. Impact of the
SLCO1B1 polymorphism on the pharmacokinetics and lipidlowering efficacy of multiple-dose
pravastatin. Clin Pharmacol Ther. 2006; 79:419–426. [PubMed: 16678544]
35. AstraZeneca. Rosuvastatin (Crestor). 2005 Ref Type: Unpublished Work.
36. Martin PD, Mitchell PD, Schneck DW. Pharmacodynamic effects and pharmacokinetics of a new
HMG-CoA reductase inhibitor, rosuvastatin, after morning or evening administration in healthy
volunteers. Br J Clin Pharmacol. 2002; 54:472–477. [PubMed: 12445025]
37. Lee E, Ryan S, Birmingham B, Zalikowski J, March R, Ambrose H, et al. Rosuvastatin
pharmacokinetics and pharmacogenetics in white and Asian subjects residing in the same
environment. Clin Pharmacol Ther. 2005; 78:330–341. [PubMed: 16198652]
38. Tamura A, Watanabe M, Saito H, Nakagawa H, Kamachi T, Okura I, Ishikawa T. Functional
validation of the genetic polymorphisms of human ABC transporter ABCG2: identification of
alleles that are defective in porphyrin transport. Mol Pharmacol. 2006; 70:287–296. [PubMed:
16608919]
Ho et al. Page 13
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
39. Sparreboom A, Gelderblom H, Marsh S, Ahluwalia R, Obach R, Principe P, et al. Diflomotecan
Pharmacokinetics in Relation to ABCG2 421C>A Genotype. Clin Pharmacol Ther. 2004; 76:38–
44. [PubMed: 15229462]
40. Morisaki K, Robey RW, Ozvegy-Laczka C, Honjo Y, Polgar O, Steadman K, et al. Single
Nucleotide Polymorphisms Modify the Transporter Activity of ABCG2. Cancer Chemother
Pharmacol. 2005; 56:161–172. [PubMed: 15838659]
41. Cui Y, Konig J, Leier I, Buchholz U, Keppler D. Hepatic uptake of bilirubin and its conjugates by
the human organic anion transporter SLC21A6. J Biol Chem. 2001; 276:9626–9630. [PubMed:
11134001]
42. Wang P, Kim RB, Chowdhury JR, Wolkoff AW. The human organic anion transport protein
SLC21A6 is not sufficient for bilirubin transport. J Biol Chem. 2003; 278:20695–20699.
[PubMed: 12670950]
Ho et al. Page 14
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Mean ± SD plasma concentrations after a single 40 mg dose of pravastatin in relation to (a)
SLCO1B1 521TC genotype and (b) ethnicity.
Ho et al. Page 15
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Mean (◊) ± SD (arrows) pravastatin AUC in relation to SLCO1B1 diplotype. Box plots
represent 25th–75th percentiles, horizontal line within plots represent the median, horizontal
lines upper and lower quartiles represent minimum and maximum values except outliers that
are represented by a single horizontal line beyond 1.5 interquartile range. AUC, area under
the curve.
Ho et al. Page 16
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Mean (◊) ± SD (arrows) serum (a) total and (b) unconjugated bilirubin concentrations in
relation to SLCO1B1 388AG genotype. Box plots represent 25th–75th percentiles,
horizontal line within plots represent the median, horizontal lines upper and lower quartiles
represent minimum and maximum values except outliers, which are represented by a single
horizontal line beyond 1.5 interquartile range.
Ho et al. Page 17
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Ho et al. Page 18
Table 1
Allele frequencies for transporter polymorphisms
Allele frequency
Transporter gene Genotype (P value)aEuropean-American African-American Total
SLCO1B1 388AG A 0.62 A 0.23 A 0.52
(P< 0.001) G 0.38 G 0.77 G 0.48
521TC T 0.85 T 0.99 T 0.90
(P=0.008) C 0.15 C 0.01 C 0.10
1463GC G 1.0 G 0.99 G 0.995
(P= 0.181) C 0.0 C 0.01 C 0.005
ABCC2 1249GA G 0.85 G 0.86 G 0.855
(P= 0.740) A 0.15 A 0.14 A 0.145
3563GA T 0.95 T 0.96 G 0.95
(P=0.685) A 0.05 A 0.04 A 0.05
4544GA G 0.95 G 0.83 G 0.91
(P= 0.021) A 0.05 A 0.17 A 0.09
ABCB11 1331TC T 0.28 T 0.54 T 0.37
(P= 0.001) C 0.72 C 0.46 C 0.63
2029AG A 1.0 A 0.79 A 0.93
(P< 0.001) G 0.0 G 0.21 G 0.07
ABCG2 421CA C 0.08 C 0.04 C 0.065
(P= 0.210) A 0.92 A 0.96 A 0.935
34GA G 0.02 G 0.09 G 0.05
(P= 0.110) A 0.98 A 0.91 A 0.95
aP-values for the association between genotype and race.
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Ho et al. Page 19
Table 2
Factors contributing to variability in pravastatin pharmacokinetics
Model covariates P-value R2 for model (%)
Gender < 0.001 11.1
BSA < 0.001 12.2
Assay sensitivity 0.004 7.8
521TC 0.031 6.5
Race 0.010 6.2
521TC and race 0.012 10.0
Gender/BSA/assay sensitivity/521TC/race < 0.001 30.7
BSA, body surface area.
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Ho et al. Page 20
Table 3
SLCO1B1 diplotype and pravastatin pharmacokinetic variables
Number Diplotype *1a/*1a *1a/*1b *1b/*1b *1a/*5 *1a/*15 *1b/*15 *15/*15 Total
69 European-American 26 23 2 1 8 7 2
38 African-Am 3 11 23 0 0 1 0
107(1.00)†† Total 29 (0.27) 343 (0.32) 25 (0.24) 1 (0.01) 8 (0.07) 8 (0.07) 2 (0.02)
AUC0–5 (ng h/ml) European-American 83.4 ± 66.3 103.2 ± 63.7 71.5 ± 58.7 58.9 120.9 ± 37.0 105.1 ± 48.8 167.0 ± 31.4 98.3 ± 60.7#
African-American 119.4 ± 82.7 62.3 ± 46.6 66.7 ± 57.8 61.8 69.5 ± 56.3
Total 87.1 ± 67.4 90.0 ± 61.1 67.1 ± 56.7 58.9 120.9 ± 37.0*99.7 ± 47.7 1 67.0 ± 31.4**
Cmax (ng/ml) European-American 45.1 ± 35.1 58.0 ± 37.6 41.5 ± 16.3 29.3 66.4 ± 24.6 54.1 ± 24.7 75.6 ± 2.1 53.3 ± 33.3#
African-American 49.6 ± 29.3 35.3 ± 27.1 36.9 ± 31.6 35.1 37.4 ± 29.2
Total 45.6 ± 34.1 50.7 ± 35.8 37.3 ± 30.5 29.3 66.4 ± 24.651.7 ± 23.9 75.6 ± 2.1
Total bilirubin (mg/dl) European-American 0.62 ± 0.23 0.50 ± 0.25 0.75 ± 0.49 0.50 0.61 ± 0.19 0.59 ± 0.29 0.4 ± 0.14
African-American 0.37 ± 0.12 0.56 ± 0.30 0.53 ± 0.33 0.40 ±
Total 0.60 ± 0.23 0.52 ± 0.27 0.55 ± 0.33 0.50 0.61 ± 0.19 0.56 ± 0.28 0.4 ± 0.14
AUC, area under the curve.
*P<0.05 vs. *1a/*1a and P= 0.001 vs. *1b/*1b.
**P<0.05 vs. *1a/*1a and P<0.05 vs. *1b/*1b.
#P<0.05 European-American vs. African-American.
P<0.05 vs. *1a/*1a and P<0.01 vs. *1b/*1b.
††P < 0.001 for the association between haplotype and race.
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Ho et al. Page 21
Table 4
Efflux transporter polymorphisms and pravastatin pharmacokinetics
MRP2 BCRP BSEP
1249GA 3563TA 4544GA 421CA 34GA 1331TC 2029AG
Total AUC Wt/Wt 89.9 ± 61.6 87.8 ± 60.4 87.8 ± 60.9 89.8 ± 61.3 88.1 ± 61.8 92.2 ± 64.9 91.9 ± 62.1
Wt/Var 80.0 ± 56.4 98.0 ± 67.4 83.3 ± 56.0 77.4 ± 58.4 85.0 ± 53.7 86.3 ± 64.8 62.5 ± 47.3
Var/Var 1 98.1 211.2 128.4 88.8 ± 55.9 80
Cmax Wt/Wt 47.5 ± 32.0 48.1 ± 33.0 48.8 ± 33.5 48.8 ± 33.7 48.2 ± 33.4 47.7 ± 30.8 49.9 ± 33.4
Wt/Var 46.8 ± 34.3 48.9 ± 33.2 41.3 ± 29.2 41.8 ± 26.5 40.7 ± 26.2 46.3 ± 34.1 34.7 ± 26.5
Var/Var 104 80 70.4 49.6 ± 32.8 32.6
European-American AUC Wt/Wt 95.1 ± 62.9 97.3 ± 60.1 97.3 ± 60.1 100.8 ± 61.2 97.0 ± 61.2 92.3 ± 56.8 98.7 ± 61.1
Wt/Var 102.9 ± 53.6 110.6 ± 73.2 110.6 ± 73.2 87.7 ± 62.3 134.4 ± 53.6 107.2 ± 67.6
Var/Var 1 98.1 93.6 ± 57.6
Cmax Wt/Wt 50.7 ± 32.9 53.7 ± 33.5 53.7 ± 33.5 55.1 ± 34.4 53.2 ± 33.7 55.8 ± 32.0 53.7 ± 33.5
Wt/Var 59.0 ± 33.8 53.2 ± 35.9 53.2 ± 35.9 46.1 ± 28.2 63.7 ± 30.4 56.3 ± 34.0
Var/Var 104 51.4 ± 34.1
African-American AUC Wt/Wt 80.4 ± 59.1 70.7 ± 57.9 65.5 ± 57.9 72.0 ± 58.0 69.8 ± 59.6 92.1 ± 72.3 72.8 ± 62.4
Wt/Var 42.5 ± 39.0 68.6 ± 50.4 66.0 ± 35.7 39.9 ± 10.3 55.4 ± 26.1 60.5 ± 51.8 62.5 ± 47.3
Var/Var 211.2 128.4 63.9 ± 40.0 80
Cmax Wt/Wt 41.8 ± 29.9 37.9 ± 29.8 37.4 ± 31.2 38.4 ± 30.1 38.0 ± 30.8 42.8 ± 30.8 39.1 ± 31.5
Wt/Var 26.8 ± 25.4 38.9 ± 29.7 33.7 ± 22.7 26.2 ± 10.84 27.0 ± 10.5 33.9 ± 30.6 34.7 ± 26.5
Var/Var 80 70.4 40.2 ± 24.8 32.6
All P values > 0.05.
Wt, wild-type; Var, variant.
Pharmacogenet Genomics. Author manuscript; available in PMC 2014 June 19.
... 48,59 Previous studies on the effect of SLCO1B1 variants on endogenous substate transportation have been limited to bilirubin and were largely inconclusive. 49,[60][61][62] However, the associations we identified between SLCO1B1 and many oxylipins motivate the need for future experimental studies to determine whether rs4149056 and other variants also impact oxylipin transportation. ...
Preprint
Full-text available
Bioactive fatty acid-derived oxylipin molecules play key roles in mediating inflammation and oxidative stress, which underlie many chronic diseases. Circulating levels of fatty acids and oxylipins are influenced by both environmental and genetic factors; characterizing the genetic architecture of bioactive lipids could yield new insights into underlying biological pathways. Thus, we performed a genome wide association study (GWAS) of n=81 fatty acids and oxylipins in n=11,584 Hispanic Community Health Study/Study of Latinos (HCHS/SOL) participants with genetic and lipidomic data measured at study baseline (58.6% female, mean age = 46.1 years, standard deviation = 13.8 years). Additionally, given the effects of central obesity on inflammation, we examined interactions with waist circumference using two-degree-of-freedom joint tests. Heritability estimates ranged from 0% to 47.9%, and 48 of the 81oxylipins and fatty acids were significantly heritable. Moreover, 40 (49.4%) of the 81 oxylipins and fatty acids had at least one genome-wide significant (p< 6.94E-11) variant resulting in 19 independent genetic loci involved in fatty acid and oxylipin synthesis, as well as downstream pathways. Four loci (lead variant minor allele frequency [MAF] range: 0.08-0.50), including the desaturase-encoding FADS and the OATP1B1 transporter protein-encoding SLCO1B1, exhibited associations with four or more fatty acids and oxylipins. The majority of the 15 remaining loci (87.5%) (lead variant MAF range = 0.03-0.45, mean = 0.23) were only associated with one oxylipin or fatty acid, demonstrating evidence of distinct genetic effects. Finally, while most loci identified in two-degree-of-freedom tests were previously identified in our main effects analyses, we also identified an additional rare variant (MAF = 0.002) near CARS2, a locus previously implicated in inflammation. Our analyses revealed shared and distinct genetic architecture underlying fatty acids and oxylipins, providing insights into genetic factors and motivating future multi-omics work to characterize these compounds and elucidate their roles in disease pathways.
... However, there is growing evidence that inter-individual variability in drug pharmacokinetics due to (in part ethnicity-dependent) transporter genetic polymorphisms or transporter-mediated drug-drug interactions is also an important determinant of the efficacy and toxicity of drugs [18][19][20][21]. For instance, statins, particularly the pharmacologically active acid forms of statins, have been suggested to be highly dependent on drug transporters for their disposition and efficacy [22,23]. The possible roles of single uptake or efflux transporters in physiology and pharmacology have been assessed in genetically modified cell lines and animal models, as well as (pre-)clinical drug-drug interaction studies [24], but still could not fully explain the variable pharmacokinetic behavior of many drugs. ...
... 37 This may potentially result in altered bioavailability and disposition of statins, methotrexates, and other drugs that are substrates of one or both of these transporters. 38 According to the 2020 US Census Bureau (https://www.census.gov), the US population has a wide range of ethnic and racial groups, including ~ 60.6% White, 18.1% Hispanic or Latino, 12.3% Black or African American, 5.5% Asian, 0.7% American Indian and Alaska Native, 0.2% Native Hawaiian and other Pacific Islander, with 0.3% reporting other races and 2.4% identifying as 2 or more races. Predicting interethnic pharmacogenetic susceptibility toward co-administered drugs as well as from unintentional exposure to environmental chemicals is an understudied but crucial dimension to clinical and regulatory decision making for precision medicine and improved patient care. ...
Article
Full-text available
Environmental health science seeks to predict how environmental toxins, chemical toxicants, and prescription drugs accumulate and interact within the body. Xenobiotic transporters of the ATP‐binding Cassette (ABC) and solute carrier (SLC) superfamilies are major determinants of the uptake and disposition of xenobiotics across the kingdoms of life. The goal of this study was to integrate drug and environmental chemical interactions (DECIs) of mammalian ABC and SLC proteins in a centralized, integrative database. We built upon an existing publicly accessible platform – the “TransPortal” – which was updated with novel data and searchable features on transporter‐interfering chemicals (TICs) from manually curated literature data. The integrated resource TransPortal‐TICBase ( https://transportal.compbio.ucsf.edu ) now contains information on 46 different mammalian xenobiotic transporters of the ABC‐ and SLC‐type superfamilies, including 13 newly added rodent and two additional human drug transporters, 126 clinical drug‐drug interactions (DDIs), and a more than quadrupled expansion of the initial in vitro chemical interaction data from 1402 to 6296 total interactions. Based on our updated database, environmental interference with major human and rodent drug transporters occurs across the ABC‐ and SLC‐type superfamilies, with kinetics indicating that some chemicals, such as the ionic liquid 1‐hexylpyridinium chloride and the antiseptic chlorhexidine, can act as strong inhibitors with potencies similar or even higher than pharmacological model inhibitors. The new integrated web portal serves as a central repository of current and emerging data for interactions of prescription drugs and environmental chemicals with human drug transporters. This archive has important implications for predicting adverse drug‐drug and drug‐environmental chemical interactions and can serve as a reference website for the broader scientific community of clinicians and researchers.
... The Individuals carrying the CC (17-fold) and TC (5-fold) genotypes have a higher risk of developing myopathy than individuals with the TT genotype [25]. The relatively high frequencies of low-activity variants (CC and TC) in European ancestry may imply a higher risk of statininduced myopathy [26]. ...
Article
Full-text available
Background Statin-induced myopathy is reported to be associated with the solute carrier organic anion transporter family member 1B1 gene single nucleotide polymorphism, c.521 T > C. There is no epidemiologic data on this gene polymorphism in several countries. Therefore, this study aimed at assessing the genotype and allele frequencies of the gene variant in three countries. Methods This study involved healthy individuals from Colombia, Mozambique, and Portugal. Genomic DNA was isolated from blood samples using the Qiamp DNA Extraction Kit (Qiagen). The isolated DNA was genotyped using novel Polymerase Chain Reaction—Restriction Fragment Length Polymorphism. Microstat and GraphPad QuickCal software were used for the Chi-square test and the evaluation of Hardy–Weinberg equilibrium respectively. Results A total of 181 individuals’ blood samples were analyzed. Overall, the TT (74.0%) genotype was the highest and the CC (7.8%) was the lowest. Country wise genotypic frequencies were Colombia 47(70.2%) TT, 12(17.9%) TC and 8(11.9%) CC; Mozambique 47(88.7%) TT, 5(9.4%) TC, and 1(1.9%) CC; and Portugal 40(65.6%) TT, 16(26.2%) TC, and 5(8.2%) CC. The reference (T) allele was highest among Mozambicans (93.4%) compared to Colombians (79.1%) and Portuguese (78.7%). Mozambicans showed statistically significant genotypic and allelic frequency differences compared to Colombians (p < 0.01) and Portuguese (p < 0.01). Conclusions Overall and country-wise, CC genotype was less frequent and it is relatively high for Colombians and Portuguese populations. This finding may imply statins risk–benefit variability associated with CC genotype among these populations that needs further understanding.
... Two of these had a genotype with expected lower transport function. Similar to various pharmacogenetic studies (Niemi et al., 2004;Ho et al., 2007;Nakanishi and Tamai, 2012), pravastatin AUC and C max were higher in those two participants (with the haplotypes SCLO1B1*5/*15 and *15/*15) than in homozygous or heterozygous carriers of wild-type alleles. ...
Article
Full-text available
Introduction: Bulevirtide is a first-in-class antiviral drug to treat chronic hepatitis B/D. We investigated the drug-drug interaction potential and pharmacokinetics of high-dose subcutaneous bulevirtide (5 mg twice daily) with organic anion transporting polypeptide 1B1 (OATP1B1) and cytochrome P450 (CYP) 3A4. Methods: This was a single-center, open-label, fixed-sequence drug-drug interaction trial in 19 healthy volunteers. Before and at bulevirtide steady state, participants ingested a single 40 mg dose of pravastatin. A midazolam microdose was applied to quantify CYP3A4 activity. Results: At bulevirtide steady state, pravastatin area under the concentration-time curve (AUC0–∞) increased 1.32-fold (90% CI 1.08-1.61). The 5 mg bulevirtide twice-daily treatment resulted in a mean AUC0-12 of 1210 h*ng/ml (95% CI 1040-1408) and remained essentially unchanged under the influence of pravastatin. CYP3A4 activity did not change to a clinically relevant extent. As expected, total bile acids increased substantially (35-fold) compared to baseline during bulevirtide treatment. All study medication was well tolerated. Discussion: The study demonstrated that high-dose bulevirtide inhibited OATP1B-mediated hepatic uptake of the marker substrate pravastatin but the extent is considered clinically not relevant. Changes in CYP3A4 activity were also not clinically relevant. In conclusion, this study suggests that OATP1B substrate drugs as well as CYP3A4 substrates may safely be used without dose adjustment in patients treated with bulevirtide. However, in patients using high statin doses and where concomitant factors potentially further increase statin exposure, caution may be required when using bulevirtide.
Chapter
There are numerous clinical risk factors associated with stroke, including atrial fibrillation, hypertension, and hypercholesterolemia. Such risks can be ameliorated with appropriate pharmacotherapy. However, there is variability in response to drugs used for these conditions, which may, at least in part, be genetically determined. For instance, genetic determinants of response to warfarin, statins, antiplatelet agents, and some antihypertensives have been described.
Chapter
Pharmacogenomics has a potential to improve and individualize treatment of cardiovascular diseases. In the past couple of decades, strong evidence emerged to inform PGx guided drug prescribing, dose selection, and clinical implementation of some common cardiovascular drug classes such as anticoagulants, antiplatelets, and statins. This chapter discusses evidence, including several clinical trials, treatment algorithms, genetic variants with demonstrated clinical impact and guidelines related to these drug and other cardiovascular drug classes.
Article
Aim To demonstrate the use of PBPK modelling to explore the impact of ethnic differences on drug PK. Method A PBPK model developed for lansoprazole was used to predict the clinical PK of lansoprazole in Japanese subjects by incorporating the physiological parameters of a Japanese population into the model. Further verification of the developed Japanese population with clinical studies involving eight other CYP substrates‐ omeprazole, ticlopidine, alprazolam, midazolam, nifedipine, cinacalcet, paroxetine and dextromethorphan was also carried out. Results The PK of lansoprazole in both Caucasian and Japanese subjects was well predicted by the model as the observed data were within the 5 th and 95 th percentiles across all the clinical studies. In age‐ and sex‐matched simulations in both the Caucasian and Japanese populations, the predicted PK (mean ± SD) of a single oral dose of 30mg lansoprazole was higher in the Japanese population in all cases, with more than 2‐fold higher AUC of 5.98 ± 6.43 mg/L.hr (95% CI: 4.72, 7.24) vs 2.46 ± 2.45 mg/L.hr (95% CI: 1.98, 2.94) in one scenario. In addition, in two out of the nine clinical DDIs of lansoprazole and the additional CYP substrates simulated using the Japanese population, the predicted DDI in Japanese was more than 1.25‐fold that in Caucasians, indicating an increased DDI liability. Conclusions By accounting for various physiological parameters that characterise a population in a PBPK model, the impact of the different identified inter‐ethnic differences on the drug’s PK can be explored, which can inform the adoption of drugs from one region to another.
Article
Full-text available
A novel human organic transporter, OATP2, has been identified that transports taurocholic acid, the adrenal androgen dehydroepiandrosterone sulfate, and thyroid hormone, as well as the hydroxymethylglutaryl-CoA reductase inhibitor, pravastatin. OATP2 is expressed exclusively in liver in contrast to all other known transporter subtypes that are found in both hepatic and nonhepatic tissues. OATP2 is considerably diverged from other family members, sharing only 42% sequence identity with the four other subtypes. Furthermore, unlike other subtypes, OATP2 did not transport digoxin or aldosterone. The rat isoform oatp1 was also shown to transport pravastatin, whereas other members of the OATP family, i.e.rat oatp2, human OATP, and the prostaglandin transporter, did not. Cis-inhibition studies indicate that both OATP2 and roatp1 also transport other statins including lovastatin, simvastatin, and atorvastatin. In summary, OATP2 is a novel organic anion transport protein that has overlapping but not identical substrate specificities with each of the other subtypes and, with its liver-specific expression, represents a functionally distinct OATP isoform. Furthermore, the identification of oatp1 and OATP2 as pravastatin transporters suggests that they are responsible for the hepatic uptake of this liver-specific hydroxymethylglutaryl-CoA reductase inhibitor in rat and man.
Article
Full-text available
Bilirubin, the end product of heme catabolism, is taken up from the blood circulation into the liver. This work identifies a high-affinity transport protein mediating the uptake of bilirubin and its conjugates into human hepatocytes. Human embryonic kidney cells (HEK293) permanently expressing the recombinant organic anion-transporting polypeptide 2 (human OATP2, also known as LST-1 or OATP-C; symbol SLC21A6) showed uptake of [3H]monoglucuronosyl bilirubin, [3H]bisglucuronosyl bilirubin, and [3H]sulfobromophthalein withK m values of 0.10, 0.28, and 0.14 μm, respectively. High-affinity uptake of unconjugated [3H]bilirubin by OATP2 occurred in the presence of albumin and was not mediated by another basolateral hepatic uptake transporter, human OATP8 (symbol SLC21A8). OATP2 and OATP8 differed by their capacity to extract substrates from albumin before transport. In comparison to the high-affinity transport by OATP2, OATP8 transported [3H]sulfobromophthalein and [3H]monoglucuronosyl bilirubin with lower affinity, withK m values of 3.3 and 0.5 μm, respectively. The organic anion indocyanine green potently inhibited transport mediated by OATP2, with a Ki value of 112 nm, but did not inhibit transport mediated by OATP8. Human OATP2 may play a key role in the prevention of hyperbilirubinemia by facilitating the selective entry of unconjugated bilirubin and its glucuronate conjugates into human hepatocytes.
Chapter
The chief attribute of the fluorogenic 5′ nuclease assay is that it is completely homogeneous. After mixing the sample and reaction components, the assay is run in a closed tube format with no postpolymerase chain reaction (PCR) processing steps. Results are obtained by simply measuring the fluorescence of the completed reactions. By eliminating post-PCR processing, allelic discrimination with fluorogenic probes reduces the time of analysis, eliminates the labor and supply costs of post-PCR steps, reduces the risk of crossover contamination, and minimizes sources of error. The assay has the sensitivity of PCR so that a minimum amount of genomic DNA is required. The use of endpoint fluorescence measurements maximizes throughput. Using a single ABI Prism® 7900HT Sequence Detection System and 24-h operation, it is possible to generate up to 250,000 SNP results per day. TaqMan® MGB probes and the entire operating system outlined below make it possible to quickly apply the 5′ nuclease assay to any allelic discrimination application where high throughput is of paramount concern.
Article
Zinc is an essential trace element with important biological functions, depending on the structural and/or catalytic role played by zinc ions in a large variety of enzymes. Zinc plays a critical role in cellular integrity, protein synthesis, nucleic acid metabolism, contributing to cell growth, proliferation, differentiation and death. The present review reports data on zinc homeostasis and metabolism, zinc absorption, intercellular trafficking, intracellular transport inside enterocytes and hepatocytes. Particular emphasis is given to data regarding the role of zinc carriers ZnTs and Zips, and to their expression in liver and gut in experimental and in human studies. The role of zinc in the gastrointestinal tract and in the liver as a powerful antioxidant and its relationship with apoptosis is discussed. Possible implications of zinc status in different disorders of the gastrointestinal tract are presented, focusing on its possible introduction in the therapy of inflammatory bowel diseases. Data on the role of zinc and zinc carriers in the evolution of liver fibrosis towards cirrhosis are also discussed. Finally, data on the ability of zinc therapy to obtain regression of liver cirrhosis in patients affected by Wilson's disease are reported, and the hypothesis that zinc could protect against liver fibrosis in chronic liver disease of different origin is presented.
Article
Pravastatin is a hydrophilic liver-specific inhibitor of the enzyme 3-hydroxy-3-methylglutaryl coenzyme A reductase. It effectively lowers plasma cholesterol and low-density lipoprotein concentrations in humans. Pharmacokinetic studies of pravastatin have been mostly performed by means of radioactively labelled drug or by measuring plasma concentrations with gas chromatography and mass spectrometry. Aim of our study was to develop a simple, but reliable method which allows the determination of pravastatin plasma concentrations under clinical routine conditions. Samples were prepared by solid-phase extraction on cyclohexyl bond elut cartridges. Chromatography was carried out on an octyl matrix. Triamcinolone acetonide was used as internal standard. The method was linear within the range of 5 to 200 microg/l pravastatin. The coefficient of variation depended on the pravastatin concentration, but was less than 10% throughout. The pharmacokinetics of pravastatin were determined in healthy individuals. Five healthy subjects received single oral doses of pravastatin (60 mg) and one of these subjects additionally received a dose of 80 mg at three different study days. In all subjects blood was sampled 0, 30, 60, 90, 120, 150, 180, 240 and 300 min after drug intake. Peak plasma concentrations of pravastatin were found between 60 min and 120 min after oral administration of 60 mg and reached values between 37 microg/l and 126 microg/l. The calculated AUCs were between 52 ng/ml x h and 311 ng/ml x h and the corresponding plasma elimination half-life times were between 95 min and 165 min. In all subjects plasma concentrations of pravastatin 5 hours after oral drug administration were near the detection limit of the method (5 microg/l). Intraindividually, there was only little variation in the kinetics of pravastatin. However, marked differences were encountered between the subjects studied. The data suggest that the determination of pravastatin plasma concentrations by means of a HPLC system can be used for routine analysis of pravastatin plasma concentrations. The obtained pharmacokinetic data in healthy individuals stand in ample agreement with the results of prior studies in which the concentrations of pravastatin were determined by other more sophisticated methods.
Article
Pravastatin, one of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) widely used in the management of hypercholesterolaemia, has unique pharmacokinetic characteristics among the members of this class. Many In vivo and In vitro human and animal studies suggest that active transport mechanisms are involved in the pharmacokinetics of pravastatin. The oral bioavailability of pravastatin is low because of incomplete absorption and a first-pass effect. The drug is rapidly absorbed from the upper part of the small intestine, probably via proton-coupled carrier-mediated transport, and then taken up by the liver by a sodium-independent bile acid transporter. About half of the pravastatin that reaches the liver via the portal vein is extracted by the liver, and this hepatic extraction is mainly attributed to biliary excretion which is performed by a primary active transport mechanism. The major metabolites are produced by chemical degradation in the stomach rather than by cytochrome P450-dependent metabolism in the liver. The intact drug and its metabolites are cleared through both hepatic and renal routes, and tubular secretion is a predominant mechanism in renal excretion. The dual routes of pravastatin elimination reduce the need for dosage adjustment if the function of either the liver or kidney is impaired, and also reduce the possibility of drug interactions compared with other statins, which are largely eliminated by metabolism. The lower protein binding than other statins weakens the tendency for displacement of highly protein-bound drugs. Although all statins show a hepatoselective disposition, the mechanism for pravastatin is different from that of the others. There is high uptake of pravastatin by the liver via an active transport mechanism, but not by other tissues because of its hydrophilicity, whereas the disposition characteristics of other statins result from high hepatic extraction because of high lipophilicity. These pharmacokinetic properties of pravastatin may be the result of the drug being given in the pharmacologically active open hydroxy acid form and the fact that its hydrophilicity is markedly higher than that of other statins. The nature of the pravastatin transporters, particularly in humans, remains unknown at present. Further mechanistic studies are required to establish the pharmacokinetic-pharmacodynamic relationships of pravastatin and to provide the optimal therapeutic efficacy for various types of patients with hypercholesterolaemia.
Article
Current routine genotyping methods typically do not provide haplotype information, which is essential for many analyses of fine-scale molecular-genetics data. Haplotypes can be obtained, at considerable cost, experimentally or (partially) through genotyping of additional family members. Alternatively, a statistical method can be used to infer phase and to reconstruct haplotypes. We present a new statistical method, applicable to genotype data at linked loci from a population sample, that improves substantially on current algorithms; often, error rates are reduced by > 50%, relative to its nearest competitor. Furthermore, our algorithm performs well in absolute terms, suggesting that reconstructing haplotypes experimentally or by genotyping additional family members may be an inefficient use of resources.