You are on page 1of 6

articles nature publishing group

Rifampin Enhances the Glucose-Lowering


Effect of Metformin and Increases OCT1 mRNA
Levels in Healthy Participants
SK Cho1,2, JS Yoon3, MG Lee1, DH Lee1,2, LA Lim1,2, K Park1, MS Park4 and J-Y Chung1

We evaluated the effect of the pregnane X receptor (PXR) agonist rifampin on metformin pharmacokinetics, organic
cation transporter 1 (OCT1) and OCT2 mRNA levels, and glucose levels, using the oral glucose tolerance test (OGTT) in
16 healthy subjects. The glucose-lowering effects of metformin were evaluated by OGTT before and after metformin
treatment on days 1 and 2 and again on days 13 and 14 after a 10-day course of rifampin. Rifampin increased the
difference in maximum glucose levels (∆Gmax) by 41.9% (P = 0.024) and the area under the concentration–time curve
(AUC) during the first 60 min after glucose ingestion (∆AUCgluc60) by 54.5% (P = 0.020). Renal clearance (CLR) of metformin
was increased by 16% (P = 0.008), but the systemic exposure was only slightly increased (13%, P = 0.049), possibly
because of increased absorption. Rifampin increased OCT1 mRNA levels 4.1-fold in peripheral blood cells (P = 0.001);
however, OCT2 mRNA was not detected. Our results suggest that rifampin increases OCT1 expression and hepatic uptake
of metformin, leading to enhanced glucose-lowering action.

The biguanide derivative metformin is the first-line oral hypogly- the concentration–time curve (AUC) in rats by upregulating Oct1
cemic drug for the treatment of type 2 diabetes. Its primary expression in the liver and Oct2 expression in the kidneys.12
action is to lower hepatic glucose production by inhibiting glu- The effects of PXR agonists on OCT expression in humans
coneogenesis.1 Metformin is eliminated unchanged in the urine, have not been reported, and, specifically, the effects of rifampin
and its renal clearance (CLR) is greater than that of creatinine, on metformin pharmacokinetics and on the clinical response to
indicating that tubular secretion is the major route of elimina- the drug are unclear. Moreover, rifampin itself appears to affect
tion.2 Metformin is a substrate of organic cation transporters blood glucose regardless of whether metformin is administered
(OCTs), which mediate drug absorption and elimination.3,4 concurrently. Several cases have been reported in which rifampin
OCT1 is located primarily in hepatocyte sinusoidal membranes,5 therapy for tuberculosis induced hyperglycemia.13–15
whereas OCT2 is localized mainly in the basolateral membrane In this study, we hypothesized that the plasma concentration
of the kidney proximal tubule.6 Uptake by OCTs in the liver and of metformin and its glucose-lowering action would be affected
kidney plays an important role in the pharmacokinetics and by rifampin, probably by altering the expression or function of
pharmacodynamics of metformin.7,8 OCTs in the liver, its primary target organ, as well as in the kid-
The tuberculosis drug rifampin is a pregnane X receptor (PXR) ney, the site of elimination. We therefore determined the OCT1
agonist in humans and a powerful inducer of cytochrome P450 and OCT2 mRNA levels along with the pharmacokinetics and
(CYP) enzymes and various transporters.9 Rifampin was found to glucose-lowering effects of metformin after rifampin treatment
reduce the glucose-lowering effects of the oral hypoglycemic agent in healthy participants.
glyburide, possibly by increasing clearance through the induc-
tion of CYP2C9 or the efflux pump P-glycoprotein.10,11 Because Results
metformin is not metabolized, it would not be susceptible to this Glucose-lowering effect of metformin after rifampin treatment
effect. However, Maeda et al. reported that the PXR agonist preg- Healthy volunteers (n = 16) underwent oral glucose tolerance
nenolone-16-carbonitrile reduced metformin blood area under tests (OGTTs) before and after receiving two doses of metformin

1Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea; 2Brain Korea 21 Project for Medical Science, Yonsei University, Seoul, Korea;
3Department of Pharmacology, Kwandong University College of Medicine, Gangneung, Korea; 4Department of Pediatrics, Yonsei University College of Medicine,

Seoul, Korea. Correspondence: J-Y Chung (jychung@yuhs.ac)


Received 12 August 2010; accepted 4 October 2010; advance online publication 26 January 2011. doi:10.1038/clpt.2010.266

416 VOLUME 89 NUMBER 3 | march 2011 | www.nature.com/cpt


articles

on days 1 and 2 and again on days 13 and 14 after a 10-day course plasma AUCs (before and after rifampin treatment) are shown in
of rifampin. Baseline serum glucose concentrations (before the Figure 2. The mean AUC from 0 to 24 h (AUCmet(0–24)) and the
first metformin dose) were similar before and after rifampin maximum metformin concentration (Cmax) were comparable
treatment; however, the glucose-lowering effects of metformin to the results of previous studies.16–18
were considerably increased by the 10-day rifampin treatment After rifampin treatment, metformin plasma concentra-
(Figure 1). The ability of metformin to reduce maximum blood tions were 23% higher at 0.5 h (P = 0.002) and 13% higher
glucose levels (ΔGmax), the area under the curve of glucose con- at 1 h (P = 0.036). As shown in Table 2, the partial AUCs
centration–time (AUC of glucose) during the first 60 min after early after metformin administration were also higher after
glucose ingestion (ΔAUCgluc60), and the AUC of glucose for the rifampin treatment (AUCmet(0–1), 18% increase, P = 0.003;
entire 180-min test were compared before and after rifampin AUCmet(0–2), 14% increase, P = 0.015; and AUCmet(0–6), 12%
treatment (Table 1). Rifampin treatment increased the mean increase, P = 0.049), but the value of AUCmet(6–24) was not
value of ΔGmax by 41.9% (31 vs. 44 mg/dl; P = 0.024) and that of significantly different after rifampin treatment (P = 0.121).
ΔAUCgluc60 by 54.5% (914 ± 510 mg/dl·min vs. 1,412 ± 555 mg/ The AUC for the full 24 h (AUCmet(0–24)) increased 13% after
dl·min; P = 0.020). The mean ΔAUCgluc was increased by 44% rifampin treatment (9,408 ± 2,410 vs. 10,672 ± 3,149 ng/
after the rifampin treatment, but the difference was not signifi- ml·h, P = 0.049), whereas Cmax was not significantly differ-
cant (Table 1). ent (1,536 ± 350 vs. 1,692 ± 114 ng/ml, P = 0.070). Rifampin
treatment did not significantly alter the half-time (t1/2) of met-
Metformin pharmacokinetics formin, but it increased the CLR of metformin by 16% (501 ± 97
Participants received two oral doses of metformin on days 1 and vs. 580 ± 101 ml/min, P = 0.008); creatinine clearance (CLCr)
2 (1,750 mg total) and again on days 13 and 14. The metformin was unchanged. In addition, the net tubular secretion (SrCLR)

a 180
Table 1 The glucose-lowering effect parameters of metformin
160 before and after rifampin treatment in healthy participants
Serum glucose concentration (mg/dI)

Before metformin (n = 16)


After metformin
140 Parameter Before rifampin After rifampin P
ΔGmax (mg/dl) 31 ± 14 44 ± 14 0.024
120
ΔAUCgluc60 (mg/dl·min) 914 ± 510 1,412 ± 555 0.020
100 ΔAUCgluc (mg/dl·min) 1,679 ± 1,155 2,378 ± 1,316 0.121
Data were evaluated by Wilcoxon signed-rank test and are expressed as mean ± SD.
80 ΔAUCgluc, difference in total area under the plasma concentration–time curve for
glucose before and after metformin treatment; ΔAUCgluc60, difference in partial area
60 under the plasma concentration–time curve for glucose (0–60 min after ingestion,
during which glucose concentration increases) before and after metformin treatment;
ΔGmax, difference in maximum glucose level before and after metformin treatment.
40
0 30 60 90 120 150 180
Time (min)
2,200
b 180
2,000
Plasma metformin concentration (ng/ml)

1,800
160
Serum glucose concentration (mg/dI)

Before metformin
1,600 Before rifampin
After metformin
140 1,400
After rifampin
1,200
120
1,000 *
100 800

600 *
80
400

60 200
0
40 0 6 12 18 24
0 30 60 90 120 150 180 Time (h)
Time (min)
Figure 2  The plasma concentration–time curve of metformin on day
Figure 1  Serum glucose levels were determined by means of oral glucose 2 (before rifampin treatment) and on day 14 (after rifampin treatment).
tolerance tests before and after metformin administration. (a) Serum glucose Metformin concentrations were measured after the second dose of
profile before the 10-day course of rifampin. (b) Serum glucose profile after metformin. Data are expressed as mean ± SD (n = 16). *P < 0.05 (before
the 10-day course of rifampin. Data are expressed as mean ± SD (n = 16). rifampin treatment vs. after rifampin treatment).

Clinical pharmacology & Therapeutics | VOLUME 89 NUMBER 3 | march 2011 417


articles

of metformin (CLR – CLCr) was higher by 21% because of the in OCT1 and OCT2 expression may account, at least in part,
effect of rifampin (398 ± 92 vs. 475 ± 98 ml/min, P = 0.005). for rifampin-induced changes in metformin’s effects. Consistent
with the rifampin-enhanced glucose tolerance, OCT1 mRNA
OCT1 and OCT2 genotypes and expressions levels were 4.1-fold higher after rifampin treatment (410 ± 260%,
For the OCT1 and OCT2 genotypes that have been reported, P = 0.001) (Figure 3). However, OCT2 mRNA was not detected
the functional significance and frequencies in Korean popula- in the peripheral blood cells.
tions18–21 were almost evenly distributed between wild-type and
variant alleles: OCT1 rs2282143 (CC = 13 and CT = 3), rs622342 Discussion
(AA = 10, AC = 5, and CC = 1), and OCT2 rs316019 (GG = 7, In this study, we evaluated the interaction between rifampin and
GT = 7, and TT = 2). In order to characterize the mechanism metformin by comparing glucose levels in healthy volunteers
through which rifampin enhances the glucose-lowering action before and after metformin treatment on days 1 and 2 and again
of metformin, OCT1 and OCT2 mRNA levels in peripheral on days 13 and 14, after a 10-day course of rifampin. Our data
blood cells were determined using real-time PCR. Metformin is show that rifampin increased the glucose-lowering effect of met-
transported primarily by OCT1 and OCT2;22 therefore, changes formin by as much as 54.5% but did not alter baseline glucose
levels. This is the first study to report a substantial drug–drug
Table 2  Pharmacokinetic parameters of metformin in healthy interaction between rifampin and metformin in humans.
participants (n = 16) before and after a 10-day course of rifampin The rifampin-induced effects were assessed 2 days after the last
Before rifampin After rifampin P dose of rifampin was taken. The half-life of rifampin is relatively
AUCmet(0–1) (ng/ml·h) 909 ± 209 1,077 ± 258 0.003 short (~3 h), and although we did not measure rifampin blood
AUCmet(0–2) (ng/ml·h) 2,340 ± 484 2,664 ± 634 0.015
levels, systemic exposure to rifampin was expected to be very
low. For that reason, the enhanced metformin action appears
AUCmet(0–6) (ng/ml·h) 625 ± 1,531 6,997 ± 2,012 0.049
to be an indirect effect of rifampin. OCT1 plays an important
AUCmet(6–24) (ng/ml·h) 3,149 ± 1,048 3,675 ± 1,275 0.121 role in metformin uptake and action in the liver; therefore, the
AUCmet(0–24) (ng/ml·h) 9,408 ± 2,410 10,672 ± 3,149 0.049 increased OCT1 mRNA levels (Figure 3) suggest that rifampin
t1/2 (h) 7.38 ± 3.09 6.81 ± 1.86 0.501 enhances the glucose-lowering action of metformin by upregu-
Cmax (ng/ml) 1,536 ± 350 1,692 ± 114 0.070 lating OCT1. This is consistent with the results of a previous
study in rats reporting that the PXR ligand pregnenolone-16-
Tmax (h) 1.63 ± 0.53 1.59 ± 0.64 0.881
carbonitrile affects the pharmacokinetics of metformin both in
CLR (ml/min) 501 ± 97 580 ± 101 0.008
vivo and in vitro by upregulating the expression of Oct1 and
SrCLR (ml/min) 398 ± 92 475 ± 98 0.005 Oct2.12 Although rifampin has been reported to induce early-
CLCr (ml/min) 109 ± 15 105 ± 15 0.173 phase hyperglycemia by boosting glucose absorption,15 its ability
Data were evaluated by Wilcoxon signed-rank test and are expressed as mean ± SD. to enhance the glucose-lowering effect of metformin may have
AUCmet(a–b), area under the plasma concentration–time curve from time point a to offset its independent hyperglycemic action.
time point b; CLCr, creatinine clearance; CLR, renal clearance; Cmax, maximum plasma Our pharmacokinetic data show that the values of CLR
concentration; SrCLR, renal clearance by tubular secretion; t1/2, elimination half-life;
Tmax, time of maximum plasma concentration. and SrCLR of metformin increased after the 10-day course
of rifampin, but the value of CLCr did not change, indicating
P = 0.001 that the transporter activity increased the renal elimination of
metformin. OCT2 is located in the basolateral membrane of
the renal proximal tubule, where it plays an important role in
−1
metformin elimination.23,24 Although we were unable to detect
−2 OCT2 mRNA in the blood, the increase in elimination of met-
Normalized mRNA expression level

−3 formin was probably due to upregulated activity of OCT2 or


∆Ct = Ct (GAPDH) – Ct (OCT1)

−4 another transporter.
−5 We observed that rifampin slightly increased systemic expo-
−6 sure to metformin as assessed by pharmacokinetic parameters
−7
(AUCs). These results are not consistent with the hypotheses of
−8
increased hepatic uptake mediated by OCT1 and increased CLR
mediated by OCT2; rather, they may be due to rifampin-induced
−9
changes in absorption kinetics. Metformin concentrations at
−10
0.5 and 1 h after administration were significantly higher after
−11 rifampin treatment. Metformin exhibits apparent absorption-
−12 dependent kinetics,25 and therefore the higher metformin con-
Before rifampin After rifampin
centrations immediately after administration may have been the
Figure 3  Rifampin treatment (10 days) increased OCT1 mRNA levels in the result of increased absorption. OCT1 and OCT3, as well as other
peripheral blood cells of 16 healthy volunteers, as determined by real-time transporters, are also located in the intestine,26 thereby suggest-
PCR. OCT, organic cation transporter. ing that their rifampin-induced upregulation may be responsible

418 VOLUME 89 NUMBER 3 | march 2011 | www.nature.com/cpt


articles

for altered metformin absorption kinetics. Recently Zhou et al. Methods


reported that the plasma membrane monoamine transporter Subjects. Sixteen healthy subjects (men, n  =  14; women, n  =  2; age
expressed in the intestine may also play an important role in 27  ±  4 years; height, 172.4  ±  7.3 cm; weight, 67.4  ±  11.2 kg; fasting
glucose, 90  ±  8 mg/dl) were recruited for participation in the study.
metformin absorption.27 Further study is needed to characterize Exclusion criteria were anemia (hemoglobin <12 g/dl), history of
the effect of rifampin on the activity and expression of intestinal drug abuse, symptomatic coronary heart disease, significant eleva-
OCTs and plasma membrane monoamine transporter. tion of hepatic enzyme level (aspartate aminotransferase or alanine
Our study has some limitations. First, OCT1 mRNA levels aminotransferase >60 IU/l), serum creatinine >1.5 mg/dl, or present-
in blood may not reflect hepatic OCT1 mRNA or protein lev- ing any one of the criteria for metabolic syndrome. Subjects who were
consuming more than 2 alcoholic drinks (at one time) twice a week,
els; however, a correlation between mRNA levels and protein smoking more than 10 cigarettes a day, or taking any medication were
expression of Oct1 and Oct2 has been reported in rats,28 and also excluded. Women of childbearing age were given urine pregnancy
it is not ethical to perform liver biopsies in healthy human tests to avoid enrollment of pregnant women.
volunteers. Second, rifampin may regulate transporters other
Clinical study procedures. The study protocol was reviewed and approved
than OCTs that are involved in metformin pharmacokinet-
by the institutional review board of Severance Hospital in the Yonsei
ics. For example, metformin is a substrate for the multidrug University Health System, Seoul, Korea (4-2009-0334). All procedures
and toxin extrusion 1 transporter, which has been reported to were carried out in accordance with the guidelines of the International
transport metformin and affect its glucose-lowering action;19,29 Conference on the Harmonisation of Technical Requirements for the
however, multidrug and toxin extrusion 1 does not appear Registration of Pharmaceuticals for Human Use—Good Clinical Practice.
to be affected by rifampin or PXR agonists in general.30 On Written informed consent for participation was obtained from all subjects
before enrollment in the study. The participants were asked to maintain
the other hand, transporters involved in absorption, such as normal physical activity at least 5 days before the study began. Dieticians
plasma membrane monoamine transporter, may be induced instructed the subjects regarding the meal plan designed to maintain a
by rifampin. Third, the genetic polymorphisms of OCT1 and carbohydrate intake of 200–250 g/day and the use of a food diary to record
OCT2 may have influenced the results. Because of the small food intake for 3 days before admission. The last meal before admission
number of subjects, a subgroup analysis could not be carried was consumed at the Clinical Trials Center at Severance Hospital. After an
overnight fast, blood was drawn to determine OCT1 and OCT2 mRNA
out; however, the mean values of glucose-lowering and phar- levels, and a 3-h OGTT (75 g glucose) was performed at 10 am (day 1).
macokinetic parameters were not very different among the The participants received a 1,000-mg oral dose of metformin (Diabex Tab;
genotype groups (data not shown). Fourth, the findings might Daewoong Pharmaceutical, Seoul, Korea) at 8 pm. After an overnight fast,
not be extrapolatable to diabetic patients because they were a 750-mg dose of metformin was administered at 8 am on day 2, followed
observed in healthy volunteers. Diabetes can differently affect by the second OGTT at 10 am. The dosing regimen was determined on
the basis of the results from a similar investigation carried out previously
not only OCT1 mRNA expression but also the transcriptional in healthy subjects.7 The time required for metformin concentration (in
activators PXR and CAR. Further studies in diabetic patients plasma) to reach maximum concentration has been reported as being
are needed. Finally, the pharmacological action of metformin ~2 h.16,21,25 The time point of the OGTT was therefore chosen to coincide
is the result of multiple factors including many that are beyond with the maximum glucose-lowering effect of metformin.
the scope of this study. The regulation of hepatic gluconeogen- Blood and urine samples were collected to determine the pharmacoki-
netics of metformin. Carbohydrate-controlled meals were provided at
esis by PXR may be mediated by numerous factors, including 1 pm on day 2, that is, 5 h after the second metformin dose. After receiv-
hepatocyte nuclear factor 4α,31 but the clinical significance of ing a 600-mg oral dose of rifampin (Rifampin Tab; Yuhan, Seoul, Korea)
these interactions is unclear. In this study, we observed that on the morning of day 3, the subjects were discharged. They continued
baseline glucose levels were not changed by rifampin, thereby taking rifampin (600 mg daily) up to day 12 (on an outpatient basis), dur-
suggesting that PXR activation itself does not significantly ing which they also maintained a food diary. The subjects were admitted
again to the Clinical Trials Center on day 13 and stayed there until day
alter serum glucose levels. Moreover, this prospective study 15. The second set of OGTT tests, metformin administration, and blood
was designed to minimize the effect of confounding factors. To and urine collection were carried out on the same schedule as that on
the best of our knowledge, this is the first study to evaluate the days 1–3.
role of OCTs in the metformin and rifampin interaction. Our
Blood and urine collection. The blood samples for determining OCT
findings suggest that metformin combination therapy with an
mRNA levels were collected before the OGTT on day 1. For OGTT analy-
OCT1 inducer (rifampin) or inhibitor (e.g., ranitidine) may sis, blood samples were collected before the ingestion of glucose and at
affect clinical outcomes. 15, 30, 45, 60, 90, 120, 150, and 180 min after ingestion. For determin-
In conclusion, we found that rifampin increased the concen- ing metformin concentrations in plasma, blood samples were collected
tration of metformin in the blood and enhanced its glucose- before the second dose of metformin and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 6,
lowering action. In addition, rifampin increased the renal 8, 10, 12, and 24 h after. After the second dose of metformin, participants
were asked to drink 240 ml of water every 4 h in order to maintain urine
tubular secretion of metformin. Rifampin–metformin inter- flow. The first portion of urine was voided, and the subsequent samples
actions in patients with tuberculosis or type 2 diabetes may were collected during the following time intervals: 0–4, 4–8, 8–12, and
affect drug safety as well as efficacy because the most toxic 12–24 h after the second metformin dose. The volume and pH of urine
side effect of metformin, namely, lactic acidosis, is a dose- were recorded before analysis. In order to calculate CLCr, serum creati-
dependent effect.32 A large-scale clinical study is necessary to nine level was determined from a blood sample (3 ml) drawn before each
of the admission times.
confirm OCT-based drug interactions in patients with tuber-
culosis and type 2 diabetes who are receiving both rifampin Metformin concentration analysis. Metformin concentrations in plasma
and metformin. and urine were determined using the highly specific and sensitive

Clinical pharmacology & Therapeutics | VOLUME 89 NUMBER 3 | march 2011 419


articles

method of liquid chromatography–tandem mass spectrometry (API Hilden, Germany). Genotyping was carried out using TaqMan
3200; Applied Biosystems Sciex, Concord, Ontario, Canada). In order allelic discrimination assays on an AB 7300 Real-time PCR System
to prepare the samples for analysis, an aliquot of the plasma or urine (Applied Biosystems). Ten microliters of PCR mixture was prepared
specimen was mixed with acetonitrile in the presence or absence of the with 5 μl of 2× TaqMan Genotyping Master Mix, 0.5 μl of 20× Drug
internal standard formoterol. The mixture was vortexed for 5 min and Metabolism Genotyping Assay Mix, 3.5 μl of DNase-free water, and
then centrifuged for 5 min at 10,000 r.p.m. An aliquot of the supernatant 1 μl of genomic DNA. Genotyping for SLC22A1 (OCT1) rs2282143
was transferred to an autosampler vial, and 1 µl was injected onto the (assay ID: C__15877554_40) and rs622342 (assay ID: C__928527_10)
column at 10 °C. The mobile phase consisted of 75% acetonitrile, 25% and for SLC22A2 (OCT2) rs316019 (assay ID: C__3111809_20)
double-distilled water, and 5 mmol/l ammonium formate aqueous solu- ­single-nucleotide polymorphisms was performed with validated
tion. The limit of quantification was 10 ng/ml for plasma and 0.5 µg/ml TaqMan genotyping assays purchased from Applied Biosystems. PCRs
for urine. The correlation coefficients (R2) were >0.994. The intraday and were as follows: initial denaturation at 95 °C for 10 min, followed by 50
interday coefficients of variation were <10%. The limit of detection for cycles of denaturation at 92 °C for 15 s and annealing/extension at 60
both blood and urine was 1 ng/ml. °C for 1 min. The allelic discrimination results were determined after
amplification by performing an end-point read. AB Sequence Detection
Analysis of glucose-lowering effect from OGTT. Participants were on a System 7300 sds software version 1.3.1 (Applied Biosystems) was used
carbohydrate-controlled diet (200–250 g/day) for 3 days before admission. for the analysis.
At the time point of the OGTT (10 am), all the subjects had fasted con-
tinuously for >14 h. Metformin lowers glucose production in patients with Statistical analysis. Measurements from the same subject before and after
diabetes1,33 and exerts the same effect in healthy subjects if serum glucose rifampin treatment were compared using the Wilcoxon signed-rank test.
levels are increased by glucose ingestion.7 The OGTT was conducted four The data were analyzed using SPSS 17.0 (SPSS, Chicago, IL). Data were
times: before and after metformin doses, before rifampin administration expressed as mean values ± SD. P < 0.05 was considered significant.
(days 1 and 2), and after rifampin administration (days 13 and 14). The
maximum glucose level (Gmax) was determined, and the area under the Acknowledgments
serum glucose concentration–time curve (AUCgluc) was calculated using This study was supported by grant A030001 of the Korea Healthcare
the trapezoidal rule. AUCgluc60 was defined as area under the curve from 0 Technology R&D Project, Ministry for Health and Welfare, Republic of Korea.
to 60 min after glucose ingestion, the period during which serum glucose We thank Yeon-Ju Shin for her excellent technical assistance with drug
concentration increases. The effect of metformin on the glucose-lower- concentration analysis and the staff of the Severance Hospital Clinical Trials
ing action was calculated as the differences in Gmax and AUCgluc values Center for their generous cooperation.
(ΔGmax, ΔAUCgluc60, and ΔAUCgluc) before and after administration of
metformin (on days 1 and 2, and again on days 13 and 14) in each subject. Conflict of Interest
The effect of rifampin on the glucose-lowering effect of metformin was The authors declared no conflict of interest.
evaluated from the ΔGmax, ΔAUCgluc60, and ΔAUCgluc values. © 2011 American Society for Clinical Pharmacology and Therapeutics

Pharmacokinetics. The pharmacokinetic parameters were calculated by 1. Hundal, R.S. et al. Mechanism by which metformin reduces glucose
noncompartmental analysis using WinNonlin 5.3 (Pharsight, Mountain production in type 2 diabetes. Diabetes 49, 2063–2069 (2000).
2. Tucker, G.T., Casey, C., Phillips, P.J., Connor, H., Ward, J.D. & Woods, H.F.
View, CA). Maximum metformin concentration (Cmax) and the time of
Metformin kinetics in healthy subjects and in patients with diabetes mellitus.
maximum concentration (Tmax) were determined, and the area under the Br. J. Clin. Pharmacol. 12, 235–246 (1981).
metformin concentration–time curves (AUCsmet) for the time periods 3. Gorboulev, V. et al. Cloning and characterization of two human polyspecific
0–1 h, 0–6 h, 6–24 h, and 0–24 h were calculated using the linear trapezoi- organic cation transporters. DNA Cell Biol. 16, 871–881 (1997).
dal rule. The elimination rate constant (ke) was estimated from the slope 4. Zhang, L., Dresser, M.J., Gray, A.T., Yost, S.C., Terashita, S. & Giacomini,
of the best-fit line determined by linear regression analysis of the log- K.M. Cloning and functional expression of a human liver organic cation
transformed concentration–time curve. The elimination half-life (t1/2) transporter. Mol. Pharmacol. 51, 913–921 (1997).
was then calculated from the equation t1/2 = ln(2)/ke. 5. Nies, A.T. et al. Expression of organic cation transporters OCT1 (SLC22A1) and
The CLR of metformin was calculated as the total amount of metformin OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver.
Hepatology 50, 1227–1240 (2009).
excreted in urine during 24 h divided by AUCmet. CLCr was calculated
6. Motohashi, H. et al. Gene expression levels and immunolocalization of organic
from the Cockcroft–Gault equation ((140 – age) × (body weight, kg) × ion transporters in the human kidney. J. Am. Soc. Nephrol. 13, 866–874 (2002).
(0.85 if female)/(72 × serum creatinine). The SrCLR of metformin was 7. Shu, Y. et al. Effect of genetic variation in the organic cation transporter 1
calculated by subtracting CLCr from CLR of metformin. (OCT1) on metformin action. J. Clin. Invest. 117, 1422–1431 (2007).
8. Song, I.S., Shin, H.J. & Shin, J.G. Genetic variants of organic cation transporter
OCT1 and OCT2 expression. Blood levels of OCT1 and OCT2 mRNA 2 (OCT2) significantly reduce metformin uptake in oocytes. Xenobiotica. 38,
were determined using real-time PCR. Total RNA was extracted using the 1252–1262 (2008).
QIAamp RNA Blood Mini Kit (Qiagen, Valencia, CA). Equal amounts 9. Li, T. & Chiang, J.Y. Mechanism of rifampicin and pregnane X receptor
of RNA (500 ng) from each sample were reverse transcribed, using inhibition of human cholesterol 7 alpha-hydroxylase gene transcription.
Am. J. Physiol. Gastrointest. Liver Physiol. 288, G74–G84 (2005).
an oligo(dT) primer and RNase H-reverse transcriptase (Invitrogen,
10. Niemi, M., Backman, J.T., Neuvonen, M., Neuvonen, P.J. & Kivistö, K.T. Effects of
Carlsbad, CA). The cDNA was amplified with human OCT1-specific and rifampin on the pharmacokinetics and pharmacodynamics of glyburide and
OCT2-specific TaqMan gene expression primer probe sets (Hs00427550_ glipizide. Clin. Pharmacol. Ther. 69, 400–406 (2001).
m1 and Hs01010723_m1, respectively), TaqMan Gene Expression Master 11. Self, T.H., Tsiu, S.J. & Fowler, J.W. Jr. Interaction of rifampin and glyburide. Chest
Mix, and the ABI 7300 real-time PCR system (Applied Biosystems, Foster 96, 1443–1444 (1989).
City, CA). The following cycling conditions were used: 1 cycle at 50 °C 12. Maeda, T. et al. Effect of pregnane X receptor ligand on pharmacokinetics of
for 2 min; 1 cycle at 95 °C for 10 min; and 50 cycles of 95 °C for 15 s fol- substrates of organic cation transporter Oct1 in rats. Drug Metab. Dispos. 35,
lowed by 60 °C for 1 min. A negative control using water as the template 1580–1586 (2007).
was included in every PCR experiment. Target gene mRNA levels were 13. Waterhouse, M., Wilson, C., White, V.L. & Chowdhury, T.A. Resolution of insulin-
requiring diabetes after cessation of chemotherapy for tuberculosis. J. R. Soc.
normalized to the endogenous control gene glyceraldehyde 3-phosphate
Med. 98, 270–271 (2005).
dehydrogenase (GAPDH: 4333764F). 14. Kihara, Y. & Otsuki, M. Interaction of gliclazide and rifampicin. Diabetes Care
23, 1204–1205 (2000).
Genotyping of OCT1 and OCT2. Genomic DNA was extracted from 15. Takasu, N. et al. Rifampicin-induced early phase hyperglycemia in humans.
peripheral whole blood using a QIAamp DNA Blood Mini kit (Qiagen, Am. Rev. Respir. Dis. 125, 23–27 (1982).

420 VOLUME 89 NUMBER 3 | march 2011 | www.nature.com/cpt


articles

16. Shu, Y. et al. Effect of genetic variation in the organic cation transporter 1, OCT1, 26. Seithel, A., Karlsson, J., Hilgendorf, C., Björquist, A. & Ungell, A.L. Variability
on metformin pharmacokinetics. Clin. Pharmacol. Ther. 83, 273–280 (2008). in mRNA expression of ABC- and SLC-transporters in human intestinal cells:
17. Chen, Y. et al. Effect of genetic variation in the organic cation transporter 2 on comparison between human segments and Caco-2 cells. Eur. J. Pharm. Sci. 28,
the renal elimination of metformin. Pharmacogenet. Genomics 19, 497–504 291–299 (2006).
(2009). 27. Zhou, M., Xia, L. & Wang, J. Metformin transport by a newly cloned proton-
18. Wang, Z.J., Yin, O.Q., Tomlinson, B. & Chow, M.S. OCT2 polymorphisms and stimulated organic cation transporter (plasma membrane monoamine
in-vivo renal functional consequence: studies with metformin and cimetidine. transporter) expressed in human intestine. Drug Metab. Dispos. 35, 1956–1962
Pharmacogenet. Genomics 18, 637–645 (2008). (2007).
19. Becker, M.L., Visser, L.E., van Schaik, R.H., Hofman, A., Uitterlinden, A.G. 28. Heemskerk, S., Wouterse, A.C., Russel, F.G. & Masereeuw, R. Nitric oxide down-
& Stricker, B.H. Genetic variation in the multidrug and toxin extrusion 1 regulates the expression of organic cation transporters (OCT) 1 and 2 in rat
transporter protein influences the glucose-lowering effect of metformin in kidney during endotoxemia. Eur. J. Pharmacol. 584, 390–397 (2008).
patients with diabetes: a preliminary study. Diabetes 58, 745–749 (2009). 29. Becker, M.L., Visser, L.E., van Schaik, R.H., Hofman, A., Uitterlinden, A.G. &
20. Kang, H.J. et al. Identification and functional characterization of genetic Stricker, B.H. Interaction between polymorphisms in the OCT1 and MATE1
variants of human organic cation transporters in a Korean population. Drug transporter and metformin response. Pharmacogenet. Genomics 20, 38–44
Metab. Dispos. 35, 667–675 (2007). (2010).
21. Song, I.S. et al. Genetic variants of the organic cation transporter 2 influence 30. Lickteig, A.J., Cheng, X., Augustine, L.M., Klaassen, C.D. & Cherrington, N.J.
the disposition of metformin. Clin. Pharmacol. Ther. 84, 559–562 (2008). Tissue distribution, ontogeny and induction of the transporters Multidrug
22. Urakami, Y., Okuda, M., Masuda, S., Saito, H. & Inui, K.I. Functional and toxin extrusion (MATE) 1 and MATE2 mRNA expression levels in mice.
characteristics and membrane localization of rat multispecific organic cation Life Sci. 83, 59–64 (2008).
transporters, OCT1 and OCT2, mediating tubular secretion of cationic drugs. 31. Wada, T., Gao, J. & Xie, W. PXR and CAR in energy metabolism. Trends
J. Pharmacol. Exp. Ther. 287, 800–805 (1998). Endocrinol. Metab. 20, 273–279 (2009).
23. Karbach, U. et al. Localization of organic cation transporters OCT1 and OCT2 in 32. Wang, D.S., Kusuhara, H., Kato, Y., Jonker, J.W., Schinkel, A.H. & Sugiyama, Y.
rat kidney. Am. J. Physiol. Renal Physiol. 279, F679–F687 (2000). Involvement of organic cation transporter 1 in the lactic acidosis caused by
24. Kimura, N., Okuda, M. & Inui, K. Metformin transport by renal basolateral metformin. Mol. Pharmacol. 63, 844–848 (2003).
organic cation transporter hOCT2. Pharm. Res. 22, 255–259 (2005). 33. Jeng, C.Y., Sheu, W.H., Fuh, M.M., Chen, Y.D. & Reaven, G.M. Relationship
25. Scheen, A.J. Clinical pharmacokinetics of metformin. Clin. Pharmacokinet. 30, between hepatic glucose production and fasting plasma glucose
359–371 (1996). concentration in patients with NIDDM. Diabetes 43, 1440–1444 (1994).

Clinical pharmacology & Therapeutics | VOLUME 89 NUMBER 3 | march 2011 421

You might also like