You are on page 1of 8

original article

Diabetes, Obesity and Metabolism 16: 215–222, 2014.


© 2013 John Wiley & Sons Ltd

Pharmacokinetics, pharmacodynamics and safety of


empagliflozin, a sodium glucose cotransporter 2 (SGLT2)

original
inhibitor, in subjects with renal impairment

article
S. Macha1 , M. Mattheus2 , A. Halabi3 , S. Pinnetti4 , H. J. Woerle2 & U. C. Broedl2
1 Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
2 Boehringer Ingelheim GmbH & Co. KG, Ingelheim, Germany
3 CRS Clinical Research Services, Kiel, Germany
4 Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany

Aims: Empagliflozin is a selective sodium glucose cotransporter 2 (SGLT2) inhibitor that inhibits renal glucose reabsorption and is being
investigated for the treatment of type 2 diabetes mellitus (T2DM).
Methods: In this open-label study, the effect of renal impairment on the pharmacokinetics, pharmacodynamics and safety of a 50 mg dose
of empagliflozin was investigated in 40 subjects, grouped according to estimated glomerular filtration rate (eGFR).
Results: Maximum empagliflozin plasma concentrations were similar in subjects with normal renal function and renal impairment. Area under
the empagliflozin concentration-time curve (AUC0 –∞ ) values increased by approximately 18, 20, 66 and 48% in subjects with mild, moderate,
severe renal impairment and renal failure/end stage renal disease (ESRD), respectively, in comparison to healthy subjects. This was attributed
to decreased renal clearance (CLR ). Urinary glucose excretion (UGE) decreased with increasing renal impairment and correlated with decreased
eGFR and CLR . Empagliflozin was well tolerated, with no increase in adverse events associated with renal impairment.
Conclusions: Renal insufficiency resulted in decreased CLR of empagliflozin, moderately increased systemic exposure and decreased UGE.
A single 50 mg dose of empagliflozin was well tolerated in subjects with normal renal function and any degree of renal impairment. The
pharmacokinetic results of this study indicate that no dose adjustment of empagliflozin is required in patients with renal impairment.
Keywords: diabetes, empagliflozin, pharmacodynamics, pharmacokinetics, renal impairment, SGLT2 inhibitor

Date submitted 8 April 2013; date of first decision 13 May 2013; date of final acceptance 11 July 2013

Introduction 30–59); stage 4 is a severe decrease in GFR (GFR 15–29) and


stage 5 is kidney failure (GFR <15 or dialysis) [9].
Diabetes is a major public health problem, with a prevalence The use of a number of anti-diabetes agents is restricted
that is expected to reach 552 million people worldwide by in patients with renal impairment [5]. Metformin is
2030 [1]. Type 2 diabetes mellitus (T2DM) accounts for 90% contraindicated in patients with renal dysfunction due to the
of all diabetes cases [2]. Most medications for the treatment risk of accumulation and lactic acidosis. Caution is advised
of T2DM act through insulin-dependent mechanisms; the with the use of insulin secretagogues in renally impaired
progressive loss of beta-cell function that is characteristic of patients. The dipeptidyl peptidase-4 (DPP-4) inhibitors
T2DM means that most patients with T2DM ultimately require saxagliptin, sitagliptin and vildagliptin (but not linagliptin) are
multiple therapies to maintain glycaemic control [3–6]. predominantly excreted renally, so dose reduction is necessary
Nephropathy is a well-established complication of poor in patients with advanced CKD [5].
glycaemic control in patients with diabetes. An estimated Empagliflozin is a potent and selective sodium glucose
10–36% of patients with T2DM have some degree of renal cotransporter 2 (SGLT2) inhibitor (IC50 : 3.1 nM; pIC50 ± s.e.:
impairment [7] and chronic kidney disease (CKD) is present 8.5 ± 0.02 nM) with a >2500-fold selectivity for SGLT2
in approximately 40% of patients with diabetes [8]. CKD has over SGLT1 [10]. By blocking SGLT2, empagliflozin blocks
been classified into five stages, where stage 1 is kidney damage reabsorption of glucose from the glomerular filtrate in the
with normal glomerular filtration rate (GFR; ml/min/1.73 m2 ) proximal tubule of the kidney, leading to urinary glucose
of ≥90; stage 2 is kidney damage with a mild decrease in GFR excretion (UGE) [11,12], which results in a reduction in fasting
(GFR 60–89); stage 3 is a moderate decrease in GFR (GFR plasma glucose and glycosylated haemoglobin (HbA1c) in
patients with T2DM [13,14].
Correspondence to: Sreeraj Macha, Clinical Pharmacokinetics and Pharmacodynamics, Single oral doses of empagliflozin (0.5–800 mg) exhibit
Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, linear pharmacokinetics in healthy subjects [11]. After multiple
USA.
E-mail: sreeraj.macha@boehringer-ingelheim.com oral doses (up to 100 mg once daily for 4 weeks) in patients
with T2DM, empagliflozin is rapidly absorbed (reaching peak
original article DIABETES, OBESITY AND METABOLISM

levels after 1.5 h) with a mean terminal elimination half-life The investigators, in cooperation with nephrology centres,
(t 1/2 ) of between 13 and 17 h [12]. Glucuronidation is the major aimed to recruit eight subjects for every group. Subjects with
metabolic pathway for empagliflozin, and no major metabolites normal renal function were matched to those in the renal
of empagliflozin were detected in human plasma (data not impairment groups by age (±5 years) and weight (±15%),
published). Approximately 11–19% of the administered dose where possible.
is excreted unchanged in urine [11]. As renal function
may affect the pharmacokinetics and pharmacodynamics
Study Design
of empagliflozin, this study was undertaken to determine
the effect of renal impairment on the pharmacokinetics, This two-centre, open-label, parallel-group study was under-
pharmacodynamics and safety of a single 50 mg oral dose taken to assess the effect of renal function on the pharma-
of empagliflozin. cokinetics, pharmacodynamics and safety of a single 50 mg
dose of empagliflozin. Subjects were screened for eligibility up
to 21 days prior to study drug administration. Following an
Materials and Methods overnight fast, subjects were admitted to the research unit and
received a 50 mg dose of empagliflozin with 240 ml of water
The study protocol was approved by the independent ethics
(day 1). Water was allowed ad libitum except for 1 h before
committee Ethikkommission Schleswig-Holstein, Bad Sege-
and 1 h after study drug administration. Following medical
berg, Germany and the German Competent Authority—the
surveillance for 24 h, subjects were discharged (day 2) and were
Federal Institute for Drugs and Medical Devices (Bundesinstitut
monitored as outpatients until they attended an end-of-study
für Arzneimittel und Medizinprodukte, BfArM), Bonn, Ger-
examination (within 14 days after the last trial procedure).
many. The study was conducted in compliance with the Decla-
ration of Helsinki (1996) and the International Conference on
Harmonization Good Clinical Practice (ICH-GCP) guidelines. Pharmacokinetic Endpoints
The study was registered with the EU Clinical Trials Register One objective of the study was to determine the effect of renal
(Eudra CT number: 2008-006086-86) and conducted at two impairment on the relative bioavailability of empagliflozin,
German centres: CRS Clinical Research Services Kiel GmbH, based on the primary endpoints of AUC0−∞ (area under the
Kiel and Profil Institut für Stoffwechselforschung GmbH, concentration-time curve of empagliflozin in plasma from time
Neuss. All participants provided written informed consent. 0 extrapolated to infinity) and Cmax (maximum concentration
of empagliflozin in plasma). Secondary pharmacokinetic
Subjects endpoints included: t max (time to peak empagliflozin plasma
levels), t 1/2 , fe0–96 (fraction of the dose excreted in urine
Male and female subjects aged 18–75 years weighing at least following drug administration), CLR,0–96 (renal clearance) and
45 kg (females only) and with a body mass index (BMI) of plasma protein binding of empagliflozin.
18–34 kg/m2 were eligible for inclusion in this study. Partici-
pants with normal renal function (eGFR >90 ml/min/1.73 m2 ;
control) were required to have T2DM. Patients with mild Pharmacodynamic Endpoint
renal impairment (eGFR 60–89 ml/min/1.73 m2 ), moderate The pharmacodynamic endpoint of the study was the
renal impairment (eGFR 30–59 ml/min/1.73 m2 ), severe renal cumulative amount of UGE over a 24 h period following drug
impairment (eGFR <30 ml/min/1.73 m2 ) or renal failure/end administration (UGE0–24 ), relative to baseline, with a baseline
stage renal disease (ESRD) (requiring dialysis) did not need measurement obtained over 24 h preceding administration of
to have T2DM. eGFR was calculated using the Modification study drug.
of Diet in Renal Disease (MDRD) formula: 186 × serum
creatinine−1.154 × age−0.203 × [0.742 if female].
Subjects were excluded from the study if they had recently Safety Endpoints
participated in a study (multiple-dose: within 2 months; single- The safety evaluation was based on monitoring vital signs,
dose: within 1 month), were abusing alcohol (males >60 g/day; 12-lead electrocardiograms (ECGs), physical examinations,
females >40 g/day) or drugs, had donated >100 ml blood in the clinical laboratory tests, adverse events (AEs) and a global
previous 4 weeks, were taking concomitant medications known assessment of tolerability made by the investigator (with
to inhibit or induce p-glycoprotein or cytochrome P450 3A, or options of ‘good’, ‘satisfactory’, ‘not satisfactory’ or ‘bad’). AEs
had any medical or laboratory results deviating from normal were assessed throughout the study, and were coded using the
and of clinical relevance. Medical Dictionary for Drug Regulatory Activities (MedDRA)
Subjects with renal impairment were excluded if they had version 12.1 (http://www.meddramsso.com). Vital signs (blood
significant diseases other than renal impairment or T2DM, pressure and pulse rate), ECG and physical examination
including moderate and severe concurrent hepatic impair- were assessed at the screening visit and at the end-of-study
ment, haemoglobin <8 g/dl indicating severe renal anaemia examination. Vital signs were also measured prior to dosing.
(erythropoietin could be used to maintain haemoglobin lev- Clinical laboratory tests (haematology, clinical chemistry, urine
els), and intake of drugs with a long half-life (>24 h) within the analysis) were conducted at the screening visit, prior to dosing,
previous month (or within 10 half-lives of that drug, if longer), and at the end-of-study examination. Urinary creatinine and
except for those being taken for the treatment of renal disease. glucose were determined in −24 to 0 h and 0 to 24 h fractions.

216 Macha et al. Volume 16 No. 3 March 2014


DIABETES, OBESITY AND METABOLISM original article
Table 1. Demographics and baseline characteristics based on the treated set.

Renal impairment Total


Normal renal function Mild Moderate Severe Renal failure/ESRD
n 8 9 7 8 8 40
Female, n (%) 7 (87.5) 6 (66.7) 3 (42.9) 1 (12.5) 4 (50.0) 21 (52.5)
Age, years
Median (range) 56 (38–67) 61 (42–69) 64 (48–74) 56 (34–74) 40 (36–70) 56 (34–74)
Weight, kg
Median (range) 81.70 (67.5–98.3) 70.20 (60.5–90.3) 78.60 (73.6–101.8) 75.10 (65.2–103.0) 77.25 (63.0–98.0) 78.30 (60.5–103.0)
BMI, kg/m2
Median (range) 28.85 (26.0–33.1) 26.80 (21.7–30.1) 28.10 (24.6–34.0) 23.85 (19.6–32.5) 26.85 (22.1–33.8) 27.65 (19.6–34.0)

ESRD, end stage renal disease; BMI, body mass index.

A
Empagliflozin plasma concentration (nmol/l)

1500 Normal
Mild
10000 Moderate
Empagliflozin plasma
concentration (nmol/l)

Severe
1000
1000 ESRD
100

10

500 1
0 24 48 72 96
Time (h)

0
0 24 48 72 96
Time (h)

B C

Figure 1. Exposure to a single oral 50 mg dose of empagliflozin in subjects with normal and impaired renal function. (A) Mean plasma concentration-time
profiles (insert: semi-log plot) (n = 40). (B) AUC0 –∞ and (C) Cmax ; midline of boxes are medians, and boundaries are 25th and 75th percentiles; whiskers
are the standard span for the quartiles (1.5 × interquartile range).

Volume 16 No. 3 March 2014 doi:10.1111/dom.12182 217


original article DIABETES, OBESITY AND METABOLISM

Sample Collection and Analysis concentrations. The AUC0−∞ value was estimated as the
sum of AUC to the last measured concentration, with the
Approximately 124 ml of blood was taken from every subject
extrapolated area given by the quotient of the last measured
over the course of the study for clinical laboratory tests (44 ml),
concentration and λz . The amount of drug (Ae ) excreted
pharmacokinetic assessments (50 ml) and determination of
unchanged in urine in each collection interval was determined
protein binding (30 ml). Blood samples for clinical laboratory
by the product of the urine concentration and the urine
testing were collected after subjects had fasted for at least 10 h.
volume. The fraction of the dose (f e ) that was excreted
For quantification of empagliflozin plasma concentrations,
unchanged in urine was determined by the quotient of the
2.7 ml of blood was taken from a forearm vein in a K3 -EDTA
sum of drug excreted over all dosing intervals and the dose
(tripotassium ethylenediaminetetraacetic acid)-anticoagulant
administered. CLR was determined as the quotient of Ae
blood drawing tube at pre-dose and 0.33, 0.67, 1, 1.5, 2, 2.5,
over AUC. Cumulative UGE was calculated using the glucose
3, 4, 6, 8, 10, 12, 14, 24, 36, 48, 72 and 96 h post-dose. Within
concentration measured in every urine sample collected
30 min of collection, the samples were centrifuged for 10 min
from −24 to 0 h and 0 to 96 h after dosing. UGE (mg) was
at 2000–4000 g and 4–8 ◦ C. The EDTA plasma obtained was
calculated as follows: glucose concentration (mg/dl) × urine
stored at −18 ◦ C until it was shipped on dry ice for analysis.
volume (dl).
Urine was collected over 24 h prior to drug administration
and at the following intervals following drug administration:
0–4, 4–8, 8–12, 12–24, 24–36, 36–48, 48–72 and 72–96 h. Statistical Analysis
Urine containers were weighed empty and at the end of every All subjects who provided at least one observation for at least
sampling period and the difference was recorded as the urine one primary endpoint without any protocol violations relevant
volume (weight was set equal to volume, i.e. 1 kg = 1 l). Urine to pharmacokinetic evaluation were included in the analysis
containers were refrigerated until 25 h after drug administration of relative bioavailability (pharmacokinetic analysis set). All
and aliquots were stored at −18 ◦ C until shipped on dry ice for subjects with a baseline UGE value (0–24 h pre-dose) and a
analysis. UGE value from 0 to 24 h post-dose without any protocol
Empagliflozin concentrations in plasma and urine were violations relevant to UGE analysis were included in the UGE
determined by validated high performance liquid chromatog- analysis set. All safety analyses were performed on all subjects
raphy, tandem mass spectrometry (HPLC-MS/MS) assays. who received study drug (treated set).
Empagliflozin and the internal standard [13 C6 ]-empagliflozin One objective of the study was to investigate the relative
were extracted from urine or plasma by supported liquid bioavailability of empagliflozin in subjects with normal renal
extraction. After evaporation under nitrogen, the residue function (R) compared with subjects with various degrees
was reconstituted and analysed using liquid chromatography of renal impairment (T1–4), by presenting point estimators
with MS/MS detection. The lower limit of quantification for [adjusted geometric mean (gMean) T/R ratios] of AUC0–∞
empagliflozin in human plasma was 1.11 nmol/l, with linear- and Cmax and their two-sided 90% confidence intervals
ity to 1110 nmol/l using a sample volume of 0.15 ml, and in (CIs). AUC0–∞ and Cmax were analysed using an analysis
human urine was 4.44 nmol/l, with linearity to 4440 nmol/l of variance (anova) model on the logarithmic scale including
using a sample volume of 0.05 ml. For both plasma and urine, a fixed effect corresponding to the renal function (normal,
results were calculated using peak area ratios and calibration mild impairment, moderate impairment, severe impairment
curves were created using weighted (1/x2 ) quadratic regression. or renal failure/ESRD). The change in UGE 0–24 h from
For determination of protein binding of empagliflozin, 10 ml baseline after drug administration was analysed by an analysis
blood was collected at pre-dose, 1.5 and 3 h post-dose. Blood of covariance (ancova) model including baseline UGE values
was centrifuged for 10 min at 4000 g and 4 ◦ C, and the plasma as a continuous covariate, and a fixed effect corresponding
was stored at −20 ◦ C until it was shipped on dry ice for analysis. to the renal function. Descriptive statistics were calculated for
The binding of empagliflozin and the internal standard [13 C6 ]- all pharmacokinetic and pharmacodynamic parameters. Safety
empagliflozin to human plasma protein was determined by analyses were descriptive in nature.
equilibrium dialysis at 37 ◦ C. As a quality control measure,
protein-binding analyses were performed using the pre-dose
plasma samples. Results
Pharmacokinetic parameters were calculated using
WinNonlinTM software (v5.01, Pharsight Corporation, Subjects
Mountain View, CA, USA). Cmax and t max values were Forty subjects (21 female and 19 male) participated in this
directly determined from the plasma concentration time open-label, parallel-group study: eight with T2DM and
profiles of each subject. The apparent terminal rate constant normal renal function (eGFR >90 ml/min/1.73 m2 ; con-
(λz ) was estimated from a regression of ln(C) versus time trol); nine with T2DM and mild renal impairment (eGFR
over the terminal log-linear drug disposition portion of 60–89 ml/min/1.73 m2 ); seven with T2DM and moderate renal
the concentration-time profiles. The t 1/2 was calculated impairment (eGFR 30–59 ml/min/1.73 m2 ); eight with severe
as the quotient of ln(2) and λz . Area under the plasma renal impairment (eGFR <30 ml/min/1.73 m2 ), four of whom
concentration-time curve to the last time point (AUC0–tz ) was also had T2DM; and eight subjects with renal failure/ESRD
calculated using the linear trapezoidal method for ascending requiring dialysis, none of whom had T2DM. Subjects in the
concentrations and the log trapezoidal method for descending group with normal renal function were matched to those in the

218 Macha et al. Volume 16 No. 3 March 2014


DIABETES, OBESITY AND METABOLISM original article
Table 2. Pharmacokinetic and pharmacodynamic parameters for empagliflozin after administration of a single oral 50 mg dose.

Renal impairment
Normal
renal function Mild Moderate Severe Renal failure/ESRD
Pharmacokinetics n=8 n=9 n=7 n=8 n=8
AUC0–∞ , nmol × h/l 10 600 (16.4) 12 700 (20.8) 13 000 (25.1) 17 700 (17.8) 16 600 (38.7)
Cmax, nmol/l 1240 (23.5) 1500 (29.4) 1290 (37.9) 1520 (31.6) 1290 (27.5)
t max , h* 1.0 (1.0–3.0) 2.5 (2.0–4.0) 2.0 (1.5–3.0) 2.0 (0.7–4.0) 2.5 (1.5–3.0)
t 1/2 , h 19.9 (58.8) 24.6 (84.5) 23.8 (87.9) 27.9 (76.8) 22.0 (74.3)
f e0–96 , % 16.1 (26.7) 11.7 (36.4) 7.7 (70.1) 3.6 (36.1) 0.3 (56.4)
CLR,0–96 , ml/min 28.5 (20.5) 18.6 (46.9) 11.8 (69.6) 4.0 (30.6) 0.5 (59.1)
Pharmacodynamics n=8 n=7 n=5 n=6 n=5
UGE0–24 , baseline, g 4.49 (2.88) 4.16 (3.08) 0.97 (0.49) 0.97 (0.43) 2.16 (2.00)
UGE0–24 , g 102.13 (7.92) 65.75 (7.31) 56.64 (17.13) 19.22 (4.04) 2.93 (1.28)
Change from baseline, g 97.64 (7.20) 61.59 (6.89) 55.67 (16.89) 18.25 (3.93) 0.78 (0.90)

Pharmacokinetic values are mean (coefficient of variation [%]) unless otherwise stated; pharmacodynamic values are mean (standard error). ESRD, end
stage renal disease; AUC0-∞ , area under the concentration-time curve of empagliflozin in plasma from time 0 extrapolated to infinity; Cmax , maximum
concentration of empagliflozin in plasma; t max , time to peak empagliflozin plasma levels; t 1/2 , terminal elimination half-life; fe0-96 , fraction of the dose
excreted in urine following drug administration; CLR, 0-96 , renal clearance; UGE, urinary glucose excretion.
*Data are median (range).

Figure 2. (A) CLR from 0 to 96 h; (B) AUC0−∞ and (C) cumulative amount of glucose excreted in urine in 24 h versus eGFR after administration of a
single oral 50 mg dose in subjects with normal and impaired renal function (n = 40).

renal impairment groups by age (±5 years) and weight (±15%). were included in the pharmacokinetic analysis set and treated
Demographic and baseline characteristics are shown in set. In total, 31 subjects were included in the UGE analysis set;
Table 1. 6 did not have baseline values and/or values for 24 h following
All 40 entered subjects received a single 50 mg oral dose of dosing (2 subjects each with mild, moderate and severe renal
empagliflozin (Boehringer Ingelheim Pharma GmbH & Co. impairment) and 3 subjects with renal failure/ESRD had no
KG, Biberach, Germany) and completed the study. All subjects urine output.

Volume 16 No. 3 March 2014 doi:10.1111/dom.12182 219


original article DIABETES, OBESITY AND METABOLISM

Table 3. Relative bioavailability of empagliflozin (50 mg qd) in subjects with impaired renal function compared with subjects with normal renal function
(n = 40).

90% CI for adjusted GMR


Renal impairment group Parameter Adjusted GMR (renal impaired/normal group) Lower limit (%) Upper limit (%)
Mild AUC0–∞ 118.2 96.2 145.4
Cmax 118.8 93.6 150.8
Moderate AUC0–∞ 119.9 96.3 149.5
Cmax 102.3 79.3 131.9
Severe AUC0–∞ 166.3 134.4 205.7
Cmax 120.7 94.4 154.3
Renal failure/ESRD AUC0–∞ 148.3 119.9 183.4
Cmax 103.8 81.2 132.6

ESRD, end stage renal disease; CI, confidence interval; GMR, geometric mean ratio.

Figure 3. Mean cumulative amounts of glucose excreted in urine after administration of a single oral 50 mg dose in subjects with normal and impaired
renal function (n = 31).

Pharmacokinetics severe renal impairment and renal failure/ESRD, respectively,


as compared with subjects with normal renal function (Tables
Empagliflozin was rapidly absorbed after oral administration
2, 3, figure 1B). The Cmax was similar in subjects with moderate
in all subjects (figure 1A). The rate of absorption was slightly
renal impairment, renal failure/ESRD and those with normal
slower in subjects with renal impairment compared with
those with normal renal function, with a median t max of renal function and was slightly (∼20%) higher in subjects with
2.0–2.5 h and 1.0 h, respectively (Table 2). After reaching mild or severe renal impairment as compared to subjects with
peak levels, empagliflozin plasma concentrations declined in normal renal function (Table 3, figure 1C).
a biphasic manner with a rapid distribution phase followed
by a slower elimination phase (figure 1A). The mean t 1/2 Protein Binding. The proportion of empagliflozin bound to
of empagliflozin increased slightly with increasing renal plasma proteins ranged from 81.0 to 85.1% at 1.5 h post-
impairment: mean t 1/2 was approximately 20 h in subjects dose, with no relevant differences over time. Renal impairment
with normal renal function and approximately 28 h in those resulted in a slight (∼4%) decrease in the degree of plasma
with severe renal impairment (Table 2). CLR of empagliflozin protein binding, from 85.1% with normal renal function to
following study drug administration decreased with increasing 81.1% with renal failure/ESRD at 1.5 h post-dose, leading to
renal impairment (Table 2, figure 2A), leading to an increase in an approximate 27% increase in the unbound fraction (f u )
the overall exposure (AUC0–∞ ) of empagliflozin (figure 2B). of empagliflozin in those with renal failure/ESRD; the f u was
There were also decreases in the mean fe0–96 with increasing 14.9% in subjects with renal failure/ESRD and 18.9% in those
renal impairment (Table 2). with normal renal function. A similar reduction in protein
binding was observed in the in vitro protein-binding analysis of
Relative Bioavailability. The AUC0–∞ increased by approx- pre-dose plasma samples, from 83.6% in subjects with normal
imately 18, 20, 66 and 48% in subjects with mild, moderate, renal function to 81.1% in subjects with renal failure/ESRD.

220 Macha et al. Volume 16 No. 3 March 2014


DIABETES, OBESITY AND METABOLISM original article
Pharmacodynamics partially to the changes in apparent volume of distribution.
However, any effect of decreased protein binding on exposure
The cumulative amount of glucose recovered in urine decreased
in subjects with renal impairment could have been offset by
with increasing renal impairment (Table 2, figures 2C, 3).
a rapid decrease in CLR , with an overall effect of increased
The total amount of glucose excreted in urine (change from
exposure in subjects with renal impairment.
baseline) over 24 h was 97.6 g in subjects with normal renal
The pharmacodynamic results of this study indicate that
function, decreasing to approximately 0.8 g in subjects with
treatment with empagliflozin leads to increases in UGE in
renal failure/ESRD (Table 2). Consistent results were observed
patients with mild and moderate renal impairment, as well
in a statistical model with adjustment for renal function and
as in those with normal renal function. This suggests that
UGE at baseline. The decrease in glucose excretion followed
clinically meaningful reductions in plasma glucose levels might
the same pattern as the decreases in the empagliflozin renal
be obtained with empagliflozin even in patients with moderate
clearance with increasing renal impairment (figures 2A, C).
renal impairment (CKD stage 3), although reductions in HbA1c
cannot be predicted solely based on UGE. A potential limitation
Safety and Tolerability of this study is that, although all subjects with normal renal
A total of 6 treatment-emergent AEs were reported by 4 of the function or mild or moderate renal impairment had T2DM,
40 subjects. All were mild in intensity. AEs were considered only four of the eight subjects with severe renal impairment and
drug-related by the investigator in two subjects: erythema and none of the participants with renal failure/ESRD had T2DM.
pruritus in a subject with moderate renal impairment, and Thus, the ability of empagliflozin to increase UGE in patients
pruritus in a subject with renal failure/ESRD. All AEs resolved with T2DM and severe renal impairment or renal failure/ESRD
without treatment by the end of the study. No effects on safety may have been underestimated.
laboratory values, vital signs or ECG parameters were observed. The increasing exposure to empagliflozin with increasing
The investigators rated the global tolerability of empagliflozin renal impairment had no effect on the incidence of AEs. AEs
as ‘good’ for all participants. were reported by one patient with normal renal function
and moderate impairment and two patients with renal
failure/ESRD. The only AEs reported during the study were
Discussion headache, nausea, erythema, pruritus and diarrhoea. No AEs
This study was undertaken to determine the effect of were serious or severe, and all patients recovered by the
different degrees of renal impairment on the pharmacokinetics, end of the trial without therapy. Single dose administration
pharmacodynamics and safety of a single dose of empagliflozin. of empagliflozin in patients with various degrees of renal
Exposure to empagliflozin increased moderately with impairment had no clinically relevant effects on laboratory
increasing renal impairment. The upper limits of 90% CIs values, vital signs or ECG parameters. The safety and efficacy
for both AUC0–∞ and Cmax were outside the standard of empagliflozin in patients with T2DM and renal impairment
bioequivalence boundaries (80–125%) in all renal impairment will be determined by the results of a dedicated 52-week Phase
groups. The effect was greater on the extent of exposure III trial (NCT01164501).
compared with peak levels (AUC0–∞ and Cmax , respectively), In conclusion, the increase in exposure to empagliflozin with
suggesting that the effect was due to decreased clearance increasing renal impairment was moderate, with no significant
of empagliflozin. Indeed, the moderate increase in systemic increase in maximum plasma concentrations. On the basis of
exposure to empagliflozin could be explained by the decrease pharmacokinetics, no dose adjustment is required in patients
in CLR of empagliflozin leading to a slight increase in t 1/2 . with renal insufficiency. Empagliflozin increased UGE in
AUC0–∞ was higher by approximately 18, 20, 66 and 48% subjects with normal renal function and mild or moderate renal
in subjects with mild, moderate, severe renal impairment and impairment and showed little to no effect on UGE in subjects
renal failure/ESRD, respectively, than in those with normal with severe renal impairment or renal failure/ESRD. A single
renal function. Peak plasma levels of empagliflozin in subjects 50 mg dose of empagliflozin was well tolerated in all subjects.
with moderate renal impairment or renal failure/ESRD were
similar to those in subjects with normal renal function, and were
approximately 20% higher in those with mild and severe renal Acknowledgements
impairment compared to subjects with normal renal function. This study was funded by Boehringer Ingelheim. The authors
The pharmacokinetic properties of empagliflozin were largely acknowledge Andreas Port for his contributions related to
unaltered by renal impairment, suggesting that no dose the planning and conduct of the studies, and his support in
adjustments are required in patients with renal impairment. preparing the clinical trial reports and manuscript. The authors
Protein binding of empagliflozin was slightly lower in would also like to thank Lois Rowland for coordinating the
subjects with renal impairment than in those with normal bioanalytical analyses, Dale Sharp for coordinating the protein-
renal function (81.1% in subjects with renal failure/ESRD vs. binding analysis, Tim Heise, Principal Investigator at the second
85.1% in subjects with normal renal function at 1.5 h post- site (Profil, Neuss), Haidar Maatouk, who was the responsible
dose), and the f u of empagliflozin was approximately 27% research physician at CRS and Leszek Nosek, who was the
higher at 1.5 h post-dose in subjects with renal failure/ESRD responsible clinical research physician at the second site (Profil,
compared with those with normal renal function. This slight Neuss). Medical writing assistance, supported financially by
increase in the free fraction of drug could have contributed Boehringer Ingelheim, was provided by Isobel Lever and Wendy

Volume 16 No. 3 March 2014 doi:10.1111/dom.12182 221


original article DIABETES, OBESITY AND METABOLISM

Morris of Fleishman-Hillard Group Ltd during the preparation American Diabetes Association (ADA) and the European Association for the
of this manuscript. The authors were fully responsible for all Study of Diabetes (EASD). Diabetes Care 2012; 35: 1364–1379.
content and editorial decisions, were involved at all stages of 6. Turner RC, Cull CA, Frighi V, Holman RR. Glycemic control with diet,
manuscript development and have approved the final version. sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus:
progressive requirement for multiple therapies (UKPDS 49). UK Prospective
Diabetes Study (UKPDS) Group. JAMA 1999; 281: 2005–2012.
Conflict of Interest 7. Coll-de-Tuero G, Mata-Cases M, Rodriguez-Poncelas A et al. Chronic kidney
S. M., M. M., S. P., H. J. W. and U. C. B. are employees of disease in the type 2 diabetic patients: prevalence and associated variables
Boehringer Ingelheim. A. H. has no conflict of interests to in a random sample of 2642 patients of a Mediterranean area. BMC Nephrol
disclose. 2012; 13: 87.
The authors meet criteria for authorship as recommended 8. Koro CE, Lee BH, Bowlin SJ. Antidiabetic medication use and prevalence of
by the International Committee of Medical Journal Editors chronic kidney disease among patients with type 2 diabetes mellitus in
the United States. Clin Ther 2009; 31: 2608–2617.
(ICMJE). S. M., M. M. and S. P. made substantial contributions
to the conception and design of the study and were involved 9. Eckardt KU, Berns JS, Rocco MV, Kasiske BL. Definition and classification of
in the analysis and interpretation of data. A. H. was involved CKD: the debate should be about patient prognosis - a postion statement
from KDOQI and KDIGO. Am J Kidney Dis 2009; 53: 915–920.
in the acquisition and interpretation of data. U. C. B. was
involved in the interpretation of the data. All authors were 10. Grempler R, Thomas L, Eckhardt M et al. Empagliflozin, a novel selective
sodium glucose cotransporter-2 (SGLT-2) inhibitor: characterisation and
involved with drafting the article or revising it critically for
comparison with other SGLT-2 inhibitors. Diabetes Obes Metab 2012; 14:
important intellectual content and all authors have approved 83–90.
the final version.
11. Seman L, Macha S, Nehmiz G et al. Empagliflozin (BI 10773), a potent and
selective SGLT-2 inhibitor, induces dose-dependent glucosuria in healthy
References subjects. Clin Pharm in Drug Dev 2013; 2: 152–161.
12. Heise T, Seewaldt-Becker E, Macha S et al. Safety, tolerability,
1. Whiting DR, Guariguata L, Weil C, Shaw J. IDF Diabetes Atlas: global pharmacokinetics and pharmacodynamics following 4 weeks’ treatment
estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res with empagliflozin once daily in patients with type 2 diabetes. Diabetes
Clin Pract 2011; 94: 311–321. Obes Metab 2013; 15: 613–621.
2. WHO. Ten facts about diabetes. Available from URL: http://www.who.int/ 13. Ferrannini E, Seman L, Seewaldt-Becker E, Hantel S, Pinnetti S, Woerle HJ.
features/factfiles/diabetes/facts/en/index4.html. Accessed 24 Septem- A phase IIb, randomised, placebo-controlled study of the SGLT2 inhibitor
ber 2012. empagliflozin in patients with type 2 diabetes. Diabetes Obes Metab 2013;
3. Cefalu WT. Evolving treatment strategies for the management of type 2 15: 721–728.
diabetes. Am J Med Sci 2012; 343: 21–26. 14. Rosenstock J, Jelaska A, Seman L, Pinnetti S, Hantel S, Woerle HJ. Efficacy and
4. DeFronzo RA. Overview of newer agents: where treatment is going. Am J safety of BI 10773, a new sodium glucose cotransporter (SGLT-2) inhibitor,
Med 2010; 123: S38–S48. in type 2 diabetes inadequately controlled on metformin. Diabetes 2011;
5. Inzucchi SE, Bergenstal RM, Buse JB et al. Management of hyperglycemia 60: A271[989-P].
in type 2 diabetes: a patient-centered approach: position statement of the

222 Macha et al. Volume 16 No. 3 March 2014

You might also like