ArticlePDF Available

Biochemical alterations and liver toxicity analysis with pioglitazone in healthy subjects

Authors:

Abstract and Figures

Pioglitazone, a member of the thiazolidinediones, is a potent, highly selective agonist for peroxisome proliferator-activated receptor gamma and is an excellent insulin sensitizer used in treating type 2 diabetes mellitus. The present study investigated the effect of pioglitazone on glucose, total cholesterol, triglyceride, low-density lipoprotein (LDL) cholesterol and high density lipoprotein (HDL) cholesterol, total proteins, albumin (ALB), alanine transaminase (ALT), and aspartate transaminase (AST) levels in 20 healthy Bengali male volunteers in a randomized, placebo-controlled study. Blood samples were collected before and 0.5-24.0 hours after a single oral dose of a 30 mg pioglitazone tablet. Plasma pioglitazone level was determined using a validated method of reverse-phase binary high-performance liquid chromatography. Blood lipid profile and levels of glucose, ALT, and AST were estimated using enzyme assay kits, plasma protein level was estimated by the biuret method, and plasma ALB level was determined colorimetrically. No significant change in blood glucose, total proteins, total cholesterol, triglyceride, HDL, and LDL levels was observed over the 24-hour assessment period, indicating no plasma biochemical alterations. There were no significant differences between baseline and 24-hour values of ALB, ALT, and AST levels, indicating a lack of liver toxicity. Our results indicate that a single dose of 30 mg of pioglitazone has no hypoglycemic or hypolipidemic effect or liver toxicity within 24 hours of treatment among healthy Bengali males.
Content may be subject to copyright.
1
Introduction
e prevalence of diabetes, especially type 2 diabetes
mellitus (T2DM), has been increasing rapidly, imposing
one of the most challenging public health problems of
the 21st century to Bangladesh and the world. ere is no
known fail-safe method of preventing T2DM. e treat-
ment goals for T2DM include eectively controlling blood
glucose level and maintaining a healthy blood pressure
and lipid prole to avert the serious complications result-
ing from a sustained tissue exposure to excess glucose.
A number of agents have been used in treating T2DM,
but more eective new drugs are necessary (Ripsin et
al., 2009). Some of the new agents include peroxisome
proliferator-activated receptor gamma PPAR-γ agonists,
such as pioglitazone and rosiglitazone. Comprehensive
information on the mechanism(s) of action, ecacy,
pharmacokinetics, pleiotropic eects, drug interactions,
and adverse eects of such drugs are essential. Benecial
or neutral eects on body weight are some of the attrac-
tive features of the new drugs. However, the higher cost
and lack of adequate long-term safety and clinical out-
come data for the agents remain of concern.
Pioglitazone is a thiazolidinedione (TZD) derivative
and a novel oral hypoglycemic agent for the management
of T2DM (Arono et al., 2000). Insulin resistance and
hyperinsulinemia play important roles on the pathogenesis
of T2DM. Hyperinsulinemia is an independent risk
factor for cardiovascular diseases (Uwaifo and Ratner,
2003; Despres et al., 1996). Pioglitazone activates the
transcription of insulin-responsive genes and thus
increases insulin sensitivity (Gillies and Dunn, 2000). e
drug stimulates the uptake of glucose and fatty acids by
RESEARCH ARTICLE
Biochemical alterations and liver toxicity analysis with
pioglitazone in healthy subjects
Sajal Kumar Saha1, Sreedam Chandra Das1, Abdullah-Al-Emran2, Mithun Sarker1,
Md Aftab Uddin2, A.K. Azad Chowdhury1, and Sitesh Chandra Bachar3
Departments of 1Clinical Pharmacy and Pharmacology,2Genetic Engineering and Biotechnology, and
3Pharmaceutical Technology, University of Dhaka, Dhaka, Bangladesh
Abstract
Pioglitazone, a member of the thiazolidinediones, is a potent, highly selective agonist for peroxisome proliferator-
activated receptor gamma and is an excellent insulin sensitizer used in treating type 2 diabetes mellitus. The present
study investigated the eect of pioglitazone on glucose, total cholesterol, triglyceride, low-density lipoprotein (LDL)
cholesterol and high density lipoprotein (HDL) cholesterol, total proteins, albumin (ALB), alanine transaminase (ALT),
and aspartate transaminase (AST) levels in 20 healthy Bengali male volunteers in a randomized, placebo-controlled
study. Blood samples were collected before and 0.5–24.0 hours after a single oral dose of a 30 mg pioglitazone tablet.
Plasma pioglitazone level was determined using a validated method of reverse-phase binary high-performance liquid
chromatography. Blood lipid prole and levels of glucose, ALT, and AST were estimated using enzyme assay kits,
plasma protein level was estimated by the biuret method, and plasma ALB level was determined colorimetrically. No
signicant change in blood glucose, total proteins, total cholesterol, triglyceride, HDL, and LDL levels was observed
over the 24-hour assessment period, indicating no plasma biochemical alterations. There were no signicant
dierences between baseline and 24-hour values of ALB, ALT, and AST levels, indicating a lack of liver toxicity. Our
results indicate that a single dose of 30 mg of pioglitazone has no hypoglycemic or hypolipidemic eect or liver
toxicity within 24 hours of treatment among healthy Bengali males.
Keywords: Biochemical alterations, Bengali population, liver toxicity, pioglitazone, PPAR-γ
Address for Correspondence: Sajal Kumar Saha, Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Curzon Hall,
Dhaka 1000, Bangladesh; Fax: +880 28615583; E-mail: sajal331@yahoo.com
(Received 20 November 2011; revised 15 January 2012; accepted 16 January 2012)
Drug and Chemical Toxicology, 2012; Early Online: 1–6
© 2012 Informa Healthcare USA, Inc.
ISSN 0148-0545 print/ISSN 1525-6014 online
DOI: 10.3109/01480545.2012.658920
Drug and Chemical Toxicology
00
00
1
6
20November2011
15January2012
16January2012
0148-0545
1525-6014
© 2012 Informa Healthcare USA, Inc.
10.3109/01480545.2012.658920
2012
Pioglitazone in healthy subjects
S. K. Saha et al.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
2 S. K. Saha et al.
Drug and Chemical Toxicology
promoting the expression of cellular glucose and fatty acid
transporters (Gillies and Dunn, 2000). Similar to other
TZDs, pioglitazone ameliorates insulin resistance without
stimulating insulin release by the pancreatic β cells, thus
lowering the risk of hypoglycemia (Madan, 2005). It has
been demonstrated that pioglitazone improves glycemic
control and glycated hemoglobin, fasting glucose and
high-density lipoprotein (HDL) levels, and signicantly
decreases triglyceride (Tg) level without eecting total
cholesterol and low-density lipoprotein (LDL) levels
(Dormandy et al., 2005).
A measurable level of pioglitazone appears in the
plasma of fasting subjects within 30 minutes of oral
administration and peak plasma concentration and is
attained within 2 hours (Eckland and Danhof, 2000;
Baba, 2001). e absolute bioavailability of the drug
ranges between 70 and 96%, with a mean value of 83%
(Hanefeld, 2001). Over 99% of plasma pioglitazone is
found to be associated with plasma proteins, mainly to
plasma albumin (ALB) (Li, 2006). After a single-dose
administration, the mean apparent volume of distribu-
tion of pioglitazone was observed at 0.63 ± 0.41 L/kg of
body weight and the drug was excreted primarily in the
form of metabolites and their conjugates (Waugh et
al., 2006). Identication of these metabolites and their
routes of excretion will be helpful in deciphering the
mechanism of pioglitazone clearance (Torii et al., 1984).
e above summary indicates that much is known on the
pharmacokinetics of the drug.
Bangladesh has over 3.2 million diabetic patients
(Wild et al., 2004) and pioglitazone has been prescribed
alone or with other drugs in Bangladesh since 1999, but
there are no data on the clinical eect of the drug on
Bengali populations. e present work was designed to
study the eect of pioglitazone on hypoglycemic activity,
hypolipidemic activity, and liver toxicity among healthy
Bangladeshi subjects.
Methods
Study subjects
Twenty healthy Bengali male adult volunteers (mean
age, 23.93 ± 2.73 year; range, 20–30; mean body weight,
61.40 ± 7.98 kg; range, 58–70; mean height, 164.93 ± 4.87
cm; range, 166–185; mean body mass index, 22.57 ± 1.47
kg/m2) were enrolled in this study. Before enrollment,
each subject was screened for good health through a
routine physical checkup and laboratory tests through
qualied healthcare providers. None of the volunteers
used any medications, including the test drug, within 2
weeks before and throughout the study. Exclusion cri-
teria included any history of signicant gastrointestinal
conditions that could potentially impair the absorption
or disposition of the drug, previous history of allergy to
any medications, donation of blood or plasma within 30
days preceding the study, and the use of the investiga-
tional agent within 30 days of the study. Volunteers were
asked to abstain from smoking and from taking alcohol
or caeine for at least 48 hours before and throughout
the study. Volunteers were informed of the risks, ben-
ets, procedures, and aims of the study as well as their
rights as research subjects. Informed signed consent was
obtained from each of the volunteers, and the study pro-
tocol was approved by the Institutional Review Board of
the Department of Clinical Pharmacy and Pharmacology
at the University of Dhaka (Dhaka, Bangladesh).
Reagent and chemicals
Pioglitazone hydrochloride (99.98% purity), for use as a
reference standard, and rosiglitazone (99.96% purity),
for use as an internal standard, were purchased from Dr.
Reddy’s Laboratories Ltd. (Hyderabad, India). Sodium
dihydrogen phosphate, disodium hydrogen phosphate,
and glacial acetic acid were of analytical reagent grade.
All solvents were of high-performance liquid chromatog-
raphy (HPLC) grade and were obtained from Scharlau
(Sentmenat, Spain). Reagent-grade water was obtained
from the Center of Excellence at the University of Dhaka
(Dhaka, Bangladesh).
Chromatography
e HPLC system consisted of a Shimadzu prominence
module with ultraviolet-visible (UV/vis) (SPD20AVP;
Shimadzu) detector (Shimadzu, Kyoto, Japan).
Chromatographic separation was carried out on a Luna
C18 (250 × 4.6 mm) column (Phenomenex, Torrence,
California, USA). e mobile phase was comprised of
acetonitrile and 20 mM of ammonium acetate buer (pH
4.5 ± 0.2; 60:40, v/v). All separations were performed at a
ow rate of 1.0 mL/min. Injection volume was 20 μL, and
the column was maintained at an ambient temperature.
Peaks were determined using a UV detector set at a wave-
length of 269 nm.
Blood collection
Venous blood samples (6.0 mL) were collected in hepa-
rinized tubes immediately before and 0.5, 1.0, 1.5, 2.0,
2.5, 3.0, 5.0, 8.0, 12.0, and 24.0 hours after drug adminis-
tration. Plasma was separated by centrifuging the tubes
at 1,000 relative centrifugal force (RCF) for 20 minutes.
Plasma samples were stored at –20°C until analysis.
Detection and quantification of analytes
e level of the drug in the plasma samples was deter-
mined using HPLC. Concentrations of the other com-
ponents of plasma samples were determined using the
following methods: total protein by the biuret method,
ALB by the colorimetric method, glucose by the glu-
cose-oxidase method, and alanine transaminase (ALT),
aspartate transaminase (AST), and lipid prole (total
cholesterol, Tg, and HDL and LDL) using appropriate
enzymatic assay kits and a biochemical analyzer.
Preparation of stock and standard solutions
Base stock solutions (200 µg/mL) of pioglitazone and the
internal standard were prepared separately in dimethyl
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
Pioglitazone in healthy subjects 3
© 2012 Informa Healthcare USA, Inc.
sulfoxide (DMSO). e stock solution was diluted as
necessary using the mobile phase as the diluent. Working
standard solutions of pioglitazone were prepared by
mixing the secondary standard solutions with drug-free
human plasma to a nal pioglitazone concentration of
0, 0.05, 0.10, 0.25, 0.50, 1.0, and 2.0 μg/mL. e working
internal standard solution with a concentration of 0.05
μg/mL was prepared by diluting the stock solution with
the mobile phase.
Preparation of samples
Plasma (1.0 mL) was mixed with 0.25 mL of 0.1 mol/L of
K2HPO4 in a 10 mL glass tube, and the tube was applied
on a vortex mixer for 30 seconds. An aliquot of 5.0 mL of
ethyl acetate was added, and the tube was applied again
to a vortex mixer for 3 minutes. e tube was centrifuged
at 1,300 RCF for 6 minutes. A sample of the supernatant
(4.0 mL, exclusively the organic phase) was removed to
a separate tube, and the uid phase was evaporated by
placing the tube in a 45°C water bath under a stream of
nitrogen gas. e dried residue was completely dissolved
in 0.1 mL of DMSO. A 20 μL volume of the solution was
injected onto the HPLC column.
Bioanalytical method validation
Linearity
e calibration curve was linear over the range of 0.05–
2.0 μg/mL in human plasma. e linear equation was
typically Y = 64,510 X –1,128.6, r2 = 0.9995 [Y = peak area
and X = concentration (μg/mL)]. e extracted recovery
was >80%, and relative standard deviation for intra- and
interday assay was less than 10%. Limit of quantication
was 0.05 μg/mL.
Accuracy and precision
Accuracy and precision (i.e., intra- and interday) was
ascertained on the basis of quality-control samples (QCs)
analysis. A result obtained from six replicate injections of
the QCs in plasma is summarized in Table 1.
Recovery
Recovery was examined from QCs for low-, medium-,
and high-concentration ranges in plasma samples.
Recovery was expressed as the percentage of analytes
recovered by the assay. In plasma, average recovery was
>80%. e high recovery conrmed the suitability of the
method for analysis of pioglitazone in the given samples.
Statistical analyses
e values for the control and test subjects were compared
using analysis of variance (paired t-test), followed by least
signicant dierence analysis using the SPSS software
bundle (Dublin, Ireland). Results were expressed as mean
± standard deviation, where P ≤ 0.05 was considered sig-
nicant and P ≤ 0.01 was considered highly signicant.
Results
e plasma concentration of pioglitazone rose rapidly and
peaked at 2.514 ± 0.735 hours (tmax) post-administration. e
peak plasma concentration (Cmax) was 1.117 ± 0.315 μg/mL
(Figure 1). e plasma drug concentration declined rapidly
for aproximately 2 hours after the Cmax and then slowly for
the next 9 hours to 0.109 ± 0.059 μg/mL at 24 hours past the
time of administration of the single dose (Figure 1A).
To investigate whether the single dose of the drug
aected the metabolic homeostasis of the subjects,
Table 1. Accuracy and precision in determining plasma pioglitazone concentrations using HPLC based on six replicate injections
reecting intra- and interday variations.
Intraday Interday
Cadded (µg/mL) Cobs (µg/mL) RSD Recovery (%) Cobs (µg/mL) RSD Recovery (%)
0.05 0.043 ± 0.01 1.98 80.75 0.039 ± 0.01 2.56 79.28
0.10 0.031 ± 0.01 1.76 82.10 0.080 ± 0.01 1.74 80.36
0.50 0.400 ± 0.02 2.20 80.64 0.390 ± 0.01 1.62 79.53
Figure 1. Pattern of changes of plasma concentrations of pioglitazone, glucose, and total proteins over a 24-hour assessment period. (A)
Change of plasma pioglitazone and glucose concentration. (B) Change of plasma pioglitazone and total protein concentration. Values at
the 0 time point indicate the concentrations of pioglitazone, glucose, and total proteins before drug administration.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
4 S. K. Saha et al.
Drug and Chemical Toxicology
plasma glucose and total protein levels were determined
concomitantly. Plasma glucose concentration increased
slightly and peaked at 8 hours post–drug administration
(most likely because of dietary reasons) before returning
to the base level (Figure 1A). Likewise, plasma protein
concentration increased slightly after drug administra-
tion and peaked at 3 hours and then decreased to baseline
(Figure 1B). e slight wavy nature of the plasma protein
concentration curve, both in test and control subjects
(Figures 2B), was most likely a result of the limitations of
the biuret method of protein assay. Overall, there was no
signicant dierence in plasma glucose and total protein
concentrations of the test subjects and the control sub-
jects (Figure 2).
To investigate whether administration of pioglitazone
caused any alterations in lipid prole, the prole at
baseline and 24 hours after administration of the drug
were examined. Table 2 shows that there was no sig-
nicant dierence in total cholesterol (P < 0.472), Tg
(P < 0.814), HDL (P < 0.600), and LDL (P < 0.098) levels in
the pre- and post-treatment samples. Finally, to investi-
gate whether administration of the drug caused any hep-
atotoxicity, levels of two important indicator enzymes in
the pretreatment and 24-hour post-treatment samples
were determined. As shown in Table 2, there was no sig-
nicant change in the levels of ALT (P < 0.485) and AST
(P < 0.053) in the two types of samples. Notably, ALB
binds the majority of plasma pioglitazone, but there was
no signicant change in plasma ALB levels (P < 0.650) in
the pretreatment and 24-hour post-treatment samples
(Table 2).
Discussion and conclusion
Success of a drug and a drug delivery system depend
on the fate of the drug in the human body (Tsuchida
and Abe, 1982). A large body of literature has identied
inappropriate pharmacokinetics as the major cause
for the failure of drug development projects (Prentis
et al., 1988). Appropriate consideration regarding
safety and ecacy of a drug requires an analysis of
the links between systemic exposure and eects of the
drug (Walker, 2004). Hereditary dierences aect drug
metabolism and thus play a role as a major determinant
of variable drug exposure and response (Flockhart and
Desta, 2009). Yet, many drugs are developed, tested, and
formulated in one country and then exported and used
in another. Besides genetic dierences, environmental
dierences may also play important roles in drug
metabolism, exposure, and response (Belle and Singh,
2008). erefore, there is a need for studying the
biodisposition of drugs in target populations residing in
diverse environments.
Here, we studied the biodisposition of pioglitazone,
an imported drug in Bangladesh, using healthy Bengali
volunteers as the test subjects. In this population, the
Cmax of the drug was observed at 1.117 ± 0.315 μg/mL
and the tmax was 2.54 ± 0.735 hours postadministration
of a single 30-mg tablet (Figure 1). Plasma drug con-
centration was negligible (0.109 ± 0.059 μg/mL) at 24
hours after administration. Wittayalertpanya et al. (2006)
examined the similar parameters on a ai population
and observed a Cmax of 1.14 ± 0.29 μg/mL at the tmax of 2.00
± 1.61 hours. Budde et al. (2003) used a single dose of
45 mg of pioglitazone on a sample of a German popula-
tion and observed a Cmax 1.329 ± 0.667 µg/ mL at the tmax of
2.0 hours (range, 1.0–4.0). ese values are comparable
to the values we observed for the Bengali population.
However, Zhang et al. (2004) observed a Cmax of 1.85 µg/
Table 2. Plasma concentrations of selected metabolites and
enzymes before and 24 hours after a single 30-mg dose of
pioglitazone administration among healthy Bengali subjects.
Parameters Baseline After 24 hours P–value
Glucose (mg/dL) 81.095 ± 5.542 81.513 ± 3.999 0.424
Total cholesterol
(mg/dL)
130.957 ± 21.998 114.042 ± 20.098 0.472
Tg (mg/dL) 100.721 ± 21.819 111.78 ± 31.285 0.814
HDL (mg/dL) 7.691 ± 1.879 7.447 ± 2.316 0.600
LDL(mg/dL) 103.049 ± 23.381 84.238 ± 24.925 0.098
Total proteins
(mg/mL)
67.71 ± 3.330 67.70 ± 3.230 0.440
ALB (g/dL) 5.045 ± 0.030 5.034 ± 0.030 0.650
ALT (U/L) 7.027 ± 1.608 7.550 ± 1.607 0.485
AST (U/L) 6.250 ± 1.927 7.602 ± 1.633 0.053
e high P-values indicate a lack of signicant dierence in each
biochemical parameter.
Figure 2. Change of plasma glucose and total protein concentrations between test and control subjects before and up to 24 hours after
administration of pioglitazone. (A) Comparison of glucose concentration changes between control and test group. (B) Comparison of
protein concentration changes between control and test group.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
Pioglitazone in healthy subjects 5
© 2012 Informa Healthcare USA, Inc.
mL at the tmax of 1.80 ± 0.6 hours using the same dose in
Chinese population. e observation reects that there
is indeed a dierence in pioglitazone metabolism among
dierent ethnic groups.
e safety of the oral administration of pioglitazone in
humans has been examined in acute settings previously,
and no signicant adverse eect has been observed
(Erdmann et al., 2007; Hanefeld et al., 2007); however,
a drop in plasma glucose level was observed by some
groups (Miyazaki et al., 2001; Bajaj et al., 2003). In the
present study, we observed no signicant change in
plasma glucose level, irrespective of the plasma drug con-
centration. Plasma glucose level changed slightly during
the 24-hour observation period, and maximum (129.471
± 7.414 mg/dL) and minimum (81.513 ± 3.99 mg/dL)
plasma glucose concentrations were recorded at 8 and
24 hours post-administration, respectively (Figure 1A).
e above values were not signicantly dierent from the
values for the control subjects (Figure 2A). e slight dif-
ference in plasma glucose level, both in control and test
groups, was most likely a result of dietary reasons. us,
our results indicate that a single dose of pioglitazone has
no signicant eect on plasma glucose concentration.
A previous report indicated that pioglitazone does not
cause hypoglycemia in nondiabetic animals (Stevenson
et al., 1991).
Because pioglitazone interacts with plasma proteins,
the drug may aect plasma protein levels (Li, 2006).
However, we observed no signicant change in total
plasma protein in the 24-hour observation period after
the administration of the drug in test and control subjects
(Figure 2B). Maximum (68.02 ± 3.33 mg/mL) and
minimum (67.39 ± 3.69 mg/mL) plasma protein levels
were observed at 3 and 5 hours post–drug administration,
respectively (Figure 1B). Drugs used in treating T2DM
may aect lipid metabolism (Reed et al., 1999). However,
we observed no signicant dierences between baseline
values and values 24 hours after administration for
total cholesterol, Tg, and HDL and LDL levels (Table 2).
Many drugs are hepatotoxic, and the toxicity is reected
by the activation of certain liver enzymes. e present
study indicated no signicant changes in the level of
two important indicator enzymes (i.e., ALT and AST)
within the 24-hour study period after administration of
pioglitazone. Although our study detected no eect of
pioglitazone in glucose and lipid metabolism and no
hepatotoxic eect among healthy Bengali males within
24 hours of administration of 30 mg of pioglitazone, it
remains to be observed whether chronic administration
of the drug may aect the above parameters of healthy
subjects and the subjects with T2DM.
Acknowledgments
e authors are grateful to Professor Ruhul H. Kudddus
of Utah Valley University (Orem, Utah, USA), for critically
reviewing the article, and the volunteers who partici-
pated in this study.
Declaration of interest
is work was supported solely through internal fund-
ing of the Departments of Clinical Pharmacy and
Pharmacology, Genetic Engineering and Biotechnology
and Pharmaceutical Technology, University of Dhaka,
(Dhaka, Bangladesh).
References
Arono, S., Rosenblatt, S., Braithwaite, S., Egan, J. W., Mathisen, A. L.,
Schneider, R. L. (2000). Pioglitazone hydrochloride monotherapy
improves glycemic control in the treatment of patients with type
2 diabetes: a 6-month randomized placebo-controlled dose-
response study. e pioglitazone 001 study group. Diabetes Care
23:1605–1611.
Baba, S. (2001). Pioglitazone: a review of Japanese clinical studies.
Curr Med Res Opin 17:166–189.
Bajaj, M., Suraamornkul, S., Pratipanawatr, T., Hardies, L. J.,
Pratipanawatr, W., Glass, L., et al. (2003). Pioglitazone reduces
hepatic fat content and augments splanchnic glucose uptake in
patients with type 2 diabetes. Diabetes 52:1364–1370.
Belle, D. J., Singh, H. (2008). Genetic factors in drug metabolism. Am
Fam Physician 77:1553–1560.
Budde, K., Neumayer, H. H., Fritsche, L., Sulowicz, W., Stompor,
T., Eckland D. (2003). e pharmacokinetics of pioglitazone
in patients with impaired renal function. Br J Clin Pharmacol
55:368–374.
Despres, J. P., Lamarche, B., Mauriege, P., Cantin, B., Dagenais,
G. R., Moorjani, S., et al. (1996). Hyperinsulinemia as an
independent risk factor for ischemic heart disease. N Engl J Med
334:952–957.
Dormandy, J. A., Charbonnel, B., Eckland, D. J. A., Erdmann, E.,
Massi- Benedetti, M., Moules, I. K, et al. (2005). Secondary
prevention of macro vascular events in patients with type 2
diabetes in the PROactive study (Prospective pioglitazone clinical
trial in macroVascular events): a randomized controlled trial.
Lancet 366:1279–1289.
Eckland, D. A., Danhof, M. (2000). Clinical pharmacokinetics
of pioglitazone. Exp Clin Endocrinol Diabetes 108 (Suppl
2):S234–S242.
Erdmann, E., Dormandy, J. A., Charbonnel, B., Massi-Benedetti, M.,
Moules, I. K., Skene, A. M. (2007). e eect of pioglitazone on
recurrent myocardial infarction in 2,445 patients with type 2
diabetes and previous myocardial infarction. J Am Coll Cardiol
49:1772–1780.
Flockhart, D. A., Desta, Z. (2009). Pharmacogenetics of drug meta-
bolism. In: Robertson, D., Williams, G. H. (Eds.), Clinical and
translational science: principles of human research (pp. 301–317).
San Diego, California, USA: Elsevier.
Gillies, P. S., Dunn, C. J. (2000). Pioglitazone. Drugs 60:333–343.
Hanefeld, M. (2001). Pharmacokinetics and clinical ecacy of
pioglitazone. Int J Clin Pract Suppl 121:19–25.
Hanefeld, M., Marx, N., Pfützner, A., Baurecht, W., Lübben, G.,
Karagiannis, E., et al. (2007). Anti-inammatory eects of
pioglitazone and/or simvastatin in high cardiovascular risk
patients with elevated high sensitivity C-reactive protein. J Am
Coll Cardiol 49:290–297.
Li, J. (2006). Peroxisome proliferator-activated receptor (PPAR)
agonists for type 2 diabetes. In: Johnson, D. S., Li, J. J. (Eds.), e
art of drug synthesis (pp. 115–127). Hoboken, New Jersey, USA:
John Wiley and Sons.
Madan, P. (2005). Eect of thiazolidinediones on lipid prole. CMAJ
173:344–345.
Miyazaki, Y., Mahankali, A., Matsuda, M., Glass, L., Mahankali, S.,
Ferrannini, E., et al. (2001). Improved glycemic control and
enhanced insulin sensitivity in type 2 diabetic subjects treated
with pioglitazone. Diabetes Care 24:710–719.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
6 S. K. Saha et al.
Drug and Chemical Toxicology
Prentis, R. A., Lis, Y., Walker, S. R. (1988). Pharmaceutical innovation
by seven UK-owned pharmaceutical companies (1964–1985). Br J
Clin Pharmacol 25:387–396.
Reed, M. J., Meszaros, K., Entes, L. J., Claypool, M. D., Pinkett, J. G.,
Brignetti, D., et al. (1999). Eect of masoprocol on carbohydrate
and lipid metabolism in a rat model of type II diabetes.
Diabetologia 42:102–106.
Ripsin, C. M., Kang, H., Urban, R. J. (2009). Management of
blood glucose in type 2 diabetes mellitus. Am Fam Physician
79:29–36.
Stevenson, R. W., McPherson, R. K., Genereux, B. E., Danbury,
B. H., Kreutter, D. K. (1991). Antidiabetic agent englitazone
enhances insulin action in nondiabetic rats without producing
hypoglycemia. Metabolism 40:1268–1274.
Torii, H. K., Yoshida, T., Tsukamoto, T., Tanayama, S., (1984).
Disposition in rats and dogs of ciglitazone, a new antidiabetic
agent. Xenobiotica 14:259–268.
Tsuchida, E., Abe, K. (1982). Interactions between macromolecules in
solutions and intermolecular complexes. In: Cantow, H. W., Asta,
G. D., DuSek, K., Ferry, J. D., Fujita, H., Gordon, M., et al. (Eds.),
Advances in polymer science (vol. 45, pp. 1–119). New York:
Springer-Verlag.
Uwaifo, G. I., Ratner, R. E. (2003). e roles of insulin resistance,
hyperinsulinemia, and thiazolidinediones in cardiovascular
disease. Am J Med 115(Suppl. 8A):12S–19S.
Walker, D. K. (2004). e use of pharmacokinetic and pharma-
codynamic data in the assessment of drug safety in early drug
development. Br J Clin Pharmacol 58:6601–6608.
Waugh, J., Keating, G. M., Plosker, G. L., Easthope, S., Robinson, D. L.
(2006). Pioglitazone: a review of its use in type 2 diabetes mellitus.
Drugs 66:85–109.
Wild, S., Bchir, M. B., Roglic, G., Green, A., Sicree, R., King, H. (2004).
Global prevalence of diabetes. Diabetes Care 27: 1047–1053.
Wittayalertpanya, S., Chompootaweep, S., aworn, N. (2006). e
pharmacokinetics of pioglitazone in ai healthy subjects. J Med
Assoc ai 89:2116–2122.
Zhang H, Wang, X. Q., Zhang, X., Zhang, Q., Yin, Q., Li, K. X. (2004).
Study on bioequivalence of pioglitazone hydrochloride tablets in
healthy Chinese volunteers. Asian J Drug Metab Pharmacokinet
4:119–122.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by 203.112.196.90 on 04/10/12
For personal use only.
... TZDs such as pioglitazone and rosiglitazone improve the survival of neurons and glial cells during ischemic damage by reducing inflammation and exhibiting antiatherosclerotic properties [15]. Unfortunately, recent research reports about the hepatotoxicity and cardiotoxicity of TZDs resulting in partial withdrawal of these compounds from the pharmaceutical market [16,17]. ...
Article
Full-text available
Amorfrutin B is a selective modulator of the PPARγ receptor, which has recently been identified as an effective neuroprotective compound that protects brain neurons from hypoxic and ischemic damage. Our study demonstrated for the first time that a 6-h delayed post-treatment with amorfrutin B prevented hypoxia/ischemia-induced neuronal apoptosis in terms of the loss of mitochondrial membrane potential, heterochromatin foci formation, and expression of specific genes and proteins. The expression of all studied apoptosis-related factors was decreased in response to amorfrutin B, both during hypoxia and ischemia, except for the expression of anti-apoptotic BCL2, which was increased. After post-treatment with amorfrutin B, the methylation rate of the pro-apoptotic Bax gene was inversely correlated with the protein level, which explained the decrease in the BAX/BCL2 ratio as a result of Bax hypermethylation. The mechanisms of the protective action of amorfrutin B also involved the inhibition of autophagy, as evidenced by diminished autophagolysosome formation and the loss of neuroprotective properties of amorfrutin B after the silencing of Becn1 and/or Atg7. Although post-treatment with amorfrutin B reduced the expression levels of Becn1, Nup62, and Ambra1 during hypoxia, it stimulated Atg5 and the protein levels of MAP1LC3B and AMBRA1 during ischemia, supporting the ambiguous role of autophagy in the development of brain pathologies. Furthermore, amorfrutin B affected the expression levels of apoptosis-focused and autophagy-related miRNAs, and many of these miRNAs were oppositely regulated by amorfrutin B and hypoxia/ischemia. The results strongly support the position of amorfrutin B among the most promising anti-stroke and wide-window therapeutics.
... Its activity depends on the presence of insulin, however, it does not stimulate insulin secretion nor inhibit glucose absorption (4), moreover, it improves dyslipidemia, hypertension, and insulin sensitivity in the diabetic patients by improving the sensitivity of hepatic and peripheral (muscle) tissue to insulin (5). It is highly bound to plasma proteins and metabolized in the liver inducing six metabolites which have pharmacological activity (hypoglycemic activity) (6). It has a slow clearance from the blood, despite the small volume of distribution with negligible renal elimination (7). ...
Article
Full-text available
Objective(s): Pioglitazone (Actos) is one of the most controversial recent oral antidiabetic drugs. It was originally authorized in the European Union in 2000, and approved as an oral monotherapy for overweight second type of diabetic patients in 2002. It belongs to the thiazolidinedione group which some of its members have been withdrawn from the market due to the hepatotoxicity or cardiotoxicity effects. This study investigates sub-chronic use of pioglitazone induced toxicity in mice by the assessment of renal and liver function tests, cardiac enzymes, and some hematological indices with histological changes of liver, kidney, heart, and bladder. Materials and Methods: 120 albino mice were divided into four groups; 30 in each. The first group (control) received water, second (diabetic) group received alloxan only, while the third and the fourth groups received alloxan with 200 and 400 mg/kg/day of pioglitazone, respectively for 90 days. Results: Prolonged use of pioglitazone induced significant abnormalities of hepatic, renal, and cardiac biomarkers and some hematological indices associated with histopathological changes in the liver, kidney, heart, and bladder that increased based on administered dose. Conclusion: Subchronic use of pioglitazone leads to hepatic, renal, cardiac, hematological, and bladder affection depending on the applied dose.
... Its activity depends on the presence of insulin, however, it does not stimulate insulin secretion nor inhibit glucose absorption (4), moreover, it improves dyslipidemia, hypertension, and insulin sensitivity in the diabetic patients by improving the sensitivity of hepatic and peripheral (muscle) tissue to insulin (5). It is highly bound to plasma proteins and metabolized in the liver inducing six metabolites which have pharmacological activity (hypoglycemic activity) (6). It has a slow clearance from the blood, despite the small volume of distribution with negligible renal elimination (7). ...
Article
Full-text available
Objective(s): Pioglitazone (Actos) is one of the most controversial recent oral antidiabetic drugs. It was originally authorized in the European Union in 2000, and approved as an oral monotherapy for overweight second type of diabetic patients in 2002. It belongs to the thiazolidinedione group which some of its members have been withdrawn from the market due to the hepatotoxicity or cardiotoxicity effects. This study investigates sub-chronic use of pioglitazone induced toxicity in mice by the assessment of renal and liver function tests, cardiac enzymes, and some hematological indices with histological changes of liver, kidney, heart, and bladder. Materials and Methods: 120 albino mice were divided into four groups; 30 in each. The first group (control) received water, second (diabetic) group received alloxan only, while the third and the fourth groups received alloxan with 200 and 400 mg/kg/day of pioglitazone, respectively for 90 days. Results: Prolonged use of pioglitazone induced significant abnormalities of hepatic, renal, and cardiac biomarkers and some hematological indices associated with histopathological changes in the liver, kidney, heart, and bladder that increased based on administered dose. Conclusion: Subchronic use of pioglitazone leads to hepatic, renal, cardiac, hematological, and bladder affection depending on the applied dose
... In common with other TZDs, PTZ ameliorates insulin resistance associated with T2DM without stimulating insulin release from pancreatic β cell, thus lowering the risk of hypoglycemia [12] . A single dose of 30 mg of PTZ has no hypoglycemic or hypolipidemic effect or liver toxicity within 24 h of treatment among healthy Bengali males [13] . The structural formulation of PTZ hydrochloride is ( ±) -5-[p-[2-(5-ethyl-2-pyridyl) ethoxy]-2, 4 thiazolidinedione hydrochloride. ...
... In common with other TZDs, PTZ ameliorates insulin resistance associated with T2DM without stimulating insulin release from pancreatic β cell, thus lowering the risk of hypoglycemia [12] . A single dose of 30 mg of PTZ has no hypoglycemic or hypolipidemic effect or liver toxicity within 24 h of treatment among healthy Bengali males [13] . The structural formulation of PTZ hydrochloride is ( ±) -5-[p-[2-(5-ethyl-2-pyridyl) ethoxy]-2, 4 thiazolidinedione hydrochloride. ...
... In common with other TZDs, PTZ ameliorates insulin resistance associated with T2DM without stimulating insulin release from pancreatic β cell, thus lowering the risk of hypoglycemia [12] . A single dose of 30 mg of PTZ has no hypoglycemic or hypolipidemic effect or liver toxicity within 24 h of treatment among healthy Bengali males [13] . The structural formulation of PTZ hydrochloride is ( ±) -5-[p-[2-(5-ethyl-2-pyridyl) ethoxy]-2, 4 thiazolidinedione hydrochloride. ...
Article
Full-text available
Objective: To assess the in vitro-in vivo correlation of immediate release formulation of pioglitazone 30 mg film coated tablet. Methods: In vitro release data were obtained for test and reference formulation using the USP paddle method (Apparatus 2) at 50 r/min and with the temperature of 37 °C in the dissolution meddium of 0.1 mol/L of hydrochloric acid of pH 1.2. Twelve healthy volunteers were administered both test and reference pioglitazone 30 mg tablet orally and blood samples were collected over 24 h period. In-vitro drug concentration were analyzed by a simple, fast and precise reverse phase binary HPLC method with UV detection to establish a correlation between in-vitro release and in-vivo absorption data. Results: Similarity factor (f2) and dissimilarity factor (f1) were determined for the time intervals of 5, 10, 15, 45, 60, 75, 90, 105 and 120 min and the obtained values were 65.17%, 59.37%, 63.62%, 66.61%, 68.89%, 70.73%, 72.27%, 73.59%, 74.65% and 75.67% for f2 and 9.43%, 9.00%, 5.42%, 3.86%, 3.07%, 2.56%, 2.20%, 1.94%, 1,82% and 1.65% for f1 at respectve time intervals. Mean dissolution time for test and referencee products were obtained at 3.06 and 3.40 min respectively. The f2 and f1 values obtained were within the acceptable range f2 (50%-100%) and f1 (<15%). Conclusions: Comparison of dissolution profiles corroborate that the test and reference formulations are similar and there is no linear in vitro-in vivo correlation.
Article
Full-text available
Here we report a jute endophyte Staphylococcus hominis strain MBL_AB63 isolated from jute seeds which showed promising antimicrobial activity against Staphylococcus aureus SG511 when screening for antimicrobial substances. The whole genome sequence of this strain, annotated using BAGEL4 and antiSMASH 5.0 to predict the gene clusters for antimicrobial substances identified a novel antimicrobial peptide cluster that belongs to the class I lantibiotic group. The predicted lantibiotic (homicorcin) was found to be 82% similar to a reported peptide epicidin 280 having a difference of seven amino acids at several positions of the core peptide. Two distinct peaks obtained at close retention times from a RP-HPLC purified fraction have comparable antimicrobial activities and LC–MS revealed the molecular mass of these peaks to be 3046.5 and 3043.2 Da. The presence of an oxidoreductase (homO) similar to that of epicidin 280- associated eciO or epilancin 15X- associated elxO in the homicorcin gene cluster is predicted to be responsible for the reduction of the first dehydrated residue dehydroalanine (Dha) to 2-hydroxypropionate that causes an increase of 3 Da mass of homicorcin 1. Trypsin digestion of the core peptide and its variant followed by ESI–MS analysis suggests the presence of three ring structures, one in the N-terminal and other two interlocking rings at the C-terminal region that remain undigested. Homicorcin exerts bactericidal activity against susceptible cells by disrupting the integrity of the cytoplasmic membrane through pore formation as observed under FE-SEM.
Data
Full-text available
Objective To assess the in vitro-in vivo correlation of immediate release formulation of pioglitazone 30 mg film coated tablet. Methods In vitro release data were obtained for test and reference formulation using the USP paddle method (Apparatus 2) at 50 r/min and with the temperature of 37 °C in the dissolution medium of 0.1 mol/L hydrochloric acid of pH 1.2. Twelve healthy volunteers were administered both test and reference pioglitazone 30 mg tablet orally and blood samples were collected over 24 h period. In vivo drug concentrations were analyzed by a simple, fast and precise reverse phase binary HPLC method with UV detection to establish a correlation between in vitro release and in vivo absorption data. Results Similarity factor (f2) and dissimilarity factor (f1) were determined for the time intervals of 5, 10, 15, 30, 45, 60, 75, 90, 105 and 120 min and the obtained values were 65.17%, 59.37%, 63.62%, 66.61%, 68.89%, 70.73%, 72.27%, 73.59%, 74.65% and 75.67% for f2 and 9.43%, 9.00%, 5.42%, 3.86%, 3.07%, 2.56%, 2.20%, 1.94%, 1.82% and 1.65% for f1 at respective time intervals. Mean dissolution time for test and reference products were obtained at 3.06 and 3.40 min respectively. f2 and f1 values obtained were within the acceptable range f2 (50%-100%) and f1 (<15%). Conclusions Comparison of dissolution profiles corroborate that the test and reference formulations are similar and there is no linear in vitro-in vivo correlation.
Article
Full-text available
Pioglitazone is an antihyperglycaemic agent that, in the presence of insulin resistance, increases hepatic and peripheral insulin sensitivity, thereby inhibiting hepatic gluconeogenesis and increasing peripheral and splanchnic glucose uptake Pioglitazone is generally well tolerated, weight gain and oedema are the most common emergent adverse events, and there are no known drug interactions between pioglitazone and other drugs. In clinical trials in patients with type 2 diabetes mellitus, pioglitazone as monotherapy, or in combination with metformin, repaglinide, insulin or a sulfonylurea, induced both long- and short-term improvements in glycaemic control and serum lipid profiles. Pioglitazone was also effective in reducing some measures of cardiovascular risk and arteriosclerosis. Pioglitazone thus offers an effective treatment option for the management of patients with type 2 diabetes. Pharmacological Properties Pioglitazone activates a specific nuclear receptor, the peroxisome-proliferator activated receptor-γ, which increases insulin sensitivity in liver, fat and skeletal muscle cells, increases peripheral and splanchnic glucose uptake and decreases hepatic glucose output. Pioglitazone is dependent on the presence of insulin in order to exert its beneficial effects and may help preserve β-cells of the islets of Langerhans, but does not act as an insulin secretagogue. Pioglitazone promotes lipid storage and redistribution from visceral to subcutaneous deposits, resulting in an increase in whole body adiposity, while promoting the differentiation of adipocytes. It also appears to have protective effects against atherosclerosis and antihypertensive actions. Following oral administration of pioglitazone in patients with type 2 diabetes, peak plasma concentrations of pioglitazone are achieved in 2–2.5 hours. Plasma concentrations are dose dependent and steady state is achieved after 4–7 days’ treatment. Bioavailability is 83% and there is no accumulation of pioglitazone or its metabolites after repeated administration. Pioglitazone is metabolised in the liver predominantly via the cytochrome P450 enzyme system. About 15–30% of a dose is renally excreted, mainly as metabolites and their conjugates, with the remainder eliminated in faeces. Therapeutic Efficacy In well designed, randomised, controlled monotherapy trials of up to 2 years’ duration in patients with type 2 diabetes, glycaemic control improved with pioglitazone 15, 30 or 45 mg/day versus baseline and placebo. Improvements in glycaemic control in pioglitazone recipients were similar to those of metformin, insulin and rosiglitazone recipients, and greater than those in recipients of acar-bose or the sulfonylureas gliclazide, glimepiride or glibenclamide. Additive effects on glycaemic profiles occurred when pioglitazone was used in combination with metformin, repaglinide, insulin or the sulfonylureas gliclazide, glipizide, glimepiride or glibenclamide (glyburide). In patients with type 2 diabetes, lipid control was also improved with pioglitazone versus baseline and placebo in monotherapy trials and in trials in combination with metformin, insulin and sulfonylureas. Furthermore pioglitazone produced greater reductions in serum triglycerides and greater increases in high-density lipoprotein-cholesterol than metformin, sulfonylureas or rosiglitazone. Pioglitazone also reduced a cardiovascular risk parameter (carotid intima-media thickness), inflammatory biomarkers of arteriosclerosis (high sensitivity C-reactive protein, matrix metalloproteinase and monocyte chemoattractant protein levels) and a secondary composite measure of the risk of macrovascular events (all-cause mortality, nonfatal myocardial infarct, stroke), but not the primary composite endpoint (all-cause mortality, nonfatal myocardial infarct [MI] including silent MI, stroke, major leg amputation, acute coronary syndrome, cardiac intervention or leg revascularisation). Tolerability Pioglitazone was generally well tolerated in patients with type 2 diabetes in clinical trials of up to 2.5 years’ duration when used as monotherapy and in combination with other drugs including metformin, a sulfonylurea, repaglinide or insulin. The most commonly reported treatment-emergent adverse events were weight gain, oedema, arthralgia, headache and decreases in haemoglobin and haematocrit levels. Hepatocellular dysfunction and of hepatic enzyme elevations of three or more times the upper limit of normal have rarely been reported, and very rarely have involved hepatic failure with and without fatal outcome, although a causal link has not been established. Overall, small reductions in mean liver enzyme levels with pioglitazone treatment have been observed.
Article
Full-text available
To elucidate the effects of pioglitazone treatment on glucose and lipid metabolism in patients with type 2 diabetes. A total of 23 diabetic patients (age 30-70 years BMI < 36 kg/m2) who being treated with a stable dose of sulfonylurea were randomly assigned to receive either placebo (n = 11) or pioglitazone (45 mg/day) (n = 12) for 16 weeks. Before and after 16 weeks of treatment, all subjects received a 75-g oral glucose tolerance test (OGTT) and hepatic peripheral insulin sensitivity was measured with a two-step euglycemic insulin (40 and 160 mU x min(-1) x m(-2) clamp performed with 3-[3H]glucose and indirect calorimetry HbA1c measured monthly throughout the study period. After 16 weeks of pioglitazone treatment, the fasting plasma glucose (FPG; 184 +/- 15 to 135 +/- 11 mg/dl, P < 0.01), mean plasma glucose during OGTT(293 +/- 12 to 225 +/- 14 mg/dl, P < 0.01), and HbA1c (8.9 +/- 0.3 to 7.2 +/- 0.5%, P < 0.01 ) decreased significantly without change in fasting or glucose-stimulated insulin/C-peptide concentrations. Fasting plasma free fatty acid (FFA; 647 +/- 39 to 478 +/- 49) microEq/l, P < 0.01) and mean plasma FFA during OGTT (485 +/- 30 to 347 +/- 33 microEq/l, P < 0.01) decreased significantly after pioglitazone treatment. Before and after pioglitazone treatment, basal endogenous glucose prodution (EGP) and FPG were strongly correlated (r = 0.67, P < 0.01). EGP during the first insulin clamp step was significantly decreased after pioglitazone treatment (P < 0.05) whereas insulin-stimulated total and nonoxidative glucose disposal during the second insulin clamp was increased (P < 0.01). The change in FPG was related to the change in basal EGP, EGP during the first insulin clamp step, and total glucose disposal during the second insulin clamp step. The change in mean plasma glucose concentration during the OGGTT was strongly related to the change in total body glucose disposl during the second insulin clamp step. These results suggest that pioglitazone therapy in type 2 diabetic patients decreases lasting and postprandial plasma glucose levels by improving hepatic and peripheral (muscle) tissue sensitivity to insulin.
Article
In this chapter, common genetic polymorphisms affecting pharmacokinetics via effects on drug metabolism are outlined and their clinical relevances are discussed. The goal of effective and safe therapy of many drugs is made difficult by large interpatient variability in response and toxicity, and this problem is a substantial burden for patients, their caretakers, and the healthcare system. For many drugs, the response to chronic administration is determined by the area under the plasma concentration time curve (AUC), during a dosing interval at steady state, a measure of drug exposure. An increasing number of drug metabolizing enzymes have been shown to result in large pharmacokinetic changes through a variety of different mechanisms. Drugs that are most affected are those that have a dominant route of clearance by a genetically polymorphic enzyme. The effects of such pharmacokinetic changes are most important in settings where clinically important pharmacodynamic change ensues. Pharmacogenetics tests that are most valuable are those for specific drugs for which the prediction of activity or adverse effects is important and difficult to anticipate given our current clinical tools and technologic capability. Although data from a variety of platforms documenting a wide range of genetic variability are being accumulated rapidly and clinical genetic tests have been recommended or implemented for drugs such as mercaptopurine, azathioprine, warfarin, irinotecan, and tamoxifen, there is still a great need for well-designed, prospective clinical trials that test pharmacogenetic approaches versus standard practice.
Chapter
Introduction Synthesis of Rosiglitazone Synthesis of Pioglitazone Synthesis of Muraglitazar References
Article
Pioglitazone is an orally administered insulin sensitising thiazolidinedione agent that has been developed for the treatment of type 2 diabetes mellitus. ▴ Pioglitazone activates the nuclear peroxisome proliferator activated receptor-γ (PPAR-γ), which leads to the increased transcription of various proteins regulating glucose and lipid metabolism. These proteins amplify the post-receptor actions of insulin in the liver and peripheral tissues, which leads to improved glycaemic control with no increase in the endogenous secretion of insulin. ▴ In placebo-controlled clinical trials, monotherapy with pioglitazone 15 to 45 mg/day has been shown to decrease blood glycosylated haemoglobin (HbA1c) levels in patients with type 2 diabetes mellitus. ▴ The addition of pioglitazone 30 mg/day to preexisting therapy with metformin, or of pioglitazone 15 or 30 mg/day to sulphonylurea, insulin or voglibose therapy, has been shown to decrease HbA1c and fasting blood glucose levels significantly in patients with poorly controlled type 2 diabetes mellitus. ▴ Pioglitazone has also been associated with improvements in serum lipid profiles in randomised placebo-controlled clinical studies. ▴ The drug has been well tolerated by adult patients of all ages in clinical studies. Oedema has been reported with monotherapy, and pooled data have shown hypoglycaemia in 2 to 15% of patients after the addition of pioglitazone to sulphonylurea or insulin treatment. There have been no reports of hepatotoxicity.
Article
Aim This study was to investigate the pharmacokinetics of pioglitazone after a single oral dose of pioglitazone hydrochloride tablets in healthy Chinese volunteers and to evaluate the bioequivalence between the test and the reference tablets. Methods Twenty healthy Chinese volunteers received a single oral dose of 30mg pioglitazone either as test or as reference tablet in a randomized, open label, two-way crossover study. Pioglitazone in human plasma was determined by a HPLC method. Results Mean maximum concentration (C max) of pioglitazone was 1.85 mg¡ ¤ L -1 at 1.8 hours for test tablets and 1.86mg¡ ¤ L -1 at 1.9 hours for reference tablets. Mean area under the plasma concentration-time curve from zero to last measured point (AUC 0-t) for test was 15.51mg¡ ¤ h¡ ¤ L -1 compared with 15.37 mg¡ ¤ h¡ ¤ L -1 for reference. The analysis of variance on C max and AUC indicated that there was no significant difference between the two formulations. All 90% confidence intervals (CIs) of the test/reference geometric mean ratio were within the bioequivalence limits. Conclusion The test tablets were bioequivalent with the reference tablets.
Article
Pioglitazone is a thiazolidinedione antidiabetic agent that increases insulin sensitivity and decreases hepatic gluconeogenesis. Administered once daily without regard to meals, it is well absorbed, and is metabolised by the hepatic cytochrome P450 enzyme system. The half-life of the drug is approximately 9 hours, but two active metabolites (M-III and M-IV) contribute to extended glucose-lowering effects. In animals, after absorption the highest concentrations are found in the liver, plasma, and kidney. The mean absolute bioavailability is 83%, t max is 1.5 (range 0.5-3.0) hours, and the absorption rate constant ranges from 0.04 to 1.17 hr -1. Mean clearance is 2.4 (range 1.72-4.17) L/hr. With single oral doses between 2 and 60 mg, C max and area-under-the-curve (AUC) increased linearly with dose: no changes were observed upon repeated administration. The AUC is not affected by food. The volume of distribution is 0.253 L/kg, probably due to extensive protein binding (>97%). Drug interaction studies have not shown inhibition or induction of any cytochrome P450 enzymes involved in drug metabolism, thus the potential for drug interactions is low. The pharmacokinetics of pioglitazone do not differ significantly between healthy volunteers and patients with type 2 diabetes. Dosage adjustment is not necessary in patients with renal failure, or in those undergoing haemodialysis. In hepatic insufficiency, volume of distribution was increased, and C max was reduced. Age and gender appear to have no significant effect on the pharmacokinetics of pioglitazone, and there do not appear to be any differences between races.
Article
Evidence-based guidelines for the treatment of type 2 diabetes mellitus focus on three areas: intensive lifestyle intervention that includes at least 150 minutes per week of physical activity, weight loss with an initial goal of 7 percent of baseline weight, and a low-fat, reduced-calorie diet; aggressive management of cardiovascular risk factors (i.e., hypertension, dyslipidemia, and microalbuminuria) with the use of aspirin, statins, and angiotensin-converting enzyme inhibitors; and normalization of blood glucose levels (hemoglobin A1C level less than 7 percent). Insulin resistance, decreased insulin secretion, and increased hepatic glucose output are the hallmarks of type 2 diabetes, and each class of medication targets one or more of these defects. Metformin, which decreases hepatic glucose output and sensitizes peripheral tissues to insulin, has been shown to decrease mortality rates in patients with type 2 diabetes and is considered a first-line agent. Other medications include sulfonylureas and nonsulfonylurea secretagogues, alpha glucosidase inhibitors, and thiazolidinediones. Insulin can be used acutely in patients newly diagnosed with type 2 diabetes to normalize blood glucose, or it can be added to a regimen of oral medication to improve glycemic control. Except in patients taking multiple insulin injections, home monitoring of blood glucose levels has questionable utility, especially in relatively well-controlled patients. Its use should be tailored to the needs of the individual patient.