Você está na página 1de 68

CHAPTER THREE

Gliclazide
Fatmah A.M. Al-Omary
College of Pharmacy, King Saud University, Riyadh, Saudi Arabia

Contents
1. Description 126
1.1 Nomenclature 126
1.2 Formulae 126
1.3 Elemental Analysis 126
1.4 Appearance 126
1.5 Uses and Applications 126
2. Methods of Preparation 131
2.1 Chemical Synthesis 131
3. Physical Characteristics 133
3.1 Melting Behavior 133
3.2 Single-Crystal X-ray Diffraction 133
3.3 Spectroscopy 133
4. Methods of Analysis 138
4.1 Compendial Methods 138
4.2 Reported Methods of Analysis 145
5. Biological Analysis 152
6. Stability 153
7. Pharmacokinetics 154
7.1 Metabolism 160
7.2 Absorption 163
7.3 Secretion 166
7.4 Excretion 169
8. Pharmacology 170
8.1 Sites and Mechanism of Action 170
8.2 Cardiovascular Effect 175
9. Adverse Effects 178
10. Drug Interactions 180
11. Toxicology 181
11.1 Toxicity 181
11.2 Subchronic toxicity 182
11.3 Chronic Toxicity 182
Acknowledgment 183
References 184

Profiles of Drug Substances, Excipients, and Related Methodology, Volume 42 # 2017 Elsevier Inc. 125
ISSN 1871-5125 All rights reserved.
http://dx.doi.org/10.1016/bs.podrm.2017.02.003
126 Fatmah A.M. Al-Omary

1. DESCRIPTION
1.1 Nomenclature
1.1.1 Systematic Chemical Names
1-(Hexahydrocyclopenta[c]pyrrol-2(1H)-yl)-3-[(4-methylphenyl)sulfonyl]
urea [1].

1.1.2 Nonproprietary Names


Diamicron; Dramion [2].

1.1.3 Proprietary Names


Gliclazide; Gliclazidum; Gliklatsidi; Gliklazid; Gliklazidas; Glyclazide [3].

1.2 Formulae
1.2.1 Empirical Formula, Molecular Weight, CAS Number [1]

C15H21N3O3S 323.4 21187-98-4

1.2.2 Structural Formula [1]

O O O
S N
N N
H H
H3C

1.3 Elemental Analysis


C 55.71%, H 6.54%, N 12.99%, O 14.84%, S 9.91%.

1.4 Appearance
A white or almost-white powder. Practically insoluble in water, slightly
soluble in alcohol, sparingly soluble in acetone, and freely soluble in
dichloromethane. Dissociation constant pKa 5.8 [3].

1.5 Uses and Applications


Gliclazide is a sulfonylurea derivative, which is used for treatment of diabetes
mellitus through inhibition of ATP-dependent potassium channels. It is
given orally in the treatment of type 2 diabetes mellitus and has the duration
Gliclazide 127

of action of 1224 h. Because its effects are less prolonged than those of
chlorpropamide or glibenclamide, it may be more suitable for elderly
patients, who are prone to hypoglycemia with longer-acting sulfonylureas.
The usual initial dose is 4080 mg daily, gradually increased, if necessary, up
to 320 mg daily. Doses of more than 160 mg daily are given in two divided
doses. A modified-release (MR) tablet is also available, and the usual initial
dose is 30 mg once daily and increased if necessary up to a maximum of
120 mg daily [3,4].

1.5.1 Antioxidant Activity


Diabetic patients exhibit an oxidative stress status that is an imbalance between
reactive oxygen species (ROS) and antioxidant defenses, in favor of the first
ones. This oxidative stress, together with formation of advanced glycation
endproducts (AGEs), is involved in diabetic complications. It could thus be
of great interest to propose antioxidant and/or anti-AGE therapeutics as
complementary treatment in these patients. Antioxidants can be classical
molecules such as vitamin E, lipoic acid, and N-acetylcysteine. Thus,
vitamin E supplementation can improve insulin efficiency and glycemic
equilibrium, as shown by the decrease of glycemia, glycated hemoglobin,
and fructosamine values. In addition, this kind of supplementation lowers
plasma lipid peroxidation and oxidizability of low-density lipoproteins,
which is involved in the atherogenesis process. Moreover, it allows to fight
against complications such as retinopathy. A second category is represented
by molecules able to fight against the effects of AGEs. They can act: either
by preventing cellular action of AGEs; this is obtained with soluble recep-
tors of advanced glycation endproducts; or by inhibiting AGE formation
(scavenging of reactive carbonyl intermediates). Nucleophilic compounds
such as pyridoxamine, tenilsetam, 2,3-diaminophenazone, OPB-9195,
and aminoguanidine can act in this way. Aminoguanidine is able to limit
the development of the main diabetes-associated complications in ani-
mals. A double-blind clinical assay has been conducted in type 2 diabetic
patients in the United States and the Canada, in order to determine if ami-
noguanidine is able to slow down the progression of diabetes-induced
nephropathy.
Bonnefont-Rousselot [5] studied and discussed about another guanidic
molecule, i.e., metformin, which is also able to scavenge AGEs, in the
last part of this review. A third category of molecules is constituted by
oral antidiabetic molecules exhibiting antioxidant properties. They are
thiazolidinediones (troglitazone) and sulfonylureas (gliclazide). Troglitazone
128 Fatmah A.M. Al-Omary

and gliclazide can thus decrease low-density lipoprotein (LDL) oxidizability


and monocyte adhesion to endothelial cells (ECs), which is an early step in
the atherogenesis process and which is stimulated by oxidized LDLs. Finally,
a prospective way is devoted to oral antidiabetic drugs exhibiting both anti-
oxidant and anti-AGE properties. A very used antidiabetic drug of interest is
metformin (dimethylbiguanide), since it can prevent diabetes complications
not only by lowering glycemia but also by inhibiting AGE formation and by
stimulating antioxidant defenses. The latter therapeutic approach constitutes
a future way in the diabetes area, in order to obtain both a better glycemic
control and a least development of diabetic complications [5].
In a separate study [6], the effects of 10 months of oral gliclazide therapy
on oxidative parameters were assessed in 44 type 2 diabetic patients.
Gliclazide, but not glibenclamide, glimepiride, glipizide, or tolbutamide,
inhibited LDL oxidation and enhanced total plasma antioxidant capacity
(TPAC). With the addition of 1 M gliclazide, oxidation lag time increased
from 53.6  2.6 to 113.6  5.1 min (P < 0.001), and TPAC increased from
1.09  0.11 to 1.23  0.11 mM (P < 0.01). Administration of either MR or
standard gliclazide to type 2 diabetic patients resulted in a fall in
8-isoprostanes, a marker of lipid oxidation, and an increase in the antioxi-
dant parameters TPAC.
Sun et al. [7] investigated the effect of insulin and gliclazide therapy on
endoplasmic reticulum (ER) stress and insulin sensitivity (IS) in the liver of
type 2 diabetic rats and found that both insulin and gliclazide therapy could
relieve ER stress and c-Jun N-terminal kinase activity and improved IS. The
effect of insulin on Bip, XBP-1s, p-c-Jun, p-IRS-1, and G6Pase protein
expressions is more obvious than that of gliclazide, which indicates that
besides lowering glucose, insulin might have protective effects of anti-
inflammation, antioxidative stress, or stimulation of lipid redistribution.
The importance of K(ATP) channels in stimulussecretion coupling of
-cells is well established although they are not indispensable for the main-
tenance of glycemic control. Drews and D ufer [8] studied a new role for
K(ATP) channels by showing that genetic or pharmacological ablation of
these channels protects -cells against oxidative stress. Increased production
of oxidants is a crucial factor in the pathogenesis of type 2 diabetes mellitus
(T2DM). T2DM develops when -cells can no longer compensate for the
high demand of insulin resulting from excess fuel intake. Instead, -cells start
to secrete less insulin and -cell mass is diminished by apoptosis. Both reduc-
tion of insulin secretion and -cell mass induced by oxidative stress are
Gliclazide 129

prevented by deletion or inhibition of K(ATP) channels. These findings may


open up new insights into the early treatment of T2DM.
There have not been yet enough studies about effects of -glucan and
gliclazide on oxidative stress created by streptozotocin in the brain and sci-
atic nerve of diabetic rats. Alp et al. [9] studied and investigated the antiox-
idant effects of gliclazide and -glucan on oxidative stress and lipid
peroxidation created by streptozotosin in brain and sciatic nerve. Also, this
study shows that in terms of these parameters, both gliclazide and -glucan
have a neuroprotective effect on the brain and sciatic nerve of the
streptozotocin-induced diabetic rat. Their conclusion was that gliclazide
and -glucan have antioxidant effects on the brain and sciatic nerve of
the streptozotocin-induced diabetic rat.
The antioxidant properties of diabenol and gliclazide with reference to
mexidol have been studied by Spasov et al. [10] in vitro on several model
systems, including chemiluminescence (CL) of lipids, CL with generation
of ROS, CL dependent on luminol oxidation by peroxy radicals, and the
Glavind DPPH free radical method. Gliclazide demonstrated dose-
dependent antioxidant activity only on the model with stable DPPH radi-
cals, where its inhibitory effect was 15 times greater than that of the
reference drug.
Experimental and clinical studies done by Sliwinska et al. [11] suggested
that gliclazide may protect pancreatic -cells from apoptosis induced by an
oxidative stress. Their findings indicate that gliclazide may protect both nor-
mal and cancer human cells against apoptosis induced by hydrogen peroxide.
It appears that the antiapoptotic effect of the drug is most likely associated
with reduction of oxidative stress.
Avogaro [12] reported that gliclazide has antioxidant properties, reduces
markers of endothelial inflammation, and prevents glucose-induced apopto-
sis of ECs. These positive antioxidant effects are not confined to the vascular
wall, but they are effective also in the -cells. These properties are important
because (i) in patients with atherosclerotic process, microvascular abnormal-
ities may hasten disease progression and (ii) slowing the microvascular com-
plications may have a potentially remarkable effect on the natural history of
macrovascular disease.
Chen et al. [13] studied and examined the effect of gliclazide on AMPK
activity in the adipose tissue of individuals with type 2 diabetes. The results
suggest that gliclazide could improve the endothelial function in diabetes,
which may be related to its antioxidant properties.
130 Fatmah A.M. Al-Omary

T2DM is associated with increased oxidative stress. Free radicals pro-


duced during this stress may damage various cellular components. Gliclazide
that possesses antioxidant properties diminishes the harmful consequences of
oxidative stress in diabetic patients. Silwinska et al. [14] studied evaluated the
action of gliclazide on DNA damage and repair in normal human peripheral
blood lymphocytes and insulinoma mouse cells (-TC-6). DNA damage
and repair were induced by hydrogen peroxide, gamma and ultraviolet radi-
ation, and MNNG (N-methyl-N 0 -nitro-N-nitrosoguanidine) in the pres-
ence or absence of gliclazide. Gliclazide protected DNA of both kinds of
cells from DNA damage induced by chemicals and radiations. These results
suggest that gliclazide may diminish the risk of free radical-related diseases
associated with T2DM and possibly cancer.
Evidence implicates hyperglycemia-derived oxygen free radicals as medi-
ators of diabetic complications. Ceriello [15] reported that hyperglycemia-
induced overproduction of superoxide seems the first and key event in
the activation of all pathways involved in the pathogenesis of diabetic com-
plications. Superoxide overproduction is accompanied by increased nitric
oxide generation and, consequently, formation of the strong oxidant per-
oxynitrite, and by poly(adenosine diphosphate ribose) polymerase activa-
tion, which in turn further activates the pathways involved in the
pathogenesis of diabetic complications. This process results in acute endo-
thelial dysfunction and activation of inflammation in diabetic blood vessels
that, convincingly, contribute to the development of diabetic complications.
Clinical evidences suggest that gliclazide works as an antioxidative drug,
independently from its ability to reduce hyperglycemia. The availability
of a compound that simultaneously decreases hyperglycemia, restores insulin
secretion, and inhibits oxidative stress produced by high glucose seems to be
an interesting therapeutic prospect for the prevention of vascular complica-
tions of diabetes.
Satyanarayana et al. [16] studied the effect of oral administration of sele-
nium on blood glucose and its influence on gliclazide-induced hypoglyce-
mia/antihyperglycemia in normal and alloxan-induced diabetic rats. The
combination of selenium with gliclazide significantly enhanced the
glucose-lowering effect of gliclazide in normal and diabetic rats.
Alper et al. [17] studied and investigated the possible effect of mono-
amine oxidase inhibitor, selegiline (L-deprenyl), in combination with
gliclazide in preventing oxidative stress in streptozotocin-induced diabetes
model in male Swiss Albino rats by measuring oxidant stress/DNA damage
and antioxidant levels. The combined treatment of deprenyl and gliclazide
Gliclazide 131

may contribute to the control of the physiopathological mechanisms under-


lying both the process of aging and T2DM by reducing oxidant stress and
DNA damage, improving antioxidant status, and increasing survival, and
may have implications for further clinical studies.
Onozato et al. [18] hypothesized that gliclazide may have a beneficial
effect on diabetic nephropathy via radical scavenging. Gliclazide reduced
oxidative stress in diabetic rats fed a high cholesterol diet with reduction
of renal NAD(P)H oxidase expression, enhanced MnSOD and eNOS
expression, and had a beneficial effect on glomerular macrophage infiltration
and mesangial expansion.
Kimoto et al. [19] studied and investigated results that suggest that
gliclazide reduces oxidative stress of -cells by hydrogen peroxide, probably
due to its radical scavenging activity. Gliclazide may be effective in
preventing -cells from the toxic action of ROS in diabetes.
Fava et al. [20] studied and evaluated the effects of gliclazide on oxidative
status and vascular response to systemic administration of L-arginine, the nat-
ural precursor of nitric oxide (NO), in T2DM patients. Gliclazide reduces
oxidative stress in T2DM patients by improving plasma antioxidant status.
This effect is associated with enhanced NO-mediated vasodilation.

2. METHODS OF PREPARATION
2.1 Chemical Synthesis
Several methods were reported for the synthesis of gliclazide. The chemical
synthesis of gliclazide was first reported by Science Union and CIE in
1970 [21]. The synthetic pathway involves the condensation of
4-methylbenzenesulfonyl urethane (I) with hexahydro-2-cyclopentano[c]
pyrrolylamine (II), in anhydrous toluene to yield gliclazide in 60% yield.

O O O
O O O
Toluene S N
S + N N N
N OEt H2N Reflux, 1 h H H
H
Gliclazide
(I) (II)
The main disadvantages of the above synthesis are the high production
cost due to the use of expensive precursors and the relative low yield.
The synthesis of gliclazide was further improved via utilization of less expen-
sive starting materials. Qian et al. [22] developed a three-step method for the
large-scale synthesis of gliclazide starting with cyclopentane-1,2-dicarboxylic
132 Fatmah A.M. Al-Omary

anhydride (III) through condensation with hydrazine hydrate to yield


N-aminocyclopentane-1,2-dicarboximide (IV), which was reduced to
hexahydro-2-cyclopentano[c]pyrrolylamine (V), and subsequent reaction
with p-toluenesulfonyl urea (VI) to furnish gliclazide in 60% overall yield
as outlined in the following scheme:
O O
N2H4H2O
O N NH2

(III) O (IV) O
O
KBH4 /AlCl3
NNH2 N NH2

O (V)
O O O
S Gliclazide
N NH2 + N NH2
H
(VI)
Che and Du [23] developed a more convenient procedure for the synthesis
of gliclazide in about 70% overall yield via condensation p-toluenesulfonyl
urea (VI) with hydrazine hydrate to yield the corresponding hydrazide
(VII), which was then reacted with cyclopentane-1,2-dicarboxylic anhydride
(III) to afford 1-(1,3-dicarbonylhexahydrocyclopentano[c]pyrrol-2(1H)-yl)3-
p-tolylsulfonylurea (VIII), and the latter was reduced to gliclazide as outlined
in the following scheme:
O O O O
O O
S S NH2
N NH2 N2H4 H2O
H N N
H H
(VI) (VII)

O
O O O O
O O O
S N
S NH2 + O N N
N N H H
H H O
O
(III) (VIII)

O
O O O
NaBH4/LiCl
S N Gliclazide
N N
H H O
Gliclazide 133

3. PHYSICAL CHARACTERISTICS
3.1 Melting Behavior
Gliclazide is a crystalline solid that exhibits a melting point of about
163169C [2].

3.2 Single-Crystal X-ray Diffraction


Gliclazide single crystals suitable for X-ray analysis were obtained by slow
evaporation of methanolic solution at room temperature. The X-ray data
were reported by Parvez et al. [24]. Gliclazide crystallizes in the P21/n
group and belongs to the monoclinic system with the following cell
dimensions: a 10.8326 A, b 14.3281 A, c 10.976 A and 107.02
degrees, V 1628.9 A3. The measured density of the crystal is
1.319 mg/m3. The X-ray data (Fig. 1) also revealed a discrete molecule
with normal molecular dimensions and that both of the five-membered
fused rings adopted envelop conformation. The molecules are linked into
chains by intermolecular hydrogen bonds involving the amino-H atoms
with the sulfonyl and carbonyl O atoms. The structural geometrical
parameter of gliclazide (bond lengths and angles) is shown in Tables 1
and 2, respectively.

3.3 Spectroscopy
3.3.1 Ultraviolet Spectroscopy
The UV absorption maximum (max) of gliclazide was determined in differ-
ent solvents. Singh et al. reported the max of gliclazide in dichloromethane at

Fig. 1 The ORTEP of gliclazide showing the atomic numbering schemes.


134 Fatmah A.M. Al-Omary

Table 1 Bond Lengths () of Gliclazide From the Single-


Crystal Structure
Bond Bond Length ()
S1O1 1.419
S1O2 1.427
S1N1 1.634
S1C2 1.758
O3C1 1.218
N1C1 1.394
N2C1 1.333
N2N3 1.414
N3C9 1.465
N3C15 1.473
C2C3 1.369
C2C7 1.376
C3C4 1.378
C4C5 1.371
C5C6 1.377
C5C8 1.497
C6C7 1.375
C9C10 1.514
C10C11 1.526
C10C14 1.547
C11C12 1.484
C12C13 1.501
C13C14 1.538
C14C15 1.501

232 nm [25]. Meanwhile, Jamadar et al. reported the max at 229.5 nm in


methanol [26], and Revathi et al. measured the max at 224 nm (224 nm)
in aqueous sodium hydroxide [27]. The spectrum of gliclazide in methanol
(Fig. 2) showed the main peak max at 226 nm.
Gliclazide 135

Table 2 The Bond Angles (in Degrees) of Gliclazide From the Single-Crystal Structure
Bond Bond Angle Bond Bond Angle
O1S1O2 119.5 C8C5C6C7 177.8
O1S1N1 108.6 C5C4C3 121.6
O2S1N1 103.2 C4C5C6 117.5
O1S1C2 108.9 C4C5C8 120.5
O2S1C2 108.6 C6C5C8 121.9
N1S1C2 107.5 C7C6C5 121.8
C1N1S1 124.6 C6C7C2 119.6
C1N2N3 121.6 N3C9C10 104.2
N2N3C9 111.3 C9C10C11 113.9
N2N3C15 109.8 C9C10C14 104.3
C9N3C15 104.1 C11C10C14 105.0
O3C1N2 123.8 C12C11C10 105.3
O3C1N1 121.8 C11C12C13 105.6
N2C1N1 114.3 C12C13C14 104.6
C3C2C7 119.5 C15C14C13 114.0
C3C2S1 119.9 C15C14C10 105.2
C7C2S1 120.6 C13C14C10 106.3
C2C3C4 119.9 N3C15C14 105.5
O1S1N1C1 48.4 C5C6C7C2 0.2
O2S1N1C1 176.2 C3C2C7C6 0.3
C2S1N1C1 69.2 S1C2C7C6 178.8
C1N2N3C9 137.7 N2N3C9C10 159.4
C1N2N3C15 107.5 C15N3C9C10 41.1
N3N2C1O3 163.2 N3C9C10C11 142.3
N3N2C1N1 19.8 N3C9C10C14 28.4
S1N1C1O3 11.2 C9C10C11C12 88.4
S1N1C1N2 171.73 C14C10C11C12 25.0
O1S1C2C3 156.5 C10C11C12C13 36.9
Continued
136 Fatmah A.M. Al-Omary

Table 2 The Bond Angles (in Degrees) of Gliclazide From the Single-Crystal
Structurecontd
Bond Bond Angle Bond Bond Angle
O2-S1C2C3 24.9 C11C12C13C14 33.7
N1S1C2C3 86.1 C12C13C14C15 97.9
O1S1C2C7 22.0 C12C13C14C10 17.5
O2S1C2C7 153.6 C9C10C14C15 5.5
N1S1C2C7 95.4 C11C10C14C15 125.6
C7C2C3C4 0.8 C9C10C14C13 115.8
S1C2C3C4 179.3 C11C10C14C13 4.4
C2C3C4C5 0.7 N2N3C15C14 157.0
C3C4C5C6 0.2 C9N3C15C14 37.7
C3-C4C5C8 178.3 C13C14C15N3 135.3
C4C5C6C7 0.3 C10C14C15N3 19.2

1.0
0.9
0.8
0.7
0.6
0.5
A

0.4
0.3
0.2
0.1
0.0
0.1
200 250 300 350 400
nm
Fig. 2 The UV spectrum of gliclazide in methanol.

3.3.2 Nuclear Magnetic Resonance Spectroscopy


3.3.2.1 1H NMR Spectrum
The 700.17 MHz 1H NMR spectrum of gliclazide in CDCl3 and the proton
assignments are shown in Figs. 3 and 4, respectively. The spectrum showed
Gliclazide 137

Fig. 3 1H NMR spectrum of gliclazide in CDCl3.

2.0
1.37
H
3.28 2.81 H 1.67
H
H H
O O O H
1.46
7.31
S N
N N H
H H 1.57
7.95 6.40 8.80
2.41

Fig. 4 1H NMR proton assignments of gliclazide.

the aromatic, CH3, and NH protons as isolated peaks. The aliphatic protons
of the fused rings were overlapped.
13
3.3.2.2 C NMR Spectrum
The 176.17 MHz 13C NMR spectrum of gliclazide in CDCl3 and the car-
bon assignments are shown in Figs. 5 and 6, respectively.

3.3.3 Infrared Spectrum


The Fourier transfer infrared (FTIR) absorption spectrum of gliclazide
(4004000 cm1) is shown in Fig. 7. The spectrum showed the main
138 Fatmah A.M. Al-Omary

13
Fig. 5 C NMR spectrum of gliclazide in CDCl3.

24.42
34.36
O O O
30.95
S N
40.36

129.40 N N
152.32

136.39
H H
128.38
144.48

21.64

13
Fig. 6 C NMR carbon assignments of gliclazide.

characteristic absorption bands at 3273 (NH), 3117, 3193 (aromatic CH),


28372949 (aliphatic perhydro-clopentapyrrole moiety), 1710 (C]O),
1596 (CC), 1347 (NC), and 10871038 cm1 (SO2).

4. METHODS OF ANALYSIS
4.1 Compendial Methods
4.1.1 British Pharmacopoeia Methods
4.1.1.1 Identification
Shake a quantity of the powdered tablets containing 0.16 g of gliclazide with
20 mL of dichloromethane, centrifuge, and evaporate the supernatant liquid
46.0 FA-1

40

1917.25
35 2369.71

2344.78

30

421.50

25

%T 1040.04 446.86
20

470.07

15
1347.38
1240.51 1122.51

10 1021.14
883.07
751.79 530.36
2836.76 1010.88 706.05 632.36
5 1709.97 1164.19
1596.24 812.30
3112.58 2867.44 1309.77
996.01 896.68 577.81
3273.15 3193.13 2949.30 1278.75 1087.46 668.15
1435.90 919.47
1.0
4000.0 3600 3200 2800 2400 2000 1800 1600 1400 1200 1000 800 600 400.0
cm1
Fig. 7 The FTIR spectrum of gliclazide in solid phase in potassium bromide.
140 Fatmah A.M. Al-Omary

to dryness. The infrared absorption spectrum of the residue is concordant


with the reference spectrum of gliclazide (RS 168).

4.1.1.2 Tests
4.1.1.2.1 Dissolution Comply with the requirements for Monographs
of the British Pharmacopoeia in the dissolution test for tablets and capsules.
Use as the dissolution medium 900 mL of phosphate buffer pH 7.4 and rotate
the paddle at 100 revolutions per minute. Withdraw a sample of 10 mL of the
medium and filter through a 0.5-m filter. Measure the absorbance of the
filtrate diluted, if necessary, with the dissolution medium to an expected con-
centration of 12.5 g/mL of gliclazide at 226 and 290 nm using the dissolu-
tion medium in the reference cell. Correct the absorbance obtained at
226 nm by subtracting the absorbance obtained at 290 nm. Calculate the total
content of gliclazide, C15H21N393S, in the medium using the absorbance of
a solution prepared by dissolving 62.0 mg of gliclazide BPCRS in 20 mL of
methanol, adding sufficient dissolution medium to produce 1000 mL and
diluting 1 volume of this solution to 5 volumes with the dissolution medium
and using the declared content of C15H21N3O3S in gliclazide BPCRS.

4.1.1.2.2 Related Substances Carry out the method for liquid chro-
matography using the following solutions without delay. For solution (1)
shake a quantity of the powdered tablets containing 0.8 g of gliclazide for
1 h with 200 mL of acetonitrile, filter, and dilute 10 mL of the filtrate to
50 mL with a mixture of 1 volume of acetonitrile and 2 volumes of water.
For solution (2) dilute 1 volume of solution (1) to 500 volumes with a mix-
ture of 45 volumes of acetonitrile and 55 volumes of water. For solution (3)
dissolve 5 mg of gliclazide BPCRS and 15 mg of 1-(3-azabicyclo[3.3.0]oct-
3-yl)-3-o-tolysulfonylurea BPCRS in 25 mL of acetonitrile, dilute to 50 mL
with water, and dilute 1 volume of the resulting solution to 20 volumes with
a mixture 45 volumes of acetonitrile and 55 volumes of water. For solution
(4) dissolve 8 mg of 1-(3-azabicyclo[3.3.0]oct-3-yl)-3-o-tolylsulfonylurea
BPCRS in 25 mL of acetonitrile, dilute to 50 mL with water, and dilute
1 volume of the resulting solution to 100 volumes with a mixture of 45 vol-
umes of acetonitrile and 55 volumes of water.
The chromatographic procedure may be carried out using (a) a stainless
steel column (25 cm  4 mm) packed with octylsilyl silica gel for chroma-
tography (4 m) (Superspher 60RP8 is suitable); (b) a mixture of 0.1 volume
of triethylamine, 0.1 volume of trifluoroacetic acid, 45 volumes of acetoni-
trile, and 55 volumes of water as the mobile phase with a flow rate of
Gliclazide 141

0.9 mL/min; and (c) a detection wavelength of 235 nm. Inject separately
20 L of each solution. For solution (1) allow the chromatography to pro-
ceed for twice the retention time of the principal peak. The test is not valid
unless the resolution factor between the peaks in the chromatogram
obtained with solution (3) is at least 1.8.
In the chromatogram obtained with solution (1) the area of any peak
corresponding to 1-(3-azabicyclo[3.3.0]oct-3-yl)-3-o-tolylsulfonylurea is
not greater than the area of the principal peak in the chromatogram obtained
with solution (4) (0.2%), the area of any other secondary peak is not greater
than the area of the principal peak in the chromatogram obtained with solu-
tion (2) (0.2%), and the sum of the areas of any other secondary peaks is not
greater than twice the area of the principal peak in the chromatogram
obtained with solution (2) (0.4%). Disregard any peak with an area less than
one quarter of the area of the peak corresponding to gliclazide in the chro-
matogram obtained with solution (2) (0.05%).

4.1.1.3 Assay
Weigh and powder 20 tablets. Carry out the method for liquid chromatogra-
phy using the following solutions. For solution (1) shake a quantity of the pow-
dered tablets containing 0.8 g of gliclazide for 1 h with 200 mL of acetonitrile,
filter, and dilute 10 mL of the filtrate to 200 mL with a mixture of 2 volumes of
acetonitrile and 3 volumes of water. For solution (2) dissolve 40 mg of
gliclazide BPCRS in 10 mL of acetonitrile and dilute to 200 mL with a mixture
of 2 volumes of acetonitrile and 3 volumes of water. For solution (3) dissolve
5 mg of gliclazide BPCRS and 15 mg of 1-(3-azabicyclo[3.3.0]oct-3-yl)-3-o-
tolylsulfonylurea BPCRS in 25 mL of acetonitrile, dilute to 50 mL with water,
and dilute 1 volume of the resulting solution to 20 volumes with a mixture
of 45 volumes of acetonitrile and 55 volumes of water. The chromatographic
procedure described under Related substances may be used.
The test is not valid unless the resolution factor between the peaks in the
chromatogram obtained with solution (3) is at least 1.8.
Calculate the content of C15N21N3O3S in the tablets using the declared
content of C15N21N3O3S in gliclazide BPCRS [1].

4.1.2 European Pharmacopoeia Methods


4.1.2.1 Identification
Infrared absorption spectrophotometry.
Preparation: KBr discs.
Comparison: gliclazide CRS.
142 Fatmah A.M. Al-Omary

4.1.2.2 Tests
Related substances. Liquid chromatography. Prepare the solutions immedi-
ately before use.
Solvent mixture: acetonitrile R, water R (45:55, v/v).
Test solution: dissolve 50.0 mg of the substance to be examined in
23 mL of acetonitrile R and dilute to 50.0 mL with water R.
Reference solution (a): dilute 1.0 mL of the test solution to 100.0 mL
with the solvent mixture. Dilute 10.0 mL of this solution to 100.0 mL
with the solvent mixture.
Reference solution (b): dissolve 5 mg of the substance to be exam-
ined and 15 mg of gliclazide impurity F CRS in 23 mL of acetonitrile
R and dilute to 50 mL with water R. Dilute 1 mL of this solution to
20 mL with the solvent mixture.
Reference solution (c): Dissolve 10.0 mg of gliclazide impurity
F CRS in 45 mL of acetonitrile R and dilute to 100.0 mL with water
R. Dilute 1.0 mL of this solution to 100.0 mL with the solvent
mixture.
Column:
2 size: l 0.25 m, 4 mm;
2 stationary phase: octylsilyl silica gel for chromatography R (5 m).
Mobile phase: triethylamine R, trifluoroacetic acid R, acetonitrile R,
water R (0.1:0.1:45:55, v/v/v/v).
Flow rate: 0.9 mL/min.
Detection: spectrophotometer at 235 nm.
Injection: 20 L.
Run time: twice the retention time of gliclazide.
Relative retention with reference to gliclazide (retention time about
16 min): impurity F about 0.9.
System suitability: reference solution (b):
2 resolution: minimum 1.8 between the peaks due to impurity F and
gliclazide.
Limits:
2 impurity F: not more than the area of the corresponding peak in the
chromatogram obtained with reference solution (c) (0.1%);
2 unspecified impurities: for each impurity, not more than the area of
the principal peak in the chromatogram obtained with reference
solution (a) (0.10%);
Gliclazide 143

2 sum of impurities other than F: not more than twice the area of the
principal peak in the chromatogram obtained with reference solution
(a) (0.2%);
2 disregard limit: 0.2 times the area of the principal peak in the chro-
matogram obtained with reference solution (0.02%).
Impurity B. Liquid chromatography as described test for related substances
with the following modification:
Test solution: dissolve 0.400 g of the substance to be examined in 2.5 mL
of dimethyl sulfoxide R and dilute to 10.0 mL with water R. Stir for
10 min, store at 4C for 30 min, and filter.
Reference solution: dissolve 20.0 mg of gliclazide impurity B CRS in
dimethyl sulfoxide R and dilute to 100 mL. with the same solvent.
To 1.0 mL of the solution, add 12 mL dimethyl sulfoxide R and dilute
to 50.0 mL with water R. 1.0 mL of this solution, add 12 mL of dimethyl
sulfoxide R dilute to 50.0 mL with water R.
Injection: 50 L.
Retention time: impurity B 8 min.
Limit:
2 impurity B: not more than the area of the corresponding peak in the
chromatogram obtained with the reference solution (2 ppm).
Heavy metals: maximum 10 ppm.
1.5 g complies with test F. Prepare the reference solution by using
1.5 mL standard lead solution (10 ppm Pb) R.
Loss on drying: maximum 0.25% determine on 1.000 g by drying in an oven
at 105C for 2 h.
Sulfated ash: maximum 0.1% determined on 1.0 g.

4.1.2.3 Assay
Dissolve 0.250 g in 50 mL of anhydrous acetic acid R. Titrate with 0.1 M
perchloric acid, determining the end-point potentiometrically.
1 mL of 0.1 M perchloric acid equivalent to 32.34 mg of C15H21N3O3S.

4.1.2.4 Impurites
Specified impurities: B, F.
Other detectable impurities (the following substances would, if present at
a sufficient level, be detected by one or other of the tests in the monograph.
They are limited by the general acceptance criterion for other/unspecified
144 Fatmah A.M. Al-Omary

impurities and/or by the general monograph Substances for pharmaceutical


use. It is therefore not necessary to identify these impurities for demonstra-
tion of compliance. See also Section 5. Control of impurities in substances
for pharmaceuticals use): A, C, D, E.
O O
S
R = N
H
H3C

A. RH: 4-methylbenzenesulfonamide,

N
O N

B. 2-Nitroso-octahydrocyclopenta[c]pyrrole,
C. RCOOC2H5: ethyl [(4-methylphenyl)sulfonyl]carbamate,

R N

D. N-[(4-Methylphenyl)sulfonyl]hexahydrocyclopenta[c]pyrrol-2(1H)-
carboxamide,

O
N
R N
H

E. 1-[(4-Methylphenyl)sulfonyl]-3-(3,3a,4,6a-tetrahydrocyclopenta[c]pyrrol-
2(1H)-yl)urea,

O O O
S N
N N
H H
CH3

F. 1-(Hexahydrocyclopenta[c]pyrrol-2(1H)-yl)-3-[(2-methylphenyl)sulfonyl]
urea,
Gliclazide 145

O
N
R N

G. N-[(1-Methylphenyl)sulfonyl]-1,4a,5,6,7,7a-hexahydro-2H-cyclopenta
[d] pyridazine-2-carboxamide [4].

4.2 Reported Methods of Analysis


Barnett et al. [28] determined if therapeutic management programs for
T2DM that include self-monitoring of blood glucose (SMBG) result in
greater reductions in glycated hemoglobin (HbA1c) compared with pro-
grams without SMBG in noninsulin requiring patients. In patients with type
2 diabetes, the application of SMBG as an adjunct to oral antidiabetic agent
therapy results in further reductions in HbA1c.
Charbonnel et al. [29] compared the effects of pioglitazone and gliclazide
on metabolic control in drug-naive patients with T2DM. Pioglitazone
monotherapy was equivalent to gliclazide in reducing HbA1c, with specific
differences between treatments in terms of mechanism of action, plasma
lipids, and adverse events.
Baksi et al. [30] reported that patients with T2DM who are inadequately
controlled on a half-maximal dose of a sulfonylurea managed by either
increasing the dose of sulfonylurea or adding another agent.
Many lines of evidence indicate that hyperinsulinemia might be associ-
ated with coronary atherosclerosis, and, currently, there are no effective
strategies for preventing this. They previously reported that high insulin
enhances neutrophil-transendothelial migration, a process that involves
increased surface presentation of platelet endothelial cell adhesion
molecule-1 (PECAM-1) through a mitogen-activated protein (MAP)
kinase-dependent event. Okouchi et al. [31] examined if antidiabetic agents,
especially K(ATP) channel blockers, might similarly protect against the
leukocyteEC interactions enhanced by high insulin. The results suggest
that the K(ATP) channel blocker, gliclazide, blocks high insulin-mediated
neutrophil migration and PECAM-1 expression. These gliclazide effects
may be mediated through the inhibition of MAP kinase activation and
are unrelated to NO production.
Salman et al. [32] compared the effect of acarbose and gliclazide on clin-
ical findings, biochemical parameters, and safety in T2DM patients insuffi-
ciently controlled with medical nutrition therapy. The results of the study
demonstrate that acarbose and gliclazide were reasonably effective in
146 Fatmah A.M. Al-Omary

improving metabolic control in patients insufficiently controlled with


diet alone, and both treatments were well tolerated. Because of its effects
on weight reduction and PP hyperinsulinemia, acarbose may be preferred
as a first-line drug, particularly in the treatment of overweight T2DM
patients.
Pugh et al. [33] assessed the efficacy of combination therapy with insulin
and sulfonylurea in the treatment of noninsulin-dependent diabetes mellitus
(NIDDM). Combined insulinsulfonylurea therapy leads to modest
improvement in glycemic control compared with insulin therapy alone.
With combined therapy, lower insulin doses may be used to achieve similar
control. Obese patients with higher fasting C-peptides may be more likely to
respond than others.

4.2.1 Chromatographic Methods


Matsuda et al. [34] used high-performance affinity chromatography (HPAC)
to examine the binding of gliclazide with the protein human serum albumin
(HSA) at various stages of modification due to glycation. This work illus-
trated how HPAC can be used to examine both the overall binding of a drug
with normal or modified proteins and the site-specific changes that can
occur in these interactions as a result of protein modification.
Sener et al. [35] investigated a standardized procedure for the assay and
identification of hypoglycemic sulfonylureas in plasma. External standards
and plasma extracts containing glibenclamide, gliquidone, glipizide, or
gliclazide were examined by high-performance liquid chromatography
(HPLC). The four sulfonylureas could be identified and measured with a
precision of 5%7% and a limit of detection (LOD) close to 12 ng in
injected external standards and 1040 ng/mL in plasma samples. This
method is suitable to study the pharmacokinetics of hypoglycemic sulfonyl-
ureas, for blood drug monitoring in diabetic patients, for diagnostic purposes
in factitious hypoglycemia, and in cases relevant to forensic medicine.
Park et al. [36] studied and evaluated the bioequivalence of two formu-
lations of gliclazide in healthy human volunteers. Bioequivalence of the two
formulations was determined in 20 healthy subjects with a single-dose, two-
period, crossover study. A new HPLC method for the pharmacokinetic
analysis of gliclazide was developed, using a semimicrocolumn to quantify
gliclazide in plasma samples. Chromatographic separation was achieved
with. This result suggests that two formulations are bioequivalent when
administered orally at a dose of 80 mg gliclazide.
Gliclazide 147

Rojanasthien et al. [37] studied the bioequivalence of MR 30 mg


gliclazide tablets in 18 healthy Thai volunteers. A test product, Glycon
MR, was compared with a reference product, Diamicron MR. The study
was performed under a single-dose, two-treatment, two-period, and two-
sequence crossover design in fasted and fed conditions with a washout
period of 2 weeks. Blood samples were collected for 72 h after drug admin-
istration. Drug plasma concentrations were determined by HPLC with a UV
detector. Analysis of pharmacokinetic characteristics was based on a
noncompartmental model.
Wang et al. [38] developed a sensitive and selective liquid
chromatographic-mass spectrometric (LCMS) method for the determina-
tion of gliclazide in human plasma. Sample treatment was based on pro-
tein precipitation with acetonitrile. The results show that AUC, Tmax,
Cmax, and T1/2 between the test formulation and the reference formulation
have no significant difference (P > 0.05). Relative bioavailability is
96.74  12.9%.
Mendes et al. [39] studied the performance of one gliclazide tablet for-
mulation (gliclazide 80 mg tablet from EMS Industria Farmac^eutica Ltda.)
in human volunteers against two reference gliclazide tablet formulations
(Diamicron 80 mg tablet from Servier do Brazil Ltda. and Diamicron
80 mg tablet from Servier (Ireland) Industries Limited). Since the 90%
Cl for Cmax, AUClast, and AUC(0infinity) ratios were all outside the 125%
interval proposed by the US Food and Drug Administration, they con-
cluded that the gliclazide test formulation was not bioequivalent to either
reference formulation. Interestingly, the pharmacokinetic parameters such
as Cmax and AUClast of both reference formulations are compatible with
neither the literature nor the profile of an immediate-release formula-
tion. In addition, both reference formulations were not bioequivalent
in themselves, indicating significant differences in reference product
formulation.
Pharmaceutical counterfeiting is becoming a serious problem in the
world, especially in developing countries including China. Isocratic
reversed-phase high-performance liquid chromatography (RP-HPLC)
method was developed for screening counterfeit medicines and adulterated
dietary supplement products. Yao et al. [40] developed the method that could
be employed to separate and determine simultaneously six antidiabetic drugs
(glipizide, gliclazide, glibenclamide, glimepiride, gliquidone, repaglinide) on
an isocratic solvent system using an Alltima C18 column (5 m,
148 Fatmah A.M. Al-Omary

150 mm  4.6 mm) with an isocratic mobile phase of methanolphosphate


buffer (pH 3.0; 0.01 mol/L) (70:30, v/v), at a flow rate of 1.0 mL/min
and at a wavelength of 230 nm. The proposed method was successfully
applied to the analysis of medicinal and dietary supplement samples pur-
chased from the local market in China.
Foroutan et al. [41] developed and validated a simple, rapid, and sen-
sitive isocratic RP-HPLC method with UV detection using a monolithic
column for the determination of gliclazide in human plasma. The assay
enables the measurement of gliclazide for therapeutic drug monitoring
with a minimum quantification limit of 10 ng/mL. The method involves
a simple, one-step extraction procedure and analytical recovery was com-
plete. The separation was carried out in reversed-phase conditions using a
Chromolith Performance (RP-18e, 100 mm  4.6 mm) column with an
isocratic mobile phase consisting of 0.01 M disodium hydrogen phosphate
bufferacetonitrile (52:48, v/v) adjusted to pH 4.0. The wavelength was
set at 230 nm. The calibration curve was linear over the concentration
range of 105000 ng/mL. The coefficients of variation for inter- and intra-
day assay were found to be less than 6.0%.
Berecka et al. [42] developed and validated a new HPLC method for the
quantitation of gliclazide and repaglinide in pharmaceutical formulations.
Determination was performed using a LiChroCART RP-18 column, a
mobile phase containing acetonitrilephosphate buffer (pH 2.1; 60:40, v/v),
and ultraviolet (UV) detection at 225 nm. Repaglinide was used as an internal
standard for gliclazide determination and gliclazide for repaglinide assay. The
method was validated with respect to linearity, precision, robustness, rugged-
ness, accuracy, and specificity. Finally, the method was applied for the quality
control of commercial gliclazide and repaglinide tablets. Total recovery
was 100.40  0.35% and 104.46 0.23% for gliclazide and repaglinide, respec-
tively (mean  SD).
Kuo and Wu [43] developed a sensitive HPLC-electrochemical detection
method for the analysis of gliclazide in human plasma. After deproteination of
100 L of plasma by acetonitrile, evaporation, and reconstitution, GL was
separated on a C18 column (150 mm  4.6 mm) by the mobile phase
(70 mM disodium tetraborate, pH 7.5, containing 26.5% of acetonitrile).
The regression equations were linear (r > 0.9990) over the range of 50 nM
to 4.00 M. The precision and accuracy of intra- and interday analysis were
less than 5.3% and 0.93% for relative standard deviation and relative error,
respectively. The LOD for plasma was 10 nM for GL [signal-to-noise
Gliclazide 149

(S/N) ratio 3, 10 L injection]. This method was applied for monitoring


blood levels with one healthy volunteer dosing with a gliclazide tablet.
Rouini et al. [44] developed a simple, rapid, and specific method for the
analysis of gliclazide in serum by a sensitive HPLC method. Only 100 L of
serum and a little sample work-up is required. A simple procedure of extrac-
tion by toluene followed by evaporation to dryness under a gentle stream of
air and dissolving the dried residue in mobile phase. The gliclazide peak was
separated from endogenous peaks on a C8 column by a mobile phase of
acetonitrilewater (45:55, v/v), pH 3. Gliclazide and internal standard (phe-
nytoin) were eluted at 6.8 and 3.8 min, respectively. The limit of quantita-
tion for gliclazide in serum was 75 ng/mL at 230 nm. The method was linear
over the range of 7510,000 ng/mL with r2 of 0.999. Mean recovery for
gliclazide and internal standard was 84.5% and 87%, respectively.
Vasudevan et al. [45] developed a simple, precise, and accurate HPLC
method for the simultaneous estimation of metformin with gliclazide and
glipizide in multicomponent dosage forms. The method was carried out on
an Inertsil C18 column. A mobile phase composed of acetonitrilewater con-
taining camphor sulfonic acid (adjusted to pH 7 using 0.1 N sodium hydrox-
ide; 75 mM) at a flow rate of 1 mL/min was used for the separation. Detection
was carried out at 225 nm. Tolbutamide was used as the internal standard.
Noguchi et al. [46] described a simple and sensitive HPLC method for a
routine assay of gliclazide in serum. Serum samples spiked with gliben-
clamide (internal standard) were applied to Bond Elut C18 cartridges. After
washing with phosphate buffer (pH 7.5) and water, the cartridge was eluted
with 60% methanol. The eluate was evaporated to dryness. The residue was
dissolved in methanol and injected onto an octadecyl silica column (5 m,
150 mm  4.6 mm I.D.). The mobile phase was 0.04 M potassium
dihydrogen phosphate (pH 4.6)acetonitrileisopropyl alcohol (5:4:1,
v/v/v). Ultraviolet detection at 227 nm was used. The minimum detectable
level of gliclazide was 20 ng/mL.

4.2.2 Calorimetric Methods


Awasthi and Kulkarni [47] developed the hollow floating beads of gliclazide.
The primary effect of this drug is to potentiate glucose-stimulated insulin
release from pancreatic islet--cells by induction of a decrease in potassium
efflux from these cells. Because of the poor aqueous solubility, its absorption
is limited. Thus, an attempt was made to improve its release profile.
Methods: The hollow drug-loaded alginate beads in combination with
150 Fatmah A.M. Al-Omary

low methoxyl pectin and hydroxypropyl methylcellulose (HPMC) were


prepared by a simple ionotropic gelation method. The beads were evalu-
ated for particle size and morphology using optical microscopy and scan-
ning electron microscopy (SEM). The FTIR spectroscopy, differential
scanning calorimetry (DSC), and X-ray diffraction analysis showed
stable character of drug in the drug-loaded hollow beads and revealed
the absence of any drugpolymer interactions. The beads remained buoy-
ant for more than 12 h. The drug release from beads followed Fickian
diffusion with swelling. Conclusion: The preliminary results of this study
suggest that the developed beads containing gliclazide could enhance drug
entrapment efficiency, reduce the initial burst release, and modulate the
drug release.
The dissolution profile of gliclazide was improved by developing floating
alginate beads using various biodegradable polymers like gelatin, pectin, and
HPMC [48]. The floating beads were prepared by a simple ionotropic
gelatin method using calcium carbonate as the gas-generating agent. The
developed beads were characterized by the FTIR spectroscopy analysis,
DSC, X-ray diffraction analysis, and SEM. The prepared beads showed
good in vitro floatation, which was dependent on the concentration of
the gas-forming agent. A significant (P < 0.05) reduction in fasting and
nonfasting blood glucose levels, a reduction in fasting plasma insulin level,
and a significant improvement in glucose tolerance were observed in animals
treated with formulations. The developed beads were suitable carriers for
improving the systemic absorption of gliclazide and maintaining reduced
blood glucose levels.
Patil and Gaikwad [49] studied the effect of polyethylene glycol 4000
(PEG 4000) on in vitro dissolution of gliclazide from solid dispersions. Ini-
tial studies were carried out using physical mixtures of the drug and the car-
rier. Solid dispersions were prepared by the melting or fusion method.
Phase and saturation solubility study, in vitro dissolution of pure drug, phys-
ical mixtures, and solid dispersions were carried out. PEG was found to
be effective in increasing the dissolution of gliclazide in solid dispersions
when compared to pure drug. FTIR spectroscopy, DSC, and X-ray diffrac-
tometry studies were carried out in order to characterize the drug in the
physical mixtures and solid dispersions. Dissolution enhancement was
attributed to decreased crystallinity of the drug and to the wetting and
solubilizing effect of the carrier from the solid dispersions of gliclazide.
In conclusion, dissolution of gliclazide can be enhanced by the use of the
hydrophilic carrier.
Gliclazide 151

Lo et al. [50] studied the solid complex of gliclazide and -cyclodextrin


(CD), which was prepared by the neutralization method, and the precipita-
tion solvent evaporation method was used to prepare gliclazide nanospheres.
FTIR spectroscopy and DSC were used to examine whether gliclazide solid
complex and gliclazide nanospheres were successfully formed in this study.
The dissolution rate of gliclazide from its nanospheres was faster than its solid
complex and pure drug. The morphology of particles for nanospheres
showed no crystal character of gliclazide. In summary, the results indicate
that nanotechnology provides better effects in solubility and dissolution rate
of gliclazide than the neutralization method.

4.2.3 Ultraviolet Spectrometric Methods


Charged aerosol detection (CAD), a new kind of universal detection
method, has been widely employed in the HPLC system. Shaodong
et al. [51] studied four kinds of antidiabetic drug standards, glipizide,
gliclazide, glibenclamide, and glimepiride by UV detection, evaporative
light scattering detection (ELSD), and the aforementioned CAD. The results
were compared with reference to linearity, accuracy, precision, and LOD.
All of the experiments were performed on a reverse phase column with
water and acetonitrile as the mobile phase. Separations were achieved under
the same chromatographic conditions for each detection method. As a result,
CAD generated nearly uniform responses compared with UV detection and
ELSD. It showed the best accuracy and LOD among three detectors and had
similar precision with UV detection at higher concentrations, while UV
detection showed a better precision at lower concentrations than did
CAD or ELSD. The LOD of CAD, in fact, can be up to two times higher
than that of ELSD. The UV and ELSD linearity was satisfactory at r2 > 0.99,
though in the case of CAD, a loglog transformation was needed. The pro-
posed methods were also applied to the real antidiabetic drugs and diabetes-
related dietary supplements.
El-Enany [52] developed an accurate, sensitive, and simple spectropho-
tometric and spectrofluorimetric method for the determination of glicla-
zide in pharmaceutical formulations and biological fluids. Both methods
are based on a coupling reaction between gliclazide and 4-chloro-7-
nitrobenzo-2-oxa-1,3-diazole in borate buffer, pH 7.8, in which a yellow
reaction product that can be measured spectrophotometrically at 400 nm.
The same product exhibited a yellow fluorescence at 470 nm upon excita-
tion at 400 nm. The absorbanceconcentration plot was rectilinear over the
range of 220 g/mL with minimum detectability (S/N ratio 2) of
152 Fatmah A.M. Al-Omary

0.2 g/mL (6.18  107 M); the fluorescenceconcentration plot was rec-
tilinear over the range of 0.22.5 g/mL with minimum detectability
(S/N ratio 2) of 0.02 g/mL (6.18  108 M). The different experimental
parameters affecting the development and stability of the color were care-
fully studied and optimized. Both methods were successfully applied to
the analysis of commercial tablets. The results were in good agreement with
those obtained with the official and reference spectrophotometric methods.

5. BIOLOGICAL ANALYSIS
Lakshmi and Rajesh [53] developed and validated an analytical method
based on the isocratic RP-HPLC for the separation and quantification of
eight antidiabetic drugs: rosiglitazone, pioglitazone, glipizide, gliclazide,
repaglinide, nateglinide, glibenclamide, and glimepiride for their application
in human plasma assay. Metformin was used as an internal standard. Analysis
was done on Onyx monolithic C18 column (100 mm 4.6 mm I.D., 5 m)
using a mixture of 0.05% formic acid in water and methanol in the ratio of
42:58 (v/v) fixed at a flow rate of 0.5 mL/min, and they were monitored at
234 nm. Separation was achieved in less than 20 min. The calibration curves
were linear in the range of 502000 ng/mL. The method was validated for its
recovery, intra- and interday precision, stability, specificity, and selectivity.
Plasma samples were prepared using solid-phase extraction of analytes.
The developed method was found to be suitable for the routine analysis of
selected antidiabetic drugs in biological matrices.
Circulating endothelial progenitor cells (EPCs) play an important role in
the development and progression of diabetic vascular complications. Chen
et al. [54] investigated the effects of gliclazide plus metformin (GLIMET)
compared with metformin alone on number and function of circulating
EPCs in T2DM patients. This study demonstrated that both metformin
monotreatment and metformin plus gliclazide combination treatment
provided with improvements in number and function of circulating
EPCs. Compared with metformin monotreatment, early use of combina-
tion therapy with GLIMET made more effective improvements in
circulating EPCs.
Wang et al. [55] developed a method for safety monitoring of natural die-
tary supplementsquality profile. It would convert passive monitoring of
synthetic drug to active and guarantee the security of natural dietary supple-
ments. Preliminary research on quality profile was completed by HPLC
and MS.
Gliclazide 153

Seedher and Kanojia [56] studied the mechanism of interaction of anti-


diabetic drugs, repaglinide and gliclazide, to HSA using a fluorescence spec-
troscopic technique. Repaglinide had much higher affinity for HSA when
compared with gliclazide. Site specificity can be useful in predicting the
competitive displacement of these drugs by other coadministered drugs,
resulting in fluctuations of the blood glucose levels in diabetic patients.
The SternVolmer analysis of quenching data indicated that the tryptophan
residues are not fully accessible to the drugs and predominantly dynamic
quenching mechanism is involved in the binding.
Hypoglycemia induced by surreptitious sulfonylurea ingestion is difficult
to distinguish from an insulin-secreting tumor. Shenfield et al. [57] described
a technique for detecting most of the common sulfonylurea drugs in the
plasma. After preliminary acidification, and extraction in ether, the residue
was reconstituted and injected onto a Versapack HPLC column. Detection
was at 230 nm. This procedure gives good separation of chlorpropamide,
glibenclamide, gliclazide, glipizide, and tolbutamide. Results were semi-
quantitative, but the sensitivity of the assay was sufficient to detect and iden-
tify clinically active concentrations of all five drugs.

6. STABILITY
Jondhale et al. [58] transformed gliclazide into a glassy state by melt
quench technique in order to improve its physicochemical properties.
Chemical stability of GLI during formation of glass was assessed by moni-
toring thin-layer chromatography, and an existence of amorphous form
was confirmed by DSC and X-ray powder diffractometry. The glass transi-
tion occurred at 67.5C. The amorphous material thus generated was exam-
ined for its in vitro dissolution performance in phosphate buffer (pH 6.8).
Surprisingly, amorphous GLI did not perform well and was unable to
improve the dissolution characteristics compared to pure drug over entire
period of dissolution studies. These unexpected results might be due to
the formation of a cohesive supercooled liquid state and structural relaxation
of amorphous form toward the supercooled liquid region, which indicated
functional inability of amorphous GLI from stability point of view. Stabili-
zation of amorphous GLI was attempted by elevation of T(g) via formation
of solid dispersion systems involving comprehensive antiplasticizing as well
as surface adsorption mechanisms. During accelerated stability studies, ter-
nary systems showed no significant reduction in drug dissolution
154 Fatmah A.M. Al-Omary

performance over a period of 3 months, indicating excellent stabilization of


amorphous GLI.
Chadha et al. [59] investigated possible differences induced in the phys-
icochemical properties within the amorphous forms prepared by different
methods. Enthalpy of solution measured by solution calorimetry was utilized
to highlight the differences prevailing within the amorphous forms and to
determine the percentage of amorphous content. Emphasis is laid on the
quantification and physical stability of these forms. Amorphization was
induced in poorly water-soluble oral hypoglycemic agents (repaglinide,
gliclazide, and glipizide), by quench cooling, vaporization under reduced
pressure, and lyophilization. The amorphous nature was evident from a halo
pattern in powder X-ray diffraction. A glass transition event is evident in
DSC thermograms of the amorphous forms of the three drugs. The amor-
phous forms showed improvement in solubility and dissolution profiles.
Subjecting these amorphous forms to different relative humidities at 25C
for 3 months and subsequent analysis showed that the amorphous form of
repaglinide prepared by quench cooling is most stable and has the potential
to be formulated without any additive, while the amorphous form of
gliclazide tends to devitrify pointing toward its unstable nature.

7. PHARMACOKINETICS
Gliclazide is readily absorbed from the gastrointestinal tract. It is
extensively bound to plasma proteins. The half-life is about 1012 h.
Gliclazide is extensively metabolized in the liver to metabolites that have
no significant hypoglycemic activity. Metabolites and a small amount of
unchanged drug are excreted in the urine [3].
Aburuz et al. [60] described the development of SPE and HPLC methods
for the simultaneous determination of metformin and glipizide, gliclazide,
glibenclamide, or glimepiride in plasma. The simultaneous determination
of these analytes is important for the routine monitoring of diabetic patients
who take combination medications and for studying the pharmacokinetics
of the combined dosage forms. In addition, this developed method can serve
as a standard method for the plasma determination of these analytes, there-
fore saving time, effort, and money.
Aggarwal et al. [61] studied the dissolution rate and bioavailability of
gliclazide by complexation with -CD in the presence of HPMC. Phase
solubility studies were performed in aqueous solutions of different
Gliclazide 155

concentrations of gliclazide alone and in the presence of some water-soluble


polymers.
Al-Kassas et al. [62] investigated the preparation of biodegradable beads
with alginate polymer by an ionotropic gelation method to take the advan-
tages of the swelling and mucoadhesive properties of alginate beads for
improving the oral delivery of gliclazide. It demonstrates that the ionic gela-
tion of alginate molecules offers a flexible and easily controllable process for
manipulating the characteristics of the beads that are important in control-
ling the release rate and consequently the absorption of gliclazide from the
gastrointestinal tract. Variations in polymer concentration, stirring speed,
internal phase volume, and the type of surfactant in the external phase were
examined systemically for their effects on the particle size, incorporation
efficiency, and flow properties of the beads.
Al-Salami et al. [63] reported that gliclazide has no hypoglycemic effect
on type 1 diabetic (T1D) rats, while monoketocholic (MKC) does, and their
combination exerted a better hypoglycemic effect than MKC alone. They
also showed that the most hypoglycemic effect was noticed when rats were
treated with probiotics and then gavaged with MKC + gliclazide. The aim of
this study was to investigate the influence of probiotics on MKC pharma-
cokinetics when coadministered with gliclazide in T1D rats. The decrease
in MKC bioavailability, when administered with gliclazide, caused by pro-
biotic treatment in healthy but not diabetic rats suggests that probiotic treat-
ment induced MKC metabolism or impaired its absorption only in healthy
animals. The different MKC bioavailability in healthy and diabetic rats was
explained by a different induction of presystemic elimination of MKC in the
gut by probiotic treatment.
Al-Salami et al. [64] also investigated the influence of sodium 3,7-
dihydroxy-12-keto-5-cholanate MKC on the ileal permeation of gliclazide
in healthy and diabetic rats treated with probiotics. In healthy rats treated
with probiotics, the degradation of MKC by bacterial polypeptides pro-
duced divalent bile salts, which resulted in reducing secretion and stimulat-
ing the absorption of gliclazide. In contrast, in diabetic rats treated with
probiotics, MKC had no effect possibly due to a difference in the metabolic
profile and resulting in no net flux.
Al-Salami et al. [65] investigated how the semisynthetic bile acid, 3,7-
dihydroxy-12-keto-5-cholanate, also known as 12-monoketocholic acid
(MKC) influences the ileal permeation of gliclazide in healthy and diabetic
rats. The lack of any net flux of gliclazide in diabetic rats suggests the lack of
action of drug transporters involved or the suppression of their expression.
156 Fatmah A.M. Al-Omary

Furthermore, MKC-induced inhibition of mucosal to serosal unidirectional


flux of gliclazide, in healthy rats, can be the result of the selective inhibition
of Mrp3.
Al-Salami et al. [66] studied the influence of probiotics on gliclazide
pharmacokinetics and the effect of both probiotics and gliclazide on blood
glucose levels in healthy and diabetic rats. The probiotic treatment of dia-
betic rats increases gliclazide bioavailability and lowers blood glucose levels
by insulin-independent mechanisms, suggesting that the administration of
probiotics may be beneficial as adjunct therapy in the treatment of diabetes.
Arno et al. [67] studied metformin/gliclazide extended-release tablets,
which were formulated with Eudragit NE30D by a wet granulation tech-
nique. Two batches were prepared in order to study the influence of the
drugpolymer ratio on the tablet formation and in vitro drug release. The
formulated tablets were characterized by disintegration time, hardness, fri-
ability, thickness, weight variation, and in vitro drug release. The percentage
of polymer, with respect to metformin/gliclazide, required to produce tab-
lets with acceptable qualities was 913.45. The quantity of metformin/
gliclazide present in the tablets and the release medium were estimated by
a validated HPLC method. The formulated tablets had acceptable physico-
chemical characters and released the drug over 68 h. The data obtained
from in vitro release studies were fitted with various kinetic models and were
found to follow Higuchi kinetics.
Asyarie and Rachmawati [68] studied the use of PEG 6000 as a matrix to
disperse gliclazide in the solid state, and the pharmacokinetic profile of this
solid dispersion was studied in rats. In conclusion, gliclazide dissolution
increased in the presence of PEG 6000.
Brendel et al. [69] studied several types of metrics based on observations
(standardized prediction error with or without simulation and normalized
prediction distribution error), based on hyperparameters (with or without
simulation), and based on the likelihood of the model. All the metrics
described were applied to evaluate a model built from two phase II studies
of gliclazide. A real phase I dataset and two datasets simulated with the real
dataset design are used as external validation datasets to show and compare
how metrics are able to detect and explain potential adequacies or inadequa-
cies of the model. In conclusion, for external model evaluation, prediction
distribution errors are recommended when the aim is to use the model to
simulate data. Metrics through hyperparameters should be preferred when
the aim is to compare two populations and metrics based on the objective
function are useful during the model building process.
Gliclazide 157

Xu et al. [70] investigated the impact of genetic polymorphisms on the


pharmacokinetics and pharmacodynamics of sulfonylurea drugs. CYP2C9
is the major enzyme involved in sulfonylurea drug metabolism. CYP2C9
variant allele carriers have a significant lower apparent clearance of these
medicines. CYP2C19 genotype is more influential for gliclazide pharmaco-
kinetics when compared to CYP2C9. Sulfonylurea pharmacodynamics is
affected by several genes. Sulfonylurea receptor 1 (SUR1, ABCC8 gene)
and K+ inward rectifier Kir6.2 (KCNJ11) have been correlated to significant
variation in sulfonylurea response. Diabetics with the SUR1 exon 33 G
allele are more sensitive to gliclazide, and the rs5210 variant of the KCNJ11
gene was associated with improved clinical efficacy of gliclazide. Carriers of
transcription factor 7-like 2 (TCF7L2) variants are more likely to fail sulfo-
nylurea therapy. On the other hand, patients with HNF-1 mutations had a
significant greater response to gliclazide when compared to those with type 2
diabetes. The Arg972 polymorphism of insulin receptor substrate 1 (IRS1)
may lead to secondary failure of sulfonylurea therapy. Calpain 10 gene
(CAPN10) polymorphism has also been linked to a sulfonylurea drug
response. In conclusion, the pharmacokinetics and pharmacodynamics of
sulfonylurea drugs are needed to variability in response to these all potential
contributing factors to variability in response to these which in turn will
provide information to optimize sulfonylurea use in people with diabetes.
Cho et al. [71] evaluated the bioequivalence of a gliclazide/metformin
combination tablet (at dose of 80/500 mg) with coadministration of metfor-
min (500 mg) and gliclazide (80 mg) as individual tablets in healthy male
Korean volunteers and concluded that the combination tablet of
gliclazide/metformin is bioequivalent to coadministration of individual tab-
lets. As a result, the combination tablets are regarded therapeutically equiv-
alent and exchangeable to the coadministration of individual tablets in
clinical practice. Moreover, the combination tablets are expected to
improve convenience and adherence to prescribed therapy and to contribute
to better blood glucose control for patients with T2DM.
Courtois et al. [72] studied oral administration of gliquidone (30 mg), gli-
benclamide (5 mg), gliclazide (80 mg), and glipizide (5 mg) on six middle-
aged (4259 years old) and six aged (7175 years old) on separate days. The
half-life of gliclazide was higher than that of the other three hypoglycemic
agents in middle-aged subjects and was the sole to be significantly increa-
sed in aged subjects. There is no obvious difference between sulfonylureas eli-
minated mainly by either the kidney (glibenclamide, gliclazide, glipizide) or
the liver (gliquidone) in terms of the influence of aging upon their clearance.
158 Fatmah A.M. Al-Omary

The pharmacokinetics and pharmacodynamics of gliclazide in 9


Caucasians and 10 Australian Aborigines with uncomplicated T2DM were
assessed by Davis et al. [73].
Delrat et al. [74] developed a new MR formulation containing 30 mg of
gliclazide to obtain a better predictable release of the active principle and to
allow once-daily dosing regimen. An absolute bioavailability study was
carried out to characterize the performance of the new formulation, and
the food effect was also investigated in a separate study. In conclusion,
after single-oral administration of a 30 mg MR tablet, gliclazide was
completely absorbed both under fasted and fed conditions. A consistent
and optimal release of gliclazide from this formulation leads to a low to
moderate overall variability of its pharmacokinetic parameters. Diamicron
30 mg MR can be given without regard to meals, i.e., before, during, or
after breakfast.
El-Maghraby and Alomrani [75] investigated the effects of binary and
ternary solid dispersions of gliclazide with PEG 6000 and/or pluronic
F68 (PL F68) on the dissolution of gliclazide. The study also investigated
the intestinal absorption in the presence of solid dispersion components.
The latter employed the in situ rabbit intestinal perfusion technique. The
ternary solid dispersion of gliclazide with both polymers resulted in
rapid drug dissolution with most drug being released in the first 5 min.
The intestinal perfusion indicated the possibility of complete drug absorp-
tion from the small intestine. This study suggested that the absorption of
gliclazide is dissolution rate limited. The presence of PEG 6000 did not alter
the intestinal absorption, but PL F68 showed a trend of enhanced intestinal
absorption of the drug. Ternary solid dispersion can thus provide rapid
absorption due to rapid dissolution and potential increase in intestinal
permeability.
Frey et al. [76] studied the relationship between the pharmacokinetics of
gliclazide and its long-term pharmacodynamic effect in a large population of
type 2 diabetic patients to identify factors predicting intersubject variability.
The study results underline the clinical interest of quickly increasing the dose
of gliclazide MR according to the response to treatment in order to achieve
effective blood glucose control.
Hermann et al. [77] studied the immediate release and modified release of
gliclazide formulation tablets, which are available on the market. The kinet-
ics of gliclazide release from these tablets concluded that the immediate
release of gliclazide formulation tablets obeys a first-order equation.
Gliclazide 159

Kim et al. [78] studied the pharmacokinetic and pharmacodynamic prop-


erties of gliclazide after an oral administration of gliclazide tablets in healthy
volunteers. Observations indicated that the maximum hypoglycemic effect
of gliclazide was reached at approximately at 1.5 h after the administration
and the effect decreased, probably because of the homeostasis mechanism, in
health volunteers.
Kobayashi et al. [79] studied the pharmacokinetics of total and free
gliclazide in healthy (n 12) and diabetic (n 12) subjects. The serum level
of gliclazide was determined by an HPLC method. The results of this study
showed that the pharmacokinetics of the total gliclazide level reflect those of
the free gliclazide in serum.
The blood level of gliclazide was determined by an HPLC, and the free
gliclazide (unbound to proteins) in the serum was separated by means of an
ultrafiltration by Kobayashi et al. [80] in the healthy (n 12) and diabetic
subjects (n 11). The binding ratio of gliclazide to the blood proteins was
about 96% during the periods of 24 h after administration of the drug. Sev-
eral pharmacokinetic parameters for the blood gliclazide were derived
from the decay curves of the blood drug levels. In conclusion, the result
showed that the pharmacokinetics of the total blood level of gliclazide
reflect the free gliclazide level; moreover, gliclazide predominantly binds
with albumin in the blood and its binding ratio is not constant, but the var-
iable according to the dose relation between the drug and the serum
protein.
Mandal et al. [81] studied the in vitroin vivo correlation (IVIVC) for
two 60 mg gliclazide extended-release formulations (fast and slow release)
given once a day to compare their plasma concentrations over time. Linear
regression analysis of the mean percentage of dose absorbed vs the mean per-
centage of in vitro release resulted in a significant correlation (r2 > 0.98) for
the two formulations. An average percent prediction error for Cmax was
4.15% for fast release and 3.99% for slow release formulation, whereas for
AUC(0infinity) it was 6.36% and 4.66% for fast release and slow release for-
mulation, respectively.
de Smet and Fischer [82] reported that administration of gliclazide 30 min
prior to meals allegedly offers the advantage that an active plasma level has
been reached when food enters the gastrointestinal tract. Alleged disadvan-
tages are higher risk of hypoglycemia and poor compliance. The absorption
rate of glibenclamide and tolbutamide was not affected by food. The study
results concerning gliclazide were contradictory. From the authors opinion,
160 Fatmah A.M. Al-Omary

the pharmacokinetic and pharmacodynamic evidence that sulfonylurea deriv-


atives should be taken 30 min before meals appears to be so limited.

7.1 Metabolism
Gliclazide is absorbed after oral administration. It is extensively metabolized
by hydroxylation, N-oxidation, and oxidation to several inactive metabo-
lites; the p-carboxy metabolite, which accounts for about 1% of the plasma
concentration, has no hypoglycemic activity but has some antithrombotic
activity. About 60%70% of a dose is excreted in the urine with less than 5%
as the unchanged drug. The p-carboxy and N-oxide metabolites account for
about 40% of the dose. About 10205 of the dose is eliminated in the feces as
metabolites. After a single-oral dose of 80 mg to 23 subjects, peak plasma
concentrations of 0.74.9 g/mL were attained in about 4 h. Following
daily oral administration of 80 mg to 144 subjects, steady-state plasma
concentrations of 0.38.2 g/mL (mean 2.5) were reported [2].
The traditional sulfonylureas with long half-lives have sustained
stimulatory effects on insulin secretion compared to the short-acting insulin
secretagogue. Wu et al. [83] studied and used the frequently sampled intra-
venous glucose tolerance test (FSIGT) to evaluate the IS, glucose sensitivity
(SG), and acute insulin response after glucose load (AIRg) after 4 months
treatment with either gliclazide or repaglinide. The design of study was
randomized crossover. 20 patients were enrolled with new-onset type 2 dia-
betes (mean age, 49.3 years). Totally three FSIGTs were performed, one
before and one after each of the two treatment periods as aforementioned.
No significant differences in fasting plasma glucose, insulin, body mass
index, blood pressure, glycated hemoglobin, or lipids were noted between
the two treatments. After the repaglinide treatment, higher AIRg, lower IS,
and lower SG were noted, but they did not reach statistical significance. The
disposal index (DI) was also not significantly different between the two treat-
ments. In conclusion, since nonsignificantly higher DI, acute insulin
response AIRg, and lower insulin sensitivity IS and SG were noted after
repaglinide treatment, it might be a better treatment for diabetes, relative
to gliclazide.
Taylor et al. [84] investigated the metabolism of gliclazide in the urine of
nine patients of different ethnic origins receiving gliclazide therapy for the
treatment of diabetes. Urine extracts were analyzed by GC/MS to quantify
and identify the metabolites excreted in urine and the metabolites compared
with the synthesized products. Metabolic profiles in all diabetic patients
Gliclazide 161

were very similar and comparable with those reported for healthy human
volunteers. In addition to the expected metabolites arising from oxidation
of the 4-methylphenyl ring, four isomeric hydroxylated products of the
azabicyclooctyl ring were identified and the structure of a fifth isomer
postulated.
H2S is an important gasotransmitter, generated in mammalian cells from
L-cysteine metabolism. As it stimulates K(ATP) channels in vascular smooth
muscle cells (VSMCs), H2S may also function as an endogenous opener of
K(ATP) channels in INS-1E cells, an insulin-secreting cell line. Yang
et al. [85] studied K(ATP) channel currents in INS-1E cells using the
whole-cell and single-channel recording configurations of the patch-
clamp technique. K(ATP) channels in INS-1E cells have a single-channel
conductance of 78 pS. These channels were activated by diazoxide and
inhibited by gliclazide. In conclusion, endogenous H2S production from
INS-1E cells varies with in vivo conditions, which significantly affects
insulin secretion from INS-1E cells. H2S stimulates K(ATP) channels in
INS-1E cells, independent of activation of cytosolic second messengers,
which may underlie H2S-inhibited insulin secretion from these cells.
Interaction among H2S, glucose, and the K(ATP) channel may constitute
an important and novel mechanism for the fine control of insulin secretion
from pancreatic -cells.
Gliclazide has been recommended for use on the basis of both its met-
abolic and nonmetabolic effects [86]. It has a clear beneficial effect on met-
abolic control in T2DM. Blood glucose and lipid levels are lowered. The
glucose-lowering effects are secondary to both enhanced insulin secretion
and a decrease in insulin resistance. The former is due to closure of a K+
adenosine triphosphate (ATP) channel in the -cell. The mechanism
whereby insulin action on the liver and muscle is potentiated remains
unknown. It does not appear to involve the insulin receptor, and although
glycogen synthase activation is enhanced, this is probably not specified. It has
proven difficult to separate the metabolic effects of gliclazide form of the
effects of improved control. The metabolic actions are probably also shared
with over sulfonylureas. Gliclazide also has beneficial effects on platelet
behavior and function and on the endothelium, in addition to improving
free radical status. These effects should be beneficial for the prevention of
diabetic microangiopathy. Some evidence has appeared for the prevention
of deterioration of diabetic retinopathy, but results are variable and more
convincing studies are required. Many of the nonmetabolic effects of
gliclazide appear to be unique to this agent. Gliclazide thus appears to be
162 Fatmah A.M. Al-Omary

a reasonable choice in the treatment of T2DM with diet failure, both from
the metabolic and from the nonmetabolic standpoint.
Babichev et al. [87] studied the analysis of pancreatic -cell receptors
binding the sulfanilamide drugs, which are widely used in therapy of
T2DM, such as glibenclamide, glipizide, and gliclazide. The study showed
that these drugs are characterized by excellent parameters of specific binding
to these receptors. The receptors were tested for two parameters: number of
binding sites and dissociation constant. Glibenclamide was the most active of
the tested drugs, the other two agents being less active. The binding of these
agents was reversible.
Shustov et al. [88] studied a group of patients with T2DM having disor-
ders in carbohydrate, lipid, and other kinds of metabolism. This increases the
risk of cardiovascular complications and atherogenesis. Therefore, it is advis-
able to use drugs preventing an excessive late phase of insulin secretion with
resultant reduction of hyperinsulinemia.
Misawa et al. [89] reported that hypoglycemic agents with a rapid onset
and short duration of action are useful for controlling postprandial hyper-
glycemia. The results suggest that in T2DM caused by, at least, insulin defi-
ciency, KAD-1229 may improve impaired insulin secretion in the early
phase and attenuate hyperglycemia without causing a sustained
hypoglycemia.
Graal and Wolffenbuttel [90] reported that T2DM is a heterogeneous
disorder characterized by defects in insulin secretion as well as reduced insu-
lin action. During aging, glucose intolerance will gradually develop, and this
is manifested primarily by an increase in the postprandial blood glucose
response while fasting blood glucose levels are often less elevated. Abnormal
-cell secretion of insulin is a main feature of this. Treatment of elderly
patients with T2DM focuses on the reduction of (hyperglycemic) com-
plaints and prevention of the development or progression of secondary com-
plications. Although regular physical activity and dietary measures, aiming at
body weight normalization, are the cornerstones of therapy, pharmacolog-
ical treatment with oral blood glucose-lowering agents often proves neces-
sary to control the hyperglycemia. Therefore, shorter-acting compounds
like tolbutamide and gliclazide have been relatively well tolerated and appear
to be the best choice to treat elderly patients. It is advisable to start with a low
dose and increase the dose, when needed, in small steps. The efficacy of sul-
fonylureas is much greater when they are taken before a meal. Because of the
fact that T2DM is a progressive disease, and residual -cell function decreases
Gliclazide 163

with time, insulin therapy may ultimately be warranted in a significant num-


ber of patients.
Shiba et al. [91] determined serum levels of gliclazide by radioimmuno-
assay in 7 healthy controls and 18 diabetic in-patients receiving single-oral
dosing and consecutive dosing over 5 days. During consecutive administra-
tion, the serum levels both at fasting and at the peak reached a plateau in
2 days and no further accumulations were observed. The steady-state peak
levels of gliclazide in the diabetic patients revealed a strongly positive cor-
relation with the dose per m2 body surface area (r 0.78, P < 0.001), and
their steady-state fasting levels correlated positively but weakly with the dose
per m2 body surface area (r 0.48, P < 0.05). Thus, measuring either the
fasting or the peak concentration of gliclazide will be useful for monitoring
drug concentration in the serum.
Schernthaner [92] studied gliclazide MR in a new formulation of given
once daily. The specifically designed hydrophilic matrix of gliclazide MR
leads to a progressive drug release that parallels the 24-h glycemic profile
in T2DM patients.

7.2 Absorption
Adibkia et al. [93] indicated that preparing gliclazidecrospovidone solid dis-
persion in the drug/carrier ratio of 1:1 using a cogrinding technique is able to
enhance the drug dissolution rate. It follows that the formulation of gliclazide
crosspovidone coground is able to improve the oral absorption of the drug.
Saharan and Choudhury [94] studied the effect of excipients on dissolu-
tion rate enhancement of gliclazide. Ordered mixtures of micronized
gliclazide with lactose, mannitol, sorbitol, maltitol, and sodium chloride
were prepared by manual shaking of glass vials containing the drug and
excipient(s). Different water-soluble excipients, addition of surfactant and
superdisintegrant, drug concentration, and carrier particle size influenced
the dissolution rate of the drug. Dissolution rate studies of the prepared
ordered mixtures revealed an increase in drug dissolution with all water-
soluble excipients. The order of dissolution rate improvement for gliclazide
was mannitol > lactose > maltitol > sorbitol > sodium chloride. Reducing
the carrier particle size decreased the dissolution rate of the drug as well
as the increase in drug concentration. Kinetic modeling of drug release data
fitted best the HixsonCrowell model, which indicates that all the ordered
mixture formulations followed the cube root law fairly well.
164 Fatmah A.M. Al-Omary

Pal and Nayak [95] developed and optimized gliclazide-loaded alginate


methylcellulose mucoadhesive microcapsules by ionotropic gelation using cen-
tral composite design. The developed and optimized alginatemethylcellulose
microcapsules are suitable for prolonged systemic absorption of gliclazide to
maintain lower blood glucose level and improved patient compliance.
Grbic et al. [96] developed a drug-specific absorption model for gliclazide
using a mechanistic gastrointestinal simulation technology (GIST) imple-
mented in GastroPlus (TM) software package. The results of the simulations
were compared with actual clinical data and revealed that the GIST model
gave accurate prediction of gliclazide oral absorption. The generated absorp-
tion model provided the target in vivo dissolution profile for IVIVC and
identification of biorelevant dissolution specification for gliclazide
immediate-release tablets. A set of virtual in vitro data were used for corre-
lation purposes. The obtained results suggest that dissolution specification of
more than 85% gliclazide dissolved in 60 min may be considered as
biorelevant dissolution acceptance criteria for gliclazide tablets.
The studies of Ishibashi and Takashina [97] recommended that gliclazide
is best taken 30 min before breakfast.
Ambrogi et al. [98] studied a new hydrotalcite-like compound used as a
matrix to improve the dissolution rate of gliclazide and to administer at the
same time micro- and oligoelements useful to improve insulin performance.
The results showed that the hybrid nanostructure could represent a prom-
ising system for improving the drug dissolution rate and to release cations
involved in the performance of insulin.
The low water solubility of gliclazide leads to a low dissolution rate and
variable bioavailability. Talari et al. [99] investigated the effect of micro-
nization on the absorption and pharmacokinetics of after oral administra-
tion in rats. Gliclazide microcrystals were prepared using solvent-change
and pH-shift methods. SEM showed considerable changes in the shape
and size of crystals using both methods. In the optimized formulation of
each method, the particle size of treated gliclazide was reduced about
30 (from 290 to 9.9 m) and 61 times (to 4.76 m) by solvent-change
and pH-shift methods, respectively. Recrystallized samples showed faster
dissolution rate than untreated gliclazide particles. Glucose-lowering
effect, Cmax, and area under the drug concentrationtime profile (area
under the curve (AUC)) were compared in diabetic and normal rats.
AUC and Cmax were increased by microcrystals in both groups of animals.
Administration of 40 mg/kg of gliclazide in the form of untreated drug and
microcrystals obtained by solvent-change and pH-shift methods caused
Gliclazide 165

12.49% and 21.04% enhancement in glucose-lowering effect of gliclazide


in diabetic rats, respectively.
Immediate-release and modified-release gliclazide formulation tablets
are available on the market. Hermann et al. [100] measured the kinetics
of gliclazide release from these tablets, and to propose a technique for pro-
ducing gliclazide matrix tablets, comparing their release kinetic profile with
the gliclazide tablets available on the market. The dissolution process of
immediate-release gliclazide formulation tablets (diabrezide, diabezidum,
F, I) obeys a first-order equation. However, the process for MR formulation
tablets (diaprel, diaprel MR, A, B, C, D G, H, J, K) proceeds according to a
zero-order equation.
Scheen [101] investigated gliclazide MR that has been launched by
Servier (Uni Diamicron 30 mg) to be given once daily. The original
hydrophilic matrix improves the biodisponsibility of gliclazide and allows
a progressive release of the drug that better parallels the 24-h glycemic
profile of patients with T2DM. Such characteristics may explain the
rather low risk of hypoglycemic episodes, and the morning administration
should contribute to improve patients compliance. As the common
formulation of gliclazide, the MR formulation is indicated in the treat-
ment of T2DM, in adult subjects, when diet, exercise, and weight loss
are insufficient to restore an adequate metabolic control. It may be used
alone or in combination with metformin, glitazones, acarbose, or insulin,
with the same general principles of use as for the classical formulation of
gliclazide.
Alkhamis et al. [102] studied increasing the solubility of gliclazide in
aqueous media. Therefore, solubilization of gliclazide in a variety of surfac-
tants was investigated. Anionic and cationic surfactants exhibited dramatic
solubilizing ability for gliclazide, whereas nonionic surfactants showed sig-
nificantly lower solubilizing ability. It was found that gliclazide solubility
increases with increasing the carbon chain length of cationic surfactants
and decreases with increasing the carbon chain length of anionic surfactants.
The solubilization data were analyzed on the basis of a pseudo-phase model
with gliclazide exhibiting moderate partition coefficients into the micellar
phase. The possible sites of solubilization of gliclazide in the micelle were
examined by studying the effect of NaCl on solubilization and by comparing
the absorption spectra of gliclazide in different solvents. The results obtained
from these two experiments indicated that gliclazide is solubilized mainly in
the inner core of the cationic surfactant micelles and in the outer regions of
the anionic surfactant micelles.
166 Fatmah A.M. Al-Omary

7.3 Secretion
An et al. [103] investigated insulin secretion function and insulin resistance in
Chinese newly diagnosed T2DM patients (obese and nonobese patients) in
order to provide evidence for clinical treatment. In newly diagnostic
T2DM, IS and insulin secretion function were decreased with the increase
of FPG, but they were different between obese and nonobese group. Insulin
secretion function was recovered better in obese group when eliminated
glucose toxicity.
Ionescu-Trgoviste et al. [104] reported that Diaprel MR can be used
safely in diabetic patients newly diagnosed, uncontrolled on diet or other
oral antidiabetic drugs, overweight, safely in those with cardiovascular dis-
ease, or in patients with a creatinine clearance 5080 mL/min.
Mokuda et al. [105] studied the difference between effects of therapeutic
dose and subtherapeutic dose of gliclazide on the glucose-induced insulin
secretion. Gliclazide in subtherapeutically low dose has different effects
on insulin secretion from in therapeutic dose, namely sharpens the insulin
secretion sensitivity to glucose with no influence on the maximal insulin
secretion. It is possible that low doses of gliclazide might be of interest in
some T2DM patients whose main pathophysiology is the blunting of insulin
secretion response to hyperglycemia.
Bataille [106] studied the insulin secretion from the -cells in the islets
of Langerhans is mainly regulated by glucose entry via its transporter. The
intracellular glucose metabolism induces a rise in the ATP/ADP ratio,
which increases the degree of closure of ATP-sensitive potassium channels
(K(ATP) channels), inducing a higher intracellular K+, which, in turn,
depolarizes the membrane and opens voltage-sensitive calcium channels.
The ensuing Ca2+ entry triggers extrusion of insulin-containing secretory
granules and, thus, hormone secretion. The analysis of the structure of the
genes encoding K(ATP) channels that are made of four Kir subunits (for-
ming the ionic pore) and four regulatory SUR subunits (that contain the
binding site for antidiabetic sulfonylureas) allowed to several subclasses of
those ionic channels to be described: insulin-secreting -cells contain the
SUR1/Kir 6.2 complex, heart and skeletal muscles contain the SUR2A/
Kir 6.2 set, and vascular smooth muscles (such as those present in coronary
arteries) have SUR2B/Kir 6.1 and nonvascular smooth muscle SUR2B/
Kir 6.2. The pharmacological specificity of each sulfonylurea depends on
the type of SUR protein present in each tissue: most of the second-
generation sulfonylureas used in diabetic clinics (e.g., glibenclamide,
Gliclazide 167

glimepiride) display almost the same affinity for SUR1, SUR2A, and
SUR2B, leading to possible harmful adverse effects in type 2 diabetic
patients with an associated cardiovascular pathology. In contrast, among
the second-generation sulfonylureas, only gliclazide displays a remarkable
specificity toward the -cell K(ATP) channels, making this drug particu-
larly safe in all situations, as it does not induce any interference with the
cardiovascular system.
Sulfonylureas primarily act by binding to the SUR subunit of the
K(ATP) channel and inducing channel closure. However, the channel is still
able to open to a limited extent when the drug is bound, so that high-affinity
sulfonylurea inhibition is not complete, even at saturating drug concentra-
tions. K(ATP) channels are also found in cardiac, skeletal, and smooth mus-
cle, but in these tissues are composed of different SUR subunits that confer
different drug sensitivities. Thus, tolbutamide and gliclazide block channels
containing SUR1 (-cell type), but not SUR2 (cardiac, smooth muscle
types), whereas glibenclamide, glimepiride, repaglinide, and meglitinide
block both types of channels. This difference has been exploited to deter-
mine residues contributing to the sulfonylurea-binding site. Sulfonylurea
block is decreased by mutations or agents (e.g., phosphatidylinositol
bisphosphate) that increase K(ATP) channel open probability. Proks et al.
[107] proposed a kinetic model that explains this effect in terms of changes
in the channel open probability and in the transduction between the drug-
binding site and the channel gate. They also clarify the mechanism by which
MgADP produces an apparent increase of sulfonylurea efficacy on channels
containing SUR1 (but not SUR2).
The high-frequency oscillatory pattern of insulin release is disturbed in
T2DM. Although sulfonylurea drugs are widely used for the treatment of
this disease, their effect on insulin release patterns is not well established. Juhl
et al. [108] assessed the impact of acute treatment and 5 weeks of sulfonylurea
(gliclazide) treatment on insulin secretory dynamics in T2DM patients. In
conclusion, gliclazide augments insulin secretion by concurrently increasing
pulse mass and basal insulin secretion without changing secretory burst fre-
quency or regularity. The data suggest a possible relationship between the
improvement in short-term glycemic control and the acute improvement
of regularity of the in vivo insulin release process.
McGavin et al. [109] studied a gliclazide once-daily MR formulation.
The hydrophilic matrix of hypromellose-based polymer in the new formu-
lation effects a progressive release of the drug, which parallels the 24-h
168 Fatmah A.M. Al-Omary

glycemic profile in untreated patients with T2DM. The formulation shows


high bioavailability, and its absorption profile is unaffected by
coadministration with food. Mean plasma glucose levels are significantly
reduced over a 24-h period in patients with T2DM treated with this
gliclazide formulation once daily, in both fasting and postprandial states.
No cardiovascular K(ATP) channel interaction has been observed at thera-
peutic concentrations. In this study, no episodes of nocturnal hypoglycemia
or hypoglycemia requiring third-party assistance were observed during
treatment with gliclazide. Episodes of symptomatic hypoglycemia were
infrequent, occurring in approximately 5% of patients.
Ligtenberg et al. [110] studied the effect of the acute administration of
gliclazide at 160 mg on insulin release during hyperglycemic clamps in type
2 diabetes patients. In conclusion, in long-standing type 2 diabetes the acute
administration of gliclazide significantly enhances the second-phase insulin
release at a moderately elevated blood glucose level.
Gliclazide improves defective insulin secretion and may reverse insulin
resistance observed in patients with NIDDM [111]. These actions are
reflected in a reduction in blood glucose levels, which are maintained during
both short- and long-term administration, and are comparable with that
achieved by other sulfonylurea agents. Gradually accumulating evidence
suggests that gliclazide may be useful in patients with diabetic retinopathy,
due to its hemobiological actions, and that addition of gliclazide to insulin
therapy enables insulin dosage to be reduced. Thus, gliclazide is an effective
agent for the treatment of the metabolic defects associated with NIDDM and
may have the added advantage of potentially slowing the progression of dia-
betic retinopathy. These actions, together with its good general tolerability
and low incidence of hypoglycemia, have allowed gliclazide to be well
placed within the array of oral hypoglycemic agents available for the control
of NIDDM.
Both repaglinide and gliclazide are insulin secretagogues widely used in
the treatment of T2DM. They stimulate insulin secretion through distinct
mechanisms and may benefit patients from different aspects. Zhang et al.
[112] evaluated the effects of repaglinide or gliclazide on glycemic control,
insulin secretion, and lipid profiles in T2DM patients and found that
repaglinide and gliclazide had similar effects on glycemic control and total
insulin secretion, while repaglinide had more effects on improvements in
-cell function and lipid metabolism.
Gliclazide 169

7.4 Excretion
Hu et al. [113] indicated that pioglitazone reduces urinary albumin excretion
by a mechanism that is at least partly independent of blood sugar control.
The correlation of urinary albumin excretion with improvement in urinary
cytokines suggests that this renoprotective effect of pioglitazone in diabetes
may be related to local reduction in cytokine activity within the kidney.
Kamikuko et al. [114] studied a case of an 80-year-old woman with dia-
betes mellitus, which was treated with gliclazide. Prior to the gliclazide
administration, her urinary excretion of albumin, serum urea nitrogen,
and serum creatinine were normal. After the medication, oliguria, edema,
and azotemia developed. On the 24th day when the edema was severe
and generalized, gliclazide administration was terminated. On the following
day, urinary volume increased suddenly (5740 mL/day). Polyuria persisted
for 5 days. Edema improved and urea nitrogen and creatinine were normal-
ized thereafter. Though the mechanism is not known, the clinical course
suggests that gliclazide is the principal causative factor in the water retention
and azotemia in this patient.
Imamura et al. [115] investigated the hypoglycemic action and disposi-
tion of gliclazide in normal and analbuminemic rats. Orally administered
gliclazide exhibited a stronger hypoglycemic action in analbuminemic rats
than in normal rats. However, the plasma concentration of the drug in
the mutant was much lower than that in the normal. This apparent discrep-
ancy may be clarified by measuring the plasma concentration of unbound
gliclazide. Analbuminemic rats gave larger values for the total body clearance
and steady-state volume of distribution of gliclazide than normal rats. The
biliary and urinary excretion rates of radioactivity after intravenous bolus
administration of [3H]-gliclazide were much greater in the mutant than
in the normal. The binding of gliclazide to serum in analbuminemic rats
was much lower than that in normal rats. Furthermore, the radioactivities
of some tissues after oral administration of [3H]-gliclazide were found to
be significantly higher in the mutant than in the normal. These results clearly
indicate that albumin plays an important role in the hypoglycemic activity
and disposition of gliclazide in rats.
Elliot et al. [116] identified the human cytochrome P450 (CYP) enzymes
responsible for the formation of the 6-hydroxy (6-OHGz), 7-hydroxy
(7-OHGz), and hydroxymethyl (MeOH-Gz) metabolites of gliclazide
and concluded that CYP2C9 is the major contributor to gliclazide metabolic
170 Fatmah A.M. Al-Omary

clearance, although CYP2C19 may also be involved in the MeOH-Gz for-


mation (the major metabolic pathway). Factors known to influence
CYP2C9 activity will provide the main source of variability in gliclazide
pharmacokinetics.

8. PHARMACOLOGY
8.1 Sites and Mechanism of Action
Gliclazide binds to the -cell sulfonyl urea receptor (SUR1). This binding sub-
sequently blocks the ATP-sensitive potassium channels K(ATP). The binding
results in closure of the channels and leads to a resulting decrease in potassium
efflux leads to depolarization of the -cells. This opens voltage-dependent cal-
cium channels in the -cell resulting in calmodulin activation, which in turn
leads to exocytosis of insulin-containing secretory granules.
The importance of K(ATP) channels in stimulussecretion coupling of
-cells is well established, although they are not indispensable for the main-
tenance of glycemic control. Drews and D ufer [8] studied a new role for
K(ATP) channels by showing that genetic or pharmacological ablation of
these channels protects -cells against oxidative stress. Increased production
of oxidants is a crucial factor in the pathogenesis of T2DM. T2DM develops
when -cells can no longer compensate for the high demand of insulin
resulting from excess fuel intake. Instead, -cells start to secrete less insulin
and -cell mass is diminished by apoptosis. Both reduction of insulin secre-
tion and -cell mass induced by oxidative stress are prevented by deletion or
inhibition of K(ATP) channels. These findings may open up new insights
into the early treatment of T2DM.
Relatively recent studies have found that blockers of sulfonylureas recep-
tor 1 (SUR1) might have cardiac ischemic protective effects. Bao et al. [117]
evaluated the effects of a selective SUR1 blocker gliclazide on cardiac func-
tion and arrhythmia after isoprenaline-induced myocardial injury in obese
rats. Blocking of the SUR1 thus exerts a protective effect on the
isoprenaline-induced myocardial injury in obese rats. That SUR1 blocker
leads to ischemic protection, suggesting a critical biological role of SUR1
in regulating the function of the cardiovascular system than previously rec-
ognized under pathophysiological conditions.
Sulfonylurea drugs exert their insulinotropic action by inhibiting
K(ATP) channels in the pancreas. However, these channels are also
expressed in myocardial and vascular smooth muscle, implicating possible
Gliclazide 171

detrimental cardiovascular effects. Engbersen et al. [118] investigated the


inhibitory potency of various widely used sulfonylurea drugs in resistance
arteries. They concluded that sulfonylurea drugs exert differential effects
on vascular smooth muscle K(ATP) channels. The results suggest that
glibenclamide and glimepiride will interact with these channels at therapeu-
tic concentrations.
The fasting of Ramadan is observed by a large proportion of Muslims
with diabetes. Recommendations for the management of diabetes during
Ramadan were studied by Ahmed and Abdu [119]. Among the sulfonyl-
ureas, gliclazide and glimepiride can be safely used during Ramadan, but
glibenclamide should be avoided due to the associated risk of hypoglycemia.
In selected patients with type 1 and type 2 diabetes, the long-acting insulin
analogues glargine and detemir, as well as the premixed insulin analogues,
can be used with the minimal risk of metabolic derangement or hypoglyce-
mia; the risk is higher in type 1 diabetes. Insulin pumps can potentially
empower patients with diabetes and enable safe fasting during the month
of Ramadan. Further, clinical trials are needed to evaluate the safety and effi-
cacy of new antidiabetic agents and new diabetes-related technologies dur-
ing Ramadan.
Vascular complications are a common factor determining morbidity
and mortality of diabetic patients. Konya et al. [120] have revealed that
gliclazide has antiplatelet activities in in vitro studies. This action was clin-
ically assessed by measuring the effects of gliclazide on plate activities and
abnormal fibrinolysis in patients with T2DM. The results indicate that
gliclazide inhibits platelet aggregation via the serotonin pathway, indepen-
dently of the metabolic control. Furthermore, in the patients with
improved glycemic control, gliclazide could inhibit ADP-induced plate
aggregation and reduce PAI-I level. Taken together, the results show that
gliclazide may be more useful for the prevention of diabetic vascular com-
plications than glibenclamide.
The results of clinical studies revealed that gliclazide may reduce the risk
of cancer in T2DM, although the mechanism of the possible protective
effect is not sufficiently explored. The increased level of DNA damage
and impaired DNA repair system in diabetes mellitus may play a substantial
role in neoplastic transformation. Gliclazide protected DNA against damage
introduced by the oxidative stress, but its action on the DNA repair mech-
anisms is unclear. Sliwinska et al. [121] assessed whether gliclazide has any
effect on the DNA repair pathways, e.g., nucleotide excision repair
(NER) and nonhomologous end joining (NHEJ). NER activity was assessed
172 Fatmah A.M. Al-Omary

in the extract of human lymphocytes and pancreatic cancer cells (PANC-1)


treated or not with gliclazide by use of an UV-irradiated plasmid as a sub-
strate and by quantitative PCR performed to evaluate the efficacy of the
removal of UV-induced lesions from the p53 gene by intact cells. The effi-
cacy of NHEJ pathway was examined by a simple and rapid in vitro assay
based on the fluorescent detection of repair products. No significant differ-
ences between the efficiency of NER and NHEJ for extracts of lymphocytes
alone and lymphocytes treated with gliclazide were observed. Contrary,
gliclazide increased the efficacy of NER (46.0% vs 84.0%, P < 0.01) and
NHEJ (58.0% vs 66.0%, P < 0.05) in PANC-1 cells. The study concluded
that gliclazide did not affect NER and NHEJ in human normal cells, but it
may stimulate DNA repair in cancer cells.
The effects of hypoglycemic drugs (gliclazide, glibenclamide) and new
sugar-lowering drug diabenol on the coagulation chain of hemostasis and
fibrinolytic system of blood were studied by Spasov et al. [122] on intact rats
and in rats with experimental diabetes mellitus. Experiments revealed the
ability of drugs to reduce thromboelastogram indices, which is probably
related to the ability of hypoglycemic drugs to inhibit the platelet aggrega-
tion and prevent the subsequent activation of the coagulation chain of
hemostasis. All drugs improve the thrombogenic potential (by decreasing
the platelet activation) and increase the activity of the fibrinolytic system
of blood. The activity of diabenol and gliclazide is more pronounced as
compared to that of glibenclamide.
Koh et al. [123] compared the effects of gliclazide, an antidiabetic agent
with antioxidant properties, and found that gliclazide treatment suppressed
dRib-induced oxidative damage in HIT-T15 cells less than N-acetyl-L-
cysteine (NAC) because gliclazide did not restore the intracellular glutathi-
one content as effectively as NAC. In addition, the elevation of intracellular
glutathione rather than free radical scavenging might be an important mech-
anism for protecting against dRib-induced oxidative damage in a -cell line.
Aravind et al. [124] determined the incidence of hypoglycemia during
Ramadan in Muslim subjects with T2DM treated with a sulfonylurea.
Five-country observational study found that nearly 20% of sulfonylurea-
treated Muslim subjects with T2DM experienced symptomatic hypoglyce-
mia while fasting during Ramadan, with variations across sulfonylureas and
countries.
Mohiuddin et al. [125] studied the in vivo effects of gliclazide and met-
formin HCl on plasma concentration of caffeine in rats. The plasma concen-
tration of caffeine was determined by UV spectrophotometry after oral
Gliclazide 173

single administration of caffeine alone and with gliclazide and metformin


HCl. The in vivo study for determination of plasma concentration of caf-
feine showed that concurrent administration of caffeine and gliclazide has
not made noticeable changes in plasma concentration of caffeine. But
administration of caffeine and metformin HCl has showed a significant
change in plasma concentration of caffeine. So, a competitive inhibition
of the binding to plasma protein by metformin HCI increases the plasma
concentration of caffeine. Thus any change in plasma concentration may
affect the pharmacological or toxic effects of the drug.
Del Guerra et al. [126] studied the effects of exposure to high glucose
levels and sulfonylurea on isolated human islet-cell function and investigated
some of the mechanisms that might be involved. In conclusion, gliclazide
and glibenclamide have different effects on the changes induced by pro-
longed exposure of human islet cells to high levels of glucose.
An extrapancreatic effect of sulfonylureas has been postulated. However,
in vivo results have been disputed because the amelioration of insulin action
that follows sulfonylurea may represent the relief from glucose toxicity
rather than a direct effect of the drug. da Tos et al. [127] studied the hypo-
glycemic action of gliclazide acutely and after 2 months of therapy in seven
T2DM patients. They suggested that gliclazide enhances the suppression of
endogenous glucose production (EGP) induced by insulin and that this
effect is greater with chronic treatment because of concomitant improve-
ment of insulin secretion.
A specific site on the ATP-sensitive potassium channels is occupied by
sulfonylureas, leading to closure of the potassium channels and subsequent
opening of calcium channels [128]. This results in exocytosis of insulin. The
meglitinides are not sulfonylureas and also occupy the sulfonylurea receptor
unit coupled to the K(ATP) channel. Glibenclamide (glyburide), gliclazide,
glipizide, and glimepiride are the primary sulfonylureas in current clinical
use for T2DM. Glibenclamide has a higher frequency of hypoglycemia than
the other agents. With long-term use, there is a progressive decrease in the
effectiveness of sulfonylureas. This loss of effect is the result of a reduction in
insulin-producing capacity by the pancreatic -cell and is also seen with
other antihyperglycemic agents. The major adverse effect of sulfonylureas
is hypoglycemia. There is a theoretical concern that sulfonylureas may affect
cardiac potassium channels resulting in a diminished response to ischemia.
There are now many choices for initial therapy of T2DM in addition to sul-
fonylureas. Metformin and thiazolidinediones affect IS by independent
mechanisms. Disaccharidase inhibitors reduce the rapid carbohydrate
174 Fatmah A.M. Al-Omary

absorption. No single agent appears capable of achieving target glucose levels


in the majority of patients with T2DM. Combinations of agents are success-
ful in lowering glycosylated hemoglobin levels more than with a single
agent. Sulfonylureas are particularly beneficial when combined with agents
such as metformin that decrease insulin resistance. Sulfonylureas can also be
given with a basal insulin injection to provide enhanced endogenous insulin
secretion after meals. Sulfonylureas will continue to be used both primarily
and as part of combined therapy for most patients with T2DM.
Gribble and Reimann [129] studied the sulfonylurea receptor subunits of
K(ATP) channels. Sulfonylureas close K(ATP) channels in pancreatic -cells
and are used to stimulate insulin release in T2DM, whereas the K(ATP) chan-
nel opener nicorandil acts as an antianginal agent by opening K(ATP) channels
in cardiac and vascular smooth muscle. The predominant type of SUR varies
between tissues: SUR1 in -cells, SUR2A in cardiac muscle, and SUR2B in
smooth muscle. Sulfonylureas and related drugs exhibit differences in tissue
specificity, as the drugs interact to varying degrees with different types of
SUR. Gliclazide and tolbutamide are -cell selective and reversible.
Glimepiride, glibenclamide, and repaglinide, however, inhibit cardiac and
smooth muscle K(ATP) channels in addition to those in -cells and are only
slowly reversible. Similar properties have been observed by recording K(ATP)
channel activity in intact cells and in Xenopus oocytes expressing cloned
K(ATP) channel subunits. While K(ATP) channels in cardiac and smooth
muscle are largely closed under physiological conditions (but open during
ischemia), they are activated by antianginal agents such as nicorandil. Under
these conditions, they may be inhibited by sulfonylureas that block SUR2-
type K(ATP) channels (e.g., glibenclamide). Care should, therefore, be taken
when choosing a sulfonylurea if potential interactions with cardiac and
smooth muscle K(ATP) channels are to be avoided.
Among sulfonylureas, gliclazide is one of the mostly prescribed drugs to
diabetic patients and is metabolized extensively by P450 CYP2C9. Among
24 CYP2C9 alleles, the *2/*2 and *3/*3 genotypes showed significantly
lower gliclazide clearances with reductions of 25% and 57%, respectively.
However, the reason for the change in drug-metabolizing activity induced
by these natural alleles is unknown. Banu et al. [130] studied the molecular
dynamics simulation and autodocking studies to provide models for
gliclazide-bound complexes of CYP2C9*2, *3, and *2/*3 mutants, which
give insight into CYP2C9gliclazide interactions and explain the reduced
enzymatic activity seen in these variants. The data show that the size of
Gliclazide 175

the substrate-access entry site is significantly reduced in mutants, which


limits the access of gliclazide to heme and the active site. The distance from
the substrate oxidation site and heme is >5 A in *3 and *2/*3. Therefore,
the addition of an active oxygen molecule by heme-Fe is hindered. The
absence of F100, F114, and F476 in the interacting amino acid pocket in
*3 reduces catalytic efficiency toward gliclazide. In *1, gliclazide is stabilized
by the formation of two hydrogen bonds with R108, while it is absent in
mutants. Further in *3 and *2/*3, the key heme-stabilizing residue, R97
stabilization, is greatly reduced. Therefore, the decreased catalytic activity
of these variants can be explained from the reduced access of the gliclazide
to heme, and the interaction between the heme and the substrate is affected
due to their instability in the active site.
Zargar et al. [131] reported that a majority of Muslim patients with
T2DM fast during the month of Ramadan, and there are no accepted guide-
lines for its management during this period. The few studies on this subject
suggest that there are important alterations in energy intake and physical
activity, and that most patients change their pattern of drug intake. This
is associated with a greater risk of hypoglycemia and ketoacidosis. The usual
pattern of eating during Ramadan, and its influence on the normal diurnal
variation of blood sugar with a regular nonfasting diabetic diet, suggests that
antidiabetic agents for use during this period need to be selected according to
their pharmacokinetic and tablet formulation characteristics. The sulfonyl-
ureas are first-line drugs in T2DM and used by a majority of patients.
A comparison of the pharmacokinetics, efficacy, and safety characteristics
of these agents suggests that a long-acting once-daily formulation of
gliclazide such as gliclazide MR, taken in the evening, may be the sulfonyl-
urea of choice during Ramadan.

8.2 Cardiovascular Effect


Juurlink et al. [132] reported that older sulfonylureas such as glyburide
(glibenclamide), but not newer ones such as gliclazide, antagonize similar
channels in myocardium, interfering with the protective effects of ischemic
preconditioning. Among older patients hospitalized for acute myocardial
infarction or percutaneous coronary intervention, treatment with glyburide
is not associated with an increased risk of future adverse cardiovascular
events relative to gliclazide, suggesting that the effect of glyburide on ische-
mic preconditioning is of little clinical relevance.
176 Fatmah A.M. Al-Omary

Batty et al. [133] studied and examined the relationship between erectile
problems in men and cardiovascular disease (CVD) mortality. In this cohort
of men with T2DM, erectile dysfunction was associated with a range of
CVD events.
Severe hypoglycemia may increase the risk of a poor outcome in patients
with T2DM assigned to an intensive glucose-lowering intervention.
Zoungas et al. [134] analyzed data from a large study of intensive glucose
lowering to explore the relationship between severe hypoglycemia and
adverse clinical outcomes. Severe hypoglycemia was strongly associated
with increased risks of a range of adverse clinical outcomes. It is possible that
severe hypoglycemia contributes to adverse outcomes, but these analyses
indicate that hypoglycemia is just as likely to be a marker of vulnerability
to such events.
Diabetes is a state of increased oxidant stress and there is evidence that
oxidation may play a role in the genesis of higher left ventricular mass.
Gliclazide has been shown to possess free radical scavenging properties.
Pan et al. [135] studied and assessed whether gliclazide may have a beneficial
effect on left ventricular mass via reducing 8-iso-prostaglandin F(2) con-
centrations, a reliable marker of oxidant injury. The results demonstrated
for the first time that in addition to its primary hypoglycemia, gliclazide
may have an additional effect on reducing left ventricular mass, possibly
through the attenuation of free radical formation.
Atherosclerotic CVD is the leading cause of premature death in patients
with diabetes. Atherosclerosis is a chronic immune-mediated disease, the
initiation, progression, and destabilization of which is driven and regulated
by inflammatory cells. One critical event in the initiation of this vascular
inflammatory disease is the adhesion of leukocytes to the activated endothe-
lium and their migration into the vessel wall. These processes are mediated
by the upregulation of adhesion molecules on ECs and an increased expres-
sion in the vascular wall of chemotactic factors to leukocytes. Monocyte
binding to ECs is increased in diabetes. One major determinant of this alter-
ation could be oxidative stress. Given the free radical scavenging activity of
gliclazide, Renier et al. [136] studied the ex vivo and in vitro effects of this
drug on human monocyte binding to ECs and the molecular mechanisms
involved in this effect. The results demonstrate that short-term administra-
tion of gliclazide to patients with T2DM normalizes the levels of plasma lipid
peroxides and monocyte adhesion in these subjects. Results show that
gliclazide, at concentrations in the therapeutic range, inhibits ex vivo and
in vitro monocyte adhesiveness to vascular cells. By doing so, this gliclazide
Gliclazide 177

could reduce monocyte recruitment into the vessel wall and thereby con-
tribute to attenuating the sustained inflammatory process that occurs in
the atherosclerotic plaque. These findings suggest that the treatment of dia-
betic patients with gliclazide may prevent or retard the development of
vasculopathies associated with diabetes.
Accumulating evidence indicates that oxidative modification of LDL plays
an important role in vascular dysfunction associated with diabetes mellitus.
Mamputu and Renier [137] investigated the effect of gliclazide on human
aortic smooth muscle cell (HASMC)-mediated LDL oxidation and HASMC
dysfunction induced by oxidatively modified LDL. Incubation of HASMCs
with native human LDL (100 g/mL) in the presence of increasing concen-
trations of gliclazide (110 g/mL) resulted in a dose-dependent decrease in
HASMC-mediated LDL oxidation. Exposure of HASMCs to gliclazide
(110 g/mL) and native LDL (100 g/mL) also led to a dose-dependent
decrease in oxidized LDL-induced human monocyte adhesion to HASMCs.
In addition, incubation of HASMCs with gliclazide dramatically reduced the
ability of oxidized LDL to stimulate the proliferation of these cells. Treatment
of HASMCs with gliclazide resulted in a marked decrease in oxidatively mod-
ified LDL-induced monocyte chemoattractant protein (MCP)-1 and human
heat-shock protein 70 (hsp 70) expression, both at the gene and protein levels.
These results show that gliclazide, at concentrations in the therapeutic range
(510 g/mL), is effective in vitro in reducing VSMC dysfunction induced by
oxidatively modified LDL. These observations suggest that administration of
gliclazide to T2DM patients could form part of the strategy for the prevention
and management of diabetic CVDs.
Jennings [138] reported that AGEs and the free radicals generated in this
process can both be implicated in the accelerated atherosclerosis and vascular
and prothrombotic microangiopathic changes typified by diabetes. The rate
of formation of free radicals is dependent on the rate of protein glycosylation
and therefore the level and duration of hyperglycemia. Glycation and oxi-
dation are inextricably linked. Increased oxidative stress due to excess free
radical activity may be central to diabetic vascular disease, since endothelial
cell damage, lipoprotein oxidation, and modification of platelet reactivity
and the arachidonic acid (AA) cascade are all properties of free radicals
and their reaction products, lipid peroxides. The importance of the demon-
stration of the mechanism whereby hyperglycemia contributes to vascular
damage opens the possibility of scavenging free radicals, which will have
effects independently of improving diabetic control. Over the past 15 years,
studies have shown that gliclazide not only lowers blood glucose but also
178 Fatmah A.M. Al-Omary

confers beneficial effects on the hemorrheologic abnormalities seen in dia-


betic vascular disease. Clinically, gliclazide reduces platelet reactivity and
stimulates endothelial prostacyclin synthesis; it also increases fibrinolysis
by its effects on tissue plasminogen activator. These effects, seen both
in vitro and in vivo, are independent of glycemic control and are not seen
with other sulfonylureas. Jennings reported that the beneficial effects of
gliclazide on platelets are related to a reduction in oxidative stress. This prop-
erty is due to gliclazides free radical scavenging ability that relates to the
unique aminoazabicyclooctane ring grafted onto the sulfonylurea. It is fully
maintained by the gliclazide modified-release preparation. In diabetes,
therefore, where increased glycation and oxidation are fundamental to
the pathogenesis of diabetic vascular disease, agents such as gliclazide with
its antioxidant activities may have an enhanced therapeutic role.

9. ADVERSE EFFECTS
Gliclazide may be suitable for use in patients with renal impairment,
but careful monitoring of blood-glucose concentration is essential. It should
not be used in patients with severe renal impairment [3].
Harashima et al. [139] studied the efficacy and safety of combination ther-
apy with sitagliptin and low dosage sulfonylureas on glycemic control and
insulin secretion capacity in Japanese T2DM and found that the combination
therapy with sitagliptin and low dosage sulfonylureas was safe and effective
for glycemic control. Glucagon loading test indicated that 1-year administra-
tion of sitagliptin and sulfonylureas preserved insulin secretion capacity.
The low-affinity sodium glucose cotransporter (SGLT2) is responsible
for most of the glucose reabsorption in the kidney and has been highlighted
as a novel therapeutic target for the treatment of diabetes. Fujimori et al.
[140] discovered sergliflozin etabonate, a novel selective SGLT2 inhibitor,
and found that selective inhibition of SGLT2 increased urinary glucose
excretion and consequently decreased plasma glucose levels. They examined
the antihyperglycemic effects of sergliflozin etabonate in normal and diabetic
rats in comparison with those of a sulfonylurea (gliclazide) and an
-glucosidase inhibitor (voglibose). Sergliflozin etabonate increased urinary
glucose excretion in a dose-dependent manner and inhibited the increase in
plasma glucose after sucrose loading independently of insulin secretion in
normal rats. Sergliflozin etabonate also improved postprandial hyperglyce-
mia in neonatal streptozotocin-induced diabetic rats, whereas gliclazide
did not improve it. In rats with mild or moderate streptozotocin-induced
Gliclazide 179

diabetes, the degree of the antihyperglycemic effects of sergliflozin etabonate


correlated with the severity of the diabetic condition.
Kamikubo et al. [114] studied an 80-year-old woman with diabetes
mellitus treated with gliclazide. Prior to the gliclazide administration, her
urinary excretion of albumin, serum urea nitrogen, and serum creatinine
were normal. After the medication, oliguria, edema, and azotemia devel-
oped. On the 24th day when the edema was severe and generalized, glicla-
zide administration was terminated. On the following day, urinary volume
increased suddenly (5740 mL/day). Polyuria persisted for 5 days. Edema
improved and urea nitrogen and creatinine were normalized thereafter.
Though the mechanism is not known, the clinical course suggests that
gliclazide is the principal causative factor in the water retention and azotemia
in this patient.
NIDDM is associated with an increased risk of macro- and microvascular
degenerative complications. Gliclazide has beneficial effects on the hem-
obiological abnormalities of NIDDM. These effects are mediated by the
azabicyclooctyl ring grafted on to its sulfonylurea core. Ziegler and Drouin
[141] reported that gliclazide reduces platelet hyperadhesion and platelet
hyperaggregability. The beneficial effects of gliclazide on thromboxane/
prostacyclin balance have been confirmed in type 2 diabetic patients after
a 3-month treatment period. Concerning fibrinolysis, gliclazide restores
low plasminogen activity to normal in NIDDM patients previously treated
with first-generation sulfonylureas. Gliclazide increases fibrinolytic potential
by increasing endothelial cell tissue plasminogen activator and prekallikrein
activity.
Mizuno et al. [142] reported that gliclazide inhibits platelet functions, but
its effective concentration is reported to be much higher in vitro than in
vivo. Impedance aggregometry was found to be more sensitive than turbi-
dimetry for detecting the inhibition of platelet aggregation and revealed
significant inhibition at 1  104 M gliclazide. Gliclazide reduced the
amount of prostaglandin I2 (PGI2) needed to inhibit ADP-induced plate-
let aggregation and the adhesiveness of platelets to a rabbit vessel wall after
their preincubation with 1  103 M gliclazide for 10 min. Gliclazide
(1  1041  102 M) had no effect on platelet cyclooxygenase activity.
Gliclazide inhibited thromboxane A2-induced platelet aggregation, but
had no effect on the aggregation triggered by addition of mixtures of AA
and inhibitors of key enzymes regulating various steps of AA metabolism
in platelets. Gliclazide had no significant effect on PGI2-stimulated cyclic
AMP (cAMP) production in platelets. These results show that the difference
180 Fatmah A.M. Al-Omary

in the effective concentrations of gliclazide reported to modulate platelet


functions in vivo and in vitro is partly due to differences in the methods used
to evaluate its effect: turbidimetry evaluates platelet aggregability, but not
other platelet functions modulated by gliclazide in vivo.
Gliclazide has been reported to decrease the platelet function and to
inhibit the progression of diabetic retinopathy in addition to having a hypo-
glycemic effect. To confirm these effects, Baba et al. [143] studied and per-
formed a double-blind randomized study using glibenclamide as a reference
drug. Thirty-eight hospitals from eight university groups in Japan performed
the study on type 2 diabetic subjects. Evaluation of blood glucose control,
platelet adhesiveness, platelet aggregation, and blood lipids over 24 weeks
was assessed by the central committee. Two hundred and eighty-nine
patients were enrolled in the study. Twelve were excluded and 277 were
statistically analyzed. Homogeneity between the two diabetic groups was
demonstrated for background factors. Forty milligrams of gliclazide was
comparable to 2.5 mg of glibenclamide in the potency of hypoglycemic effi-
cacy. Funduscopic aggravations were observed in a statistically smaller num-
ber of cases in the gliclazide group than in the glibenclamide group, and in
evaluation of serum lipids, the gliclazide group was also superior to the
glibenclamide group. No significant difference between the two groups
was found in platelet adhesiveness and aggregation. Gliclazide is a useful drug
in the therapy of diabetes mellitus.

10. DRUG INTERACTIONS


Thiazide diuretics are known to aggravate the diabetic state, so caution
should be taken when administering thiazide diuretics to patients on
gliclazide treatment. Blood sugar control may also be adversely affected
where interaction between gliclazide and barbiturates, glucocorticoids, or
estrogens occurs.
The hypoglycemic effect of gliclazide may be potentiated by insulin,
biguanides, sulfonamides, salicylates, coumarins derivatives, chlorampheni-
col, monoamine oxidase inhibitors, -blockers, oxyphenbutazone, phenyl-
butazone, clofibrate, cimetidine, and ethanol.
Acute alcohol intoxication potentiates the hypoglycemic action of sul-
fonylurea agents. Disulfiram-like reactions with characteristic flushing of
the face, throbbing headache, giddiness, tachypnea, tachycardia, or angina
pectoris may also occur. Chronic alcohol abuse may result in the increased
Gliclazide 181

metabolism of sulfonylurea drugs, shortening the plasma half-life and dura-


tion of action.
There may be an interaction between gliclazide and any of the following
studied by Kosaka [144]:
ACE inhibitors (e.g., enalapril)
Alcohol
Anticoagulants (e.g., warfarin, heparin)
Azole antifungal drugs (e.g., miconazole, clotrimazole)
Barbiturates (e.g., phenobarbital, thiopental)
-Blockers (e.g., metoprolol, propranolol)
Chlorpromazine
Clarithromycin
Corticosteroids (e.g., prednisone)
Danazol
Disopyramide
Diuretics (e.g., thiazides, furosemide)
Fibrates (e.g., fenofibrate)
H2 receptor antagonists (e.g., ranitidine, famotidine, cimetidine)
Monoamine oxidase inhibitors (e.g., selegiline, phenelzine)
Nicotinic acid
Nonsteroidal antiinflammatory drugs (e.g., ibuprofen, naproxen)
Oral contraceptives
Other antidiabetic drugs (e.g., insulin, metformin)
Phenylbutazone
Probenecid
Salbutamol
Salicylates (e.g., acetylsalicylic acid)
Terbutaline
Tuberculosis medications (e.g., isoniazid, ethambutol)

11. TOXICOLOGY
11.1 Toxicity
LD50 3000 mg/kg (orally in mice). Gliclazide and its metabolites may
accumulate in those with severe hepatic and/or renal dysfunction. Jerums
[145] studied the symptoms of hypoglycemia including dizziness, lack of
energy, drowsiness, headache, and sweating.
182 Fatmah A.M. Al-Omary

11.2 Subchronic toxicity


The maximum tolerated dose in dog is between 150 and 200 mg/kg daily.

11.2.1 Four-Week Oral Toxicity Study in the Beagle Dog


Groups of four Beagle dogs (two males, two females) were treated for 30 days
with 0, 15, 30, 45, or 90 mg/kg/day. At the dose of 90 mg/kg, two animals
died as a result of prolonged hypoglycemic coma following 2 weeks of treat-
ment. All others showed normal behavior, with the exception of an increase
in the weight of the liver. No evidence was found for any change in bio-
chemical (apart from the fall in blood glucose), hematological, and histo-
pathological parameters were studied by Shimizu et al. [146].

11.2.2 Two-Month Oral Toxicity Study in the Guinea Pig


Groups of 10 guinea pigs (5 males, 5 females), were treated 6 days out of 7 for
2 months with 0, 25, 50, or 100 mg/kg/day. Only male animals in the
50 mg/kg group showed delayed weight gain. All others had normal bio-
chemical, hematological, and histopathological results.

11.3 Chronic Toxicity


11.3.1 Six-Month Study in the Sprague-Dawley Rat
Groups of 20 rats (10 males, 10 females) weighing 300 g were treated for
6 days out of 7 for 6 months with 0, 25, 100, or 200 mg/kg/day. Seven
deaths occurred as a result of technical problems. All other animals showed
normal behavior and hematological results. From a biochemical standpoint,
blood urea decreased significantly in the male rats as did blood glucose in the
males of the 100 mg/kg/day group. Histological examination showed an
increase in the weight of the liver and kidneys in male animals, not accom-
panied by any histological lesion. A 6-month rat study carried out in Japan
with higher doses (50, 100, 200, 400, and 800 mg/kg) indicates a possible
higher sensibility in the female to the product: slight increases in liver
enzymes together with slight decreases in erythrocytes counts, hematocrit
values, and hemoglobin concentrations at doses of 200 mg/kg and higher.

11.3.2 Six-Month Study in the Beagle Dog


Groups of six dogs (three males, three females) were treated daily for
6 months with 15 or 30 mg/kg of gliclazide or 30 mg/kg of gliclazide or
50 mg/kg of tolbutamide [146].
Gliclazide 183

From a clinical standpoint


Three deaths (one at 15 mg/kg, two at 30 mg/kg) in the gliclazide group
as a result of hypoglycemic coma.
One convulsion, four cases of severe gastrointestinal disturbances in the
tolbutamide group.
Weight changes and food consumption were similar with both drugs.
From a laboratory standpoint
40% fall in blood glucose in animals treated with gliclazide.
Signs of hepatotoxicity in the tolbutamide group.
From a histological standpoint
Increase in weight of the liver in the three deaths of the gliclazide group.
Increase in the weight of the liver and lesions of toxic hepatitis in five
animals out of six of the tolbutamide group.

11.3.3 Twelve-Month Oral Toxicity Study in the Beagle Dog


Groups of eight dogs (four males, four females) were treated for 12 months
with 0, 12, or 24 mg/kg/day of gliclazide.
Four animals in each group were sacrificed after 90 days:
there were no deaths;
no evidence of any modification in behavior and body weight;
significant fall in blood glucose;
fluctuation in certain parameters (liver enzymes, lipid profile, creatinine);
at autopsy.
Swelling of the renal and hepatic parenchyma at the highest dose and slight
increase in the weight of the thyroid and slight decrease in the weight of the
pituitary gland.

11.3.4 Twelve-Month Oral Toxicity Study in the Rhesus Monkey


Groups of eight rhesus monkeys (four males, four females) were treated daily
for 12 months with 0, 20, 60, or 180 mg/kg of gliclazide:
No evidence was found of any modification in weight gain nor food
consumption;
Significant fall in blood glucose;
Irregular rise in some liver enzymes in some animals;
No abnormality by histopathological examination.

ACKNOWLEDGMENT
The author wishes to thank Mr. Tanvir Ahmed Butt for his secretarial assistance in typing of
this profile.
184 Fatmah A.M. Al-Omary

REFERENCES
[1] British Pharmacopoeia, Monograph: Medicinal and Pharmaceutical Substances, vols.
I and II, Medicines & Health Products Regulatory Agency, UK, 2013, pp. 10221023.
[2] A.C. Moffat, D. Osslteon, B. Widdop (Eds.), Clarks Analysis of Drugs and Poison,
third ed., Pharmaceutical Press, London, 2004, p. 640.
[3] J.E.F. Reynold (Ed.), Martindale: The Extra Pharmacopeia, 32nd ed., Royal
Pharmaceutical Society, London, 1999, p. 320.
[4] European Pharmacopoeia, Monograph, seventh ed., The Council of Europe,
Strasburg, 2008, pp. 20962097.
[5] D. Bonnefont-Rousselot, Antioxidant and anti-age therapeutics: evaluation and per-
spectives, J. Soc. Biol. 195 (2001) 391398.
[6] R.C. OBrien, M. Luo, N. Balazs, J. Mercuri, In vitro and in vivo antioxidant properties
of gliclazide, J. Diabetes Complicat. 14 (2000) 201206.
[7] W.P. Sun, Y. Bi, H. Liang, M.Y. Cai, X. Chen, Y.H. Zhu, L.Z. Liao, J.P. Weng, The
effects of insulin and gliclazide therapy on endoplasmic reticulum stress and insulin sen-
sitivity in liver of type 2 diabetic rats, Zhonghua Nei ke Za Zhi 51 (2012) 638641.
[8] G. Drews, M. D ufer, Role of K(ATP) channels in -cell resistance to oxidative stress,
Diabetes Obes. Metab. 14 (2012) 120128.
[9] H. Alp, S. Varol, M.M. Celik, M. Altas, O. Evliyaoglu, O. Tokgoz, M.H. Tanriverdi,
E. Uzar, Protective effects of beta glucan and gliclazide on brain tissue and sciatic nerve
of diabetic rats induced by streptozosin, Exp. Diabetes Res. 2012 (2012) 30342.
[10] A.A. Spasov, V.A. Kosolapov, N.I. Chepliaeva, Comparative characteristics of antiox-
idant properties of hypoglycemic agents diabenol and gliclazide, Eksp. Klin. Farmakol.
74 (2011) 1416.
[11] A. Sliwinska, A. Rogalska, M. Szwed, J. Kasnicki, Z. Jozawiak, J. Drzewoski,
Gliclazide may have an antiapoptotic effect related to its antioxidant properties in
human normal and cancer cells, Mol. Biol. Rep. 39 (2012) 52535267.
[12] A. Avogaro, Treating diabetes today with gliclazide MR: a matter of numbers,
Diabetes Obes. Metab. 14 (2012) S14S19.
[13] L.L. Chen, F. Yu, T.S. Zeng, Y.F. Liao, Y.M. Li, H.C. Ding, Effects of gliclazide on
endothelial function in patients with newly diagnosed type 2 diabetes, Eur. J.
Pharmacol. 659 (2011) 296301.
[14] A. Sliwinska, J. Blasiak, J. Drzewoski, Effect of gliclazide on DNA damage in human
peripheral blood lymphocytes and insulinoma mouse cells, Chem. Biol. Interact.
162 (2006) 259267.
[15] A. Ceriello, Effects of gliclazide beyond metabolic control, Metabolism 55 (2006)
S10S15.
[16] S. Satyanarayana, J.R. Sekhar, K.E. Kumar, L.B. Shannika, B. Rajanna, S. Rajanna,
Mol. Cell. Biochem. 283 (2006) 123127.
[17] G. Alper, S. Irer, E. Duman, O. Caglayan, C. Ylmaz, Effect of I-deprenyl and
gliclazide on oxidant stress/antioxidant status and DNA damage in a diabetic rat model,
Endocr. Res. 31 (2005) 199212.
[18] M.L. Onozato, A. Tojo, A. Goto, T. Fujita, Radical scavenging effect of gliclazide in
diabetic rats fed with a high cholesterol diet, Kidney Int. 65 (2004) 951960.
[19] K. Kimoto, K. Suzuki, T. Kizaki, Y. Hitomi, H. Ishida, H. Katsuta, E. Itoh,
T. Ookawara, K. Suzuki, K. Honke, H. Ohno, Gliclazide protects pancreatic -cells
from damage by hydrogen peroxide, Biochem. Biophys. Res. Commun. 303 (2003)
112119.
[20] D. Fava, M. Cassone-Feldetta, O. Laurenti, O. De Luca, A. Ghizelli, G. De Mattia,
Gliclazide improves anti-oxidant status and nitric oxide-mediated vasodilation in Type
2 diabetes, Diabet. Med. 19 (2002) 752757.
[21] L. Beregi, P. Hugon, J. Duhault, N-Phenylsulphonyl-N-(3-azabicyclo-alkyl)urea
derivatives, 1970, US Patent No. 3501495.
Gliclazide 185

[22] C. Qian, Y. Liu, X. Chen, Improved synthesis of 1-[hexahydrocyclopenta[c]pyrrol-2


(1H)-yl]-3-(4-methylbenzenesulfonyl)urea, J. Chem. Res. 11 (2008) 635636.
[23] D. Che, X. Du, Method for preparing gliclazide and its intermediates, PCT Int. Appl.
2011, 2011054312.
[24] M. Parvez, M.S. Arayne, M.K. Zaman, N. Sultana, Gliclazide, Acta Crystallogr.
C55 (1999) 7475.
[25] P. Singh, R. Kumar, H. Singh, Application of UV spectrophotometric method for
analysis of gliclazide in pharmaceutical dosage forms, Int. J. Pharm. Pharm. Sci.
3 (2011) 259260.
[26] S.A. Jamadar, S.P. Mulye, P.S. Karekar, Y.V. Pore, K.B. Burade, Development and
validation of UV spectrophotometric method for the determination of Gliclazide in
tablet dosage form, Der. Pharma. Chem. 3 (2011) 338343.
[27] R. Revathi, V.S. Saravanan, P.M. Raj, T. Ethiraj, V. Ganesan, Spectrophotometric
estimation of gliclazide in bulk and pharmaceutical dosage forms, Int. Res. J. Pharm.
1 (2010) 227281.
[28] A.H. Barnett, A.J. Krentz, K. Strojek, J.J. Sieradzki, F. Azizi, M. Embong, S. Imamoglu,
J. Perusicova, V. Uliciansky, G. Winkler, The efficacy of self-monitoring of blood glu-
cose in the management of patients with type 2 diabetes treated with a gliclazide mod-
ified release-based regimen. A multicentre, randomized, parallel-group, 6-month
evaluation (DINAMIC 1 study), Diabetes Obes. Metab. 10 (2008) 12391247.
[29] B.H. Charbonnel, D.R. Matthews, G. Schernthaner, M. Hanefeld, P. Brunetti,
A long-term comparison of pioglitazone and gliclazide in patients with Type 2 diabetes
mellitus: a randomized, double-blind, parallel-group comparison trial, Diabet. Med.
22 (2005) 399405.
[30] A. Baksi, R.E. James, B. Zhou, J.J. Nolan, Comparison of uptitration of gliclazide
with the addition of rosiglitazone to gliclazide in patients with type 2 diabetes inad-
equately controlled on half-maximal doses of a sulphonylurea, Acta Diabetol.
41 (2004) 6369.
[31] M. Okouchi, N. Okayama, H. Omi, K. Imaeda, T. Fukutomi, A. Nakamura, M. Itoh,
The antidiabetic agent, gliclazide, reduces high insulin-enhanced neutrophil-
transendothelial migration through direct effects on the endothelium, Diabetes Metab.
Res. Rev. 20 (2004) 232238.
[32] S. Salman, F. Salman, I. Satman, Y. Yilmaz, E. Ozer, A. Seng ul, H.O. Demirel,
K. Karsida, N. Dincca, M.T. Yilmaz, Comparison of acarbose and gliclazide as
first-line agents in patients with type 2 diabetes, Curr. Med. Res. Opin. 16 (2001)
296306.
[33] J.A. Pugh, M.L. Wagner, J. Sawyer, G. Ramirez, M. Tuley, S.J. Friedberg, Is combi-
nation sulfonylurea and insulin therapy useful in NIDDM patients? A metaanalysis,
Diabetes Care 15 (1992) 953959.
[34] R. Matsuda, J. Anguizola, K.S. Joseph, D.S. Hage, High-performance affinity chro-
matography and the analysis of drug interactions with modified proteins: binding of
gliclazide with glycated human serum albumin, Eur. J. Drug Metab. Pharmacokinet.
37 (2012) 99108.
[35] A. Sener, A.G. Akkan, W.J. Malaisse, Standardized procedure for the assay and iden-
tification of hypoglycemic sulfonylureas in human plasma, Acta Diabetol. 32 (1995)
6468.
[36] J.Y. Park, K.A. Kim, S.L. Kim, P.W. Park, Quantification of gliclazide by semimi-
cro high-performance liquid chromatography: application to a bioequivalence study
of two formulations in healthy subjects, J. Pharm. Biomed. Anal. 35 (2004)
943949.
[37] N. Rojanasthien, T. Autsavakitipong, B. Kumsorn, M. Manorot, S. Teekachunhatean,
Bioequivalence study of modified-release gliclazide tablets in healthy volunteers,
ISRN Pharmacol. 2012 (2012) 375134.
186 Fatmah A.M. Al-Omary

[38] C.Y. Wang, W. Zhang, B.R. Xiang, L.Y. Yu, P.C. Ma, Liquid chromatography-mass
spectrometry method for the determination of gliclazide in human plasma and appli-
cation to a pharmacokinetic study of gliclazide sustained release tablets,
Arzneimittelforschung 58 (2008) 653658.
[39] G.D. Mendes, L.D. Moreira, S. Pereira Ados, A. Borges, F. Yui, F.D. Mendes, G. de
Nucci, A bioequivalence study of gliclazide based on quantification by high-
performance liquid chromatography coupled with electrospray tandem mass spec-
trometry, Int. J. Clin. Pharmacol. Ther. 45 (2007) 175185.
[40] J. Yao, Y.Q. Shi, Z.R. Li, S.H. Jin, Development of RP-HPLC method for screening
potentially counterfeit antidiabetic drugs, J. Chromatogr. B Analyt. Technol. Biomed.
Life Sci. 853 (2007) 254259.
[41] S.M. Foroutan, A. Zarghi, A. Shafaati, A. Khoddam, Application of monolithic col-
umn in quantification of gliclazide in human plasma by liquid chromatography,
J. Pharm. Biomed. Anal. 42 (2006) 513516.
[42] A. Berecka, A. Gumieniczek, H. Hopkala, Development and validation of a new
high-performance liquid chromatography method for the determination of
gliclazide and repaglinide in pharmaceutical formulations, J. AOAC Int. 89 (2006)
319325.
[43] C.Y. Kuo, S.M. Wu, High-performance liquid chromatography with electrochem-
ical detection for analysis of gliclazide in plasma, J. Chromatogr. A 1088 (2005)
131135.
[44] M.R. Rouini, A. Mohajer, M.H. Tahami, A simple and sensitive HPLC method for
determination of gliclazide in human serum, J. Chromatogr. B Analyt. Technol.
Biomed. Life Sci. 785 (2003) 383386.
[45] M. Vasudevan, J. Ravi, S. Ravisankar, B. Suresh, Ion-pair liquid chromatography
technique for the estimation of metformin in its multicomponent dosage forms,
J. Pharm. Biomed. Anal. 25 (2001) 7784.
[46] H. Noguchi, N. Tomita, S. Naruto, S. Nakano, Determination of gliclazide in serum
by high-performance liquid chromatography using solid-phase extraction,
J. Chromatogr. 583 (1992) 266269.
[47] R. Awasthi, G.T. Kulkarni, Development of novel gastroretentive drug delivery sys-
tem of gliclazide: hollow beads, Drug Dev. Ind. Pharm. 40 (2013) 398408.
[48] R. Awasthi, G.T. Kulkarni, Development of novel gastroretentive floating particulate
drug delivery system of gliclazide, Curr. Drug Deliv. 9 (2012) 437451.
[49] M.P. Patil, N.J. Gaikwad, Preparation and characterization of gliclazide-polyethylene
glycol 4000 solid dispersions, Acta Pharm. 59 (2009) 5765.
[50] Y.K. Lo, C.J. Chen, T.R. Tsai, T.M. Cham, Comparison of the solubility and disso-
lution rate between gliclazide solid complex and its nanospheres, Drug Dev. Ind.
Pharm. 33 (2007) 301309.
[51] J. Shaodong, W.J. Lee, J.W. Ee, J.H. Park, S.W. Kwon, J. Lee, Comparison of ultra-
violet detection, evaporative light scattering detection and charged aerosol detection
methods for liquid-chromatographic determination of anti-diabetic drugs, J. Pharm.
Biomed. Anal. 51 (2010) 973978.
[52] N. El-Enany, Spectrofluorimetric and spectrophotometric determination of gliclazide
in pharmaceuticals by derivatization with 4-chloro-7-nitrobenzo-2-oxa-1,3-diazole,
J. AOAC Int. 86 (2003) 209214.
[53] K.S. Lakshmi, T. Rajesh, Separation and quantification of eight antidiabetic drugs on a
high-performance liquid chromatography: its application to human plasma assay,
ISRN Pharm. 2011 (2011) 521353.
[54] L.L. Chen, Y.F. Liao, T.S. Zeng, F. Yu, H.Q. Li, Y. Feng, Effects of metformin plus
gliclazide compared with metformin alone on circulating endothelial progenitor cell in
type 2 diabetic patients, Endocrine 38 (2010) 266275.
Gliclazide 187

[55] J. Wang, L.P. Wang, D.J. Yang, B. Chen, A new method for safety monitoring of
natural dietary supplementsquality profile, Zhonghua Yu Fang Yi Xue Za Zhi
42 (2008) 489493.
[56] N. Seedher, M. Kanojia, Reversible binding of antidiabetic drugs, repaglinide and
gliclazide, with human serum albumin, Chem. Biol. Drug Des. 72 (2008) 290296.
[57] G.M. Shenfield, J.S. Boutagy, C. Webb, A screening test for detecting sulfonylureas in
plasma, Ther. Drug Monit. 12 (1990) 393397.
[58] S. Jondhale, S. Bhise, Y. Pore, Physicochemical investigations and stability studies of
amorphous gliclazide, AAPS PharmSciTech 13 (2012) 448459.
[59] R. Chadha, S. Bhandari, P. Arora, R. Chhikara, Characterization, quantification and
stability of differently prepared amorphous forms of some oral hypoglycaemic agents,
Pharm. Dev. Technol. 18 (2013) 504514.
[60] S. Aburuz, J. Millership, J. McElnay, The development and validation of liquid chro-
matography method for the simultaneous determination of metformin and glipizide,
gliclazide, glibenclamide or glimperide in plasma, J. Chromatogr. B Analyt. Technol.
Biomed. Life Sci. 817 (2005) 277286.
[61] S. Aggarwal, P.N. Singh, B. Mishra, Studies on solubility and hypoglycemic activity of
gliclazide beta-cyclodextrin-hydroxypropylmethylcellulose complexes, Pharmazie
57 (2002) 191193.
[62] R.S. Al-Kassas, O.M. Al-Gohry, M.M. Al-Faadhel, Controlling of systemic absorption
of gliclazide through incorporation into alginate beads, Int. J. Pharm. 341 (2007)
230237.
[63] H. Al-Salami, G. Butt, I. Tucker, S. Golocorbin, M. Mikov, Probiotics decreased the
bioavailability of the bile acid analog, monoketocholic acid, when coadministered with
gliclazide, in healthy but not diabetic rats, Eur. J. Drug Metab. Pharmacokinet.
37 (2012) 99108.
[64] H. Al-Salami, G. Butt, I. Tucker, M. Mikov, Influence of the semisynthetic bile acid
MKC on the ileal permeation of gliclazide in-vitro in healthy and diabetic rats treated
with probiotics, Methods Find. Exp. Clin. Pharmacol. 30 (2008) 107113.
[65] H. Al-Salami, G. Butt, I. Tucker, M. Mikov, Influence of the semisynthetic bile acid
(MKC) on the ileal permeation of gliclazide in healthy and diabetic rats, Pharmacol.
Rep. 60 (2008) 532541.
[66] H. Al-Slami, G. Butt, J.P. Fawcett, I.G. Ticker, S. Golocorbin-Kon, M. Mikov,
Probiotic treatment reduces blood glucose levels and increases systemic absorption
of gliclazide in diabetic rats, Eur. J. Drug Metab. Pharmacokinet. 33 (2008)
101106.
[67] E.A. Arno, P. Anand, K. Bhaskar, S. Ramachandran, M. Saravanan, R. Vinod,
Eudragit NE30D based metformin/gliclazide extended release tablets: formulation,
characterisation and in-vitro release studies, Chem. Pharm. Bull. (Tokyo) 50 (2002)
14951498.
[68] S. Asyarie, H. Rachmawati, In vivo and in vitro evaluation of a solid dispersion system of
gliclazide: PEG 6000, PDA J. PharmSciTech 61 (2007) 400410.
[69] K. Brendel, E. Comets, C. Lffont, C. Laveille, F. Mentre, Metrics for external model
evaluation with an application to the population pharmacokinetics of gliclazide,
Pharm. Res. 23 (2006) 20362049.
[70] H. Xu, M. Murray, A.J. McLachlan, Influence of genetic polymorphisms on the phar-
macokinetics and pharmacodynamics of sulfonylurea drugs, Curr. Drug Metab.
10 (2009) 643658.
[71] H.Y. Cho, H. Yoon, Y.C. Lim, Y.B. Lee, Pharmacokinetics and bioequivalence eval-
uation of gliclazide/metformin combination tablet and equivalent doses of gliclazide
and metformin in healthy Korean subjects, Int. J. Clin. Pharmacol. Ther. 47 (2009)
770779.
188 Fatmah A.M. Al-Omary

[72] P. Courtois, A. Sener, C. Herbaut, A. Turc, W.J. Malaisse, Pharmacokinetics of


gliquidone, glibenclamide, gliclazide and glipizide in middle-aged and aged subjects,
Res. Commun. Mol. Pathol. Pharmacol. 103 (1999) 211222.
[73] T.M. Davis, F. Daly, J.P. Walsh, K.F. Ilett, J.P. Beilby, I.J. Dusci, P.H. Barrett, Phar-
macokinetics and pharmacodynamics of gliclazide in Caucasians and Australian aborig-
ines with type 2 diabetes, Br. J. Clin. Pharmacol. 49 (2000) 223230.
[74] P. Delrat, M. Paraire, R. Jochemsen, Complete bioavailability and lack of food-effect
on pharmacokinetics of gliclazide 30 mg modified release in healthy volunteers,
Biopharm. Drug Dispos. 23 (2003) 151157.
[75] G.M. El-Maghraby, A.H. Alomrani, Effect of binary and ternary solid dispersions on
the in vitro dissolution and in-situ rabbit intestinal absorption of gliclazide, Pak. J.
Pharm. Sci. 24 (2011) 459468.
[76] N. Frey, C. Laveille, M. Paraire, M. Francillard, N.H. Holford, R. Jochemsen, Pop-
ulation PKPD modelling of the long-term hypoglycaemic effect of gliclazide given as a
once-a-day modified release (MR) formulation, Br. J. Clin. Pharmacol. 55 (2003)
147157.
[77] T.W. Hermann, R. Dobrucki, S. Piechocki, M. Resztak, R. Reh, Pharmaceutical
availability of gliclazide from selected matrix formulation tablets, Med. Sci. Monit.
11 (2005) BR181BR188.
[78] H. Kim, M. Yun, K.I. Kwon, Pharmacokinetic and pharmacodynamic characteriza-
tion of gliclazide in healthy volunteers, Arch. Pharm. Res. 26 (2003) 564568.
[79] K. Kobayashi, M. Kimura, T. Sakoguchi, A. Hase, A. Matsuoka, S. Kaneko, Pharma-
cokinetics of gliclazide in healthy and diabetic subjects, J. Pharm. Sci. 73 (1984)
16841687.
[80] K. Kobayashi, M. Mimura, T. Sakoguchi, Y. Kitani, M. Hata, A. Matsuoka, Influence
of blood proteins on biomedical analysis. III. Pharmacokinetics and protein binding of
gliclazide, J. Pharmacobiodyn. 4 (1981) 436442.
[81] U. Mandal, K.K. Ray, V. Gowda, A. Ghosh, T.K. Pal, In-vitro and in-vivo correlation
for two gliclazide extended-release tablets, J. Pharm. Pharmacol. 59 (2007) 971976.
[82] P.A. de Smet, H.R. Fischer, Timing of administration of sulfonylurea derivatives,
Ned. Tijdschr. Geneeskd. 144 (2000) 12061209.
[83] C.Z. Wu, D. Pei, A.T. Hsieh, K. Wang, J.D. Lin, L.H. Lee, Y.M. Chu, F.C. Hsiao,
C. Pei, T.L. Hsia, Comparison of insulin sensitivity, glucose sensitivity, and first phase
insulin secretion in patients treated with repaglinide or gliclazide, Arch. Pharm. Res.
33 (2010) 411416.
[84] A.R. Taylor, R.D. Brownsill, H. Grandon, F. Lefoulon, A. Petit, W. Luijten,
P.G. Kopelman, B. Walther, Synthesis of putative metabolites and investigation of
the metabolic fate of gliclazide, [1-(3-azabicyclo(3,3,0)oct-3-yl)-3-(4-methylphen-
ylsulfonyl)urea], in diabetic patients, Drug Metab. Dispos. 24 (1996) 5564.
[85] W. Yang, G. Yang, X. Jia, L. Wu, R. Wang, Activation of KATP channels by H2S in
rat insulin-secreting cells and the underlying mechanisms, J. Physiol. 569 (2005)
519531.
[86] K.G. Alberti, Gliclazide: review of metabolic and vascular action, Diabete Metab.
20 (1994) 341348.
[87] V.N. Babichev, M.P. Saveleva, M.I. Balabolin, Characterization of pancreatic beta
cell receptors binding sulfanilamide drugs, Probl. Endokrinol. (Mosk.) 40 (1994)
4750.
[88] S.B. Shustov, B.V. Romashevski, Iu.Sh. Khalimov, Olu Stepanova, A.G. Lysenko,
Reclid effects on carbohydrate, lipid metabolism, lipid peroxidation and hemodynamics
in patients with diabetes mellitus type 2, Klin. Med. (Mosk.) 78 (2000) 4548.
[89] K. Misawa, K. Ichikawa, K. Ojima, S. Hamano, T. Kitamura, H. Momatsu, Effect of
KAD-1229, a nonsulfonylurea hypoglycemic agent, on plasma glucose and insulin in
streptozotocin-induced diabetic dogs, Pharmacology 62 (2001) 6572.
Gliclazide 189

[90] M.B. Graal, B.H.G. Wolffenbuttel, The use of sulphonylureas in the elderly, Drugs
Aging 15 (1999) 471481.
[91] T. Shiba, H. Kaninuma, K. Suzuki, R. Hagura, A. Kawai, H. Katagiri, H. Sando,
W. Shirakawa, K. Kosaka, N. Kuzuya, Serum gliclazide concentration in diabetic
patients. Relationship between gliclazide dose and serum concentration, Diabetes
Res. Clin. Pract. 2 (1986) 301306.
[92] G. Schernthaner, Gliclazide modified release: a critical review of pharmacodynamic,
metabolic, and vasoprotective effects, Metabolism 52 (2003) 2934.
[93] K. Adibkia, H. Babaei, S. Asnaashari, S. Andalib, A. Khorrami, H. Ghavimi,
V. Jadidinia, H. Hajiloo, M. Barzegar-Jalali, Serum concentrations and hypoglycemic
effect of gliclazide: crosspovidone solid dispersion on streptozotocin induced diabetic
rats, Drug Res. (Stuttg.) 63 (2013) 9497.
[94] V.A. Saharan, P.K. Choudhury, Dissolution rate enhancement of gliclazide by ordered
mixing, Acta Pharm. 61 (2011) 323334.
[95] D. Pal, A.K. Nayak, Development, optimization, and antidiabetic activity of
gliclazide-loaded alginate-methyl cellulose mucoadhesive microcapsules, AAPS
PharmSciTech 12 (2011) 14311441.
[96] S. Grbic, J. Parojcie, S. Ibric, Z. Djuric, In-vitro, in-vivo correlation for gliclazide
immediate-release tablets based on mechanistic absorption simulation, AAPS
PharmSciTech 12 (2011) 165171.
[97] F. Ishibashi, S. Takashina, The effect of timing on gliclazide absorption and action,
Hiroshima J. Med. Sci. 39 (1990) 79.
[98] V. Ambrogi, L. Perioli, V. Ciarnelli, M.M. Nocchetti, C. Rossi, Effect of gliclazide
immobilization into layered double hydroxide on drug release, Eur. J. Pharm.
Biopharm. 7 (2009) 285291.
[99] R. Talari, J. Varshosaz, S.A. Mostafavi, A. Nokhodchi, Gliclazide microcrystals pre-
pared by two methods of in situ micronization: pharmacokinetic studies in diabetic and
normal rats, AAPS PharmSciTech 11 (2010) 786792.
[100] T.W. Hermann, R. Dobrucki, S. Piechocki, M. Resztak, R. Reh, Pharmaceutical
availability of gliclazide from selected matrix formulation tablets, Med. Sci. Monit.
11 (2005) 181188.
[101] A.J. Scheen, Medication of the month. Gliclazide modified release (Uni Diamicron),
Rev. Med. Liege 58 (2003) 641645.
[102] K.A. Alkhamis, H. Allaboun, W.Y. Al-Momani, Study of the solubilization of
gliclazide by aqueous micellar solutions, J. Pharm. Sci. 92 (2003) 839846.
[103] Y.L. An, Y. Gao, O. Zhu, G. Ning, W.P. Jia, O. Huang, W. Xu, C.J. Li, Z.G. Zhou,
B.Y. Shi, Z.Y. Shan, L.L. Chen, Z.R. Xu, H. Wang, L. Yan, H.J. Zhong, O.H. Ji,
H. Li, Y.M. Xue, M.X. Zhing, W. Gu, X.H. Guo, L.L. Gao, J. Hong, F. Liu,
D.J. Zou, Y.B. Li, X.Y. Zhang, Z. Li, G. Guo, X.J. Zhao, H.N. Wu, Y.Z. Wang,
W.G. Zhao, S.L. Zhang, J. Wu, N.Y. Zhang, F.Y. Yang, Y. Zhang, X.R. Zhou,
Y.Z. Ren, G.W. Li, Features of insulin secretion and insulin resistance in newly diag-
nosed type 2 diabetes of different body mass indices, Zhonghua Yi Xue Za Zhi
89 (2009) 11171121.
[104] C. Ionescu-Trgoviste, L. Gavril, O.M. Bradescu, C. Guja, NIDDM: once-daily
intervention for type 2 diabetes mellitus: diaprel MR, Rom. J. Intern. Med.
42 (2004) 431440.
[105] O. Mokuda, H. Tanaka, T. Hayashi, H. Ooka, R. Okazaki, Gliclazide at a lower con-
centration than therapeutic dose increases the sensitivity of insulin secretion to glucose
in perfused rat pancreas, Diabetes Metab. 30 (2004) 193196.
[106] D. Bataille, Molecular mechanisms of insulin secretion, Diabetes Metab. 28 (2002).
4S74S13.
[107] P. Proks, F. Reimann, N. Green, F. Gribble, F. Ashcroft, Sulfonylurea stimulation of
insulin secretion, Diabetes 51 (2002) S368S376.
190 Fatmah A.M. Al-Omary

[108] C.B. Juhl, N. Prksen, S.M. Pincus, A.P. Hansen, J.D. Veldhuis, O. Schmitz, Acute
and short-term administration of a sulfonylurea (gliclazide) increases pulsatile insulin
secretion in type 2 diabetes, Diabetes 50 (2001) 17781784.
[109] J.K. McGavin, C.M. Perry, K.L. Goa, Gliclazide modified release, Drugs 62 (2002)
13571364.
[110] J.J. Ligtenberg, W.D. Reitsma, T.W. van Haeften, Gliclazide mainly affects insulin
secretion in second phase of type 2 diabetes mellitus, Horm. Metab. Res. 33 (2001)
361364.
[111] K.J. Palmer, R.N. Brogden, Gliclazide. An update of its pharmacological properties
and therapeutic efficacy in non-insulin-dependent diabetes mellitus, Drugs
46 (1993) 92125.
[112] H. Zhang, P. Bu, Y.H. Xie, J. Luo, M.X. Lei, Z.H. Mo, E.Y. Liao, Effect of
repaglinide and gliclazide on glycaemic control, early-phase insulin secretion and lipid
profiles in, Chin. Med. J. 124 (2011) 172176.
[113] Y.Y. Hu, S.D. Ye, L.L. Zhao, M. Zheng, F.Z. Wu, Y. Chen, Hydrochloride
pioglitazone decreases urinary cytokines excretion in type 2 diabetes, Clin.
Endocrinol. 73 (2010) 739743.
[114] K. Kamikubo, R. Takami, T. Suwa, A. Sugiyama, N. Horibe, S. Sakata,
Possible gliclazide-induced water retention with azotemia, Intern. Med. 33 (1994)
163164.
[115] Y. Imamura, K. Kimachi, S. Nagase, M. Otagiri, Hypoglycemic action and disposition
of gliclazide in normal and analbuminemic rats, Arch. Int. Pharmacodyn. Ther.
304 (1990) 4454.
[116] D.J. Elliot, Suharjono, B.C. Lewis, E.M. Gillam, D.J. Birkett, A.S. Gross, J.O. Miners,
Identification of the human cytochromes P450 catalysing the rate-limiting pathways of
gliclazide elimination, Br. J. Clin. Pharmacol. 654 (2007) 450457.
[117] Y. Bao, X. Sun, Y. Yerong, L. Shuyuan, W. Yang, Blockers of sulfonylureas receptor 1
subunits may lead to cardiac protection against isoprenaline-induced injury in obese
rats, Eur. J. Pharmacol. 690 (2012) 142148.
[118] R. Engbersen, R. Masereeuw, M.A. van Gestel, H.L. Siero, M.M. Moons, P. Smits,
F.G. Russel, Differential effects of sulfonylurea derivatives on vascular ATP-sensitive
potassium channels, Eur. J. Pharmacol. 681 (2010) 7579.
[119] M.H. Ahmed, T.A. Abdu, Diabetes and Ramadan: an update on use of glycemic ther-
apies during fasting, Ann. Saudi Med. 31 (2011) 402406.
[120] H. Konya, Y. Hasegawa, T. Hamaguchi, K. Satani, A. Umrehara, T. Katsuno,
T. Ishikawa, M. Miuchi, K. Kohri, A. Suehiro, E. Kakishita, J. Miyagawa, M. Namba,
Effects of gliclazide on platelet aggregation and the plasminogen activator inhibitor
type 1 level in patients with type 2 diabetes mellitus, Metabolism 59 (2010) 12941299.
[121] A. Sliwinska, T. Sliwinski, J. Kasznicki, J. Drzewoski, Effect of gliclazide on nucleotide
excision repair (NER) and non-homologous DNA end joining (NHEJ) in normal and
cancer cells, J. Physiol. Pharmacol. 61 (2010) 347353.
[122] A.A. Spasov, A.F. Kucheriavenko, O.A. Salaznikova, Effect of hypoglycemic drugs on
hemostasis, Eksp. Klin. Farmakol. 73 (2010) 2628.
[123] G. Koh, M.K. Kim, E.J. Yang, D.H. Lee, Gliclazide does not fully prevent 2-deoxy-D-
ribose-induced oxidative damage because it does not restore glutathione content in a
pancreatic -cell line, Oxid. Med. Cell. Longev. 2012 (2012) 390678.
[124] S.R. Aravind, K. Al Tayeb, S.B. Ismail, N. Shehadeh, G. Kaddaha, R. Liu, R. Balshaw,
N. Lesnikova, O. Heisel, C.J. Girman, B.J. Musser, M.J. Davies, H.L. Katzeff,
S.S. Engel, L. Radican, Hypoglycaemia in sulphonylurea-treated subjects with type
2 diabetes undergoing Ramadan fasting: a five-country observational study, Curr.
Med. Res. Opin. 27 (2011) 12371242.
Gliclazide 191

[125] M. Mohiuddin, A.T. Azam, M.S. Amran, M.A. Hossain, In-vivo effects of gliclazide
and metformin on the plasma concentration of caffeine in healthy rats, Pak. J. Biol.
Sci. 12 (2009) 734737.
[126] S. Del Guerra, V. DAleo, R. Lupi, M. Masini, M. Bugliani, U. Boggi, F. Filipponi,
P. Marchetti, Effects of exposure of human islet -cells to normal and high glucose
levels with or without gliclazide or glibenclamide, Diabetes Metab. 35 (2009)
293298.
[127] V. da Tos, A. Maran, S. Vigili de Kreutzenberg, S. Marchetto, F. Tadiotto, M. Bettio,
M.C. Marescotti, A. Tiengo, S. Del Prato, Mechanisms of acute and chronic hypogly-
cemic action of gliclazide, Acta Diabetol. 37 (2000) 201206.
[128] M. Rendell, The role of sulphonylureas in the management of type 2 diabetes mellitus,
Drugs 64 (2004) 13391358.
[129] F.M. Gribble, F. Reimann, Differential selectivity of insulin secretagogues: mecha-
nisms, clinical implications, and drug interactions, J. Diabetes Complicat. 17 (2003)
1115.
[130] H. Banu, N. Renuka, G. Vasanthakumar, Reduced catalytic activity of human
CYP2C9 natural alleles for gliclazide: molecular dynamic simulation and docking stud-
ies, Biochimie 93 (2011) 10281036.
[131] A. Zargar, A. Basit, H. Mahtab, Sulphonylureas in the management of type 2 diabetes
during the fasting month of Ramadan, J. Indian Med. Assoc. 103 (2005) 444446.
[132] D.N. Juurlink, T. Gomes, B.R. Shah, M.M. Mamdani, Adverse cardiovascular events
during treatment with glyburide (glibenclamide) or gliclazide in a high-risk popula-
tion, Diabet. Med. 29 (2012) 15241528.
[133] G.D. Batty, Q. Li, S. Czernichow, B. Neal, S. Zoungas, R. Huxley, A. Patel, B.E. de
Galan, M. Woodward, P. Hamet, S.B. Harrap, N. Poulter, J. Chalmers, Erectile dys-
function and later cardiovascular disease in men with type 2 diabetes: prospective
cohort study based on the ADVANCE (Action in Diabetes and Vascular Disease: Pre-
terax and Diamicron Modified-Release Controlled Evaluation) trial, J. Am. Coll. Car-
diol. 23 (2010) 19081913.
[134] S. Zoungas, A. Patel, J. Chalmers, B.E. de Galan, Q. Li, L. Billot, M. Woodward,
T. Ninomiya, B. Neal, S. MacMahon, D.E. Grobbee, A.P. Kengne, M. Mare,
S. Heller, Severe hypoglycemia and risks of vascular events and death, N. Engl. J.
Med. 363 (2010) 14101418.
[135] N.H. Pan, T.M. Lee, M.S. Lin, C.L. Huang, N.C. Chang, Association of gliclazide
and left ventricular mass in type 2 diabetic patients, Diabetes Res. Clin. Pract.
74 (2006) 121128.
[136] G. Renier, J.C. Mamputu, O. Serri, Benefits of gliclazide in the atherosclerotic pro-
cess: decrease in monocyte adhesion to endothelial cells, Metabolism 52 (2003) 1318.
[137] J.C. Mamputu, G. Renier, Gliclazide decreases vascular smooth muscle cell dysfunc-
tion induced by cell-mediated oxidized low-density lipoprotein, Metabolism
50 (2001) 688695.
[138] P.E. Jennings, Vascular benefits of gliclazide beyond glycemic control, Metabolism
49 (2000) 1720.
[139] S.I. Harashima, M. Ogura, D. Tanaka, T. Fukushima, Y. Wang, T. Koizumi, M. Aono,
Y. Murata, M. Seike, N. Inagaki, Sitagliptin add-on to low dosage sulphonylureas: effi-
cacy and safety of combination therapy on glycaemic control and insulin secretion
capacity in type 2 diabetes, Int. J. Clin. Pract. 66 (2012) 465476.
[140] Y. Fujimori, K. Katsuno, K. Ojima, I. Nakashima, S. Nakano, Y. Ishikawa-Takemura,
H. Kusama, M. Isaji, Sergliflozin etabonate, a selective SGLT2 inhibitor, improves gly-
cemic control in streptozotocin-induced diabetic rats and Zucker fatty rats, Eur. J.
Pharmacol. 609 (2009) 148154.
192 Fatmah A.M. Al-Omary

[141] O. Ziegler, P. Drouin, Hemobiological properties of gliclazide, J. Diabetes Complicat.


8 (1994) 235239.
[142] A. Mizuno, M. Yano, J. Isobe, K. Shima, Inhibitory action of gliclazide on platelet
functions, Diabetes Res. Clin. Pract. 7 (1989) 141147.
[143] S. Baba, S. Nakagawa, K. Takebe, Y. Goto, H. Maezawa, R. Takeda, N. Sakamoto,
I. Fukui, Comparison of gliclazide and glibenclamide treatment in non-insulin-
dependent diabetes, Tohoku J. Exp. Med. 141 (1983) 693706.
[144] K. Kosaka, Clinical comparative study on the therapeutic effects of oral hypoglycaemic
agents in patients with diabetic retinopathy, J. Jpn. Diab. Soc. 26 (5) (1983) 531540.
[145] G. Jerums, Lack of effect of gliclazide on early diabetic nephropathy and retinopathy: a
two-year controlled study, Diabetes Res. Clin. Pract. 3 (1987) 7180.
[146] M. Shimizu, J. Tsuboi, B. Fujitani, Pharmacological studies on gliclazide: effects of
gliclazide on platelet aggregation, adhesion and blood coagulation, Pharmacometrics
12 (2) (1976) 295302.

Você também pode gostar