Vous êtes sur la page 1sur 25

Review

The import ance of synt het ic drugs


f or t ype 2 diabet es drug discovery

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Maliheh Safavi, AlirezaForoumadi & Mohammad Abdollahi

1.

Int roduct ion

2.

Pat hophysiology of T2DM

3.

Overview of current synt het ic


drugs in the treat ment of
T2DM

4.

New synt het ic compounds as


DPP-4 inhibitors

5.

New synt het ic compounds as


SGLT2 inhibit ors

7.

Conclusion

8.

Expert opinion

T ehran University of Medical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences


Research Center, T ehran, I ran

Int roduct ion: Type 2 diabet es mellit us (T2DM ) is a major met abolic,
mult i-causal and het erogeneous disorder w hich causes signif icant morbidit y and mortality w it h considerable burden t o healt hcare resources. The
number of deat hs due t o T2DM highlight s t he insuf ficiency of t he current ly available drugs for cont rolling t he disease and it s complications
and more needs t o be done.
Areas covered: This paper review s t he updated pat hobiology of T2DM t hat
should be t arget ed in drug discovery. Furt her, the art icle provides discussion
on t he mechanism of act ion, side ef fect s and structure of the currently available synt het ic drugs. The authors specifically evaluat e t wo new er classes of
anti-diabet ic agent s: dipept idyl pept idase IV (DPP-4) and sodium-glucose
t ransport er-2 (SGLT2). They also present inf ormat ion on new er synt het ic compounds. The art icle also highlights the key int eract ions bet w een synt hetic
compounds and DPP-4 act ive site residues f or rat ional drug design.
Expert opinion: Numerous ant i-hyperglycaemic drugs are current ly available
but many are limit ed by their adverse ef f ect s. The ident ificat ion of t he 3D
structure of DPP-4 has opened new avenues for design, t hus aiming t o produce drugs t hat direct ly exploit the st ruct ural characterist ics of t his binding
sit e. Furt her, st ruct ural- and ligand-based screening techniques have been
developed for designing novel DPP-4 and SGLT2 inhibitors. There has also
been progress w it h t he design and development of novel T2DM t herapeut ics
including: PPARa/ dual agonists, Sirtuin 1 activat ors, glycogen phosphorylase
inhibit ors and prot ein t yrosine phosphat ase 1B inhibitors. Finding new t argets and synt hesis met hods is st ill essential but it is becoming accept ed that
no diabet ic t herapy is best suit ed w ith each pat ient responding dif f erently.
Keywords: anti-diabeticagents, dipeptidyl peptidaseI V inhibitors, sodium-glucosetransporter2 inhibitors, syntheticdrugs, type2 diabetes
Expert Opin. DrugDiscov. (2013) 8(11):1339-1363

1.

Int roduction

Diabetes comprises a group of metabolic disorders characterised by chronic


hyperglycaemiawith disorders in themetabolism of carbohydrate, fat and protein
that result in defects in secretion and action of insulin [1]. Dysfunction and failure
of various organs, especially theeyes, kidneys, nerves, heart andthebloodvessels are
theusual complications of diabetes [2,3]. Diabetes is mainly divided into four main
types including insulin-dependent diabetes mellitus (type 1), non-insulin-dependent diabetes mellitus (type2), gestational diabetes and other specifictypes [1,4].
T ype2 diabetes mellitus (T 2DM) is very common andaccounts for ~ 90 --95%
of all diabeticcases. T heworldwideincreasein T 2DM becomes amost important
health concern [5]. T 2DM is usually accompaniedwith insulin resistancein theskeletal muscles and the liver, and the reduction in insulin production by the pancreas [6]. Genetic susceptibility and various environmental factors [7,8] are also
involved in T 2DM. Patients with T 2DM are at risk of vascular complications
10.1517/17460441.2013.837883 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X
All rights reserved: reproduction in whole or in part not permitted

1339

M . Saf avi et al.

Art icle highlight s.


.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

The incidence of T2DM continues to grow rapidly


worldwide and is associated with multiple comorbidities.
Peripheral insulin resistance and b-cell failure represent
the core pathophysiological defects in T2DM.
Currently available synthetic drug therapies include
metformin, sulphonylurea and other insulin
secretagogues, thiazolidinediones, a-glucosidase
inhibitors, GLP1 agonists, dopamine-2 agonists, bile acid
sequestrants, DPP-4 and SGLT2 inhibitors.
Two latter classes (DPP-4 and SGLT2 inhibitors) are novel
and promising drugs that target novel mechanisms.
Finding new drug targets and new faster and cheaper
synthesis methods are the main goals of T2DM
drug discovery.

This box summarises key points contained in the article.

such as coronary artery disease, stroke, hypertension,


nephropathy, peripheral vascular disease, neuropathy and
retinopathy [9-13]. I t has been suggested that higher glucose
levels may be a risk factor for dementia [14]. Although
T 2DM was traditionally seen in individuals over the age of
40, recent data from several countries confirm that T 2DM
occurs in younger people even in childhood [15,16]. Cases of
T 2DM that occurs before30 years of ageusually developdiabeticnephropathy, renal failure, blindness andatherosclerotic
vascular diseasein their 30s [17].
Although thereareseveral therapeuticoptions availablefor
the management of diabetes, most of them respond only in
theshort-to-medium term. Further, most of current therapies
areassociated with an increased risk of adverseeffects such as
weight gain (sulphonylureas, thiazolidinediones and insulin),
hypoglycaemia (sulphonylureas and insulin), gastrointestinal
intolerance (metformin) and myocardial infarction (rosiglitazone) [18,19]. Researchers are trying to find therapies better
than thethreeoldest classes, for example, insulin, sulphonylureas and biguanides but thenewer ones havenot shown more
potency andwereoften less effectivein loweringglycaemia[19].
Alternativetothesesyntheticagents, newinsulin analogues,
inhaled insulin and many medicinal plants arebeing investigated for possiblebenefits in diabetes [20-30]. Moreover, insulin--mimeticcomplexes havebeen synthesized in thethought
that these complexes affect both diabetes and its complications [31,32]. Of course, concerns of safety and tolerability of
current treatments and theprogressivenatureof T 2DM call
for newclasses of medicines. T his reviewprovides adiscussion
of current syntheticpharmacological agents andnewsynthetic
compounds for T 2DM.
2.

Pat hophysiology of T2DM

Unlikesimplecharacterisation of type1 diabetes, thepathogenesis of T 2DM is morecomplex and still remains amatter
of argument. T hemost problemis that thespecificaetiologies
1340

have not yet been clearly elucidated. For instance, unlike


type 1 diabetes, autoimmune destruction of cells does not
occur and ketoacidosis seldom occurs [33]. As mentioned earlier, peripheral insulin resistance and b-cell dysfunction are
rather involved in T 2DM [34]. Insulin resistance and the
related consequences called insulin resistance syndrome
(IRS) or the metabolic syndrome are common in T 2DM,
deemed mainly linking to patients genetic background [35].
Insulin resistance is also linked to obesity [36], dyslipidemia,
hypertension and increased cardiovascular risk [37].
H owever, risk alleles in some loci seem having aprimary
impact on insulin sensitivity. Peroxisome proliferatoractivated receptor-g (PPAR-g), glucokinase regulator and
insulin-likegrowth factor 1 havebeen provedas insulin resistance locus from T 2DM susceptibility loci [38]. Obesity and
lifestyleareindependent risk factor for T 2DM [1,39]. According to short-term studies, numerous benefits of weight loss
can be achieved in overweight or obese patients with
T 2DM. T hefindingof recent study indicated that theintensivelifestyleintervention do not reducetherateof cardiovascular morbidity andmortality in overweight or obesepatients
with T 2DM [40]. Gastricbypass induces substantial and sustained weight loss and is a highly effective treatment for
obesity-related diabetes. Laboratory datasuggest that gastric
bypass exhibits reprogramming of intestinal glucosemetabolism which renders theintestineamajor tissuefor glucosedisposal, contributingto theimprovement in glycaemiccontrol
after surgery [41].
Several studies haveindicated that obesity correlates with a
low-grade inflammation of the white adipose tissue. T his
results from activation of some pro-inflammatory signalling
pathways which end upwith insulin resistance, impaired glucose tolerance and diabetes [39,42]. Recent data indicate that
whiteadiposetissuein obesepatients is infiltrated by macrophages [43]. Several pro-inflammatory factors, such as
T NF-a and I L-6, are derived not only from adipocytes but
also from infiltrated macrophages [39]. T he role of cytokines
and insulin signalling pathways should not bemissed as this
interaction results in change of insulin action [44,45]. Normally, insulin acts through binding to and activation of its
cell-surface receptors. Such receptors are made up of two a
subunits and two b subunits, which are disulfide linked
into a2b2 heterotetrameric complex. T he actions of insulin
are initiated when the receptor is bound to the extracellular
a subunits, causing phosphorylation of intracellular tyrosine
kinase domain of the b subunits and inducing a conformational change [46-48]. Phosphorylation of the insulin
receptor can activate insulin receptor substrate complexes
(IRS-1 --IRS-4). T heactivated I RS binds to thep85 regulatory subunit of phosphatidylinositol (PI ) 3-kinasethat results
in activation of p110 catalytic subunit [47,49,50]. PI (3,4,5)
P3 which is produced by enzymaticactivity of PI 3-kinaseis
akey lipid second messenger in various metabolic effects of
insulin [51]. PI (3,4,5)P3 mediates thesignal transduction to
downstream molecules including Akt and atypical protein

Expert Opin. Drug Discov. (2013) 8(11)

Import ance of synt het ic drugs f or T2DM drug discovery

Obesity & life


style

Genes

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Insulin
resistance

Impaired
glucose
tolerance

Type 2
diabete
mellitus

Thiazolidinediones
biguanides

cell
dysfunction
(decreased insulin
secretion)

Sulphonylureas
nonsulphonylureas
GLP-1 analogs
DPP4 inhibitors

Decreased
glucose
transport (muscle
and adipose tissue)

Sulphonylureas

Increased
hepatic glucose
production

Biguanides

Figure 1. Pat hobiology and t arget s f or current drugs in T2DM .

kinaseC. T hesearethekey signallingmolecules in theactivation of glucoseuptake, inhibition of apoptosis and synthesis


of proteins [49,52-54].
I t is thought that in adiposeandretinal tissues, phosphorylation on specific serine sites (Ser307) on I RS-1 reduces
the tyrosine phosphorylation of the insulin receptor [48,55].
T hecytokineT NF-a plays amajor rolethrough phosphorylation of the IRS-1 protein on Ser307 site. T NF-a-induced
inhibition of I RS-1 signallingcan reduceAkt phosphorylation
and stop the insulin signalling pathway [39,48]. Also, it has
been shown that T NF-a causes marked downregulation of
the adipocytes and muscle cells insulin-regulable glucose
transporter [56].
Accordingto recent studies, thecirculatingI L-6 correlates
with insulin sensitivity in obese humans [57]. T his means
that both T NF- a and IL-6 and other adipocyte-specificsignalling elements such as resistin and leptin can alter insulin
sensitivity by provoking diverse key steps in the process of
insulin action [36,58]. Any defect in insulin action or theI RS
triggers b-cells in away to cause T 2DM [59]. T he interplay
between insulin resistance and b-cell dysfunction remains
highly complex [60]. Studies of insulin-resistant animals illustratean important rolefor expansion of theb-cell mass and
enhanced b-cell function as the compensatory mechanisms [61]. I n most obese and insulin-resistant individuals,
b-cell compensation reduces dueto hyperglycaemiaand lipid
toxicity [62,63]. Continuous declineof b-cell function usually

ends up in b-cell exhaustion and finally b-cell failure and


diabetes [60].
Numerous studies have demonstrated that enhanced proinflammatory cytokines, freeradicals andoxidativestress arecentral events tothedevelopment of diabeticcomplications [13,64-66].
Dominant causeof oxidativestress in diabetes is glucoseautoxidation that leads to production of free radicals [67]. Oxidative
stress causes b-cell death viainduction of mitochondrial stress
duringdevelopment of diabetes. In pancreaticb cells, important
targets for an oxidant insult are AT P-dependent potassium
(K AT P) channels and cell metabolism [68]. T heefficacies of different approaches in thereduction of diabetes-inducedoxidative
stress have been studied and usage of antioxidants as the
supplement to drug regimen of T 2DM patients has been
recommended [69-79]. Briefly, thepathobiology of T 2DM which
areconsidered as targets for classicand current syntheticdrugs
aresummarised in Figure 1.

Overview of current synthet ic drugs in t he


treatment of T2DM

3.

3.1

Biguanides

T heclass of biguanides includes themetformin andtwowithdrawn agents phenformin and buformin. T he reason for
removing phenformin and buformin from the market was
the occurrence of fatal lactic acidosis [80,81]. I ntroduced in
the market in 1950, metformin is a well-accepted first-line

Expert Opin. Drug Discov. (2013) 8(11)

1341

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

M . Saf avi et al.

choice for the treatment of T 2DM due to its good efficacy,


low priceand low rateof adverseeffects especially in long-term
use[82-85]. Metformin reduces fastingplasmaglucoseconcentrations by reducing rates of hepatic glucose production through
a reduction in gluconeogenesis and glycogenolysis [86-88].
Metformin also affects peripherally and improves skeletal myocyte glucose uptake, reduces the overall plasma free fatty acid
(FFA) concentration and induces mild weight loss through
reduction of caloricintake[89,90].
Activation of adenosine monophosphate-activated protein
kinase(AMPK) is required for metformins inhibitory effects
on glucoseproduction by hepatocytes andacetyl-CoA carboxylase activity and its inducing effects on glucose uptake by
skeletal muscles andfatty acidoxidation. I t shouldbenoticed
that AMPK is amajor cellular regulator of lipid and glucose
metabolism [91,92]. Metformin, by improvinginsulin sensitivity, alsoimproves avariety of other factors relatedtoincreased
cardiovascular risk. I t reduces the rate of myocardial infarction and all-cause mortality [93]. H owever, gastrointestinal
intolerance, such as nausea, abdominal pain and diarrhoea,
happens as asideeffect in about 30% of theusers that limits
the compliance of patients to consume top effective
doses [94,95]. Although rare, fatal lactic acidosis might be
observed in someusers [96,97] and thus it should not beused
in patients with liver disease [95], renal dysfunction [97] and
in thosewith aslight degreeof creatinineelevation [98]. T he
structure of biguanide(red highlight in metformin structure
in Table 1) is conventionally represented in a wrong tautomericform which was correctedin 2005 [99]. T hey aremoderately strongbases andform H Cl salts quitereadily. T his aspect
is being exploited in the preparation of oral formulations of
metformin with desired properties [99,100].
3.2

Sulphonylureas

Sulphonylureas as thefast insulin secretagogues aretheoldest


available class of oral glucose-lowering agents which were
introduced in themarket in the1940s and approved for use
in 1994 for T 2DM beforemetformin [85,95,101].
T hese drugs specially stimulate insulin secretion in a
glucose-independent manner by bindingto aregulatory protein called sulphonylureas receptor on thepancreaticb-cells.
After binding to the receptors, they trigger closure to K AT P
channels, membrane depolarisation and influx of calcium
through voltage-dependent channels, which subsequently
ends up in insulin secretion [102-104]. I n addition to their
pancreatic effects, sulphonylureas can enhance insulinstimulated peripheral glucose utilisation through triggering
insulin action on adiposetissueglucosetransport andlipogenesis and skeletal muscleglycogen synthase [105,106].
T his class of oral drugs include the second-generation
agents glipizide, gliclazide, glibornuride, gliquidone, glisoxepide, glyclopyramide and glibenclamide as well as the
first-generation agents acetohexamide, chlorpropamide, tolazamideandtolbutamide[107]. T hethird-generation sulphonylurea glimepiride is as effective as second-generation
1342

sulphonylureas and appears to have several clinical advantages over conventional sulphonylureas [108]. In clinical studies, glimepiride provides more stable blood glucose control,
lowers risk of hypoglycaemiaand induces less weight gain in
comparison to other sulphonylureas [108,109]. Owingto glimepiride pancreatic tissue specificity, its use may be safer in
patients with cardiovascular disease. Cardiovascular sideeffects
of thesecompounds result from their effect on K AT P channels
present in extra-pancreatictissues of thecardiacand vascular
smooth muscle [110,111].
All sulphonylureas contain a central S-phenyl sulphonylureastructure (red highlight in the glimepiride structure in
the Table 1) [107]. T hepharmacokineticand pharmacological
differences among the available sulphonylureas are a consequence of substitutions at the para-position on the benzene
ringandtheother at anitrogen residuein theureamoiety [112].
First-generation sulphonylureas (e.g., tolbutamide, acetohexamide, tolazamideand chlorpropamide) haverelatively small,
polar, hydrophilicsubstitutions. Second-generation sulphonylureas have large, non-polar, lipophilic substitutions that
penetrate cell membranes more easily, giving them greater
potency [107,112].

3.3

N=lurea insulin secret agogues

Anti-diabetic and insulinotropic properties of the nonsulphonylureamoiety of glibenclamide, subsequently named


meglitinide, have been discovered > 30 years ago. Repaglinide, nateglinide and mitiglinide are proposed as nonsulphonylurea insulinotropic agents exhibiting structural
analogy with meglitinide [113,114]. Repaglinide and nateglinide have been approved for treatment of T 2DM and
have been released into market in 1998 and 2001, respectively [115]. Repaglinide and nateglinide have a mechanism
of action that is similar to that of sulphonylureas [104].
T hey are short-acting insulin secretagogues with high
affinity and rapid association--dissociation kinetic activity
to theK AT P at theouter membraneof b-cells. T his property
of these compounds results in restoration of early phase
insulin secretion [107,113]. T he most common adverse events
of repaglinide and nateglinide are upper respiratory tract
infection, sinusitis, constipation, arthralgia, headache and
vomiting [116].
Nateglinideis D-phenylalaninederivative(phenylalanineis
highlightedas redin theTable1) which blocks K AT P channels
in thepancreaticb-cells tostimulateinsulin release[117,118]. I n
therapeuticconcentrations, nateglinidewith nosulphonylurea
or benzamido moiety induces high selectivity for thepancreaticK AT P subtypeover thecardiovascular subtype[119]. Nateglinide sensitizes pancreatic b-cells to limited amount of
glucoseandreduces theglucoseconcentration neededtostimulateinsulin secretion [118]. I n contrast to other insulin secretagogues, nateglinide has a low propensity to cause
hypoglycaemia because of its selective, early phase insulin
secretion and transient effect [120].

Expert Opin. Drug Discov. (2013) 8(11)

Expert Opin. Drug Discov. (2013) 8(11)

Disaccharidase inhibitors

Thiazolidinediones

Non-sulphonylureas

Sulphonylureas

Biguanide

Drug class

CH3

HO

Acarbose

HO

HO

HO

N
H
HO

NH2

H3C

OH

Rosiglitazone

H
N

Nateglinide

NH

NH

Glimepiride

H
N

N
HN
H
O S O

NH2

Metformin

CH3

OH O
HO

OH

NH

OH O
HO

Drug st ruct ure

Table 1. Current ant i-diabet ic synt het ic drug classes.

OH

OH OH

Inhibits intestinal aglucosidase enzymes

Lowers insulin resistance in


peripheral tissue by
activating PPAR-g

Increases insulin secretion in


the pancreas

Stimulates insulin release


from pancreas and reduces
post-absorptive rates of
endogenous glucose
production

Reduces hepatic glucose


production, increases
peripheral glucose utilisation
and insulin sensitivity

M echanism of act ion

Gastrointestinal disturbance
such as flatulence,
abdominal distension,
stomach rumble and
diarrhoea

Weight gain, fluid retention


and heart failure

Mild gastrointestinal
complaints and weight gain

Very rare adverse effects


compared to other
sulphonylureas side effects
such as hypoglycaemia and
weight gain

Gastrointestinal complaints
and lactic acidosis

Side ef f ect s

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Balfour et al. 1993,


Clissold and Edwards
1988 [141,238]

Yki-Jarvinen 2004,
Defronzo 2009 [125,128]

Chachin et al. 2003,


Campbell 2005 [119,120]

Korytkowski 2004,
Becic et al. 2003 [107,237]

Bharatam et al. 2005,


Lipska et al. 2011 [99,236]

St udy

Import ance of synt het ic drugs f or T2DM drug discovery

1343

1344

Expert Opin. Drug Discov. (2013) 8(11)

Dopamine-2 agonists

SGL2 inhibitors

DPP-4 inhibitors

GLP1 agonists

Drug class
O
NH

H
O

COOH

OH

CH3

OH

OH

Br

Bromocriptine

HN

H
N

O
N

HO

Invokana (canagliflozin)

HO

OH

Vildagliptin

HN

Liraglutide

His

Ala

Glu

N
O

CH3SO3H

Thr Ser Asp Val Ser Ser Tyr Leu Glu Gly Gln Ala Ala Lys Glu
Phe
Phe
Thr
Gly Arg Gly Arg Val Leu Trp Ala Ile
Gly

CH3

Drug st ruct ure

Table 1. Current ant i-diabet ic synt het ic drug classes (cont inued).

Resets abnormally elevated


hypothalamic drive for
increased plasma glucose,
triglyceride and FFA levels

Inhibits SGLT2 in the kidneys

Inhibits DPP-4 that improves


glycaemic control

Stimulates insulin
biosynthesis and secretion,
inhibits glucagon secretion
and slows gastric emptying

M echanism of act ion

Fatigue, nausea, vomiting,


dizziness and headache

Low incidence of
hypoglycaemia and genital
infections in females

Few gastrointestinal
disturbance

Nausea and vomiting

Side ef f ect s

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Keche 2010,
Kumar et al.
2012 [181,182]

Nomura et al. 2010,


Rosenstock et al.
2012 [179,241]

Gupta et al. 2009,


Panina 2007 [168,240]

Wajcberg and Amarah


2010, Madsbad
2009 [159,239]

St udy

M . Saf avi et al.

Import ance of synt het ic drugs f or T2DM drug discovery

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

3.4

Thiazolidinediones

T roglitazone was the first thiazolidinedione approved as a


glucose-lowering therapy for patients with T 2DM in
1997 [98,121]. T roglitazonewas subsequently withdrawn from
use, in March 2000, becauseof causing severehepatictoxicity [122]. T wo currently available PPAR-g agonists, rosiglitazoneand pioglitazone, wereapproved in 1999. T his class of
agents also known as glitazones likethebiguanidemetformin
do not increase insulin secretion but rather increase insulin
sensitivity in muscle and adipose tissue and in the liver by
activatingPPAR-g and affectinggeneregulation in thetarget
cells [123-125].
PPAR-g is essential for normal metabolism of lipids such as
adipocytedifferentiation and proliferation as well as fatty acid
uptake and storage [126,127]. T hiazolidinediones exert their
insulin-sensitisingactions by promotinggenesis of small adipocytes andredirect fat fromnon-adiposetissues, such as liver fat,
to (subcutaneous) adiposedepots [125,128]. Although thiazolidinediones may enhanceinsulin sensitivity by keepingfat where
it belongs and sparing other tissues such as the liver, skeletal
muscle and possibly b-cells from lipotoxicity, indirect effects
may also beinvolved viaalteration of genetranscription such
as adiponectin. Adiponectin, an adipocytokine, produced
exclusively by adiposetissueincreases insulin sensitivity [129-131].
T he potential of thiazolidinediones to lower glucose production by theliver or improveglucosetransport into muscle
andadiposetissuehavebeen shown [14,132]. T hemain adverse
events associated with thethiazolidinediones class areweight
gain and fluid retention that may besevereenough to exacerbateor precipitateheart failure[133,134]. T hesedrugs alsocause
slight decrease in the haemoglobin level and haematocrit,
probably without clinical consequence [125]. Pioglitazoneand
rosiglitazone are licensed for use as the second-line therapy
to metformin and glyburide, agents that have demonstrated
efficacy in decreasing the microvascular and macrovascular
complications associated with T 2DM [14].
T hiazolidine-2,4-dionestructure(red highlight in therosiglitazonestructurein theTable1) is common toall thiazolidinediones [135]. T hiazolidinedioneringhas been investigatedas
potent polar head group which is acritical binding motif in
the active site of PPAR ligand-binding domain. H owever,
these polar head groups are prone to racemisation under
physiological conditions [136].
3.5 Disaccharidase inhibit ors (a-glucosidase
inhibit ors)

T hea-glucosidaseinhibitors acarboseandmiglitol aretwoof


these agents which werereleased in the market in 1996 [95].
I nitial investigations of voglibose as another disaccharidase
inhibitor havenot been proceeded further [137,138]. Absorption
of carbohydrates requires eventual breakdown of disaccharides
intomonosaccharides by thea-glucosidaseenzymein thebrush
border of thesmall intestine. Disaccharidaseinhibitors, such as
acarbose and miglitol, inhibit digestion of carbohydrates by

affecting the breakdown of disaccharides to monosaccharides


in the intestinal epithelium. As a consequence, delayed and
decreased absorption of thesugars happen. T hea-glucosidase
inhibitors decreaseboth postprandial blood glucose and postprandial insulin levels andin that way theseimprovesensitivity
to insulin and releasethestress on b-cells [139].
T he safety of a-glucosidase inhibitors may be a definite
helpin elderly patients with T 2DM [140]. T heefficacy of acarboseandmiglitol is limitedby theadversereactions causedby
alargeamount of non-absorbeddisaccharides in theintestinal
tract with the attendant symptoms of abdominal bloating,
diarrhoea and flatulence. T hese compounds do not induce
hypoglycaemiaor weight gain [139,141].
Accordingto somestudies, acarbosedoes not directly alter
insulin resistancebut may lower postprandial plasmainsulin
levels, fasting blood glucose, glycosylated haemoglobin,
plasmatriglycerides and/or cholesterol concentrations [141,142].
Acarboseis only minimally absorbedfrom thegut andthereby
considered as anon-absorbableinhibitor that makes this drug
with no systemicadverseeffects even after long-term administration. T he symptoms, which is due to undigested carbohydrates, occur in ~ 30 --60% of patients and tend to decrease
with timeand seem to bedose-dependent [142,143]. Acarboseis
composedof an acarviosin moiety with amaltoseat thereducing terminus. Acarviosin is asugar composed of cyclohexitol
unit linked to a 4-amino-4,6-dideoxy-D-glucopyranose unit,
which is part of the acarbose and its derivatives (highlighted
as red in theTable 1) [144].
3.6

Glucagon-like pept ide 1 agonists

In April 2005, the first glucagon-like peptide 1 (GLP-1)


agonist, exenatide, was approved for the treatment of
T 2DM [145]. In 2010, other analogueliraglutidewas approved
for useas an adjunct todiet andexercisetoimproveglycaemic
control in adults with T 2DM [146]. Several additional
GLP-1 agonists, includingexenatidelong-actingrelease[15,147]
albiglutide and taspoglutide, are under development and in
various stages of clinical trials [148].
GLP-1 is agut-derivedincretin hormone, which is secreted
by the more distally located intestinal L cells. Incretin hormones are a group of gastrointestinal hormones released in
response to nutrient ingestion, which cause an increase in
theamount of insulin released even beforeelevation of blood
glucose[149]. I ncretin hormone, GLP-1 stimulates insulin biosynthesis and secretion in responseto meal ingestion, inhibits
glucagon secretion, slows gastric emptying, reduces appetite
andpromotes theregeneration andproliferation of pancreatic
b-cells [149-151].
SinceGLP-1, gastricinhibitory polypeptide(GI P) andglucagon are all pivotal in glucose homeostasis, the G-proteincoupled receptors represent important drugtargets in T 2DM.
One novel diabetes treatment strategy is activation of GLP-1
receptors andinhibition of theglucagon signal [152]. T hestructureof thehuman glucagon class B G-protein-coupledreceptor
has been reported more recently. T he distinct structural

Expert Opin. Drug Discov. (2013) 8(11)

1345

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

M . Saf avi et al.

features andlarger bindingpocket of this receptor providenew


insights into the molecular details of peptide ligand binding
andamorereliablestructural templatefor thedesign of specific
and potent small molecules for thetreatment of T 2DM [153].
T heshort half-life(t1/2 ~ 1 --1.5 min) of GLP-1, because
of their rapid inactivation by dipeptidyl peptidase I V
(DPP-4) in the circulation, is a major difficulty for its
use [149,154]. Exenatide is asynthetic form of exendin-4 that
occurs naturally in thesalivaof theGilamonster (alargevenomous lizard native to southwestern United States) and its
amino acid sequence shows 53% homology to human
GLP-1 [155]. Exendin-4 and GLP-1 exhibit similar insulinotropic potencies in both in vitro and in vivo models, while
exendin-4 has abetter efficacy andalonger duration of action
than thenativeGLP-1 in rodents andhumans [156]. Exenatide
andliraglutideexhibit increasedresistancetoDPP-4 degradation and thus providepharmacological levels of GLP-1. Exenatideas short-acting GLP-1 receptor agonists (t1/2 ~ 2.4 h)
is recommendedfor twicedaily dosing[154]. Nausea, vomiting
and hypoglycaemia were the most frequently reported side
effects and wereindifferent todosewhileheadacheand nasopharyngitis wereseen moreat lower dose[147]. Liraglutide, but
not short-acting exenatide, was associated with thyroid
C-cell hyperplasiaand tumours in rodents, probably due to
continuous high-doseGLP-1 agonist exposure[157]. Rarecases
of acute pancreatitis in association with use of incretinbased classes have been reported but the most of ensuing
reports showed inconsistent results [85,158]. Liraglutide is a
long-acting GLP-1 analogue (t1/2 ~ 24 h), with 96% structural identity to human GLP-1. T headdition of aC16 fatty
acid side chain using a -glutamic acid spacer at the -amino
group of lysine-26 (fatty acid palmitateis highlighted as red
in the Table 1), which allows non-covalent binding to albumin, increases the stability through formation of heptamer
mediated by thefatty acid sidechain [159].
3.7

DPP-4 inhibit ors

DPP-4 inhibitors are promising new class of anti-diabetics


that are extremely studied [10]. I n October 2006, the first
DPP-4 inhibitor, sitagliptin, was introduced in the market.
Indeed, several other drugs such as vildagliptin, saxagliptin,
alogliptin, linagliptin and anagliptin have been approved in
certain countries for the treatment of T 2DM. Other candidates havebeen demonstrated to bein an advanced stageof
clinical trials for T 2DM [160-163].
I nhibition of DPP-4, aserine protease, enhances endogenous GLP-1 activity by decreasingtherateof GLP-1 degradation, which represents apromisingapproach to thetreatment
of T 2DM [164]. DPP-4 inhibitors increase circulating
GLP-1 and GI P levels in humans, which leads to increased
glucose-dependent secretion of insulin and decreased blood
glucose, haemoglobin A1C and glucagon levels [165,166].
DPP-4 is a766 residue N-terminal dipeptidyl exopeptidase
that specifically cleaves an aminoacidsequencehavingproline
or alanine at the N-terminal penultimate (P1) position but
1346

may also cleave substrate with non-preferred amino acids at


this position [167].
DPP-4 inhibitors include diverse structural types. Many
DPP-4 inhibitors have five-membered heterocyclic rings
such as pyrrolidine, cyanopyrrolidine, thiazolidineandcyanothiazolidineas aprolinemimeticin theP1 part. Vildagliptin
structure as a cyanopyrrolidine derivatives are presented in
the Table 1 (cyanopyrrolidinering is highlighted in red) [18].
Despite the positive results of vildagliptin in clinical trials,
oneof theissues encounteredwith theuseof 2-cyanopyrrolidinederivatives is their stability in solution dueto theparticipation of cyano group in an intramolecular cyclisation
process leadingto inactiveproducts [168].
H owever, at this time, the tolerance and safety profile of
DPP-4 inhibitors are considered as excellent. Possible
increased risk of acute pancreatitis in the short term and
occurrence of chronic pancreatitis in the longer term are
among concerns attributed to increase in GLP-1 levels.
Non-consistent results regarding a possible effect of GLP-1
and DPP-4 inhibitors on theexocrinepancreas werereported
in various animal models [169]. In view of likely toxic side
effects associated with the inhibition of other members of
DPP family (inhibition of which was linkedtotoxicity in animal studies), it seems necessary to design selectiveinhibitors
targeting DPP-4 over DPP-8 and DPP-9 [170]. Achieving
desired selectivity toward theinhibition of DPP-4 over other
related peptidases such as DPP-8 and DPP-9 and longactingpotential for maximal efficacy arethemain challenges.
3.8

Sodium-glucose t ransport er-2 inhibitors

Canagliflozin from the new class of medications called


sodium-glucose transporter-2 (SGLT 2) inhibitors was
approved by FDA in March 2013 [171]. Although there are
several candidates, SGLT 2 inhibitors such as dapagliflozin
and BI 10773 are now in various stages of clinical development, and thephlorizine, sergliflozin and remogliflozin have
been discarded [172].
Kidney plays akey role in glucose homeostasis, primarily
by the reabsorption of filtered glucose especially by the
SGLT 2 located in the proximal convoluted tubule. SGLT 1,
the other SGLT s isoform, is the key transporter for glucose
absorption in thegastrointestinal tract andplays only aminor
role in the kidney [173,174]. T he expression of SGLT 2 and
other renal glucose transporters were elevated in diabetic
patients [175]. SGLT 2 inhibitors, with agreater selectivity for
SGLT 2 versus SGLT 1, offer a considerable advantage as
potential anti-diabetic medications, because of their ability
to inhibit renal glucose reabsorption and subsequent plasma
glucose-lowering effect without inducing excessive insulin
secretion [172,176].
Canagliflozin, an oral selectiveSGLT 2 inhibitor improves
glycaemic control in T 2DM by reducing renal threshold
for glucose reabsorption and increasing the urinary glucose
excretion ending up in weight loss [177]. Various SGLT 2
inhibitors have been proposed based on the glucoside

Expert Opin. Drug Discov. (2013) 8(11)

Import ance of synt het ic drugs f or T2DM drug discovery

structure(glucosideringis highlighted in red in canagliflozin


structurein theTable 1). Since, theO-linkageof thestructure
of SGLT 2 inhibitors is a metabolic target for b-glucosidase
enzymes that can restrict the activity of SGLT 2 inhibitors
in vivo, newer candidate with aC-glucoside linkage such as
canagliflozin have been synthesized [178]. Canagliflozin as a
C-glucoside bearing a heteroaromatic ring has improved
metabolicstability in comparison to O-glucoside [179].

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

3.9

Bile acid sequest rant s/ dopamine-2 agonist s

From this category, two classes of drugs were already


approved for other diseases. Colesevelam hydrochloride is a
bile acid sequestrant that had been originally approved for
the treatment of hypercholesterolaemia in the 2000s and
approved in January 2008 to improve glycaemic control in
T 2DM adults. T he exact mechanism influencing glucose
metabolism remains unexplained [180].
T he bromocriptine mesylate, a quick release formulation
by trade name Cycloset, as an adjunct to diet and exercise
has been approved to improve glycaemic control in adults
with T 2DM on 2009 [181]. Bromocriptinemesylateis asympatholytic D2 dopamine agonist which can reverse many of
the metabolic alterations associated with insulin resistance
and obesity viaresettingdopaminergicand sympathetictone
within thecentral nervous system [182].
Clinically, bromocriptine is used in treating Parkinsons
diseasethrough activity at dopaminereceptors andin treating
hyperprolactinaemia and acromegaly. Bromocriptine was
usedfor 30 years for other indications andbasedon its activity
in modulating central glucose and energy metabolism pathways has been approved for thetreatment of T 2DM [181,183].
Bromocriptine is a semisynthetic derivative that is derived
from a natural ergot alkaloid but bromocriptine mesylate
has been chemically designated as 2-bromoergocryptine
monomethanesulfonate (salt) [182]. More information is
presented in the Table 1.
4. New synt het ic compounds as
DPP-4 inhibitors

T heDPP-4 is responsiblefor thedegradation of theincretin


hormones GL P-1 and GI P and has therefore astrong influence on insulin secretion and glucose homeostasis [184]. In
contrast to therapy with GLP-1 analogues, becauseof limitations dueto its swift inactivation, DPP-4 inhibitors increase
effective incretin levels into amore physiological range [185].
So, in therecent years, DPP-4 inhibitors havebeen noted as
valuable agents for treatment of T 2DM. Lack of selectivity
toward several closely related proline-specificenzymes seems
apossiblecauseof toxicity [186]. In this review, thediscovery
of some synthetic compounds which may be useful in
approach tonewDPP-4 inhibitors as anewanti-diabeticclass
are presented. Structurally, two distinctive classes of DPP-4
inhibitors havebeen reported [168]. Peptidomimeticinhibitors
includesitagliptin, vildagliptin and saxagliptin (Figure 2) and

the non-peptidomimetic inhibitors include alogliptin and


linagliptin (Figure 3).
4.1

Pept idomimet ic inhibit ors

T heb-methylphenylalanine-derived amides havebeen shown


tobepotent DPP-4 inhibitors, exhibitingsuboptimal selectivity and pharmacokinetics [187,188]. A series of phenylalanine
and cyclohexylalanine derivatives also known as fluorinated
pyrrolidine amides [10] were designed and assayed for their
inhibitory potency against theDPP-4 enzymeas well as their
selectivity over the related proline-specific enzymes quiescent
cell prolinedipeptidase(QPP) (DPP-II), DPP-8 and DPP-9.
T he phenylalanine series afforded compounds such as compound (1) that werepotent and selective. Amongcyclohexylalanine derivatives, the acetamide b-methyl substitute (2) was
themost potent with better oral bioavailability [189].
T he effect of substitution on the imidazopiperidinebased b-amino acid derivatives inhibitory activity against
DPP-4, DPP-8 and DPP-9 was evaluated in another study.
Introduction of asubstituent at the4-position of theimidazopiperidineunit (3) produces compounds havingDPP-4 IC50
values < 10 nM [190].
A series of novel azobicyclo[3.3.0] octanederivatives substitutedwith pyrrolidine-2-carbonitrileweresynthesizedandevaluated against DPP-4, DPP-8 and DPP-9. Among them,
compound (4) exhibited good DPP-4 activity, high selectivity,
moderate pharmacokinetic profiles and excellent in vivo efficacy in an oral glucosetolerancetest (oGT T ) in lean mice[191].
Anagliptin is apotent and highly selectiveDPP-4 inhibitor
which has been approved in Japan for the treatment of
T 2DM in 2012 [162,192]. A series of pyrazolo[1,5-a]pyrimidines
were found to be novel DPP-4 inhibitors. Compounds were
evaluated in vitrofor inhibition of human recombinant DPP-4
and also screened for selectivity over dipeptidyl peptidase
8 and9. Structure--activity relationships (SARs) for compounds
showed N-[2-(amino)-2-methylpropyl]-2-methylpyrazolo
[1,5-a]pyrimidine-6-carboxamidehydrochloride(5) (anagliptin
hydrochloride salt) as a potent and selective DPP-4 inhibitor
with uniquepharmacological profile [193].
I t is noteworthy that many known DPP-4 inhibitors have
the P-1--P-2 fragment, wherethe P-1 sitecontains aproline
mimic [10,194]. Pyrrolidine derivatives (vildagliptin and saxagliptin) have been widely explored as DPP-4 inhibitors due
to DPP-4s specificity for substrate having an aminoterminal prolineat C-2 [10]. T heir cyanopyrrolidinemoieties
bind to the S1 sub-site and form a covalent bond between
thenitrilegroupandhydroxyl of Ser630 in thecatalytictriad.
Further, their hydroxy adamantyl groups bind to theS2 subsite[195]. Vildagliptin was chemically unstableduetointramolecular cyclisation between thenitrilegroupandtheP-2 basic
aminemoiety. Saxagliptin has an improved chemical stability
by introduction of acyclopropyl ring(cis-4,5-methanobridge)
to theprolinenitrilewhich minimises cyclisation [160,196]. T he
DPP-4 inhibitors possessing the electrophilic trap such as a
nitrile group have low selectivity against other related prolyl

Expert Opin. Drug Discov. (2013) 8(11)

1347

M . Saf avi et al.

F
F

NH2

HO

HN

NH2

OH

CF3

Saxagliptin [244,245]
DPP-4 IC50: 26 nM
DPP8/DPP4 > 400 fold
DPP9/DPP4 > 75 fold

Sitagliptin [243]
DPP-4 IC50: 18 nM
QPP IC50 > 100 M
DPP8 IC50: 48 M

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

CH3
O

CH3

H3 C

N
NH3
TFA-

NH3
Cl-

NH2

N
H CH

CN
N

H
N
3

CN

CH3
CF3

CH3

HCl

N
N

CH3

N
N
N
N
H

CH3

5 [193]
DPP-4 IC50: 3.8 nM
DPP-8 IC50: 68 nM
DPP-9 IC50: 60 nM
F
F

NH2

N
N
H

6 [194]
DPP-4 IC50: 0.37 nM
DPP-8 IC50: 72.4 nM
DPP-9 IC50: 105 nM
F

CH3
N

4 [191]
DPP-4 IC50: 0.009 M
DPP-8 IC50: 17.38 M
DPP-9 IC50: 5.7 M

3 [190]
DPP-4 IC50: 0.0058 M
QPP IC50: 15 M
DPP-8 IC50: 46 M
DPP-9 IC50 > 100 M

CF3

2 [189]
DPP-4 IC50: 0.016 M
DPP-8 IC50: 25 M
DPP-9 IC50 > 100 M

CH3

H
N

CH3

CH3

1 [189]
DPP-4 IC50: 0.025 M
QPP IC50 > 100 M
DPP-9 IC50 > 100 M
DPP-8 IC50 > 100 M

HN

F
N

CH3

Vildagliptin [246]
DPP-4 IC50: 3.5 nM
QPP IC50 > 500 M

CH3
N

CN

O
N

7 [197]
DPP-4 IC50: 0.37 nM
DPP-8 IC50: 260 nM
DPP-9 IC50: 540 nM

O
N
H

N
N

N
CF3

8 [163]
DPP-4 IC50: 0.031 M
DPP-8 IC50: 78.5 M
DPP-9 IC50: 41.6 M

Figure 2. Schemat ic represent at ion of chemical st ruct ure of some examples of t he pept idomimet ic DPP-4.

1348

Expert Opin. Drug Discov. (2013) 8(11)

Import ance of synt het ic drugs f or T2DM drug discovery

F
O
CH3

N
N

N
N

CH3

NH3+TFA-

CN

N
N

NH2

CH3

N
N

NH2

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Alogliptin [246]
DPP-4 IC50 < 10 nM
DPP-8 IC50 > 100 M
DPP-9 IC50 > 100 M

Linagliptin [247]
DPP-4 IC50: 1 nM
QPP IC50 > 100 M
DPP-8 IC50: 40 M
DPP-9 IC50 > 10 M

N
N
N

NH2

NH2
CH3

Cl

O
N

N
CH3

Cl

O2
S

N
O

CH3

NH2

10 [203]
DPP-4 IC50: 0.001 M
DPP-4 inhib. in rat: 80%
Muscarinicreceptor M1 IC50: 1.190 M

12 [206]
DPP-4 IC50: 0.055 M
QPP IC50: 63 M
DPP-8 IC50 > 100 M
DPP-9 IC50 > 100 M

11 [204]
DPP-4 K i: 0.006 m
DPP-8 K i: > 30 M
DPP-9 K i: > 30 M

H3 C

Cl

CH3

NH2
H

9 [201]
DPP-4 IC50: 1.2 nM
QPP IC50: > 100 M
DPP-8 IC50 > 100 M
DPP-9 IC50 > 19 M

O
N

HN

O
CH3

NH2

N
N

O
C

CO2NH2

CH3

NH2

OH

13 [206]
DPP-4 IC50: 0.018 M
QPP IC50: 22 M
DPP-8 IC50: 18 M
DPP-9 IC50: 27 M

14 [207]
DPP-4 IC50: 1.3 nM
QPPIC50: 20 M
DPP-8 IC50 > 60 M
DPP-9 IC50 > 60 M

15 [208]
DPP-4 IC50: 0.48 nM
QPP IC50 > 10 M
DPP-8 IC50 > 100 M
DPP-9 IC50 > 100 M

Figure 3. Schemat ic represent at ion of chemical st ruct ure of some examples of t he non-pept idomimet ic DPP-4 inhibit ors.

peptidases, DPP-8 and DPP-9 [197]. T he key interactions of


thevildagliptin and saxagliptin structures with DPP-4 active
siteresidues arepresented in Table 2.
Yoshidaet al. designed novel pyrrolidinederivatives without electrophilicnitrilemoiety focused on thesubstituent at
the g-position of proline moiety of prolylthiazolidine core
structuretowards increasing theaffinity to theS2 sub-siteof
DPP-4 [194,198,199]. Accordingtoprevious reports, introducing
an electron-deficient 4-arylpiperazineresults in highly potent

and long-lasting inhibitors [200]. Fused bicyclic heteroarylpiperazinesubstituted at theg-position of theprolinestructure


in the investigation of L-prolylthiazolidines lacking theelectrophilic nitrile was previously explored. Compound (6)
(2-trifluoroquinolyl) was the most potent, long-lasting and
selective DPP-4 inhibitor. X-ray crystal structure determination of compound (6) indicates that compound (6) made
many interactions with the active site of DPP-4 (Table 2).
T he SAR study of fused bicyclic heteroarylpiperazine parts

Expert Opin. Drug Discov. (2013) 8(11)

1349

M . Saf avi et al.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Table 2. Key int eract ions bet w een inhibit or st ruct ures and DPP-4 act ive site residues.
DPP-4 inhibit or

Involved inhibit or st ruct ure

Int eract ion t ype

Linagliptin

Amino group on the piperidine

Hydrogen bonding interactions

Saxagliptin

C-6 carbonyl of the xanthine


Quinazoline group
Uracil group
Amino group (primary)

Hydrogen bonding interactions


Stacking interactions
Stacking interactions
Hydrogen bonding interactions

Carbonyl group
Hydroxyl group on the
adamantyl moiety
Imidate nitrogen
4,5-Methanopyrrolidine ring

Hydrogen bonding interactions


Hydrogen bonding interactions
Hydrogen bonding interactions
van der Waals interactions

Nitrile of the cyanopyrrolidine


b-Amino group

Covalent bond
Hydrogen bonding interactions

Carbonyl group
Triazolopiperazine
Trifluoromethyl group on
the triazolopiperazine
Trifluorobenzyl group

Hydrogen bonding interactions


Stacking interactions
Ionic Bonds

Amino group
Carbonyl group
Hydroxyl group on the
adamantyl moiety
Imidate nitrogen
Nitrile of the cyanopyrrolidine
Amino group of the proline
Carbonyl group
Quinolyl ring

Salt bridge interactions


Hydrogen bonding interactions
Hydrogen bonding interactions

Ser630, His740, Trp659,


Tyr631, Tyr662
and Tyr666
Glu205, Glu206
Asn710
His126 and Ser209

Hydrogen bonding interactions


Covalent bond
Salt bridge interactions
Hydrogen bonding interactions
Stacking interactions
CH--p interaction
Imperfect interaction

Tyr547
Ser630
Glu205 and Glu206
Asn710
Phe357
Arg358
Tyr585

Salt bridge interactions


Hydrogen bonding interactions
Hydrogen bonding interactions
Hydrogen bonding interactions
CH--p interaction
Hydrophobic interactions
Salt bridge interactions
-Interactions

Glu205 and Glu206


Asn710
Ser209 and Arg358
Val207
Phe357
Phe357
Glu205 and Glu206
Tyr585 and Arg356

Hydrogen bonding interactions


Hydrophobic interaction
Hydrogen bonding interactions

Glu205, Glu206
and Tyr662
Phe357
Lys554

Hydrogen bonding interactions

Tyr631

Hydrophobic interaction
Salt bridge interactions

Tyr547
Glu205 and Glu206

Hydrogen bonding interactions

Tyr631

Salt bridge interaction

Lys554

Stacking interactions

Tyr547

Sitagliptin

Vildagliptin

7 (teneligliptin)

14

15

1350

Trifluoromethyl on the
quinolyl ring
Amino group of the proline
Carbonyl group
Phenyl on the pyrazolyl ring
Piperazinyl ring
Pyrazolyl ring
Amino group
Trifluoromethyl group on
the triazolopiperazine
Amino group on the
quinoline ring
2-Isobutyl group
2-Oxo group on the
piperazin-2,5-dione
5-Oxo group on the
piperazin-2,5-dione
Piperazin-2,5-dione ring
Amino group at the piperidine
moiety (primary)
Carbonyl group on the
quinoline ring
Carboxyl group on the
benzene ring
3H-imidazo[4,5-c]quinolin4(5H)-one moiety

Hydrophobic interactions

Expert Opin. Drug Discov. (2013) 8(11)

DPP-4 residue
Glu205, Glu206
and Tyr662
Tyr631
Trp629
Tyr547
Glu205, Glu206
and Tyr662
Asn710
Tyr 547
Tyr 547
Val711, Val656, Tyr662,
Tyr666, Trp659
and Tyr547
Ser630
Glu205, Glu206
and Tyr662
Tyr547
Phe357
Arg358 and Ser209

St udy
Eckhardt et al.
2007 [202]

Metzler et al.
2008 [242]

Zhu et al. 2013,


Kim et al.
2005 [163,243]

Nabeno et al.
2013 [195]

Yoshida et al.
2012 [194]

Yoshida et al.
2012 [197]

Zhu et al.
2013 [163]
Maezaki et al.
2011 [207]

Ikuma et al.
2012 [208]

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Import ance of synt het ic drugs f or T2DM drug discovery

of previous study revealed that the non-linear (L-shaped)


structurewas moresuitablethan thelinear (I -shaped) onein
DPP-4 inhibitory activity. Moreover, theX-ray crystal structuredetermination of compound(6) in complex with human
DPP-4 indicated that interaction between the quinolyl ring
and the S2 extensive sub-site plays an important role for
DPP-4 inhibition [194]. H ence, the introduction of another
non-linear structure, a linked bicyclic heteroaryl group on
thepiperazineor piperidinemoiety, insteadof afusedbicyclic
heteroaryl group, was addressed in thelatter study which led
to discovery of 3-[(2S,4S)-4-[4-(3-methyl-1-phenyl-1H -pyrazol-5-yl)piperazin-1-yl]pyrrolidin-2-ylcarbonyl]thiazolidine
(7), as highly potent, selective, long-lasting and orally active
DPP-4 inhibitor. T heX-ray co-crystal structureof compound
(7) in DPP-4 demonstratedthat thecharacteristicfiverings of
compound (7) fit into the active site of DPP-4 and the key
interaction between the phenyl substituent on the pyrazolyl
ring and the S2 extensive sub-siteof DPP-4 not only raised
potency, but also increased selectivity (Table 2). Compound
(7) significantly inhibitedtheincreaseof plasmaglucoselevels
after an oral glucoseloadin Z ucker fatty rats. Compound(7)
(teneligliptin) has been approved for thetreatment of T 2DM
in Japan on June2012 [197].
T he crystal structures of DPP-4 in complex with bound
sitagliptin and alogliptin have shown that a sub-pocket
formed by the catalytic residue Ser630 and nearby residues
is usually occupied by ahydrophobicring group such as the
trifluorobenzyl groupof sitagliptin or thebenzonitrileof alogliptin. T wo acidicresidues --Glu205 andGlu206 --in theS2
sub-site of DPP-4 forms interactions with amine group of
sitagliptin (Table 2). T hese interactions between sitagliptin
andDPP-4 givealargeunoccupiedspacearoundtheright terminal portion (the fused heterocyclic ring) which could be
modified to generateanew series of DPP-4 inhibitors. With
this rationale, well-established click chemistry was used to
quickly explorethe SAR of thelinker and theterminal portion of sitagliptin. A series of 4-(2,4,5-trifluorophenyl)
butane-1,3-diamines weredesigned and elaborated as DPP-4
inhibitors. T heresults showed that compound (8) had desirable efficacy, selectivity and pharmacokinetics properties. It
is noteworthy that crystal structures of DPP-4 in complex
with bound compound (8) revealed that the trifluoromethyl
heterocycle is rotated ~ 180 and directed towards a subpocket different from thesub-sitebound by sitagliptin, providing clues for the design of new DPP-4 inhibitors. T he
key interaction between compound (8) and DPP-4 active
sitearepresented in Table 2 [163].
4.2

Non-pept idomimet ic inhibit ors

Monocyclic, bicyclic, bicycliclactams andtriazolopyridazinessubstituted 3-aminopiperidines were synthesized and evaluated for in vitro inhibition of human DPP-4, QPP, DPP-8
and DPP-9. Bicyclic lactams series were potent (DPP-4
I C50: 1.2 --8 nM) andin general hadgood-to-excellent selectivity. Compound (9) was found to be the most potent

DPP-4 inhibitor with excellent selectivity but poor pharmacokineticproperties [201].


T he X-ray crystal structure of linagliptin complexed with
DPP-4 indicatedthemanner in which theseresidues provided
a good fit (Table 2) [202]. A class of 3,5-dihydro-imidazo
[4,5-d]pyridazin-4-ones with different substituent provided.
In addition to in vitro DPP-4 inhibitory activity, the effects
of compounds on themuscarinicreceptor M1 and inhibition
of DPP-4 in rats was tested. I t should benoticed that significant M1 receptor inhibition was acharacteristicof someearly
xanthines. Compound (10) seems to beapotential candidate
showingvery potent DPP-4 inhibitor activity in vitroas well
as in vivoand only poor affinity to M1 receptors. T helonglasting strong DPP-4 inhibition (> 70% 24 h post-administration) of compound (10) is particularly noteworthy. T herefore, further works on this series, and compound (10) in
particular, will behelpful in thedevelopment of better drugs
for T 2DM [203].
Pyrazolopyrimidines areaclass of compounds under investigation. A series of pyrazolopyrimidines with different substituents on the pyrazole ring which include alkyl, aryl,
substituted aryl and carboxylic ester groups were tested
in vitro against purified human DPP-4. Favourable
DPP-4 activity, selectivity over DPP-8 andDPP-9 andample
opportunity for further optimisation aroundthepyrazolering
were the three desirable characteristics of this class of compounds. A 5-aminomethyl imidazolopyrimidines derivative
compound (11) was themost potent against DPP-4 with no
significant DPP-8/DPP-9 inhibition. T he oGT T in ob/ob
mice by compound (11) showed dose-dependent increase in
plasmainsulin levels [204].
While substituted 4-amino cyclohexylglycineanalogues in
the a-amino acid series showed good activity against
DPP-4 [205], thesubstitutedcyclohexaneandpiperidinederivatives wereprepared and assessed for DPP-4 inhibitory activity and selectivity profiles against other proline-specific
peptidases. In the cyclohexyl with sulphonamide substituent
series, N-methyl-(2,5-dimethylisoxazol-yl)sulphonamide (12)
showed the most promising potency against DPP-4 and
excellent selectivity against the off-target enzymes but poor
pharmacokinetic. AmongtheN-substitution piperidinederivatives, 3-(5-aminocarbonylpyridyl piperidine) (13) displayed
excellent DPP-4 activity with good selectivity versus other
prolineenzymes [206].
As mentioned in Table 2, many of the reported DPP-4
inhibitors includingvildagliptin and saxagliptin makeacovalent interaction with theSer630 residuein theS1 pocket. Sitagliptin and linagliptin form non-covalent interactions with
the S1, S2 or S2 extensive sub-sites in addition to the
S1 and S2 sub-sites. It appears that excess interactions may
increase DPP-4 inhibition beyond the level yielded by the
fundamental interactions with the S1 and S2 sub-sites and
are more effective than forming a covalent bond with
Ser630 in the S1 subunit [195,207]. Design of novel noncovalent inhibitors of DPP-4 using structural information

Expert Opin. Drug Discov. (2013) 8(11)

1351

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

M . Saf avi et al.

derivedfromDPP-4 structures co-crystallisedwith small molecules originatingfromdifferent chemical classes is nowavital


investigation in medicinal chemistry.
Based on the X-ray co-crystal structure of some compounds, thehydrogen bondinginteraction with Lys554 may
beapplicablein thenovel design of DPP-4 inhibitors. H ence,
Maezaki et al. developed anovel series of non-covalent quinoline-basedinhibitors totarget thesidechain of Lys554. Synthesis and evaluation of designed compounds against DPP-4,
QPP, DPP-8 and DPP-9 enzyme and determination of
absorption, distribution, metabolism and excretion and pharmacokineticprofiles revealed 1-[3-(aminomethyl)-4-(4-methylphenyl)-2-(2-methylpropyl) quinolin-6-yl]piperazine-2,5-dione
compound (14) -- a potent, selective and orally active
DPP-4 inhibitor with long-lasting ex vivoactivity in dogs and
potent glucose-loweringeffects in rats. A dockingstudy of compound (14) suggested that an efficient fit into thebindingsite
and a hydrogen-bonding interaction with the side chain of
Lys554 are important in enhancing the inhibitory activity.
Also, in comparison with other members of this series, it seems
that compound(14) achievedexcellent activity by takingadvantageof thedesirablepiperazin-2,5-dionesubstituent and forming hydrophobic and two hydrogen-bonding interactions,
includingtheonewith Lys554 (Table 2) [207].
I kuma et al. found a potent DPP-4 inhibitor by highthroughput screening of laboratory chemical library and
proper optimisation based on results of published DPP-4
enzyme-inhibitor docking study. Further docking studies
werecarried out to design astrategy for optimisation of lead
compound with 3H -imidazo[4,5-c]quinolin-4(5H )-one as
skeleton. Compound (15) with 2-chloro-5-fluorobenzylsubstituent on theimidazoquinolinegroup was found as potent
and highly selective DPP-4 inhibitor [208]. T he results of
dockingstudy areshown in Table 2.

New synthet ic compounds as


SGLT2 inhibit ors

5.

Inhibition of glucose reabsorption and increasing urinary


glucose excretion lead to anegative energy balance, making
these series of compounds a unique promising therapeutic
class with potential to reduce body weight in T 2DM [172].
Only sugars in ahexoseand theD-configuration aretranslocatedby SGLT . T hehydrogen bondingbetween SGLT 2 and
hydroxy groups at C-2, C-3 and C-4 and hydrophobicinteractions with theC-6 methylenegroupof thepyranosearethe
key interactions in determiningsugar specificity [209,210]. T he
b-glucosides with largeringstructures such as aromaticaglucones and the aglucones themselves are well accommodated
at the sugar-binding site and not translocated. T hey act as
inhibitors of the transport [174,209]. Various O-, C-, N- and
S-glucosides have been synthesized with high affinity and
high specificity for SGLT 2 that arein thedifferent stages of
clinical development [209]. T heb-glucosidephlorizin, thefirst
SGLT 2 inhibitors, is a O-aryl glycoside and non-selective
1352

competitive inhibitor with two moieties to bind with SGLT :


aglucosemoiety connectedwith an oxygen atom(O-glucoside)
and a hydrophobic aglycone moiety [209,211]. T he firstgeneration SGLT 2 inhibitors with O-glycosidic bond were
discontinued due to their short half-lives in vivo resulting
from glycosidase cleavage [211,212]. Second-generation SGLT 2
inhibitors adopted other forms of glycoside linkage resistant
to glycosidaseactivity such as C-glucosidestructure [213].
5.1

C-glycoside analogues of SGLT2 inhibit ors

Many C-aryl glycoside analogues containing conjugated aryl


moiety with glucosemoiety by C--C bond instead of instable
C--O--C bond have been designed and synthesized [179,214].
Among these analogues, dapagliflozin has been used as a
lead compound as many syntheticderivatives havebeen further madeon its SAR data. A series of fluorinated analogues
of dapagliflozins were designed and synthesized by incorporation of a gem-difluoromethylene group at C-4 position.
T hein vitroinhibitory activities of theseseries against human
SGLT 2 showed that some of the analogues (compound 16)
with CF2 at C-4 are better SGLT 2 inhibitors compared
with dapagliflozin (Figure 4) [215].
T hestructural similarity of D-xyloseandD-glucoseandthe
positiveresults of O-xylosides andN-b-D-xylosides [216] ledto
thedesign of anew series of C-linked indolylxylosides. SARs
studies indicatedthat ap-cyclopropylphenyl groupin thedistal position and substituents at the 7-position of the indole
moiety were necessary for optimum inhibitory activity. T he
pharmacokinetic and animal studies demonstrated that the
most potent compound7-substitutedindolylxlyosides bearing
adistal p-cyclopropylphenyl group(17) is metabolically stable
with significant efficacy on lowering blood glucose levels of
streptozotocin-induced diabeticrats [217].
I kegai et al. investigation led to the discovery of other
C-glucoside SGLT 2 inhibitors with azulene motifs [218].
Although azuleneis not acommon motif in medicinal chemistry of anti-diabeticdrugdiscovery, asearch of theliterature
indicates that certain azulene derivatives possess significant
pharmacological and therapeutic activity [219-221]. H ence, a
series of C-glucosides with azulenerings in theaglyconemoiety was synthesized; SAR of azulene-derived C-glucosideand
theinhibitory activities towards hSGLT 1 and hSGLT 2 were
explored. Incorporation of aphenolic hydroxyl group at the
central benzene ring afforded a more potent and selective
SGLT 2 inhibitor (18), which exerted astrong and sustained
anti-hyperglycaemic effect in rodent diabetic models.
A monocholinesalt of compound(18) (YM543) was selected
as aclinical candidatefor usein treatingT 2DM [218].
A new class of potent and selective SGLT 2 inhibitors, by
modifyingtheglyconesidechain incorporatingastructurally
novel dioxabicyclo-[3.2.1]octane ring system, was prepared [222]. At first, a series of C-5-spirocyclic C-glycoside
weresynthesized with relatively good potency and selectivity
for human SGLT 2 but suboptimal pharmacokinetics [223].
T he medicinal chemistry strategy by innovative chemistry

Expert Opin. Drug Discov. (2013) 8(11)

Import ance of synt het ic drugs f or T2DM drug discovery

HO

CH3

OH

OH

Cl

HO

HO

OH

OH

CH3

HO

HO

HO

O
O

HO

OH

OH
OH

OH

Phlorizin [215]
SGLT2 IC50: 46.03 nM

Dapagliflozin [214]
SGLT2 EC50: 1.1 nM
SGLT1/SGLT2: 1200 fold

Sergliflozin-A [249]
SGLT2 K i: 2.39 nM
SGLT1/SGLT2: 296 fold

CH3

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Cl

HO

O
F

HO
F

OH
F

HO

OH

HO
OH

OH

HO

OH
OH

16 [215]
SGLT2 IC50: 0.35 nM

17 [217]
SGLT2 EC50: 47 nM
SGLT1/SGLT2: 6 fold
NH

CH3
Cl
HO

HO

18 [218]
SGLT2 IC50: 8.9 nM
SGLT1/SGLT2: 280 fold

HO

CH3

(CH3)3

HO

OH

HO

OH

OH
OH

19 [222]
SGLT2 IC50: 0.887 nM
SGLT1/SGLT2 > 2234 fold
HO

HO

20 [225]
SGLT2 EC50: 3.85 M

S
OH

21 (pseudo-sergliflozin) [227]
SGLT2 IC50: 2.45 nM
SGLT1/SGLT2 > 200000 fold

Cl

N N
O

OH
HO

N
OH

OH

22 [228]
Inhibition rate of blood glucose
levels in oGTT: 74.85%

23 [216]
SGLT2 EC50: 161 nM
SGLT1/SGLT2: 1.3 fold

Figure 4. Schemat ic represent at ion of chemical st ruct ures of some SGLT2 inhibit ors t oget her w it h t heir experiment al
inhibit ory and select ivit y act ivit ies.

that allowed difficult, yet very desirable, targets to besynthesized in an analogue-friendly fashion and thedevelopment of
a pharmacokinetics/pharmacodynamics (PKPD) model
brought more hope. T hese efforts led to deprioritisation of
theC-5-spirocyclicC-glycosideSGLT 2 inhibitors and focusing on thedioxabicyclo[3.2.1]octaneclass. It is believed that
the bridged ketal system would confer rigidity with potentially positive impact on potency and selectivity. Moreover,
introduction of ahydroxymethyleneH -bond donor group at

C-5 (in place of the spirocycle) brought more anticipation


to reducetherateof human PhaseI I metabolism and further
improvement of the potency. T hese efforts supported the
advancement of compound (19) (PF-04971729) into clinical
development that is being evaluated for the treatment of
T 2DM [222].
Vyas et al. performed aligand-based 3D quantitativeSARs
(QSARs) study on C-aryl glucoside SGLT 2 inhibitors
(180 analogues), which belong to various diversified

Expert Opin. Drug Discov. (2013) 8(11)

1353

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

M . Saf avi et al.

structures such as benzisothiazole, indolizine-b-D-glucopyranoside, thiazolylmethylphenyl, pyridazine, thiazole and


pyrimidinylmethylphenyl glucoside analogues. Comparative
molecular fieldanalysis (CoMFA) andcomparativemolecular
similarity indices analysis (CoMSI A) were used to further
explore the structural requirements of C-aryl glucoside analogues for SGLT 2 inhibition. CoMFA and CoMSIA contour
map analysis offered enough information to understand the
importance of the substituent at particular positions with
respect to steric, electrostatic, hydrophobic, hydrogen-bond
donor andacceptor properties for better activity of C-aryl glucoside SGLT 2 inhibitors. For example analysis of CoMFA and
CoMSI Acontour plots revealedtheimportanceof hydrophobic
and hydrogen-bond donor and acceptor properties [224].
5.2

SGLT2 inhibit ors w it h ot her st ruct ures

T he application of ligand-based virtual screening strategy


comprising the combination of pharmacophore model with
shape-based scoring and structure clustering analysis led to
the discovery of non-glycosideSGLT 2 inhibitors. A total of
7989 compounds matchingall thefeatures describedby pharmacophoremodel werethen subjectedtoshape-basedscoring
andstructureclusteringanalysis. T otally, 80 compounds with
high scoreand structurally diversescaffolds wereselected for
biological testing, and out of these, threecompounds showed
inhibition of SGLT 2 with EC50 < 10 M. T henon-glycoside
compound (20) with EC50 = 3.85 M was the most potent
compound [225].
Most SGLT 2 inhibitors in clinical trials aretaken fromtwo
distinct series: C-aryl glycosides (e.g., dapagliflozin) and
O-aryl glycosides (e.g., sergliflozin and remogliflozin) [223].
T he abovementioned compounds were C-glucoside derivatives but showed higher cancer risk [226]. I t is thought that
themetabolicinstability of theO-glucosideSGLT 2 inhibitors
can be solved by modifying the sugar core of sergliflozin-A.
Several small-moleculecarbohydratemimics such as pseudosergliflozin weresynthesized by aregion- and stereo-selective
allylicsubstitution reaction. T hen theSGLT 2/SGLT 1 inhibitory activities of these newly synthesized compounds were
studied. T he endocyclic oxygen atom of sergliflozin-A was
replaced by a methylene unit to render the molecule free
from glycosidase degradation. I n this way, compound (21)
was found as apotent and selectiveinhibitor of SGLT 2 that
considered alead compound for further development [227].
A series of thiadiazole-based S-glycosides weresynthesized
from D-glucose, D-galactoseandavariety of phenylaceticacids
and then evaluated in vivo with an oGT T test. Of these,
5-benzyl-1,3,4-thiadiazol-2-yl
1-thio-b-D-glucopyranoside
(22) was the most efficacious to suppress the blood glucose
excursion during oGT T with the inhibition rates of blood
glucoselevels equal to 74.85% [228].
Since, N-linked glycosides were expected to have greater
metabolic stability compared to O-glucosides, a novel series
of N-linked b-D-xylosides were synthesized and evaluated
for inhibitory activity against SGLT 2 in a cell-based assay.
1354

Among them, the 4-chloroindolyl-N-xyloside with the


4-cyclopropylbenzyl at thedistal position (23) was found the
most potent inhibitor with no selectivity for SGLT 2 over
SGLT 1. According to pharmacokinetic data, compound
(23) was metabolically stable with alow clearanceand good
oral bioavailability in SpragueDawley rats [216].
7.

Conclusion

Morerecently, theneedtodevelopnewdrugs for T 2DM as a


multi-causal andcomplicateddiseasehas been emphasized, as
theworldwideincidenceof this diseaseis increasingdramatically. Various drugoptions arenow availablefor themanagement of T 2DM, but almost all areassociatedwith restrictions
and most do not address all aspects of thedefects in diabetic
patients. T herecently introduced DPP-4 and SGLT 2 inhibitors effectively lower the blood glucose without weight gain
and risk of hypoglycaemia.
T he co-crystal structures of human DPP-4 with some
approvedinhibitors areavailable. So, in recent years, newsyntheticDPP-4 inhibitors with diversechemical structures have
been found, according to the X-ray crystal structure of
DPP-4 and computer-modellingstudies. Remarkablecreativity andthoughts havebeen demonstratedby medicinal chemists in designing novel and potent series of DPP-4 inhibitors.
Various SGLT 2 inhibitors based on theglucosidestructureof
phlorizin, sergliflozin, dapagliflozin, canagliflozin and other
inhibitors in clinical trial havesincebeen proposed. Alternative
candidate SGLT 2 inhibitors that have also been considered
includemodified sugar rings, N-glucosides, S-glucosides, nonglycoside and, more recently, bridged ketal ring, azulene
motifs. T heother promisingagents targetingnovel molecular
mechanisms are under development. T he hope is to discover
more selective and effective anti-diabetic agents in the
near future.
8.

Expert opinion

Despitearapid increasein thenumber of drugs availableto


treat hyperglycaemia, diabetes still remains a major global
concern. Although threeoldest classes comprisinginsulin, sulphonylureas and the biguanides are effective in improving
outcome, they are unable to sustain adequate glycaemic
control over time in the diabetic patients. A variety of antihyperglycaemic drugs with different and complementary
mechanisms of action are currently available. Moreover, the
use of current medications is often limited by their side
effects, mostly sudden hypoglycaemia, weight gain, gastrointestinal effects or oedema. T hesefactors necessitatetheongoing quest for novel agents that would address fundamental
defects of T 2DM and haveminimal adverseeffects.
Among the main anti-diabetic drug classes, DPP-4 and
SGLT 2 inhibitors are novel and promising therapy for
T 2DM. T he details of synthesis backgrounds of many new
DPP-4 inhibitors, in the recent years, are described in this

Expert Opin. Drug Discov. (2013) 8(11)

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Import ance of synt het ic drugs f or T2DM drug discovery

review. In this review, thestructures of somecompounds with


biological properties are also presented. Phenylalanine and
cyclohexylalanine derivatives and imidazopiperidine-based
b-aminoacidderivatives weredesigned basedon theobservation that modification of thephenylalanine, cyclohexylalanine
or piperazinemoiety improved theDPP-4 potency by several
folds. Also the 3-fluoropyrrolidine analogues obtained by
replacingthepyrrolidinering compounds (1) and (2) showed
more activity compared to parent compound [189,190]. Since
bicyclo[3.3.0]octane derivative with pyrrolidine-2-carbonitrile
was previously reported to beapotent DPP-4 inhibitor [191],
azobicyclo[3.3.0]octanecompounds weresynthesized. T hediscovery of anagliptin was accordingto studies showedthat pyrazolo[1,5-a]pyrimidine functions as a bioisostere conveying
much metabolicstability and safety [193].
A costly component of drug discovery approaches is
structure-basedscreening(docking), which is adesign strategy
for newchemical entities, or optimisation of leadcompounds
identified by other methods, using the 3D structure of the
DPP-4. T he 3D structure of DPP-4 could be obtained by
X-ray or nuclear magneticresonancestudies or from homology models. T he protein data bank is a repository for the
3D structural data of proteins. T he crystal structures of
DPP-4 have been previously disclosed and the discovery of
teneligliptin performedby aidof mentionedin silicomethods
which comprise computationally assessed ligand-binding
interactions with DPP-4. Non-peptidomimetic inhibitors of
DPP-4 such as 3-aminopiperidines with bicycliclactams substituents wereoriginated from sitagliptin --apeptidomimetic
DPP-4 inhibitor, X-ray crystallographic structure along with
molecular modelling [201]. Also non-covalent quinoline-based
inhibitors which target thesidechain of Lys554 weredesigned
based on the X-ray co-crystal structure of some compounds
containing hydrogen bonding interaction with Lys554 [207].
Numerous DPP-4 inhibitors that werediscoveredand/or optimised usingin silicomethods havereached thelevel of clinical
studies or havegained approval of usein somecountries.
Stability and selectivity of SGL T 2 inhibitors is an
important issue in drug design. T he SARs studies of
phlorizin and other candidates led to discovery of another
selectiveand potent SGL T 2 inhibitor with excellent pharmacokinetic properties. Discovery of dioxabicyclo[3.2.1]
octane was supported by analogue-friendly fashion and
PK PD model [222].

T heX-ray crystal structureof SGLT 2 is yet not available;


thus, to explorethestructural requirements for SGLT 2 inhibition, the ligand-directed drug model was used to screen
activecompounds as templates. Ligand-based screeningtechniques mainly focus on comparingmolecular similarity analyses of compounds with known and unknown moiety. T he
3D QSAR methods of CoMFA and CoMSI A models have
been developed as ligand-based approaches. As mentioned,
CoMFA and CoMSI A models weresuccessfully used in providing useful information for designing new SGLT 2 inhibitors [224]. Computer-aided drug design plays an important
role in drug discovery and development and provides useful
insights into experimental findings and mechanism of action
and new suggestions for molecular structures to synthesize.
In this way, large databases of compounds can be tested
in silicowith cheaper and faster techniques beforesubmitting
to in vitroexpensivesynthesis [229].
Most classicanti-diabeticdrugs target themajor pathophysiological defects in T 2DM, namely insulin resistance and
impaired insulin secretion, while the new developing drugs
focus on other options such as SGLT 2 inhibitors that affect
the kidney through insulin-independent mechanisms.
Recently, PPARa/ dual agonists [230], SI RT 1 activators [231,232],
glycogen phosphorylase inhibitors [233] and protein tyrosine
phosphatase 1B inhibitors [233-235] were luckily designed for
thetreatment of T 2DM.
T here is no doubt that T 2DM and its related complications are associated with oxidative stress that plays arole in
thepathogenesis of T 2DM. Antioxidant (natural or synthetic)
therapy can protect b-cells from apoptosis and might positively work in control of hyperglycaemia by activating the
production and releaseof insulin [13,66,74].
Finding new targets and new faster and cheaper synthesis
methods arethemain goal in T 2DM drugdiscovery. Nosingle diabetes treatment is best for everyone as each person
responds in adifferent way tomedication. T ogether, theavailable and rising drug candidates should provide physicians
with abroad rangeof pharmacological choices to successfully
individualisepatient treatment.

Declarat ion of int erest


T heauthors stateno conflict of interest and havereceived no
payment in preparation of this manuscript.

Expert Opin. Drug Discov. (2013) 8(11)

1355

M . Saf avi et al.

Bibliography
Papers of special notehavebeen highlighted as
either of interest ( ) or of considerableinterest
( ) to readers.
1.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

2.

3.

Alberti KG, Z immet PZ . Definition,


diagnosis and classification of diabetes
mellitus and its complications. Part 1:
diagnosis and classification of diabetes
mellitus provisional report of aWH O
consultation. Diabet Med
1998;15:539-53
BeletateV, El Dib RP, Atallah AN. Z inc
supplementation for theprevention of
type2 diabetes mellitus.
CochraneDatabaseSyst Rev
2007(1):CD005525
KuzuyaT , NakagawaS, Satoh J, et al.
Report of theCommitteeon the
classification and diagnosticcriteriaof
diabetes mellitus. Diabetes Res
Clin Pract 2002;55:65-85

4.

KuzuyaT , MatsudaA. Classification of


diabetes on thebasis of etiologies versus
degreeof insulin deficiency.
Diabetes Care1997;20:219-20

5.

American diabetes association. Diagnosis


and classification of diabetes mellitus.
Diabetes Care2008;31(Suppl 1):S55-60

6.

7.

8.

9.

10.

11.

1356

Sollu M, Banji D, Banji OJF,


KumbalaVCS. Appraisal on causes and
treatment of diabetes mellitus. Arch Appl
Sci Res 2010;2(5):239-60
McCarthy MI . Genomics,
type2 diabetes, and obesity. N Engl
J Med 2010;363:2339-50

12.

13.

14.

H avaleSH , Pal M. Medicinal chemistry


approaches to theinhibition of
dipeptidyl peptidase-4 for thetreatment
of type2 diabetes. Bioorg Med Chem
2009;17:1783-802
Comprehensive review which focuses
on the journey of drug discovery of
DPP-4 inhibitors for oral delivery.
H osseini A, Abdollahi M. Diabetic
neuropathy and oxidativestress:

McLennan SV, Abdollahi M, T wigg SM.


Connectivetissuegrowth factor, matrix
regulation, and diabetickidney disease.
Curr Opin Nephrol H ypertens
2013;22:85-92

23.

Rahimi R, Nikfar S, Larijani B,


Abdollahi M. A review on theroleof
antioxidants in themanagement of
diabetes and its complications.
Biomed Pharmacother 2005;59:365-73

Farshchi A, Nikfar S, Abdollahi M.


Concerns on theuseof chromium in
type2 diabetes mellitus; needs to
conduct major meta-analysis.
I nt J Pharmacol 2012;8:472

24.

Ghamarian A, Abdollahi M, Su X, et al.


Effect of chicory seed extract on glucose
tolerancetest (GT T ) and metabolic
profilein early and latestagediabetic
rats. Daru 2012;20:56

25.

H asani-Ranjbar S, Larijani B,
Abdollahi M. A systematicreview of
I ranian medicinal plants useful in
diabetes mellitus. Arch Med Sci
2008;4:285-92

CranePK, Walker R, H ubbard RA,


et al. Glucoselevels and risk of
dementia. N Engl J Med
2013;369(6):540-8

15.

Bastaki S. Diabetes mellitus and its


treatment. Int J Diabetes Metab
2005;13:111-34

16.

Salari P, Nikfar S, Abdollahi M. No


superiority of exenatideover insulin in
diabeticpatients in terms of weight
reduction or incidenceof adverseeffects:
ameta-analysis. I nt J Pharmacol
2011;7:749-56

26.

H asani-Ranjbar S, Larijani B,
Abdollahi M. Recent updateon animal
and human evidences of promising
anti-diabeticmedicinal plants:
amini-review of targetingnew drugs.
Asian J Anim Vet Adv 2011;6:1271-5

17.

Freeman JS. T heincreasing epidemiology


of diabetes and review of current
treatment algorithms. J Am
Osteopath Assoc2010;110:eS2-6

27.

H irsch I B. Insulin analogues. N Engl


J Med 2005;352:174-83

28.

Khanavi M, T aheri M, Rajabi A, et al.


Stimulation of hepaticglycogenolysis and
inhibition of gluconeogenesis arethe
mechanisms of antidiabeticeffect of
Centaureabruguieranassp. belangerana.
Asian J Anim Vet Adv 2012;7:1166-74

29.

Noor A, Bansal VS, Vijayalakshmi MA.


Current updateon anti-diabetic
biomolecules from key traditional Indian
medicinal plants. Curr Sci
2013;104:721-5

30.

Sarkhail P, Abdollahi M, Fadayevatan S,


et al. Effect of Phlomis persicaon
glucoselevels and hepaticenzymatic
antioxidants in streptozotocin-induced
diabeticrats. Pharmacogn Mag
2010;6:219-24

31.

Nakai M, Sekiguchi F, ObataM, et al.


Synthesis and insulin-mimeticactivities
of metal complexes with 3hydroxypyridine-2-carboxylicacid.
J Inorg Biochem 2005;99:1275-82

32.

Sakurai H , Adachi Y. T hepharmacology


of theinsulinomimeticeffect of zinc
complexes. Biometals 2005;18:319-23

33.

SangheraDK, Blackett PR.


T ype2 diabetes genetics: beyond gwas.
J Diabetes Metab 2012;3(189):6948

18.

MonikaG, Sarbjot S, Punam G.


Dipeptidyl peptidase-4 inhibitors: anew
approach in diabetes treatment. Int J
Drug Dev Res 2009;1:146-56

19.

Nathan DM, BuseJB, Davidson MB,


et al. Medical management of
hyperglycemiain type2 diabetes:
aconsensus algorithm for theinitiation
and adjustment of therapy: aconsensus
statement of theAmerican Diabetes
Association and theEuropean Association
for theStudy of Diabetes. Diabetes Care
2009;32:193-203

Mostafalou S, Abdollahi M. Pesticides


and human chronicdiseases: evidences,
mechanisms, and perspectives.
T oxicol Appl Pharmacol
2013;268:157-77
H adjibabaieM, Gholami K, Khalili H ,
et al. Comparativeefficacy and safety of
atorvastatin, simvastatin and lovastatin in
themanagement of dyslipidemic
type2 diabeticpatients. T herapy
2006;3:759-64

A review of thepotential benefits of


pentoxifyllinein diabeticand
non-diabeticproteinuria. J Pharm
Pharm Sci 2011;14:128-37

therapeuticperspectives. OxideMed
Cell Longev 2013;2013:168039

20.

21.

22.

Momtaz S, Abdollahi M. An updateon


pharmacology of Saturejaspecies; from
antioxidant, antimicrobial, antidiabetes
and anti-hyperlipidemicto reproductive
stimulation. Int J Pharmacol
2010;6(4):454-61
Abdollahi M, Z uki AB, Goh YM, et al.
Effects of Momordicacharantiaon
pancreatichistopathological changes
associated with streptozotocin-induced
diabetes in neonatal rats.
H istol H istopathol 2011;26:13-21
Badri S, Dashti-Khavidaki S,
Lessan-Pezeshki M, Abdollahi M.

Expert Opin. Drug Discov. (2013) 8(11)

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

Import ance of synt het ic drugs f or T2DM drug discovery

34.

Edelman SV. T ypeI I diabetes mellitus.


Adv I ntern Med 1998;43:449-500

35.

Mercado MM, McLenithan JC,


Silver KD, Shuldiner AR. Genetics of
insulin resistance. Curr Diab Rep
2002;2:83-95

36.

Goldstein BJ. Insulin resistanceas the


coredefect in type2 diabetes mellitus.
Am J Cardiol 2002;90(5):3-10

37.

Bloomgarden Z T . Insulin resistance:


current concepts. Clin T her
1998;20:216-31

38.

WatanabeRM. T hegenetics of insulin


resistance: wheres Waldo?
Curr Diab Rep 2010;10:476-84

39.

Bastard JP, Maachi M, Lagathu C, et al.


Recent advances in therelationship
between obesity, inflammation, and
insulin resistance. Eur CytokineNetw
2006;17:4-12
A review that helps to understand the
pathophysiological mechanisms at the
root of insulin resistance and T2DM.

47.

48.

49.

T aniguchi CM, Emanuelli B, Kahn CR.


Critical nodes in signalling pathways:
insights into insulin action. Nat Rev Mol
Cell Biol 2006;7:85-96
Walker RJ, Anderson NM, Jiang Y, et al.
Roleof beta-adrenergicreceptor
regulation of T NF-alphaand insulin
signaling in retinal Muller cells.
Invest Ophthalmol Vis Sci
2011;52:9527-33
Reiter CE, Wu X, SandirasegaraneL,
et al. Diabetes reduces basal retinal
insulin receptor signaling: reversal with
systemicand local insulin. Diabetes
2006;55:1148-56

50.

WhiteMF. I RS proteins and the


common path to diabetes. Am J Physiol
Endocrinol Metab 2002;283:413-22

51.

Khan AH , Pessin JE. Insulin regulation


of glucoseuptake: acomplex interplay of
intracellular signalling pathways.
Diabetologia2002;45:1475-83

52.

SasaokaT , H ori H , WadaT , et al.


SH 2-containing inositol phosphatase
2 negatively regulates insulin-induced
glycogen synthesis in L6 myotubes.
Diabetologia2001;44:1258-67

Saeidi N, Meoli L, Nestoridi E, et al.


Reprogramming of intestinal glucose
metabolism and glycemiccontrol in rats
after gastricbypass. Science
2013;341(6144):406-10

53.

SasaokaT , WadaT , T suneki H . Lipid


phosphatases as apossibletherapeutic
target in cases of type2 diabetes and
obesity. Pharmacol T her
2006;112:799-809

42.

Das U N. Obesity, metabolicsyndrome


X, and inflammation. Nutrition
2002;18:430-2

54.

43.

Cinti S, Mitchell G, Barbatelli G, et al.


Adipocytedeath defines macrophage
localization and function in adipose
tissueof obesemiceand humans.
J Lipid Res 2005;46:2347-55

WadaT , SasaokaT , Funaki M, et al.


Overexpression of SH 2-containing
inositol phosphatase2 results in negative
regulation of insulin-induced metabolic
actions in 3T 3-L1 adipocytes viaits
5-phosphatasecatalyticactivity.
Mol Cell Biol 2001;21:1633-46

44.

Kroder G, Bossenmaier B, Kellerer M,


et al. T umor necrosis factor-alpha- and
hyperglycemia-induced insulin resistance.
Evidencefor different mechanisms and
different effects on insulin signaling.
J Clin I nvest 1996;97:1471-7

40.

Look AH EAD Research Group.


Cardiovascular effects of intensive
lifestyleintervention in type2 diabetes.
N Engl J Med 2013;369(2):145-54

41.

45.

Lagathu C, Bastard JP, Auclair M, et al.


Chronicinterleukin-6 (IL -6) treatment
increased I L-6 secretion and induced
insulin resistancein adipocyte: prevention
by rosiglitazone. Biochem Biophys
Res Commun 2003;311:372-9

46.

Pessin JE, Saltiel AR. Signaling pathways


in insulin action: molecular targets of
insulin resistance. J Clin I nvest
2000;106:165-9

55.

56.

57.

H otamisligil GS, Budavari A, Murray D,


Spiegelman BM. Reduced tyrosinekinase
activity of theinsulin receptor in obesitydiabetes. Central roleof tumor necrosis
factor-alpha. J Clin I nvest
1994;94:1543-9
Long SD, PekalaPH . Regulation of
GLUT 4 mRNA stability by tumor
necrosis factor-alpha: alterations in both
protein binding to the3 untranslated
region and initiation of translation.
Biochem Biophys Res Commun
1996;220(3):949-53
Bastard JP, Maachi M, Van Nhieu JT ,
et al. AdiposetissueI L-6 content
correlates with resistanceto insulin
activation of glucoseuptakeboth in vivo

Expert Opin. Drug Discov. (2013) 8(11)

and in vitro. J Clin Endocrinol Metab


2002;87(5):2084-9
58.

T rayhurn P, Wood IS. Adipokines:


inflammation and thepleiotropicroleof
whiteadiposetissue. Br J Nutr
2004;92:347-55

59.

KasugaM. I nsulin resistanceand


pancreaticbetacell failure. J Clin I nvest
2006;116:1756-60

60.

Cerf ME. Betacell dysfunction and


insulin resistance.
Front Endocrinol (Lausanne) 2013;4:37

61.

Prentki M, Nolan CJ. I slet betacell


failurein type2 diabetes. J Clin Invest
2006;116:1802-12

62.

Poitout V, Robertson RP.


Glucolipotoxicity: fuel excess and
beta-cell dysfunction. Endocr Rev
2008;29:351-66

63.

Xiang AH , Kawakubo M, T rigo E, et al.


Decliningbeta-cell compensation for
insulin resistancein H ispanicwomen
with recent gestational diabetes mellitus:
association with changes in weight,
adiponectin, and C-reactiveprotein.
Diabetes Care2010;33:396-401

64.

Faghihi T , Radfar M, Barmal M, et al.


A randomized, placebo-controlled trial of
selenium supplementation in patients
with type2 diabetes: effects on glucose
homeostasis, oxidativestress, and lipid
profile. Am J T her
2013;Published online29 Apr 2013;
doi:10.1097/MJT .0b013e318269175f

65.

Navaei-Nigjeh M, Rahimifard M,
Pourkhalili N, et al. Multi-organ
protectiveeffects of cerium oxide
nanoparticle/selenium in diabeticrats:
evidencefor moreefficiency of
nanocerium in comparison to metal form
of cerium. Asian J Anim Vet Adv
2012;7:605-12

66.

T abatabaei-Malazy O, Larijani B,
Abdollahi M. A novel management of
diabetes by means of strongantioxidants
combination. J Med H ypotheses Ideas
2013;7:25-30

67.

Pourkhalili N, H osseini A,
Nili-Ahmadabadi A, et al. Biochemical
and cellular evidenceof thebenefit of a
combination of cerium oxide
nanoparticles and selenium to diabetic
rats. World J Diabetes 2011;2:204-10

68.

Drews G, Krippeit-Drews P, Dufer M.


Oxidativestress and beta-cell
dysfunction. Pflugers Arch
2010;460:703-18

1357

M . Saf avi et al.

69.

70.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

71.

Abdollahi M, Fooladian F, Emami B,


et al. Protection by sildenafil and
theophyllineof lead acetate-induced
oxidativestress in rat submandibular
gland and saliva. H um Exp T oxicol
2003;22:587-92
AstaneieF, Afshari M, Mojtahedi A,
et al. T otal antioxidant capacity and
levels of epidermal growth factor and
nitricoxidein blood and salivaof
insulin-dependent diabeticpatients.
Arch Med Res 2005;36:376-81
Farshchi A, Nikfar S, Seyedifar M,
Abdollahi M. Effect of chromium on
glucoseand lipid profiles in patients with
type2 diabetes; ameta-analysis review of
randomized trials. J Pharm Pharm Sci
2013;16:99-114

72.

H osseini A, Abdollahi M. I t is timeto


formulatean antioxidant mixturefor
management of diabetes and its
complications: noticefor pharmaceutical
industries. Int J Pharm 2012;8:60-1

73.

Kamyab S, H ejrati S, Khanavi M,


Mohammadirad A. H epaticmechanisms
of theWalnut antidiabeticeffect in mice.
Cent Eur J Biol 2010;5:304-9

74.

Larijani B, Afshari M,
Astanehi-Asghari F, et al. Effect of
short-term carvedilol therapy on salivary
and plasmaoxidativestress parameters
and plasmaglucoselevel in typeI I
diabetes. T herapy 2006;3:119-23

75.

76.

77.

78.

1358

Milani E, Nikfar S, Khorasani R, et al.


Reduction of diabetes-induced oxidative
stress by phosphodiesteraseinhibitors in
rats. Comp Biochem Physiol C
T oxicol Pharmacol 2005;140:251-5
Mohseni Salehi Monfared SS, Larijani B,
Abdollahi M. Islet transplantation and
antioxidant management:
acomprehensivereview.
World J Gastroenterol 2009;15:1153-61
Radfar M, Larijani B, H adjibabaieM,
et al. Effects of pentoxifyllineon
oxidativestress and levels of EGF and
NO in blood of diabetictype-2 patients;
arandomized, double-blind placebocontrolled clinical trial.
Biomed Pharmacother 2005;59:302-6
Sarkhail P, Rahmanipour S,
Fadyevatan S, et al. Antidiabeticeffect of
Phlomis anisodonta: effects on hepatic
cells lipid peroxidation and antioxidant
enzymes in experimental diabetes.
Pharmacol Res 2007;56:261-6

79.

80.

81.

82.

Vosough-Ghanbari S, Rahimi R,
Kharabaf S, et al. Effects of Satureja
khuzestanicaon serum glucose, lipids and
markers of oxidativestress in patients
with type2 diabetes mellitus:
adouble-blind randomized controlled
trial. Evid Based Complement
Alternat Med 2010;7:465-70

90.

Stumvoll M, Nurjhan N, Perriello G,


et al. Metaboliceffects of metformin in
non-insulin-dependent diabetes mellitus.
N Engl J Med 1995;333:550-4

91.

Bolzano K. Biguanides: reasons for


withdrawal of drugs and remaining
indications. ActaMed Austriaca
1978;5:85-8

Merrill GF, Kurth EJ, H ardieDG,


Winder WW. AI CA ribosideincreases
AMP-activated protein kinase, fatty acid
oxidation, and glucoseuptakein rat
muscle. Am J Physiol 1997;273:1107-12

92.

Luft D, Schmulling RM, Eggstein M.


Lacticacidosis in biguanide-treated
diabetics: areview of 330 cases.
Diabetologia1978;14:75-87

Z hou G, Myers R, Li Y, et al. Roleof


AMP-activated protein kinasein
mechanism of metformin action.
J Clin I nvest 2001;108:1167-74

93.

Palumbo PJ. Metformin: effects on


cardiovascular risk factors in patients
with non-insulin-dependent diabetes
mellitus. J Diabetes Complications
1998;12:110-19

94.

Garber AJ, Duncan T G, Goodman AM,


et al. Efficacy of metformin in typeII
diabetes: results of adouble-blind,
placebo-controlled, dose-responsetrial.
Am J Med 1997;103:491-7

95.

Sheehan MT . Current therapeutic


options in type2 diabetes mellitus:
apractical approach. Clin Med Res
2003;1:189-200
An informative review on the
management of T2DM from the
initiation of oral agents through dual
(potentially triple) therapy onto
insulin initiation.

Ansari G, Mojtahedzadeh M, Kajbaf F,


et al. H ow does blood glucosecontrol
with metformin influenceintensive
insulin protocols?Evidencefor
involvement of oxidativestress and
inflammatory cytokines. Adv T her
2008;25:681-702

83.

Defronzo RA, Goodman AM. Efficacy of


metformin in patients with
non-insulin-dependent diabetes mellitus.
T heMulticenter Metformin Study
Group. N Engl J Med 1995;333:541-9

84.

H olman RR, Paul SK, Bethel MA, et al.


10-year follow-up of intensiveglucose
control in type2 diabetes. N Engl J Med
2008;359:1577-89

85.

Morsink LM, Smits MM, Diamant M.


Advances in pharmacologictherapies for
type2 diabetes. Curr Atheroscler Rep
2013;15:302
Overview of recent advances in
pharmacological therapy for T2DM,
focusing on agents that recently have
been approved and those for which
approval is pending.

86.

Bailey CJ, T urner RC. Metformin.


N Engl J Med 1996;334:574-9

87.

Cusi K, Consoli A, Defronzo RA.


Metaboliceffects of metformin on
glucoseand lactatemetabolism in
noninsulin-dependent diabetes mellitus.
J Clin Endocrinol Metab
1996;81:4059-67

96.

Stang M, Wysowski DK, Butler-Jones D.


I ncidenceof lacticacidosis in metformin
users. Diabetes Care1999;22:925-7

97.

Stolar MW, H oogwerf BJ, Gorshow SM,


et al. Managing type2 diabetes: going
beyond glycemiccontrol. J Manag
CarePharm 2008;14:s2-19

98.

Rendell MS, Kirchain WR.


Pharmacotherapy of type2 diabetes
mellitus. Ann Pharmacother
2000;34:878-95

99.

Bharatam PV, Patel DS, Iqbal P.


Pharmacophoricfeatures of biguanide
derivatives: an electronicand structural
analysis. J Med Chem 2005;48:7615-22

88.

H undal RS, Krssak M, Dufour S, et al.


Mechanism by which metformin reduces
glucoseproduction in type2 diabetes.
Diabetes 2000;49:2063-9

100. Setter SM, Iltz JL, T hams J,


Campbell RK. Metformin hydrochloride
in thetreatment of type2 diabetes
mellitus: aclinical review with afocus on
dual therapy. Clin T her
2003;25:2991-3026

89.

Del Prato S, Marchetto S, PipitoneA,


et al. Metformin and freefatty acid
metabolism. Diabetes Metab Rev
1995;11:33-41

101. Del Prato S, Pulizzi N. T heplaceof


sulfonylureas in thetherapy for
type2 diabetes mellitus. Metabolism
2006;55:20-7

Expert Opin. Drug Discov. (2013) 8(11)

Import ance of synt het ic drugs f or T2DM drug discovery

102. Bryan J, CraneA, Vila-Carriles WH ,


et al. Insulin secretagogues, sulfonylurea
receptors and K(AT P) channels.
Curr Pharm Des 2005;11:2699-716

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

103. GribbleFM, Reimann F. Differential


selectivity of insulin secretagogues:
mechanisms, clinical implications, and
drug interactions.
J Diabetes Complications 2003;17:11-15
104. Perfetti R, Ahmad A. Novel sulfonylurea
and non-sulfonylureadrugs to promote
thesecretion of insulin.
T rends Endocrinol Metab
2000;11:218-23
105. Bak JF, Pedersen O. Gliclazideand
insulin action in human muscle.
Diabetes Res Clin Pract 1991;14(2):61-4
106. Pedersen O, H other-Nielsen O, Bak J,
et al. Effects of sulfonylureas on
adipocyteand skeletal muscleinsulin
action in patients with
non-insulin-dependent diabetes mellitus.
Am J Med 1991;90(6A):22-8
107. Korytkowski MT . Sulfonylureatreatment
of type2 diabetes mellitus: focus on
glimepiride. Pharmacotherapy
2004;24:606-20
108. Jasik M. Glimepiridein daily practice.
Przegl Lek 2003;60:409-12
109. Basit A, Riaz M, Fawwad A.
Glimepiride: evidence-based facts, trends,
and observations (GIFT S). VascH ealth
Risk Manag 2012;8:463-72
110. BijlstraPJ, Lutterman JA, Russel FG,
et al. Interaction of sulphonylurea
derivatives with vascular AT P-sensitive
potassium channels in humans.
Diabetologia1996;39:1083-90
111. LeeT M, Chou T F. Impairment of
myocardial protection in type2 diabetic
patients. J Clin Endocrinol Metab
2003;88:531-7
112. Campbell RK. Glimepiride: roleof a
new sulfonylureain thetreatment of
type2 diabetes mellitus.
Ann Pharmacother 1998;32:1044-52
113. Dornhorst A. Insulinotropicmeglitinide
analogues. Lancet 2001;358:1709-16
114. MalaisseWJ. Pharmacology of the
meglitinideanalogs: new treatment
options for type2 diabetes mellitus.
T reat Endocrinol 2003;2:401-14
115. Li J, T ian H , Li Q, et al. Improvement
of insulin sensitivity and beta-cell
function by nateglinideand repaglinide
in type2 diabeticpatients - a

randomized controlled double-blind and


double-dummy multicentreclinical trial.
Diabetes Obes Metab 2007;9:558-65
116. Rosenstock J, H assman DR,
Madder RD, et al. Repaglinideversus
nateglinidemonotherapy: arandomized,
multicenter study. Diabetes Care
2004;27:1265-70
117. H u S, Wang S, Fanelli B, et al.
Pancreaticbeta-cell K(AT P) channel
activity and membrane-binding studies
with nateglinide: acomparison with
sulfonylureas and repaglinide.
J Pharmacol Exp T her 2000;293:444-52
118. McLeod JF. Clinical pharmacokinetics of
nateglinide: arapidly-absorbed,
short-actinginsulinotropicagent.
Clin Pharmacokinet 2004;43:97-120
119. Chachin M, YamadaM, FujitaA, et al.
Nateglinide, aD-phenylalaninederivative
lacking either asulfonylureaor
benzamido moiety, specifically inhibits
pancreaticbeta-cell-typeK(AT P) channels.
J Pharmacol Exp T her
2003;304:1025-32
120. Campbell I W. Nateglinidecurrent and
futurerolein thetreatment of patients
with type2 diabetes mellitus. I nt J
Clin Pract 2005;59:1218-28

128. Defronzo RA. Banting Lecture. From the


triumvirateto theominous octet: anew
paradigm for thetreatment of
type2 diabetes mellitus. Diabetes
2009;58:773-95
129. MaedaN, T akahashi M, Funahashi T ,
et al. PPARgammaligands increase
expression and plasmaconcentrations of
adiponectin, an adipose-derived protein.
Diabetes 2001;50:2094-9
130. MatsudaM, ShimomuraI , SataM, et al.
Roleof adiponectin in preventing
vascular stenosis. T hemissing link of
adipo-vascular axis. J Biol Chem
2002;277:37487-91
131. Phillips LS, Grunberger G, Miller E,
et al. Once- and twice-daily dosing with
rosiglitazoneimproves glycemiccontrol
in patients with type2 diabetes.
Diabetes Care2001;24:308-15
132. Park KS, Ciaraldi T P, Abrams-Carter L,
et al. T roglitazoneregulation of glucose
metabolism in human skeletal muscle
cultures from obesetypeI I diabetic
subjects. J Clin Endocrinol Metab
1998;83:1636-43

121. Spencer CM, Markham A. T roglitazone.


Drugs 1997;54:89-101

133. I dris I , Gray S, Donnelly R.


Rosiglitazoneand pulmonary oedema:
an acutedose-dependent effect on human
endothelial cell permeability.
Diabetologia2003;46:288-90

122. BaeMA, RheeH , Song BJ. T roglitazone


but not rosiglitazoneinduces G1 cell
cyclearrest and apoptosis in human and
rat hepatomacell lines. T oxicol Lett
2003;139:67-75

134. PageRL , Gozansky WS, Ruscin JM.


Possibleheart failureexacerbation
associated with rosiglitazone: casereport
and literaturereview. Pharmacotherapy
2003;23:945-54

123. Nolan JJ, Ludvik B, Beerdsen P, et al.


Improvement in glucosetoleranceand
insulin resistancein obesesubjects treated
with troglitazone. N Engl J Med
1994;331:1188-93

135. Papoushek C. T heglitazones:


rosiglitazoneand pioglitazone. J Obstet
Gynaecol Can 2003;25:853-7

124. Suter SL, Nolan JJ, WallaceP, et al.


Metaboliceffects of new oral
hypoglycemicagent CS-045 in NIDDM
subjects. Diabetes Care1992;15:193-203
125. Yki-Jarvinen H . T hiazolidinediones.
N Engl J Med 2004;351:1106-18
126. Okuno A, T amemoto H , T obeK, et al.
T roglitazoneincreases thenumber of
small adipocytes without thechangeof
whiteadiposetissuemass in obese
Z ucker rats. J Clin Invest
1998;101:1354-61
127. Picard F, Auwerx J. PPAR (gamma) and
glucosehomeostasis. Annu Rev Nutr
2002;22:167-97

Expert Opin. Drug Discov. (2013) 8(11)

136. Gim H J, Cheon YJ, Ryu JH , Jeon R.


Design and synthesis of benzoxazole
containingindoleanalogs as peroxisome
proliferator-activated receptor-gamma/
deltadual agonists. Bioorg Med
Chem Lett 2011;21:3057-61
137. Matsumoto K, Yano M, MiyakeS, et al.
Effects of vogliboseon glycemic
excursions, insulin secretion, and insulin
sensitivity in non-insulin-treated
NI DDM patients. Diabetes Care
1998;21:256-60
138. Shinozaki K, Suzuki M, Ikebuchi M,
et al. I mprovement of insulin sensitivity
and dyslipidemiawith anew
alpha-glucosidaseinhibitor, voglibose, in
nondiabetichyperinsulinemicsubjects.
Metabolism 1996;45:731-7

1359

M . Saf avi et al.

139. Scheen AJ. I s therearolefor


alpha-glucosidaseinhibitors in the
prevention of type2 diabetes mellitus?
Drugs 2003;63:933-51
140. Johnston PS, Lebovitz H E, Coniff RF,
et al. Advantages of alpha-glucosidase
inhibition as monotherapy in elderly
type2 diabeticpatients. J Clin
Endocrinol Metab 1998;83:1515-22

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

141. Balfour JA, McT avish D. Acarbose.


An updateof its pharmacology and
therapeuticusein diabetes mellitus.
Drugs 1993;46:1025-54
142. Santeusanio F, Compagnucci P.
A risk-benefit appraisal of acarbosein the
management of non-insulin-dependent
diabetes mellitus. DrugSaf
1994;11:432-44
143. Reuser AJ, Wisselaar H A. An evaluation
of thepotential side-effects of
alpha-glucosidaseinhibitors used for the
management of diabetes mellitus. Eur J
Clin Invest 1994;24(3):19-24
144. GengP, Qiu F, Z hu Y, Bai G. Four
acarviosin-containing oligosaccharides
identified from Streptomyces
coelicoflavus Z G0656 arepotent
inhibitors of alpha-amylase.
Carbohydr Res 2008;343:882-92
145. van Genugten RE, van RaalteDH ,
Diamant M. Does glucagon-likepeptide1 receptor agonist therapy add valuein
thetreatment of type2 diabetes?Focus
on exenatide. Diabetes Res Clin Pract
2009;86(Suppl 1):S26-34
146. Parks M, Rosebraugh C. Weighing risks
and benefits of liraglutidetheFDAs
review of anew antidiabetictherapy.
N Engl J Med 2010;362:774-7
147. Nikfar S, Abdollahi M, Salari P. T he
efficacy and tolerability of exenatidein
comparison to placebo; asystematic
review and meta-analysis of randomized
clinical trials. J Pharm Pharm Sci
2012;15:1-30
148. Nauck MA, Baranov O, Ritzel RA,
Meier JJ. Do current incretin mimetics
exploit thefull therapeuticpotential
inherent in GLP-1 receptor stimulation?
Diabetologia
2013;published online9 Jun 2013;
doi:10.1007/s00125-013-2953-6
149. Drucker DJ, Nauck MA. T heincretin
system: glucagon-likepeptide-1 receptor
agonists and dipeptidyl
peptidase-4 inhibitors in type2 diabetes.
Lancet 2006;368:1696-705

1360

150. H olst JJ. T hephysiology of glucagon-like


peptide1. Physiol Rev 2007;87:1409-39
151. Pospisilik JA, Stafford SG, Demuth H U,
et al. Long-term treatment with the
dipeptidyl peptidaseI V inhibitor P32/
98 causes sustained improvements in
glucosetolerance, insulin sensitivity,
hyperinsulinemia, and beta-cell glucose
responsiveness in VDF (fa/fa) Z ucker
rats. Diabetes 2002;51:943-50
152. Cho YM, Merchant CE, Kieffer T J.
T argeting theglucagon receptor family
for diabetes and obesity therapy.
Pharmacol T her 2012;135(3):247-78
153. Siu FY, H eM, deGraaf C, et al.
Structureof thehuman glucagon class B
G-protein-coupled receptor. Nature
2013;499(7459):444-9
154. Garber AJ. L ong-acting glucagon-like
peptide1 receptor agonists: areview of
their efficacy and tolerability.
Diabetes Care2011;34(Suppl 2):S279-84
155. Bond A. Exenatide(Byetta) as anovel
treatment option for type2 diabetes
mellitus. Proc(Bayl Univ Med Cent)
2006;19:281-4
156. Parkes DG, Pittner R, JodkaC, et al.
I nsulinotropicactions of exendin-4 and
glucagon-likepeptide-1 in vivo and in
vitro. Metabolism 2001;50:583-9
157. BjerreKL, Madsen LW, Andersen S,
et al. Glucagon-likePeptide-1 receptor
agonists activaterodent thyroid C-cells
causing calcitonin releaseand C-cell
proliferation. Endocrinology
2010;151:1473-86
158. Franks AS, LeePH , GeorgeCM.
Pancreatitis: apotential complication of
liraglutide?Ann Pharmacother
2012;46:1547-53
159. WajcbergE, Amarah A. Liraglutidein
themanagement of type2 diabetes.
Drug Des Devel T her 2010;4:279-90
160. Augeri DJ, Robl JA, Betebenner DA,
et al. Discovery and preclinical profileof
Saxagliptin (BMS-477118): ahighly
potent, long-acting, orally active
dipeptidyl peptidaseI V inhibitor for the
treatment of type2 diabetes.
J Med Chem 2005;48:5025-37
161. Gallwitz B. Small molecule
dipeptidylpeptidaseIV inhibitors under
investigation for diabetes mellitus
therapy. Expert Opin Investig Drugs
2011;20:723-32
162. Saisho Y, I toh H . Dipeptidyl
peptidase-4 inhibitors and angioedema:

Expert Opin. Drug Discov. (2013) 8(11)

aclass effect?Diabet Med


2013;30:149-50
163. Z hu L , Li Y, Qiu L, et al. Design and
synthesis of 4-(2,4,5-T rifluorophenyl)
butane-1,3-diamines as Dipeptidyl
PeptidaseIV I nhibitors. ChemMedChem
2013;8:1104-16
164. Kim W, Egan JM. T heroleof incretins
in glucosehomeostasis and diabetes
treatment. Pharmacol Rev
2008;60:470-512
165. H olst JJ, Vilsboll T , Deacon CF. T he
incretin system and its rolein
type2 diabetes mellitus.
Mol Cell Endocrinol 2009;297:127-36
166. Mentlein R, Gallwitz B, Schmidt WE.
Dipeptidyl-peptidaseIV hydrolyses
gastricinhibitory polypeptide,
glucagon-likepeptide-1(7-36)amide,
peptidehistidinemethionineand is
responsiblefor their degradation in
human serum. Eur J Biochem
1993;214:829-35
167. SomeyaY, T aharaA, Nakano R, et al.
Pharmacological profileof ASP8497, a
novel, selective, and competitive
dipeptidyl peptidase-IV inhibitor, in vitro
and in vivo. Naunyn Schmiedebergs
Arch Pharmacol 2008;377:209-17
168. GuptaR, Walunj SS, T okalaRK, et al.
Emergingdrug candidates of dipeptidyl
peptidaseI V (DPP I V) inhibitor class for
thetreatment of T ype2 Diabetes.
Curr Drug T argets 2009;10:71-87
169. Scheen A. Gliptins (dipeptidyl
peptidase-4 inhibitors) and risk of acute
pancreatitis. Expert Opin Drug Saf
2013;12(4):545-57
170. Lankas GR, Leiting B, Roy RS, et al.
Dipeptidyl peptidaseIV inhibition for
thetreatment of type2 diabetes:
potential importanceof selectivity over
dipeptidyl peptidases 8 and 9. Diabetes
2005;54:2988-94
171. Babu A. Canagliflozin for thetreatment
of type2 diabetes. Drugs T oday (Barc)
2013;49:363-76
172. Ghosh RK, Ghosh SM, ChawlaS,
JasdanwalaSA. SGLT 2 inhibitors: anew
emerging therapeuticclass in the
treatment of type2 diabetes mellitus.
J Clin Pharmacol 2012;52:457-63
173. Gerich JE. Roleof thekidney in normal
glucosehomeostasis and in the
hyperglycaemiaof diabetes mellitus:
therapeuticimplications. Diabet Med
2010;27:136-42

Import ance of synt het ic drugs f or T2DM drug discovery

174. Wright EM, H irayamaBA, Loo DF.


Activesugar transport in health and
disease. J I ntern Med 2007;261:32-43

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

175. RahmouneH , T hompson PW,


Ward JM, et al. Glucosetransporters in
human renal proximal tubular cells
isolated from theurineof patients with
non-insulin-dependent diabetes. Diabetes
2005;54:3427-34
176. Neumiller JJ, WhiteJR Jr,
Campbell RK. Sodium-glucose
co-transport inhibitors: progress and
therapeuticpotential in type2 diabetes
mellitus. Drugs 2010;70:377-85
177. Elkinson S, Scott LJ. Canagliflozin: first
global approval. Drugs 2013;73:979-88
178. H ardman T C, Dubrey SW.
Development and potential roleof
type-2 sodium-glucosetransporter
inhibitors for management of
type2 diabetes. Diabetes T her
2011;2:133-45
179. NomuraS, Sakamaki S, H ongu M, et al.
Discovery of canagliflozin, anovel
C-glucosidewith thiophenering, as
sodium-dependent glucosecotransporter
2 inhibitor for thetreatment of
type2 diabetes mellitus. J Med Chem
2010;53:6355-60
180. Bays H E, Goldberg RB, T ruitt KE,
Jones MR. Colesevelam hydrochloride
therapy in patients with type2 diabetes
mellitus treated with metformin: glucose
and lipid effects. Arch Intern Med
2008;168:1975-83
181. KecheY. Bromocriptinemesylate: food
and Drug Administration approved new
approach in therapy of non-insulin
dependent diabetes mellitus with poor
glycemiccontrol. J Pharm Bioallied Sci
2010;2:148-50
182. Kumar KA, Kumar SP, Janardan S.
Bromocriptine: anovel approach for
management of type2 diabetes mellitus.
JDDT 2012;2:151-5
183. ViaMA, ChandraH , Araki T , et al.
Bromocriptineapproved as thefirst
medication to target dopamineactivity to
improveglycemiccontrol in patients
with type2 diabetes. Diabetes Metab
Syndr Obes 2010;3:43-8
184. Green BD, Flatt PR, Bailey CJ.
Dipeptidyl peptidaseIV (DPP IV)
inhibitors: anewly emerging drug class
for thetreatment of type2 diabetes.
Diab VascDis Res 2006;3:159-65

185. Inzucchi SE, McGuireDK. New drugs


for thetreatment of diabetes: part II :
incretin-based therapy and beyond.
Circulation 2008;117:574-84
186. Rummey C, Metz G. H omology models
of dipeptidyl peptidases 8 and 9 with a
focus on loop predictions near theactive
site. Proteins 2007;66:160-71
187. Edmondson SD, Mastracchio A,
Duffy JL, et al. Discovery of potent and
selectiveorally bioavailable
beta-substituted phenylalaninederived
dipeptidyl peptidaseI V inhibitors.
Bioorg Med Chem Lett
2005;15:3048-52
188. Xu J, Wei L, Mathvink R, et al.
Discovery of potent and selective
phenylalaninebased dipeptidyl peptidase
IV inhibitors. Bioorg Med Chem Lett
2005;15:2533-6
189. Duffy JL, Kirk BA, Wang L, et al.
4-aminophenylalanineand
4-aminocyclohexylalaninederivatives as
potent, selective, and orally bioavailable
inhibitors of dipeptidyl peptidaseIV.
Bioorg Med Chem Lett
2007;17:2879-85
190. Chen P, Caldwell CG, Mathvink RJ,
et al. Imidazopiperidineamides as
dipeptidyl peptidaseI V inhibitors for the
treatment of diabetes. Bioorg Med
Chem Lett 2007;17:5853-7
191. Cho T P, Gang LZ , Long YF, et al.
Synthesis and biological evaluation of
bicyclo[3.3.0] octanederivatives as
dipeptidyl peptidase4 inhibitors for the
treatment of type2 diabetes. Bioorg Med
Chem Lett 2010;20:3521-5
192. FurutaS, Smart C, H ackett A, et al.
Pharmacokinetics and metabolism of
[14C]anagliptin, anovel dipeptidyl
peptidase-4 inhibitor, in humans.
Xenobiotica2013;43:432-42
193. Kato N, OkaM, MuraseT , et al.
Discovery and pharmacological
characterization of N-[2-({2-[(2S)-2cyanopyrrolidin-1-yl]-2-oxoethyl}amino)2-methylpropyl]-2-methyl pyrazolo[1,5-a]
pyrimidine-6-carboxamidehydrochloride
(anagliptin hydrochloridesalt) as a
potent and selectiveDPP-IV inhibitor.
Bioorg Med Chem 2011;19:7221-7
194. YoshidaT , Akahoshi F, SakashitaH ,
et al. Fused bicyclic
heteroarylpiperazine-substituted
L-prolylthiazolidines as highly potent
DPP-4 inhibitors lackingtheelectrophilic

Expert Opin. Drug Discov. (2013) 8(11)

..

nitrilegroup. Bioorg Med Chem


2012;20:5033-41
Example of original report of the
design of new DPP-4 inhibitors
according to previous structure-based
screening techniques.

195. Nabeno M, Akahoshi F, KishidaH ,


et al. A comparativestudy of thebinding
modes of recently launched dipeptidyl
peptidaseI V inhibitors in theactivesite.
Biochem Biophys Res Commun
2013;434:191-6
..
A study describing co-crystal structure
of vildagliptin with DPP-4 and the
relationships between six inhibitors
binding interactions with DPP-4 and
their inhibitory activity.
196. Magnin DR, Robl JA, Sulsky RB, et al.
Synthesis of novel potent dipeptidyl
peptidaseI V inhibitors with enhanced
chemical stability: interplay between the
N-terminal amino acid alkyl sidechain
and thecyclopropyl group of alphaaminoacyl-l-cis-4,5-methanoprolinenitrilebased inhibitors. J Med Chem
2004;47:2587-98
197. YoshidaT , Akahoshi F, SakashitaH ,
et al. Discovery and preclinical profileof
teneligliptin (3-[(2S,4S)-4-[4-(3-methyl1-phenyl-1H -pyrazol-5-yl)piperazin-1-yl]
pyrrolidin-2-y lcarbonyl]thiazolidine):
ahighly potent, selective, long-lasting
and orally activedipeptidyl peptidaseI V
inhibitor for thetreatment of
type2 diabetes. Bioorg Med Chem
2012;20:5705-19
198. SakashitaH , Akahoshi F, KitajimaH ,
et al. [(S)-gamma-(Arylamino)prolyl]
thiazolidinecompounds as anovel series
of potent and stableDPP-IV inhibitors.
Bioorg Med Chem 2006;14:3662-71
199. SakashitaH , Akahoshi F, YoshidaT ,
et al. Lead optimization of [(S)-gamma(arylamino)prolyl]thiazolidinefocused on
gamma-substituent: indolinecompounds
as potent DPP-I V inhibitors.
Bioorg Med Chem 2007;15:641-55
200. YoshidaT , SakashitaH , Akahoshi F,
H ayashi Y. [(S)-gamma-(4-Aryl-1piperazinyl)-l-prolyl]thiazolidines as a
novel series of highly potent and
long-lasting DPP-I V inhibitors.
Bioorg Med Chem Lett
2007;17:2618-21
201. Cox JM, H arper B, Mastracchio A, et al.
Discovery of 3-aminopiperidines as
potent, selective, and orally bioavailable
dipeptidyl peptidaseI V inhibitors.

1361

M . Saf avi et al.

Bioorg Med Chem Lett


2007;17:4579-83

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

202. Eckhardt M, Langkopf E, Mark M, et al.


8-(3-(R)-aminopiperidin-1-yl)-7-but-2ynyl-3-methyl-1-(4-methyl-quinazolin-2ylmethyl)-3,7-dihydropurine-2,6-dione
(BI 1356), ahighly potent, selective,
long-acting, and orally bioavailable
DPP-4 inhibitor for thetreatment of
type2 diabetes. J Med Chem
2007;50:6450-3
203. Eckhardt M, H auel N, H immelsbach F,
et al. 3,5-Dihydro-imidazo[4,5-d]
pyridazin-4-ones: aclass of potent
DPP-4 inhibitors. Bioorg Med
Chem Lett 2008;18:3158-62
204. BriganceRP, Meng W, FuraA, et al.
Synthesis and SAR of azolopyrimidines
as potent and selectivedipeptidyl
peptidase-4 (DPP4) inhibitors for
type2 diabetes. Bioorg Med Chem Lett
2010;20:4395-8
205. ParmeeER, H eJ, Mastracchio A, et al.
4-Amino cyclohexylglycineanalogues as
potent dipeptidyl peptidaseIV inhibitors.
Bioorg Med Chem Lett 2004;14:43-6
206. Chen P, Caldwell CG, Ashton W, et al.
Synthesis and evaluation of [(1R)-1amino-2-(2,5-difluorophenyl)ethyl]
cyclohexanes and 4-[(1R)-1-amino-2(2,5-difluorophenyl)ethyl]piperidines as
DPP-4 inhibitors. Bioorg Med
Chem Lett 2011;21:1880-6
207. Maezaki H , Banno Y, Miyamoto Y, et al.
Discovery of potent, selective, and orally
bioavailablequinoline-based dipeptidyl
peptidaseIV inhibitors targeting Lys554.
Bioorg Med Chem 2011;19:4482-98
208. IkumaY, H ochigai H , KimuraH , et al.
Discovery of 3H -imidazo[4,5-c]quinolin4(5H )-ones as potent and selective
dipeptidyl peptidaseI V (DPP-4)
inhibitors. Bioorg Med Chem
2012;20:5864-83
209. KinneRK, CastanedaF. SGLT
inhibitors as new therapeutictools in the
treatment of diabetes.
H andb Exp Pharmacol
2011(203):105-26
210. Kipp H , Kinne-Saffran E, Bevan C,
KinneRK. Characteristics of renal Na
(+)-D-glucosecotransport in theskate
(Rajaerinacea) and shark (Squalus
acanthias). Am J Physiol
1997;273:R134-42
211. Kurosaki E, OgasawaraH . I pragliflozin
and other sodium-glucosecotransporter-2

1362

(SGLT 2) inhibitors in thetreatment of


type2 diabetes: preclinical and clinical
data. Pharmacol T her 2013;139:51-9
212. I saji M. SGLT 2 inhibitors: molecular
design and potential differences in effect.
Kidney I nt Suppl 2011;S14-19
213. I mamuraM, Nakanishi K, Suzuki T ,
et al. Discovery of Ipragliflozin
(ASP1941): anovel C-glucosidewith
benzothiophenestructureas apotent and
selectivesodium glucoseco-transporter 2
(SGLT 2) inhibitor for thetreatment of
type2 diabetes mellitus.
Bioorg Med Chem 2012;20:3263-79
214. Meng W, Ellsworth BA, Nirschl AA,
et al. Discovery of dapagliflozin:
apotent, selectiverenal
sodium-dependent glucosecotransporter
2 (SGLT 2) inhibitor for thetreatment of
type2 diabetes. J Med Chem
2008;51:1145-9
215. Chen Z H , Wang RW, Qing FL.
Synthesis and biological evaluation of
SGL T 2 inhibitors:
gem-difluoromethylenated Dapagliflozin
analogs. T etrahedron Lett
2012;53:2171-6
216. Yao CH , Song JS, Chen CT , et al.
Discovery of novel N-beta-Dxylosylindolederivatives as
sodium-dependent glucosecotransporter
2 (SGLT 2) inhibitors for the
management of hyperglycemiain
diabetes. J Med Chem 2011;54:166-78
217. Yao CH , Song JS, Chen CT , et al.
Synthesis and biological evaluation of
novel C-indolylxylosides as
sodium-dependent glucoseco-transporter
2 inhibitors. Eur J Med Chem
2012;55:32-8
218. I kegai K, I mamuraM, Suzuki T , et al.
Synthesis and biological evaluation of
C-glucosides with azulenerings as
selectiveSGLT 2 inhibitors for the
treatment of type2 diabetes mellitus:
discovery of YM543. Bioorg Med Chem
2013;21:3934-48
219. Chen CH , LeeO, Yao CN, et al. Novel
azulene-based derivatives as potent
multi-receptor tyrosinekinaseinhibitors.
Bioorg Med Chem Lett
2010;20:6129-32
220. RekkaE, Chrysselis M, Siskou I,
Kourounakis A. Synthesis of new azulene
derivatives and study of their effect on
lipid peroxidation and lipoxygenase
activity. Chem Pharm Bull (T okyo)
2002;50:904-7
Expert Opin. Drug Discov. (2013) 8(11)

221. Z hang LY, Yang F, Shi WQ, et al.


Synthesis and antigastriculcer activity of
novel 5-isoproyl-3,8-dimethylazulene
derivatives. Bioorg Med Chem Lett
2011;21:5722-5
222. Mascitti V, Maurer T S, Robinson RP,
et al. Discovery of aclinical candidate
from thestructurally uniquedioxabicyclo[3.2.1]octaneclass of
sodium-dependent glucosecotransporter
2 inhibitors. J Med Chem
2011;54:2952-60
villeC,
223. Mascitti V, Robinson RP, Pre
et al. Syntheses of C-5-spirocyclic.
C-glycosideSGLT 2 inhibitors.
T etrahedron Lett 2010;51:1880-3
224. Vyas VK, Bhatt H G, Patel PK, et al.
CoMFA and CoMSIA studies on C-aryl
glucosideSGL T 2 inhibitors as potential
anti-diabeticagents. SAR QSAR
Environ Res 2013;24:519-51
..
The study explores ligand-based 3D
QSAR (CoMFA and CoMSIA) on Caryl glucoside SGLT2 inhibitors as
potential anti-diabetic agents.
225. Wu JS, Peng YH , Wu JM, et al.
Discovery of non-glycosidesodiumdependent glucoseco-transporter 2
(SGL T 2) inhibitors by ligand-based
virtual screening. J Med Chem
2010;53:8770-4
226. Sheridan C. SGLT 2 inhibitors raceto
enter type-2 diabetes market.
Nat Biotechnol 2012;30:899-900
227. Shing T K, Ng WL, Chan JY, Lau CB.
Design, syntheses, and sar studies of
carbocyclicanalogues of sergliflozin as
potent sodium-dependent glucose
cotransporter 2 inhibitors. Chem I nt
Ed Engl
2013;published online19 Jun 2013;
doi:10.1002/anie.201302543
228. Gao Y, Z hao G, Liu W, et al.
T hiadiazole-based thioglycosides as
sodium-glucoseco-transporter 2 (sglt2)
inhibitors. Chin J Chem 2010;28:605-12
229. Liao C, Sitzmann M, PuglieseA,
Nicklaus MC. Softwareand resources for
computational medicinal chemistry.
FutureMed Chem 2011;3:1057-85
230. Liu L, MaY, Wang RL, et al. Find
novel dual-agonist drugs for treating
type2 diabetes by means of
cheminformatics. DrugDes Devel T her
2013;7:279-88
231. KitadaM, KumeS, Kanasaki K, et al.
Sirtuins as possibledrug targets in

Import ance of synt het ic drugs f or T2DM drug discovery

type2 diabetes. Curr Drug T argets


2013;14:622-36
232. MilneJC, Lambert PD, Schenk S, et al.
Small moleculeactivators of SIRT 1 as
therapeutics for thetreatment of
type2 diabetes. Nature2007;450:712-16

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Serials Unit - Library on 11/04/13
For personal use only.

233. Kun S, Nagy GZ , T oth M, et al.


Synthesis of variously coupled conjugates
of D-glucose, 1,3,4-oxadiazole, and
1,2,3-triazolefor inhibition of glycogen
phosphorylase. Carbohydr Res
2011;346:1427-38
234. Panzhinskiy E, Ren J, Nair S.
Pharmacological inhibition of protein
tyrosinephosphatase1B: apromising
strategy for thetreatment of obesity and
type2 diabetes mellitus.
Curr Med Chem 2013;20:2609-25
235. Patel D, Jain M, Shah SR, et al.
Discovery of potent, selectiveand orally
bioavailabletriaryl-sulfonamidebased
PT P1B inhibitors. Bioorg Med
Chem Lett 2012;22:1111-17
236. LipskaKJ, Bailey CJ, Inzucchi SE. Use
of metformin in thesetting of
mild-to-moderaterenal insufficiency.
Diabetes Care2011;34:1431-7
237. BecicF, KapicE, BecicE.
Glimepiride--an oral antidiabeticagent.
Med Arh 2003;57:125-7
238. Clissold SP, Edwards C. Acarbose.
A preliminary review of its
pharmacodynamicand pharmacokinetic
properties, and therapeuticpotential.
Drugs 1988;35:214-43
239. Madsbad S. Liraglutideeffect and action
in diabetes (LEAD ) trial. Expert Rev
Endocrinol Metab 2009;4:119-29

240. PaninaG. T heDPP-4 inhibitor


vildagliptin: robust glycaemiccontrol in
type2 diabetes and beyond.
Diabetes Obes Metab 2007;9:32-9
241. Rosenstock J, Aggarwal N, Polidori D,
et al. Dose-ranging effects of
canagliflozin, asodium-glucose
cotransporter 2 inhibitor, as add-on to
metformin in subjects with
type2 diabetes. Diabetes Care
2012;35:1232-8
244. Barnett A. DPP-4 inhibitors and their
potential rolein themanagement of
type2 diabetes. I nt J Clin Pract
2006;60:1454-70
245. T ahrani AA, PiyaMK, Barnett AH .
Saxagliptin: anew DPP-4 inhibitor for
thetreatment of type2 diabetes mellitus.
Adv T her 2009;26:249-62
243. Kim D, Wang L, Beconi M, et al. (2R)4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro
[1,2,4]triazolo[4,3-a]pyrazin-7(8H )- yl]1-(2,4,5-trifluorophenyl)butan-2-amine:
apotent, orally activedipeptidyl
peptidaseI V inhibitor for thetreatment
of type2 diabetes. J Med Chem
2005;48:141-51
246. Villhauer EB, Brinkman JA, Naderi GB,
et al. 1-[[(3-hydroxy-1-adamantyl)amino]
acetyl]-2-cyano-(S)-pyrrolidine: apotent,
selective, and orally bioavailable
dipeptidyl peptidaseI V inhibitor with
antihyperglycemicproperties.
J Med Chem 2003;46:2774-89
247. Feng J, Z hang Z , WallaceMB, et al.
Discovery of alogliptin: apotent,
selective, bioavailable, and efficacious

Expert Opin. Drug Discov. (2013) 8(11)

inhibitor of dipeptidyl peptidaseIV.


J Med Chem 2007;50:2297-300
248. T homas L, Eckhardt M, Langkopf E,
et al. (R)-8-(3-amino-piperidin-1-yl)-7but-2-ynyl-3-methyl-1-(4-methylquinazolin-2-ylm ethyl)-3,7-dihydropurine-2,6-dione(BI 1356), anovel
xanthine-based dipeptidyl peptidase
4 inhibitor, has asuperior potency and
longer duration of action compared with
other dipeptidyl peptidase-4 inhibitors.
J Pharmacol Exp T her 2008;325:175-82
242. Metzler WJ, Yanchunas J, Weigelt C,
et al. I nvolvement of DPP-IV catalytic
residues in enzyme-saxagliptin complex
formation. Protein Sci 2008;17:240-50
249. Katsuno K, Fujimori Y, T akemuraY,
et al. Sergliflozin, anovel selective
inhibitor of low-affinity sodium glucose
cotransporter (SGLT 2), validates the
critical roleof SGLT 2 in renal glucose
reabsorption and modulates plasma
glucoselevel. J Pharmacol Exp T her
2007;320:323-30

Affiliation
Maliheh Safavi 1, AlirezaForoumadi 1 &
Mohammad Abdollahi 1,2

Author for correspondence


1
T ehran University of Medical Sciences,
Faculty of Pharmacy andPharmaceutical Sciences
Research Center, T ehran, Iran
2
Professor and Dean, T ehran University of
Medical Sciences, Faculty of Pharmacy and
pharmaceutical Sciences Research Center,
Department of T oxicology and Pharmacology,
T ehran 1417614411, Iran
E-mail: Mohammad@T UMS.Ac.Ir

1363

Vous aimerez peut-être aussi