You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/280060532

Antidiabetic Drugs: Mechanisms of Action and Potential Outcomes on Cellular


Metabolism

Article  in  Current Pharmaceutical Design · July 2015


DOI: 10.2174/1381612821666150710145753 · Source: PubMed

CITATIONS READS
21 15,052

6 authors, including:

Maria João Meneses Branca Maria Silva


Universidade NOVA de Lisboa Universidade da Beira Interior
23 PUBLICATIONS   77 CITATIONS    161 PUBLICATIONS   4,073 CITATIONS   

SEE PROFILE SEE PROFILE

Rosália Sá Pedro F Oliveira


University of Porto University of Aveiro
80 PUBLICATIONS   795 CITATIONS    464 PUBLICATIONS   4,185 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Investigation of the molecular and cellular process involved in Selenium and a new form of Thioamide ability to prevent Doxorubicin toxicity in Sertoli and sperm cells View
project

ATIVIDADE FÍSICA, ESTILO DE VIDA E PROMOÇÃO DA SAÚDE View project

All content following this page was uploaded by Maria João Meneses on 16 September 2015.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.ae
3606 Current Pharmaceutical Design, 2015, 21, 3606-3620

Antidiabetic Drugs: Mechanisms of Action and Potential Outcomes on Cellular


Metabolism

Maria J. Meneses1,2, Branca M. Silva1, Mário Sousa2,3, Rosália Sá2, Pedro F. Oliveira1,2 and Marco G. Alves1*

1
CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; 2Department of
Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Insti-
tute of Biomedical Sciences, University of Porto – UMIB/ICBAS/UP; 3Centre for Reproductive Genetics Professor
Alberto Barros, Porto, Portugal

Abstract: Diabetes mellitus (DM) is one of the most prevalent chronic diseases and has been a leading cause of
death in the last decades. Thus, methods to detect, prevent or delay this disease and its co-morbidities have long
been a matter of discussion. Nowadays, DM patients, particularly those suffering with type 2 DM, are advised to
alter their diet and physical exercise regimens and then proceed progressively from monotherapy, dual therapy, and
multi-agent therapy to insulin administration, as the disease becomes more severe.
Although progresses have been made, the pursuit for the “perfect” antidiabetic drug still continues. The complexity
of DM and its impact on whole body homeodynamics are two of the main reasons why there is not yet such a drug. Moreover, the mo-
lecular mechanisms by which DM can be controlled are still under an intense debate. As the associated risks, disadvantages, side effects
and mechanisms of action vary from drug to drug, the choice of the most suitable therapy needs to be thoroughly investigated. Herein we
propose to discuss the different classes of antidiabetic drugs available, their applications and mechanisms of action, particularly those of
the newer and/or most widely prescribed classes. A special emphasis will be made on their effects on cellular metabolism, since these
drugs affect those pathways in several cellular systems and organs, promoting metabolic alterations responsible for either deleterious or
beneficial effects. This is a crucial property that needs to be carefully investigated when prescribing an antidiabetic.
Keywords: Antidiabetics, cell metabolism, diabetes mellitus, drug therapy, insulin resistance.

INTRODUCTION treat other diseases, particularly obesity, which is a major cause for
In the last decades, the number of diabetic patients has alarm- the establishment of diabetes mellitus (DM). Thus, the complexity
ingly increased, particularly due to the increased rates of type 2 of compounds available, their modes of action and their biological
diabetes mellitus (T2DM). Besides the health problems, this is as- activities are hot topics of debate for multiple areas of the human
sociated with severe economic and sociologic problems. The treat- health, particularly for cardiovascular, renal, neurologic and even
ment of diabetic individuals involves high costs and the amount of cancer pathologies.
money spent every year in their treatment is increasing. There are DM is a multifactorial disease, which indicates that a complex
several antidiabetic drugs in the market that can be used either in analysis is needed when searching for new targets to treat this dis-
monotherapy or in combination. The mechanisms of action for each ease or the mode of action of compounds with potential antidiabetic
compound are different and may vary depending on several condi- activity. Moreover, several of these compounds, if not all, have the
tions, including the doses. The antidiabetic drugs aim to control ability to alter cellular metabolism in a way that may be of benefit
glucose metabolism and, in a non-specialized approach, we can in some organs, but cause damage in others. This is a tricky prob-
affirm that their gold objective is to lower blood glucose levels. lem that hampers the development of new drugs and obliges to a
Therefore, most of their mechanisms of action are intimately linked constant monitoring of how the patients take each compound alone
with glucose metabolism. Unfortunately, most of these compounds or how combination therapies may evolve. In the last years, thou-
compensate loss of insulin sensitivity, of insulin action or of insulin sands of compounds have emerged and there is an extensive litera-
secretion, as well as other mechanisms responsible for the disease, ture on each. Herein we propose to discuss the mode of action of
but are unable to avoid or treat some of the deleterious effects. In the most used families of antidiabetic drugs and how they are cur-
addition, this is a field of research in constant change with the de- rently used. Since these compounds act on insulin sensitivity, insu-
velopment of new products and intense research. lin action or insulin secretion, they also alter cellular metabolism.
Glycemic control in diabetic patients is sometimes quite diffi- Thus, we also aim to discuss some mechanisms by which these
cult to attain. Therefore, it is urgent that the scientific community compounds act on modulating cells metabolism.
provides new and better options to improve the use of the current
DIABETES MELLITUS IN BRIEF
available drugs and to highlight the most suitable therapies. Of
particular relevance is the study of the pathophysiological altera- DM is one of the most prevalent chronic diseases and a leading
tions that diabetic individuals suffer and how the available drugs cause of morbidity and mortality on developed and in developing
may affect those processes. There are several options for the treat- countries. It is estimated that 382 million of people have DM and,
ment of diabetic patients, with distinct modes of action. Notably, according to a recent report, the number of diabetic patients will
some of those compounds were developed or are in development to reach 592 million by the year 2035 [1]. Therefore, it urges the study
of this pathology and its consequences to cells, organs and whole
body. DM is described as a group of metabolic disorders character-
*Address correspondence to this author at the CICS-UBI, Health Sciences ized by chronic hyperglycemia resulting from defects in insulin
Research Centre, University of Beira Interior, Av. Infante D. Henrique,
6201-506 Covilhã, Portugal; Tel: +351 275329077; Fax: +351 275329099;
action, insulin secretion, or both [2]. This deficiency leads to dis-
E-mail: alvesmarc@gmail.com turbances in the metabolism of carbohydrates, fat and protein,

1873-4286/15 $58.00+.00 © 2015 Bentham Science Publishers


Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3607

which causes systemic complications and co-morbidities namely complex carbohydrates present in the meal [22]. Importantly, it
cardiovascular and renal failure. The most predominant types of presents some adverse effects. The most common are flatulence,
DM are type 1 diabetes mellitus (T1DM) and T2DM. T1DM is a diarrhoea, and abdominal discomfort caused by altered bacterial
chronic autoimmune disease caused by the pathogenic action of T metabolism of disaccharides in the colon [23, 24]. Some other dis-
lymphocytes on insulin-producing -cells [3]. This leads to a dra- advantages include negligible effect on cholesterol, and potential
matic reduction or even elimination of insulin production. Thus, elevation in liver hepatic enzymes. Presently, there are three avail-
patients with T1DM are dependent on exogenous insulin to survive able -glucosidase inhibitors used as antidiabetic drugs: acarbose
[4]. Typically, the symptoms of T1DM appear in patients with less [25], miglitol [26, 27] and voglibose [23, 28]. While acarbose is
than 30 years, being known as juvenile-onset diabetes. The inci- widely available, miglitol was approved by Food and Drug Admini-
dence of T1DM has been globally rising and, if present trend con- stration and voglibose is only available in Japan.
tinues, it is predicted a doubling of new cases in European children Acarbose was the first -glucosidase inhibitor described [29]
younger than 5 years between 2005 and 2020. Additionally, it is and is the most prescribed one [30]. It is a pseudotetrasaccharide of
expected that the prevalence of cases in individuals younger than 15 microbial origin (Actinoplanes utahensis) [31], consisting of a mal-
years of age will rise by 70% during that period [5]. tose molecule linked to acarvosine with a nitrogen bound between
T2DM is characterized by hyperglycemia that results from insu- the first and second glucose unit [19, 20, 27]. This modification of a
lin resistance plus variable degrees of insufficient insulin secretion natural tetrasaccharide is important for its high affinity for active
[2]. Although new lifestyles of modern societies seem to be the centres of -glucosidases of the small intestine brush border, as
triggering pathogenic elements, genetic factors are also reported to well as for its stability [22]. Acarbose is mostly effective against
be involved in the pathogenesis of T2DM [6]. In early stages of the glucoamylase, being less effective against sucrase, maltase, and
disease, insulin production is disproportionately low for the degree dextrinase [32]. It also inhibits -amylase, but has no effect on -
of insulin sensitivity, which is usually increased. Thus, the ability of glucosidases [20]. Absorption of acarbose is negligible because it is
pancreatic -cells to produce adequate amounts of insulin in post- mostly degraded by intestinal bacteria and by amylases present in
prandial glycemia is profoundly compromised. These cells’ func- the small intestine [27, 33]. The recommended dose is 50 mg three
tional inability is the main determinant of hyperglycemia and is times a day, before the main meals, increasing until a maximum
known to progress over time [7]. As a result, most patients with dose of 100 mg three times daily [20].
T2DM will gradually need more complex therapeutic procedures to Miglitol is the first pseudomonosaccharide -glucosidase in-
control hyperglycemia. Normally, the alterations start in diet and hibitor derived from 1-deoxynojirimycin and presents a structure
exercise regimens and progress to monotherapy, dual therapy, or very similar to glucose [34]. Unlike acarbose, it is almost com-
multi-agent therapy followed by insulin administration in combina- pletely absorbed in the upper section of the small intestine, has low
tion or not with other antidiabetic drugs [8]. Thus, the demand for tissue penetration and is excreted unchanged by the kidneys [35]. It
antidiabetic drugs is very high and new products are regularly pre- is mostly effective against sucrase, inhibiting also glucoamylase,
sented. Although its safety is assured, these drugs have multiple isomaltase, lactase and trehalase [36]. The normal administration of
modes of action and effects throughout the body. Furthermore, the miglitol is usually 50 mg though it may be increased to 100 mg
complexity of T2DM led to the establishment of an intermediate three times per day after a few weeks, which is the maximal rec-
state, often called as pre-diabetes. In this prodromal stage of T2DM, ommended daily dosage [20, 27].
the patients present glycemic values higher than normal, but lower
than T2DM thresholds [9, 10]. Moreover, pre-diabetic patients pre- Voglibose is a valiolamine derivative [37], which is produced
sent impaired fasting glycemia and/or impaired glucose tolerance by Streptomyces Hygroscopicus [38]. Similarly to acarbose, vogli-
[11]. This pre-diabetic condition may be triggered by sedentary bose is slowly and poorly absorbed, being rapidly excreted [39].
lifestyle and the ingestion of high fat and saturated fatty acid diets Like the other -glucosidases inhibitors [40], voglibose also stimu-
[11, 12]. As a consequence, several metabolic changes in organs lates an increase of endogenous glucagon-like peptide-1 (GLP-1),
and cellular systems occur [13]. These patients have increased risk an incretin hormone with antihyperglycemic properties [39, 41].
for T2DM as well as for cardiovascular disease [14]. However, this Voglibose should be administered in a single oral dose of 0.2 mg
pre-diabetic stage does not imply the progression to T2DM, which three times a day and, if not sufficient, the dose can be uptitrated to
can be prevented or delayed by lifestyle and drug-based interven- 0.3 mg [20, 41].
tions [15, 16] illustrating the relevance of an early diagnostic and of Biguanides
the study of this prodromal stage of T2DM.
Several glucose-lowering guanidine derivatives were intro-
ANTIDIABETIC DRUGS duced in the 1920s, due to the finding that Galega officinalis, a
traditional herb historically used as treatment for DM, was rich in
Inhibitors of -Glucosidases guanidine [42]. These agents were almost forgotten as insulin be-
Carbohydrates are central constituents of western diets [17]. came widely available and used [43]. It was not until the 1950s that
Complex carbohydrates are enzymatically degraded to monosac- biguanides were re-investigated for the treatment of DM. In the late
charides by the action of -galactosidases [18], -amylase and - 1950s, three biguanides with antidiabetic action were reported:
glucosidases [19]. Thus, inhibitors of intestinal -glucosidase en- phenformin [44], buformin [43] and metformin [45]. The use of
zymes modulate the rate of digestion of complex carbohydrates and phenformin and buformin has been discontinued in many countries
disaccharides by competitively and reversibly inhibiting - due to a high incidence of lactic acidosis [46], leaving metformin as
glucosidases present in the brush border membrane of enterocytes the main biguanide used worldwide [43, 47]. Indeed, metformin has
that line the intestinal villi [19, 20]. Consequently, the digestion of been used to treat T2DM for over 50 years. It is usually described
carbohydrates and absorption of monosaccharides in the proximal as the first line treatment to this disorder, along with diet and exer-
jejunum are decreased or incomplete in the distal jejunum and il- cise [48]. Metformin is an insulin-sensitizing drug that exerts its
eum (Table 1). Therefore, the rise in postprandial plasma glucose antihyperglycemic effects by blocking liver gluconeogenesis [49]
levels is diminished and/or delayed. -glucosidase inhibitors allow through regulation of the gluconeogenic flux, rather than direct
the pancreatic -cell more time to augment insulin secretion in re- inhibition of gluconeogenic gene expression [50]. It also increases
sponse to the increase in plasma glucose level [21, 22]. Due to its skeletal muscle uptake of glucose [51] and reduces the absorption
mechanism of action, -glucosidase inhibitors should be taken at of glucose in the intestinal mucosa [52] (Table 1). Despite almost
the beginning of the main meals. Notably, its effectiveness in de- 60 years of research, the exact mechanisms of metformin action
creasing postprandial glycemia will be determined by the amount of remain to be unveiled [53]. Nevertheless, it is known that met-
3608 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

formin has no effect on stimulating insulin secretion in pancreatic duration of the glucose lowering effect of repaglinide is shorter than
-cell [43]. In recent years, studies provided evidence that met- that of some sulfonylureas. Consequently, the risk of having hypo-
formin acts as an inhibitor of complex I of the electron transport glycemia is lower [82]. Repaglinide should be taken before each
chain [54, 55], inducing activation of AMP-activated protein kinase meal at least twice daily and the maximum daily dosage is 16 mg
(AMPK) sensitive signalling [51]. AMPK acts as a regulator of [83]. After oral administration, repaglinide is rapidly absorbed [84]
glucose and lipid metabolism, as well as of cellular energy, through and has a bioavailability of approximately 63% [85]. It is metabo-
phosphorylation of some key proteins [56]. The increase in its ac- lised in the liver [86] to inactive metabolites and is predominantly
tivity results in several biological alterations including: the stimula- excreted via the bile into the faeces, thus being a viable option for
tion of glucose uptake in muscle; fatty acid oxidation in liver and patients with renal failure [84, 87]. However, cautions should be
muscle; inhibition of hepatic glucose production, cholesterol and taken in patients with liver disease since the pharmacokinetics of
triglyceride synthesis; and lipogenesis [57]. Another suggested this drug may be significantly modified [88]. Protein binding can be
effect for metformin is that it increases plasma levels of GLP-1, an reduced due to administration of other drugs. Although it can be
incretin hormone with antihyperglycemic properties, and induces used in combination with metformin [76] or thiazolidinediones
islet incretin receptor gene expression, through a mechanism that is [89], patients receiving multiple drugs must be carefully monitored
dependent on peroxisome proliferator-activated receptor  (PPAR) [61].
[58]. Metformin has a main advantage over other biguanides, which Nateglinide, a D-phenylalanine derivative [90], quickly and
is the very low probability to promote lactic acidosis [59]. How- reversibly binds to the SUR 1 like sulfonylureas and has specificity
ever, it has some disadvantages, such as adverse gastrointestinal for pancreatic SURs [80]. Thus, the binding site differs from that of
effects [59]. Although considered the first-line pharmacological repaglinide. Nateglinide inhibits the ATP-dependent potassium
agent for T2DM individuals, in many patients the administration of channels faster than repaglinide but, on the other hand, there is a
this drug is insufficient to reach glycemic control and a second more rapid reversal of the inhibition of the channel when taking
agent is added to the treatment [20, 60, 61]. nateglinide in comparison with repaglinide [91]. Like repaglinide,
nateglinide is effective in combination with metformin [92] or thia-
Sulfonylureas
zolidinediones [93]. It should be taken before each meal (60 mg)
An incidental observation of hypoglycemia episodes during regardless of the meal composition [94]. Nateglinide is rapidly
treatment with sulfonamides in the 1940s, led to the concept and absorbed in a dose-dependent manner and is mostly excreted in
development of sulfonylureas as agents to stimulate insulin secre- urine [95].
tion [62]. By 1955, sulfonylureas became the first pharmacological
option to treat non-insulin-dependent DM, besides insulin injections GLP-1 Receptor Agonists and DPP-4 Inhibitors
[63]. The first sulfonylureas developed were tolbutamide, chlor- In the early 1990s, it was reported that the incretin hormone
propamide, acetohexamide and tolazamide [64, 65]. These first- GLP-1 could be a potential target in the treatment of T2DM due to
generation agents have been largely set aside by the newer second- its antihyperglycemic properties, in contrast to another incretin
generation agents gliclazide, glipizide and glibenclamide (gly- glucose-dependent insulinotropic polypeptide (GIP) [96]. GLP-1 is
buride) [65]. The most recent sulfonylurea, glimepiride, is consid- secreted from endocrine L-cells [97] in the small intestine and is
ered by some researchers as a second generation of sulfonylureas released after meal ingestion, especially those rich in fats and car-
[64], while others classify it as third-generation [66]. Sulfonylureas bohydrates [98]. Once released, GLP-1 exerts direct effects in sev-
perform their secretagogue action by activating -cell sulfonylurea eral organs, due to interaction with its receptor located in organs
receptor 1 (SUR 1), leading to closure of ATP-dependent potassium such as pancreas, brain, heart, lung, stomach, intestine, and kidney
channels [67, 68] (Table 1). Thus, the potassium flow across the [99]. On pancreatic -cells, this interaction leads to activation of
plasma membrane is stopped leading to depolarization, which opens adenylate cyclase and production of cAMP, that mediates its stimu-
voltage sensitive calcium channels. Consequently, there is an up- latory effect on insulin secretion via protein kinase A [100]. How-
take of extracellular calcium, activating a cytoskeletal system, ever, GLP-1 stimulates insulin secretion via several other mecha-
which causes translocation of secretory granules to the cell surface nisms, including a direct inhibition of ATP-dependent potassium
and extrusion of insulin through exocytosis [69, 70]. Exocytosis channels. As sulfonylureas, this leads to an increase in intracellular
results in the fusion of the secretory granule with the plasma mem- levels of calcium and a rise in mitochondrial ATP synthesis which
brane, leading to the release of insulin into the extracellular space to leads to a further membrane depolarization. Finally, it causes insu-
reach the capillary blood flow [71, 72]. Hence, sulfonylureas ad- lin granule exocytosis from pancreatic -cells [101]. It was previ-
ministration must be carefully monitored since it can lead to hypo- ously shown that GLP-1 inhibits gastric emptying [102] and in-
glycemia, weight gain [73] and hyperinsulinemia [59]. duces satiety [103] (Table 1). Furthermore, it may also increase -
cell mass, through inhibition of apoptosis and promotion of
Meglitinides
neogenesis and proliferation. Thus, GLP-1 has several effects that
Meglitinides are insulin secretagogues with a similar mecha- are of potential value in the treatment of T2DM. Still, GLP-1 is
nism of action to sulfonylureas, acting on ATP-dependent potas- rapidly degraded and inactivated by the enzyme dipeptidyl pepti-
sium channels [61] (Table 1). Therefore, they fail to have any effect dase-4 (DPP-4) [104]. DPP-4 cleaves the two N-terminal amino
in patients who have already been treated with maximal therapeutic acids of GLP-1, and the resultant metabolite lacks the insulinotropic
dosage of sulfonylureas [74]. Considering this, they offer an alter- and glucagonostatic effects of GLP-1. Consequently, two therapeu-
native to sulfonylurea treatment, but have almost the same disad- tic strategies have emerged to take advantage of the beneficial ef-
vantages and a more complex dosing schedule [59]. To date, three fects of GLP-1: the use of GLP-1 receptor agonists, which are more
meglitinides have been used in clinical practice: nateglinide [75], resistant to the action of DPP-4 [105]; and the development of DPP-
repaglinide [76] and mitiglinide [77]. 4 inhibitors, which prevent inactivation of GLP-1 [106, 107].
Repaglinide, a carbamoylmethyl benzoic acid derivative, was DPP-4 inhibitors competitively and reversibly inhibit DPP-4
the first meglitinide analogue to become available for clinical use providing up to 90% inhibition of its activity during a 24-hour pe-
[78]. It promotes insulin secretion by closing ATP-dependent potas- riod [108]. Therefore, DPP-4 inhibitors promote insulin secretion
sium channels in the membrane of pancreatic -cells, but is ineffec- and inhibit glucagon secretion [109], being these actions dependent
tive in the absence of extracellular calcium [79]. Repaglinide binds on the presence of glucose (Table 1). DPP-4 inhibition also in-
to a nearby location of the receptor site for sulfonylureas drugs creases -cell mass, as demonstrated in rodent models of T2DM
[80], but generates a more rapid reaction [81]. Nevertheless, the [110, 111]. In vitro and animal studies have revealed that GLP-1
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3609

also increases -cell mass by stimulating islet cell neogenesis and blood glucose-lowering effect evolves gradually over a period of
by inhibiting apoptosis of islets [112, 113]. Thus, the pharmacol- weeks in a dose-dependent manner [132]. Pioglitazone administra-
ogical potential of these drugs is very relevant. Since 2006, there tion must be started at 15 mg once a day to a maximum daily dos-
are some DPP-4 inhibitors being marketed, namely sitagliptin, vil- age of 45 mg [121]. Although pioglitazone is not reported to cause
dagliptin, saxagliptin, linagliptin, anagliptin, teneligliptin and alo- hepatoxicity, it is debated if it might elevate the risk of bladder
gliptin. Although they share the same mechanism of action, they are cancer through an unknown mechanism [133]. Some evidences
a heterogeneous group that diverges in terms of half-life, systemic suggest an effect associated with crystal formation and bladder
exposure, bioavailability, protein binding, metabolism, presence of irritation, rather than a pharmacologic effect through PPAR [126].
active metabolites and excretion routes [114, 115]. These differ- However, there is some controversy since other studies reported
ences are extremely important when considering patients that be- that pioglitazone should not be associated with an increased risk of
sides T2DM have renal or hepatic impairment, as well as when bladder cancer [134]. Notably, pioglitazone is reported to possess
combined therapy is considered. multiple beneficial effects on lipid metabolism [135], immune func-
Importantly, these two drug classes present some interesting tion as well as in endothelial function [136].
differences that must be taken into consideration when choosing the Rosiglitazone belongs to the thiazolidinediones class but has a
antidiabetic class to use. While DPP-4 inhibitors allow the increase different side chain from those of troglitazone and pioglitazone
of GLP-1 physiological levels, GLP-1 receptor agonists increase the [137]. It reaches a 99% oral bioavailability and is extensively me-
pharmacological levels of GLP-1 leading to different side effects of tabolised in the liver [138]. Thus, rosiglitazone is contraindicated
these classes [105, 108]. for patients with liver disease. The major excretion routes of rosigli-
tazone are via the urine and faeces [138]. To achieve a significant
Thiazolidinediones antihyperglycemic efficacy, rosiglitazone should be given once or
Thiazolidinediones, also termed glitazones, are a class of oral twice a day at a starting dosage of 4 mg/day, which may be in-
antidiabetic agents that were originally developed in the early 1980s creased, if required, to 8 mg/day, in combination with diet and ex-
as antioxidants [116]. The blood glucose-lowering potential of this ercise [139]. Intriguingly, the risk of bladder cancer appears to be
class of drugs was observed after the synthesis of ciglitazone, the specifically associated with pioglitazone, and not with rosiglitazone
first thiazolidinedione. This effect was particularly pronounced in [140]. At a cardiovascular level, it has been suggested, based on a
animals with genetic insulin resistance [117]. Thiazolidinediones systematic review and meta-analysis of observational studies, that
were considered insulin sensitizers [118], as biguanides. This was rosiglitazone presents a higher risk of complications (e.g. conges-
established after observing that glycemia improved in the absence tive heart failure and myocardial infarction) when comparing with
of increasing insulin levels and that it did not affect insulin- pioglitazone [141].
deficient animals. However, due to liver toxicity, ciglitazone and
englitazone were never subjected to clinical studies [119]. Troglita- Sodium-Dependent Glucose Co-Transporter 2 (SGLT2) Inhibi-
zone, the first marketed thiazolidinedione, was introduced in Japan tors
and USA in 1997, but was subsequently withdrawn due to hepatox- Sodium-dependent glucose co-transporters (SGLTs) are a large
icity side effects [120]. Today, two thiazolidinediones are available class of proteins that facilitate the transport of sugars and sodium
for clinical use: rosiglitazone and pioglitazone. These drugs are across the plasma membrane of cells from an extensive variety of
very similar in their effect on hyperglycemia, side effects and tissues [142]. More specifically, two members of SGLT family
mechanism of action. Interestingly, both are also available in com- mediate reabsorption of glucose in the kidney. SGLT2 is a high-
bination with other antidiabetic drugs such as metformin or glime- capacity and low-affinity transporter, responsible for nearly 90% of
piride [121]. Importantly, an adverse effect of this class is fluid the active renal glucose reabsorption, while SGLT1 reabsorbs the
retention, making thiazolidinediones contraindicated in patients remaining 10% [143]. SGLTs promote reabsorption of glucose
with heart failure, which is one of the leading causes of death from the proximal tubules, which is then passively diffused into the
among T2DM patients [33] (Table 1). In addition, the therapeutic circulation via glucose transporters (GLUTs). SGLT2 is almost
effect is only observed after 3 to 4 months of therapy, having a slow exclusively expressed in renal proximal tubules, unlike SGLT1 that
onset of action compared with the other available pharmacological is also found in the gastrointestinal tract, and its inhibition is there-
treatments [59]. fore unlikely to affect other organs [144]. Thus, inhibition of
Thiazolidinediones are potent synthetic activators of the nuclear SGLT2 limits renal glucose reabsorption, promoting its urinary
receptor peroxisome proliferator-activated receptor  (PPAR) excretion and the reduction of plasma glucose levels (Table 1).
[122]. PPAR is abundantly expressed in key target tissues for insu- Thereby, SGLT2 inhibitors use a novel mechanism of action, since
lin action such as adipose tissue, but is also present in muscle, liver, they do not interfere with insulin secretion [145].
endothelium and pancreatic -cells [123]. It heterodimerizes with Phlorizin has played a pivotal role in clarifying the mechanism
retinoid X receptor and binds to nuclear responsive elements, thus of renal glucose reabsorption. It is a potent inhibitor of both,
modulating the transcription of genes that play a role in the metabo- SGLT1 and SGLT2, through a nonselective inhibition [146]. How-
lism of glucose and lipids [124]. Notably, this stimulation promotes ever, subsequent studies have shown that it cannot be used as an
differentiation of pre-adipocytes, which enhance local effects of antidiabetic therapy due to a poor intestinal absorption and, conse-
insulin. It is also discussed that signals resultant from the adipose quently, a low bioavailability [146]. Moreover, phlorizin also suf-
tissue, e.g adiponectin or leptin, may mediate the improvement in fers -glucosidase degradation and hydrolysis in the gut, producing
skeletal glucose disposal induced by thiazolidinediones [125]. phloretin [143]. This metabolite inhibits glucose transporters,
Pioglitazone, a potent PPAR agonist [126], increases insulin- namely GLUT2, leading to gastrointestinal side effects, such as
stimulated glucose uptake in peripheral tissues as well as insulin diarrhoea, and impairment in glucose absorption [147].
sensitivity in hepatic and adipose tissue [127]. It also causes a mi- Dapagliflozin was the first SGLT2 inhibitor to be approved for
nor activation of PPAR, which is related with anti-inflammatory clinical use in adults with T2DM by European Medicine Agency
effects, as well as with the decrease of plasma triglyceride levels and the second to be approved by Food and Drug Administration. It
[128]. Pioglitazone has an oral bioavailability of approximately presents resistance to -glucosidase in the gastrointestinal tract,
83%, which is not modified by the presence of food on the gastroin- which makes it appropriate for oral administration. In addition, it
testinal tract [129]. Furthermore, it is rapidly absorbed and exten- has 1200-fold selectivity for SGLT2 over SGLT1 [148]. An in vitro
sively metabolized by hydroxylation and oxidation in the liver, study showed that dapagliflozin exhibited approximately 30 times
forming active and inactive metabolites [130, 131]. Pioglitazone greater potency versus SGLT2 than phlorizin, and about 4fold less
3610 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

potency versus phlorizin against SGLT1 [149]. By reducing the metabolic pathways involved in processes related to conservation of
tubular glucose reabsorption, dapagliflozin results in urinary glu- energy from glucose to ATP. First, the glucose is oxidized through
cose excretion [150]. Moreover, total urinary glucose losses are glycolysis, resulting in two molecules of pyruvate [168] (Fig. 1).
proportional to glucose concentration in plasma, so that glucose The first limiting step of glycolysis is usually described as being
elimination in urine is greater in patients with hyperglycemia. Usu- mediated by the irreversible conversion of fructose-6-phosphate to
ally, 10 mg of dapagliflozin per day will increase the amount of fructose-1,6-biphosphate [169]. This reaction is catalysed by phos-
glucose excreted in the urine of a patient with T2DM by 50–80 phofructokinase 1, whose activity is also known to be related with
g/day [151]. Interestingly, it can be used as monotherapy, or in the energy status of the cell [168, 170]. Pyruvate may then follow
combination with other antidiabetic drugs, such as metformin [152]. one of three pathways [171]. One of those is the Krebs cycle, being
necessary for the entry of pyruvate into the mitochondria [172]. In
Exogenous Insulin the Krebs cycle, which originates six molecules of carbon dioxide,
Insulin is a hormone secreted by the pancreatic -cells into the NAD+ and FAD+ become reduced and carry electrons to the respira-
blood, in response to increased glucose concentration [153]. Be- tory chain [173]. Here, energy is conserved in ATP and GTP mole-
sides the direct involvement with glucose metabolism (Table 1), cules. Another pathway is the conversion of pyruvate to lactate by
insulin has a controlling influence on the metabolism of fats and lactate dehydrogenase [171] and the last one is the conversion of
proteins [154]. When the body is unable to produce insulin or de- the end product of glycolysis into alanine by alanine aminotrans-
velops insulin resistance, one of the possible treatments is the ad- ferase [173]. Lactate was, for several times, considered a dead-end
ministration of an exogenous supply of this hormone [155]. Usu- waste product of glycolysis [174]. Nowadays, it is known that lac-
ally, when insulin is injected into the body, its molecules form hex- tate is the main energy source of several cells, including for the
amers with zinc. For diffusion through interstitial fluid and penetra- developing male germ cells [175]. Lactate has also been reported to
tion into the bloodstream, these hexamers need to dissociate into act in wound healing processes through its effect on down-
dimers and monomers [156]. However, pharmacokinetics (follow- regulation of ADP-ribosylation. In cases of injury and sepsis, lac-
ing administration via subcutaneous injection) of the structurally tate accumulation may relate to an adrenaline surge and a resulting
unchanged regular insulin preparations makes it very difficult to stimulation of the sodium-potassium ATPase pump [174]. Another
maintain normoglycemia during a whole day [157]. Therefore, metabolite that has been suggested to play crucial roles beyond the
various types of insulin have been developed with diverse action classic relevance in glycolysis, is alanine. Indeed, alanine is a major
times in accordance with the patient needs: rapid acting insulin; gluconeogenic substrate constituting the glucose-alanine cycle and
short acting insulin; intermediate acting insulin and long acting a transporter of nitrogen generated by protein breakdown back to
insulin. Rapid acting insulin analogues have a molecular structure the liver for excretion as urea [176].
that slightly differs from human insulin, with one or two amino acid
modification. These changes allow them to be absorbed more Krebs Cycle
quickly than human insulin, once they destabilize insulin hexameri- Krebs cycle, also known as citric acid cycle or tricarboxylic
zation [158]. To achieve an optimum level of insulin, these rapid acid cycle, is composed by a series of biochemical reactions where
acting analogues can be used just before meals or immediately after the components form essential connections with crucial pathways
meals [159]. Short acting insulin analogues possess lower propen- such as gluconeogenesis, lipogenesis and amino acid metabolism.
sity to form hexamers than regular insulin, being absorbed more Krebs cycle is essential in all oxidative organisms and provides
quickly. Thus, short acting insulin analogues achieve peak plasma precursors for anabolic processes and reducing factors that drive the
concentrations about twice as high and within approximately half generation of energy [177]. This cycle oxidizes the pyruvate de-
the time compared to regular insulin [160]. These analogues should rived from glycolysis and acetyl-CoA to produce NADH and
be administrated 20 to 30 minutes before meals to cover the insulin FADH2 for ATP synthesis in the respiratory chain [173] (Fig. 1).
needs [159]. Intermediate acting insulin analogues have a reduced The activity of Krebs cycle is mainly controlled by the regulation of
absorption rate compared to regular insulin due to the addition of two enzymes: pyruvate dehydrogenase, which mediates the trans-
protamine or zinc to the insulin preparation, hindering the hexamers formation of pyruvate into acetyl-CoA [178], and isocitrate dehy-
dissociation [159]. These analogues cover insulin needs for about drogenase, which catalyzes the third step of the cycle (oxidative
half the day or overnight [161] and are often combined with rapid decarboxylation of isocitrate, producing alpha-ketoglutarate), while
or short acting insulin [162]. Long acting insulin analogues have the converting NAD+ to NADH in the mitochondria [179].
longest duration of action of all insulin analogues, once they have
an activity duration of up to 36 hours [159]. The basic principle of Lipogenesis
these preparations is shifting the isoelectric point of human insulin Lipogenesis is the pathway in which acetyl-CoA is converted
from pH 5.4 toward more neutral to make insulin less soluble on the into fatty acids (Fig. 1). It is through lipogenesis that simple sugars
administration site. This is achieved by adding positively charged are converted into fatty acids, which are combined with glycerol
amino acids to the regular insulin [163]. and stored as triglycerides. One source of acetyl-CoA is the citrate
produced during the Krebs cycle. It may be then exported from
CELLULAR METABOLISM: GENERAL CONCEPTS AND mitochondria to cytosol by citrate carrier and converted into acetyl-
RELEVANT PATHWAYS TO ANTIDIABETIC DRUG AC- CoA through ATP citrate lyase [180].
TION
Fatty acid synthesis is catalysed by the fatty acid synthase com-
Glycolysis plex [181]. This complex requires malonyl-CoA, a coenzyme A that
is formed by irreversible carboxylation of acetyl-CoA, which com-
Foods containing carbohydrates are digested to glucose, which
bines with the acyl carrier protein to form malonyl acyl carrier pro-
is then absorbed into the bloodstream. Then, glucose enters the cells
tein [182]. The fatty acid synthesis continues in a cyclical serie of
through GLUTs by facilitated diffusion [166, 167]. But there are
reactions to form palmitate. Then, three fatty acid molecules are
other ways for attaining this sugar. When glucose is not available in
combined with glycerol 3-phosphate to be stored as triglycerides.
plasma, it can also be obtained through glycogenolysis (Fig. 1).
Lipogenesis is very responsive to either hormonal or nutritional
Once a glucose molecule has passed from blood into a cell, it is
changes. One of the major nutrients responsible for stimulating
gradually transformed in a controlled sequence of biochemical
lipogenesis is glucose. It is a substrate for lipogenesis by being
steps. These steps, commonly known as metabolic pathways, are
converted to acetyl-CoA and increases lipogenesis by stimulating
supported by enzymes, which may or may not need cofactors de-
the release of insulin and inhibiting the release of glucagon from the
rived from vitamins to function properly. There are three main
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3611

Table 1. Summary of some antidiabetic drugs, its mechanism of action, main advantages and disadvantages.

Antidiabetic class Antidiabetic Drug Mechanism of action Advantages Disadvantages References

-glucosidase Acarbose Inhibit carbohydrates degradation Weight neutral Gastrointestinal side-effects [23-25]
inhibitors Miglitol in intestinal villi Negligible effect on cholesterol
Voglibose Potential elevations in liver
function tests

Biguanides Metformin Block liver gluconeogenesis Long-term safety Adverse gastrointestinal effects [49, 50, 164]
 skeletal muscle uptake of glucose Weight neutral or Probability of lactic acidosis
 the absorption of glucose in the loss Contraindicated for patients
intestinal mucosa Low risk of with liver or heart failure
 plasma levels of GLP-1 hypoglycemia

Sulfonylureas Glicazide  insulin secretion by activating - Long-term safety Risk of hypoglycemia [65-67, 69]
Glibenclamide cell SUR 1 Risk of weight gain
Glimepiride

Meglitinides Nateglinide Binds to -cell SUR 1 Faster insulin Risk of weight gain [59, 61, 77,
Repaglinide response Risk of hypoglycemia 87]

Mitiglinide Complex dosing schedule

GLP-1 receptor Exenatide Binds to GLP-1 receptor, causing: Weight loss Gastrointestinal side-effects [102, 103,
agonists Liraglutide  insulin secretion, delayed gastric Low risk of Administration by subcutane- 105, 112]
emptying, and satiety. hypoglycemia ous injection

DPP-4 inhibitors Sitagliptin  incretin (GIP and GLP-1) con- Weight neutral Gastrointestinal side-effects [109, 110,
Saxagliptin centrations 112, 114]
Alogliptin  insulin secretion
 glucagon secretion

Thiazolidinedi- Pioglitazone Activators of the PPAR Low risk of Risk of weight gain [122, 123,
ones Rosiglitazone  differentiation of pre-adipocytes hypoglycemia Risk of oedema 126]

 insulin sensitivity in muscle, Risk of heart failure


hepatic and adipose tissue
 glucose uptake in peripheral
tissues

SGLT2 inhibitors Dapagliflozin Limits renal glucose reabsorption Weight loss Risk of genitourinary tract [145, 150]
Canagliflozin Low risk of infections
hypoglycemia

Exogenous insulin Rapid acting Activates the insulin receptor Numerous for- Risk of weight gain [159, 165]
Short acting  hepatic glucose output mulations and Risk of hypoglycemia
delivery systems
Intermediate acting Administration by subcutane-
Long acting ous injection

, increase; , decrease; DPP-4, dipeptidyl peptidase-4; GLP-1, glucagon like peptide 1; GIP, glucose-dependent insulinotropic polypeptide; PPAR, peroxisome proliferator-
activated receptor ; SGLT2, sodium dependent glucose co-transporter 2; SUR 1, sulfonylurea receptor 1

pancreas [183]. On the other hand, insulin is perhaps the most im- this pathway was not accomplished until the discovery of coenzyme
portant hormone influencing lipogenesis [180, 183]. One of the A and the recognition that acetyl-CoA is the product of -oxidation
several mechanisms is the recruitment of glucose transporters to the [185].
plasma membrane, increasing the uptake of glucose to the cell Fatty acids are activated to acyl-CoA after transport across the
[183]. plasma membrane. Subsequently, acyl-CoA is transported across
the mitochondrial membrane [186] (Fig. 1). Once inside the mito-
-oxidation chondria, it is dehydrogenated by acyl-CoA dehydrogenase to yield
-oxidation is the process by which fatty acids are oxidized, a 2,3-enoyl-CoA. The latter enters then in a series of steps catalysed
thus providing a source of energy [184]. Although studies on - by mitochondrial trifunctional protein (MTP). MTP is an enzyme
oxidation have begun in the first 1900s, a complete elucidation of complex composed of four alpha-subunits (harboring enoyl-CoA
3612 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

Glucose
GLUT
Lactate

LDH
Glucose Glycolysis Mitochondrion
Glucose-6-P Pyruvate
PFK PDH Fatty Acids
Glucose-1-P ALT
Glycogenolysis Alanine

Glycogen
Lipogenesis
Citrate Acetyl-CoA Fatty Acids
Pyruvate

PDH
Fatty Acyl-CoA
Acetyl-CoA Citrate

-oxidation ACAD

Krebs MTP
Nucleus Acetyl-CoA + 2,3 Enoyl CoA
cycle
Shortened Acyl-CoA

Fig. (1). Representative diagram of some pathways of cellular metabolism: glycogenolysis, glycolysis, Krebs cycle, lipogenesis and -oxidation. Abbreviation:
GLUT, glucose transporter; -P, -phosphate; PFK, phosphofructokinase; LDH, lactate dehydrogenase; ALT, alanine aminotransferase; PDH, pyruvate dehydro-
genase; ACAD, acyl-coenzyme A dehydrogenase; MTP, mitochondrial trifunctional protein.

hydratase and 3-hydroxyacyl-CoA dehydrogenase) and four beta- Kato and collaborators. After 12 weeks of treatment with acarbose
subunits (harboring long-chain 3-ketoacyl-CoA thiolase) [185, or nateglinide, the postprandial endothelial dysfunction in newly
186]. The final product of these steps is a shortened acyl-CoA and diagnosed T2DM patients was improved and acarbose was more
acetyl-CoA (Fig. 1). effective than nateglinide [192]. It was then hypothesized that acar-
bose may improve endothelial function by inhibiting postprandial
MECHANISMS OF ACTION OF ANTIDIABETIC DRUGS hyperglycemia, without increasing postprandial insulin secretion,
AND POTENTIAL IMPLICATIONS ON CELLULAR ME- while nateglinide may improve endothelial function less signifi-
TABOLISM cantly by inhibiting postprandial hyperglycemia with an increase in
postprandial insulin secretion [192]. More studies are needed to
-glucosidase Inhibitors consolidate these assumptions and unveil the relevance of these
Voglibose not only inhibits -glucosidase enzymes, but also mechanisms to cellular metabolism in the several systems through-
increases total GLP-1 plasma levels, both by stimulating the secre- out the body.
tion of GLP-1 from gut L-cells, and by decreasing plasma DPP-4
activity. As a result, a 1.8-fold increase in plasma active GLP-1 Biguanides
levels after voglibose treatment has been reported in db/db mice, an One of the most prescribed and used antidiabetic drugs is met-
animal model of T2DM [187]. Moreover, voglibose (1-4mM) is formin, however its exact mechanism of action remains unknown.
reported to possess a strong inhibitory effect on melanin production Interestingly, some studies discuss possible pathways of action for
in melanoma cells by blocking the proper N-glycan processing of this drug and a common point in the majority of them is AMPK
tyrosinase, a glycoprotein involved in melanogenesis, resulting in a [51, 164, 193]. Studies showed data consistent with a model where
dramatic reduction of this protein [188]. Notably, voglibose has increased phosphorylation and activation of AMPK lead to the ef-
also been used in a case of nonalcoholic steatohepatitis, improving fects on glucose and lipid metabolism observed after treatment with
patient’s hepatitis. The real mechanisms remain unclear but it may metformin [194, 195]. It has been suggested that activation of
be related with a decreased production of free radicals, thereby AMPK leads to phosphorylation and inactivation of acetyl-CoA
causing a reduction in the mRNA and protein expression of the carboxylase, inhibiting the rate-limiting step of lipogenesis [194,
hepatic inflammatory cytokine, TNF- [189]. 196] (Fig. 2). Reduced synthesis of malonyl-CoA, the acetyl-CoA
Shimabukuro and collaborators have demonstrated that a single carboxylase product, is also predicted to relieve inhibition of car-
administration of acarbose [190] or nateglinide [191] improves nitine palmitoyltransferase 1, resulting in increased fatty acid oxida-
postprandial endothelial dysfunction, the initial stage of atheroscle- tion [195, 197] (Fig. 2). These effects may contribute to the met-
rosis, in T2DM. However, they neither assessed nor compared the formin’s in vivo ability to lower triglycerides [197]. It was also
long-term effects of both agents on postprandial endothelial func- demonstrated, using an AMPK inhibitor, that AMPK activation is
tion. These effects were later investigated in a comparative study by required for inhibition of hepatocyte glucose production by met-
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3613

formin [193]. Moreover, an association between increased glucose Thiazolidinediones


uptake and AMPK activation was observed in isolated skeletal Pioglitazone, an agonist of PPAR, increased the expression of
muscles, and it was suggested that metformin’s effect in augment- glucose transporters GLUT1 and GLUT4 in 3T3-F442A preadipo-
ing muscle insulin action in vivo may be attributed to AMPK as cyte cultures treated with pioglitazone (1 mol/L) and insulin (1
well [193]. mg/L). This increase was mainly due to the stabilization of GLUT1
Another study showed that AMPK activation is required for the and GLUT4 transporter messenger RNA transcripts [210]. The
glucose-lowering action of metformin in vivo but with genetic evi- increase on GLUT4 expression was also observed in epididymal fat
dence. The genetic deletion of liver kinase B1, one of the upstream cells in an animal model of insulin-resistant T2DM (KKAy mice).
activators of AMPK, did not impair AMPK activation in muscle, This augment was registered after a treatment with pioglitazone 20
yet it eliminated the effect of metformin on serum glucose levels. mg/kg/day for 4 days. Besides, treatment with pioglitazone partially
This led the authors to hypothesize that in mice, metformin primar- corrected the reduction on mRNA encoding hexokinase II levels in
ily decreases blood glucose concentrations by decreasing hepatic epididymal fat and muscle in this animal model [211]. In addition,
gluconeogenesis [198]. Interestingly, it was recently reported that pioglitazone also increases protein phosphatase-1, the enzyme that
metformin could be considered as a suitable antidiabetic drug for activates glycogen synthase, activity in hepatocytes from streptozo-
male patients of reproductive age with T2DM. The authors showed tocin-induced diabetic Sprague Dawley rats treated with insulin 0.1
that besides not having deleterious effects on human Sertoli cells, M and pioglitazone 5 M [212]. An induction of adipose tissue
the cells that support spermatogenesis [49, 173, 199], metformin secretion was also observed after treatment with 3 M pioglitazone,
presented a possible antioxidant activity. Moreover, metformin particularly of the high molecular weight adiponectin [213]. Fur-
stimulated lactate production by those testicular cells [49], which thermore, pioglitazone decreased angiotensin II-induced connective
provides nutritional support [199] and has an anti-apoptotic effect tissue growth factor expression and proliferation in atrial fibro-
in developing germ cells [200]. blasts. This effect might be at least in part related with its inhibitory
Besides T2DM, metformin is also considered a therapeutic effect on transforming growth factor-1 / Smad2/3, as well as on
option for other diseases related with insulin resistance, such as transforming growth factor-1 / tumor necrosis factor receptor as-
polycystic ovary syndrome (PCOS) [53], an endocrinopathy charac- sociated factor 6 / transforming growth factor--associated kinase 1
terized by hyperandrogenism, menstrual irregularity/anovulation, signaling pathway [214].
and polycystic-appearing ovaries on ultrasound [201, 202]. How- Concerning the female reproductive system, pioglitazone is
ever, the specific molecular pathways where metformin acts on the reported to counterbalance the inhibition of FSH-induced follicular
ovary remain elusive [164]. As in the case of T2DM, it is thought development and steroidogenesis by TNF-. These results suggest
that AMPK is involved in metfomin’s mechanism of action in that, in patients with PCOS, pioglitazone may directly act on ovar-
PCOS. AMPK is present in all cell compartments of the ovary and ian functions through PPAR activation [215]. Pioglitazone can
its expression could be different depending on the maturation stage. also suppress hepatic fetuin-A expression, a hepatokine that induces
In vitro studies with primary cells of theca interna from the rat insulin resistance [216], in vitro and in vivo. This seems to be a
showed that the addition of metformin was associated with in- characteristic restricted to thiazolidinedione derivatives compared
creased activity of AMPK and inhibition of insulin stimulated pro- to other antidiabetic drugs, such as metformin [217]. Thiazolidin-
liferation [203]. ediones were also found to have a renoprotective effect. This effect
In addition, there are several studies associating metformin to may be related to a decreased albuminuria and proteinuria [218].
reduced cancer development and progression [164]. The different Since functional PPAR has been identified in renal glomerular and
mechanisms associated to this metformin action are fundamentally tubular segments [219], thiazolidinediones can also protect against
focused on inhibition of growth stimuli and alterations of metabolic renal injury through these receptors at the kidney. The action of
processes, which control cancer cell growth. This characteristic pioglitazone in cell metabolism remains completely unknown and
began to be observed in epidemiological studies, but was later con- further studies are needed to unveil how these mechanisms alter
firmed by in vitro and in vivo studies. In fact, it has been demon- cells and organs metabolic phenotype.
strated that metformin has an antiproliferative action in various cell
Meglitinides
lines and animal models [50]. After its uptake into the cells, met-
formin seems to cause a reduction in ATP via inhibition of the mi- Meglitinides are also known for their action in glucose metabo-
tochondrial respiratory chain complex 1 leading to activation of lism. Interestingly, it was found that repaglinide is more effective in
AMPK [204] (Fig. 2). AMPK activation then disrupts the expres- lowering glucose levels when there is a greater concentration of this
sion of genes involved in gluconeogenesis, protein synthesis, lipo- sugar [220]. Furthermore, in the absence of glucose, repaglinide
genesis and possibly angiogenesis [205] (Fig. 2). Moreover, treat- had no effect on insulin secretion in isolated mouse [221] or rat
ment with metformin may decrease serum levels of insulin and [222] pancreatic islet cells. Another study showed that repaglinide
insulin-like growth factor, therefore reducing the stimulus for cell (10 uM) has no significant effect on basal insulin release. However,
growth. In fact, a decrease in insulin and insulin-like growth factor, in the presence of glucose (>5 mmol/L) repaglinide was reported to
whose receptors are expressed on many cancer cells, induced by stimulate insulin release in a concentration-dependent manner. In
caloric restriction has been shown to reduce the incidence of cancer contrast to nateglinide, which mainly stimulates first phase insulin
in in vivo animal models [206]. Numerous studies have revealed release, repaglinide does not clearly induce biphasic insulin secre-
growth-inhibiting effects of metformin in breast, endometrial, lung, tion in perfused islets [223, 224]. Unlike sulfonylureas, repaglinide
liver, gastric, and medullary thyroid cancer cell lines [207]. More- did not inhibit glucose-stimulated proinsulin biosynthesis in iso-
over, studies of the effects of some chemotherapeutics (cisplatin, lated rat -cells [225]. Notably, the insulinotropic action of repa-
carboplatin and paclitaxel) on endometrial [208] and ovarian [209] glinide was not attributable to any significant increase in the me-
cancer cell lines have suggested a role for metformin as adjuvant tabolism of exogenous or endogenous nutrients [223]. Regarding
therapy. On endometrial cancer cells, it has been described that glucagon release, neither repaglinide nor nateglinide had significant
metformin acted by a downregulation of glyoxalase I, an enzyme effects in studies using isolated perfused rat pancreas. On the other
that is related with glycometabolism. Moreover, glyoxalase I is hand, somatostatin release, which is a regulatory peptide that inhib-
overexpressed in endometrial cancer cells and its gene silencing its glucagon secretion [226], was augmented by both meglitinide
enhances the sensitivity of endometrial cancer cells to chemothera- analogues [227].
peutic drugs [208].
3614 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

Metformin
OCT

Metformin
Lipogenesis
ACAC
Acetyl-CoA Malonyl-CoA Fatty Acids

Fatty Acids
CPTI

Citrate Fatty Acyl-CoA


ATP I
-oxidation ACAD
Krebs MTP
AMPK Acetyl-CoA +
cycle 2,3 Enoyl CoA
Shortened Acyl-CoA

Gluconeogenesis,
protein synthesis and
lipogenesis related genes
Nucleus

Fig. (2). Possible metformin mechanism of action in cells. Metformin enters the cell through organic cation transporter (OCT) and may lead to the phosphory-
lation and inactivation of acetyl-CoA carboxylase (ACAC), which is responsible for the rate-limiting step of lipogenesis. The reduced synthesis of malonyl-
CoA is also predicted to relieve inhibition of carnitine palmitoyltransferase 1 (CPTI), resulting in increased -oxidation. Moreover, metformin seems to cause
a reduction in ATP via inhibition of the mitochondrial respiratory chain complex 1 (I) leading to activation of AMP-activated protein kinase (AMPK). AMPK
activation then disrupts the expression of genes involved in gluconeogenesis, protein synthesis and lipogenesis.

fects on human adipose cell precursors, inhibiting proliferation and


GLP-1 Receptor Agonists and DPP-4 Inhibitors
differentiation. Furthermore, it stimulated the expression of insulin-
Exenatide, a GLP-1 receptor agonist, induced an increase in a sensitizing adipokines. These effects could contribute to the actions
series of vasodilators in patients with T2DM on treatment with oral of GLP-1 receptor agonists on body weight and insulin sensitivity
hypoglycemic agents and insulin. These patients were started on [232]. However, there is a lack of studies focused on the effects of
exenatide 5 g twice a day and the dose was increased to 10 g liraglutide in other cellular systems.
twice daily a week later for a period of 12 weeks [228]. The in-
The effects of DPP-4 inhibitors, more specifically saxagliptin,
crease in vasodilators may contribute to the anti-hypertensive ac-
in cardiovascular system are controversial. An in vivo study with
tions of exenatide in patients with T2DM on treatment with oral
CETP-ApoB100 transgenic mice, a transgenic mouse model of
hypoglycemic agents and insulin. Exenatide has also been studied
human atherosclerosis, treated with saxagliptin (10 mg/kg/day)
for the treatment of Parkinson’s disease [229, 230]. A 12-month
showed that saxagliptin treatment reversed endothelial dysfunction
treatment with exenatide improved motor and cognitive measures
and reduced atherosclerotic lesion area [233]. However, a study
compared with the control group [230]. After 12 months of cessa-
involving 16.492 patients with T2DM showed that although DPP-4
tion of this treatment, treated patients maintained the advantage in
inhibition with saxagliptin did not increase or decrease the rate of
comparison with the placebo group [229]. However, more studies
ischemic events but the rate of hospitalization for heart failure was
are necessary to assure these neuroprotective effects, and the over-
increased [234]. Thus, even though saxagliptin improves glycemic
all effect of exenatide on organs and systems. However, these stud-
control, other approaches are necessary to reduce cardiovascular
ies provide clear evidence that exenatide may exert its protective
risk in patients with T2DM. However, saxagliptin was found to be a
effects by modulating crucial metabolic pathways though the
suitable antidiabetic for T2DM patients with renal failure. These
mechanisms remain unknown.
patients were treated with saxagliptin 2.5 mg once daily and the
Liraglutide, another GLP-1 receptor agonist, increased insulin extent of adverse events and hypoglycemic events were similar
sensitivity and attenuated high fat diet-induced insulin resistance between placebo and treated groups [235]. More studies are needed
after a treatment of 6 weeks (0.1 mg/kg, twice daily). A reduction in to consolidate these findings and expose the mode of action of
-cell mass was observed in liraglutide-treated control and high fat DPP-4 inhibitors in cells and organs.
diet-fed mice at 6 weeks, and was associated with a lower -cell
proliferation rate after 1 week of treatment [231]. An in vitro study SGLT2 Inhibitors
showed that liraglutide (10–100nM) significantly inhibited adipose In vivo studies showed that dapagliflozin acutely induces renal
stem cells proliferation and viability. Moreover, the glucose uptake glucose excretion in normal and diabetic rats, improves glucose
was significantly reduced in a dose dependent manner and the tolerance in normal rats, and reduces hyperglycemia in Zucker dia-
treatment with liraglutide reduced intracellular lipid accumulation betic fatty rats after single oral doses ranging from 0.1 to 1.0 mg/kg.
in differentiating adipose stem cells. Thus, liraglutide exerted ef- Moreover, dapagliflozin treatment once a day over 2 weeks signifi-
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3615

cantly lowered fasting and fed glucose levels at doses ranging from cardiovascular diseases, which are the leading cause of death
0.1 to 1.0 mg/kg and resulted in a significant increase in glucose worldwide.
utilization rate accompanied by a significant reduction in glucose
production [236]. In a randomized, double-blind, placebo- ABBREVIATIONS
controlled, parallel-group, 28-day study, in 29 subjects with T2DM DM = Diabetes Mellitus
not optimally controlled, canagliflozin (100 mg once daily or 300 T2DM = Type 2 Diabetes Mellitus
mg twice a day) was well tolerated without evidence for glucose
malabsorption, and the patients presented improved glycemic con- T1DM = Type 1 Diabetes Mellitus
trol [237]. In fact, after the 28 days of treatment with canagliflozin, GLP-1 = Glucagon-like peptide-1
the renal threshold for glucose excretion was reduced; urinary glu- AMPK = AMP-activated protein kinase
cose excretion was increased [237]. However, canagliflozin was
reported to increase the risk of genital mycotic infections [238]. PPAR = Peroxisome proliferator-activated receptor 
This is one of the newest classes of antidiabetics and thus the out- SUR 1 = Sulfonylurea receptor 1
comes on cellular metabolism remain unknown. It is a field of re- GIP = Glucose dependent insulinotropic polypeptide
search that will be on the spotlight for the next years.
DPP-4 = Dipeptidyl peptidase-4
CONCLUSION AND FUTURE PERSPECTIVES PPAR = Peroxisome proliferator-activated receptor 
The development of an effective and safe antidiabetic involves SGLT = Sodium dependent glucose co-transporters
a large investment. Every year new advances are made and new GLUT = Glucose transporters
compounds are presented as a gold standard to maintain glycemic
control in diabetic individuals. The newer classes and the high de- MTP = Mitochondrial trifunctional protein
mand for therapy lead to an increase in the sales of antidiabetics. PCOS = Polycystic ovary syndrome
Besides the obvious safety issue, the side effects are also on the TNF- = Tumor necrosis factor 
spotlight. There are several problems that may arise from the use of
antidiabetics, namely the higher risk of hypoglycemias with some CONFLICT OF INTEREST
drugs. This is a delicate situation since severe hypoglycemias lead The authors confirm that this article content has no conflict of
to serious complications, such as myocardial infarction or demen- interest.
tias. Moreover, transient deprivation of insulin may induce several
health problems, as for instance in the reproductive potential of ACKNOWLEDGEMENTS
males [239, 240] or in the muscular system [241, 242]. This work was supported by the “Fundação para a Ciência e a
There are several potential new targets to develop a safe and Tecnologia”–FCT co-funded by Fundo Europeu de Desen-
effective antidiabetic drug. One of the most relevant topics in this volvimento Regional - FEDER via Programa Operacional Factores
subject is focused on genetic studies. Indeed, many authors suggest de Competitividade - COMPETE/QREN to UMIB (PEst-
that there is an urgent need for gene expression analyses to unveil OE/SAU/UI0215/2014); CICS-UBI (Pest-C/SAU/UI0709/2014);
specific molecular signatures associated with the development and PF Oliveira (PTDC/QUI-BIQ/121446/2010 and Programa Ciência
establishment of DM. In fact, in the recent years, large investments 2008) and MG Alves (SFRH/BPD/80451/2011).
have been made in genome-wide association studies with the expec-
tation of identify novel therapeutic modalities or allow selection of REFERENCES
patients who would respond better to therapeutic interventions. [1] Guariguata L, Whiting DR, Hambleton I, et al. Global estimates of
These and other studies may reveal genes strongly linked with the diabetes prevalence for 2013 and projections for 2035. Diabetes
disease that may serve as therapeutic targets for the treatment of Res Clin Pract 2014; 103: 137-49.
DM and its complications. Moreover, several natural products have [2] American Diabetes Association. Diagnosis and classification of
been reported to possess antidiabetic activity. In fact, there are sev- diabetes mellitus. Diabetes Care 2008; 31 Suppl 1: S55-60.
eral plant-derived products in pharmaceutical industry. Since the [3] Mandrup-Poulsen T, Pickersgill L, Donath MY. Blockade of inter-
leukin 1 in type 1 diabetes mellitus. Nat Rev Endocrinol 2010; 6:
ancient times, natural products are used for the treatment of DM. 158-66.
Therefore, it is possible that nature can be a valuable source of [4] van Belle TL, Coppieters KT, von Herrath MG. Type 1 diabetes:
compounds to be used in the treatment of DM. There are many etiology, immunology, and therapeutic strategies. Physiol Rev
compounds under investigation that may come to the market in the 2011; 91: 79-118.
next years. [5] Patterson CC, Dahlquist GG, Gyurus E, et al. Incidence trends for
In addition, new therapeutic applications have been reported for childhood type 1 diabetes in Europe during 1989-2003 and pre-
dicted new cases 2005-20: a multicentre prospective registration
many antidiabetics. This is consistent with their mechanisms of study. Lancet 2009; 373: 2027-33.
action that may be broader than initially reported. Many of these [6] McCarthy MI. Genomics, type 2 diabetes, and obesity. N Engl J
compounds may also modulate cellular metabolism in several sys- Med 2010; 363: 2339-50.
tems and organs, thus promoting crucial metabolic alterations that [7] Stumvoll M, Tataranni PA, Stefan N, et al. Glucose allostasis.
promote either deleterious or beneficial effects. This is a crucial Diabetes 2003; 52: 903-9.
side effect that must be carefully investigated when prescribing a [8] Colberg SR, Sigal RJ, Fernhall B, et al. Exercise and type 2 diabe-
compound to a diabetic individual. Moreover, the polypharmacy tes the American College of Sports Medicine and the American
regime, that some diabetic individuals use, should be monitored Diabetes Association: joint position statement. Diabetes Care 2010;
since it may expose the individual to important side effects. 33: e147-e67.
[9] Tabák AG, Herder C, Rathmann W, et al. Prediabetes: a high-risk
In conclusion, the multiple antidiabetic drugs available in the state for diabetes development. Lancet 2012; 379: 2279-90.
market create a stimulating field of research in constant expansion, [10] Alves MG, Martins AD, Rato L, et al. Molecular mechanisms
especially since the incidence of DM is alarmingly increasing, beyond glucose transport in diabetes-related male infertility. Bio-
among all age ranges. As the need for knowledge in this particular chim Biophys Acta 2013; 1832: 626-35.
area rises, the scientific community faces new challenges. On one [11] Valensi P, Schwarz E, Hall M, et al. Pre-diabetes essential action: a
hand, new compounds are demanded, but on the other hand plei- European perspective. Diabetes Metab 2005; 31: 606-20.
[12] Rato L, Alves MG, Dias TR, et al. High-energy diets may induce a
otropic effects of the marketed compounds make these drugs suit- pre-diabetic state altering testicular glycolytic metabolic profile and
able to be used in specific groups of patients, such as patients with male reproductive parameters. Andrology 2013; 1: 495-504.
3616 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

[13] Bernardino RL, Martins AD, Socorro S, et al. Effect of prediabetes [39] Goke B, Fuder H, Wieckhorst G, et al. Voglibose (AO-128) is an
on membrane bicarbonate transporters in testis and epididymis. J efficient alpha-glucosidase inhibitor and mobilizes the endogenous
Membr Biol 2013; 246: 877-83. GLP-1 reserve. Digestion 1995; 56: 493-501.
[14] American Diabetes Association. Diagnosis and classification of [40] Lee EY, Kaneko S, Jutabha P, et al. Distinct action of the alpha-
diabetes mellitus. Diabetes Care 2010; 33 Suppl 1: S62-9. glucosidase inhibitor miglitol on SGLT3 and GLP1 secretion. J
[15] Perreault L, Kahn SE, Christophi CA, et al. Regression from pre- Endocrinol 2014: JOE-14-0555.
diabetes to normal glucose regulation in the diabetes prevention [41] Dabhi AS, Bhatt NR, Shah MJ. Voglibose: an alpha glucosidase
program. Diabetes Care 2009; 32: 1583-8. inhibitor. J Clin Diagn Res 2013; 7: 3023-7.
[16] Morris DH, Khunti K, Achana F, et al. Progression rates from [42] Oubre AY, Carlson TJ, King SR, et al. From plant to patient: an
HbA1c 6.0–6.4% and other prediabetes definitions to type 2 diabe- ethnomedical approach to the identification of new drugs for the
tes: a meta-analysis. Diabetologia 2013; 56: 1489-93. treatment of NIDDM. Diabetologia 1997; 40: 614-7.
[17] Salas-Salvadó J, Martinez-Gonzalez M, Bullo M, et al. The role of [43] Bailey CJ. Biguanides and NIDDM. Diabetes Care 1992; 15: 755-
diet in the prevention of type 2 diabetes. Nutr Metab Cardiovasc 72.
Dis 2011; 21: B32-B48. [44] Mc KJ, Kuwayti K, Rado PP. Clinical experience with DBI (phen-
[18] Gray GM, Santiago NA. Intestinal beta-galactosidases. I. Separa- formin) in the management of diabetes. Can Med Assoc J 1959; 80:
tion and characterization of three enzymes in normal human intes- 773-8.
tine. J Clin Invest 1969; 48: 716-28. [45] Gottlieb B, Auld WH. Metformin in treatment of diabetes mellitus.
[19] Bischoff H. Pharmacology of alpha-glucosidase inhibition. Eur J Br Med J 1962; 1: 680-2.
Clin Invest 1994; 24 Suppl 3: 3-10. [46] Misbin RI. Phenformin-associated lactic acidosis: pathogenesis and
[20] Derosa G, Maffioli P. Alpha-Glucosidase inhibitors and their use in treatment. Ann Intern Med 1977; 87: 591-5.
clinical practice. Arch Med Sci 2012; 8: 899-906. [47] Williams RH, Palmer JP. Farewell to phenformin for treating dia-
[21] Kumar S, Narwal S, Kumar V, et al. -glucosidase inhibitors from betes mellitus. Ann Intern Med 1975; 83: 567-8.
plants: A natural approach to treat diabetes. Pharmacogn Rev 2011; [48] Stumvoll M, Nurjhan N, Perriello G, et al. Metabolic effects of
5: 19. metformin in non-insulin-dependent diabetes mellitus. N Engl J
[22] Lebovitz HE. Alpha-Glucosidase inhibitors. Endocrinol Metab Clin Med 1995; 333: 550-4.
North Am 1997; 26: 539-51. [49] Alves MG, Martins AD, Vaz CV, et al. Metformin and male repro-
[23] Kumar RV, Sinha VR. Newer insights into the drug delivery ap- duction: effects on Sertoli cell metabolism. Br J Pharmacol 2014;
proaches of alpha-glucosidase inhibitors. Expert Opin Drug Deliv 171: 1033-42.
2012; 9: 403-16. [50] Viollet B, Foretz M. Revisiting the mechanisms of metformin
[24] Patil SB, Ghadyale VA, Taklikar SS, et al. Insulin secretagogue, action in the liver. Ann Endocrinol (Paris) 2013; 74: 123-9.
alpha-glucosidase and antioxidant activity of some selected spices [51] Musi N, Hirshman MF, Nygren J, et al. Metformin increases AMP-
in streptozotocin-induced diabetic rats. Plant Foods Hum Nutr activated protein kinase activity in skeletal muscle of subjects with
2011; 66: 85-90. type 2 diabetes. Diabetes 2002; 51: 2074-81.
[25] Breuer HW. Review of acarbose therapeutic strategies in the long- [52] Ikeda T, Iwata K, Murakami H. Inhibitory effect of metformin on
term treatment and in the prevention of type 2 diabetes. Int J Clin intestinal glucose absorption in the perfused rat intestine. Biochem
Pharmacol Ther 2003; 41: 421-40. Pharmacol 2000; 59: 887-90.
[26] Scott LJ, Spencer CM. Miglitol. Drugs 2000; 59: 521-49. [53] Meneses MJ, Sousa M, Alves MG, et al. The antidiabetic drug
[27] Sels J-PJE, Huijberts MSP, Wolffenbuttel BHR. Miglitol, a new - metformin and male reproductive function: an overview. Int J Diab
glucosidase inhibitor. Exp Opin Pharmacother 1999; 1: 149-56. Cardiov Dis Res 2015; 3: 1-2.
[28] Chen X, Zheng Y, Shen Y. Voglibose (Basen, AO-128), one of the [54] Andrzejewski S, Gravel SP, Pollak M, et al. Metformin directly
most important alpha-glucosidase inhibitors. Curr Med Chem 2006; acts on mitochondria to alter cellular bioenergetics. Cancer Metab
13: 109-16. 2014; 2: 12.
[29] Hollander P. Safety profile of acarbose, an -glucosidase inhibitor. [55] Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts
Drugs 1992; 44: 47-53. its anti-diabetic effects through inhibition of complex 1 of the mi-
[30] Kim J-S, Kwon C-S, Son KH. Inhibition of alpha-glucosidase and tochondrial respiratory chain. Biochem J 2000; 348 Pt 3: 607-14.
amylase by luteolin, a flavonoid. Biosci Biotechnol Biochem 2000; [56] Coughlan KA, Valentine RJ, Ruderman NB, et al. AMPK activa-
64: 2458-61. tion: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr
[31] Wang Y-J, Liu L-L, Feng Z-H, et al. Optimization of media com- Obes 2014; 7: 241-53.
position and culture conditions for acarbose production by Actino- [57] Winder WW, Hardie DG. AMP-activated protein kinase, a meta-
planes utahensis ZJB-08196. Worl J Microb Biot 2011; 27: 2759- bolic master switch: possible roles in type 2 diabetes. Am J Physiol
66. 1999; 277: E1-10.
[32] Wehmeier U, Piepersberg W. Biotechnology and molecular biol- [58] Maida A, Lamont BJ, Cao X, et al. Metformin regulates the in-
ogy of the -glucosidase inhibitor acarbose. Appl Microbiol Bio- cretin receptor axis via a pathway dependent on peroxisome prolif-
technol 2004; 63: 613-25. erator-activated receptor-alpha in mice. Diabetologia 2011; 54:
[33] Krentz AJ, Bailey CJ. Oral antidiabetic agents: current role in type 339-49.
2 diabetes mellitus. Drugs 2005; 65: 385-411. [59] Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hy-
[34] Yadav N, Panwar MS. Formulation and evaluation of matrix tablets perglycemia in type 2 diabetes: a patient-centered approach: posi-
of miglitol using different grades of HPMC. Asian J Pharmacol tion statement of the American Diabetes Association (ADA) and
2014; 8: 237. the European Association for the Study of Diabetes (EASD). Dia-
[35] Ahr HJ, Boberg M, Brendel E, et al. Pharmacokinetics of miglitol. betes Care 2012; 35: 1364-79.
Absorption, distribution, metabolism, and excretion following ad- [60] Ahren B. Emerging dipeptidyl peptidase-4 inhibitors for the treat-
ministration to rats, dogs, and man. Arzneimittelforschung 1997; ment of diabetes. Expert Opin Emerg Drugs 2008; 13: 593-607.
47: 734-45. [61] Rendell M. The role of sulphonylureas in the management of type 2
[36] Deshpande MC, Venkateswarlu V, Mantri AH, et al. Targeting diabetes mellitus. Drugs 2004; 64: 1339-58.
enteral endocrinal L-cells with dietary carbohydrates, by increasing [62] Levine R. Sulfonylureas: background and development of the field.
the availability of miglitol in the intestinal lumen, leads to multi- Diabetes Care 1984; 7 Suppl 1: 3-7.
fold enhancement of plasma glucagon-like peptide-1 levels in non- [63] Loubatières A. The hypoglycemic sulfonamides: history and devel-
diabetic canines. Drug Dev Ind Pharm 2011; 37: 506-17. opment of the problem from 1942 to 1955. Ann N Y Acad Sci
[37] Horii S, Fukase H, Matsuo T, et al. Synthesis and alpha-D- 1957; 71: 4-11.
glucosidase inhibitory activity of N-substituted valiolamine deriva- [64] DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus.
tives as potential oral antidiabetic agents. J Med Chem 1986; 29: Ann Intern Med 1999; 131: 281-303.
1038-46. [65] Skillman TG, Feldman JM. The pharmacology of sulfonylureas.
[38] Kameda Y, Asano N, Yoshikawa M, et al. Valiolamine, a new Am J Med 1981; 70: 361-72.
alpha-glucosidase inhibiting aminocyclitol produced by Streptomy- [66] Tsunekawa T, Hayashi T, Suzuki Y, et al. Plasma adiponectin
ces hygroscopicus. J Antibiot (Tokyo) 1984; 37: 1301-7. plays an important role in improving insulin resistance with glime-
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3617

piride in elderly type 2 diabetic subjects. Diabetes Care 2003; 26: comparison with sulfonylureas and repaglinide. J Pharmacol Exp
285-9. Ther 2000; 293: 444-52.
[67] Stumvoll M, Goldstein BJ, van Haeften TW. Type 2 diabetes: [92] Marre M, Van Gaal L, Usadel KH, et al. Nateglinide improves
principles of pathogenesis and therapy. Lancet 2005; 365: 1333-46. glycaemic control when added to metformin monotherapy: results
[68] Sturgess N, Cook D, Ashford MJ, et al. The sulphonylurea receptor of a randomized trial with type 2 diabetes patients. Diabetes Obes
may be an ATP-sensitive potassium channel. Lancet 1985; 326: Metab 2002; 4: 177-86.
474-5. [93] Fonseca V, Grunberger G, Gupta S, et al. Addition of nateglinide
[69] Ahren B. Are sulfonylureas less desirable than DPP-4 inhibitors as to rosiglitazone monotherapy suppresses mealtime hyperglycemia
add-on to metformin in the treatment of type 2 diabetes? Curr Diab and improves overall glycemic control. Diabetes Care 2003; 26:
Rep 2011; 11: 83-90. 1685-90.
[70] Rorsman P. The pancreatic beta-cell as a fuel sensor: an electro- [94] Anderson D, Shelley S, Kellett N, et al. The effect of nateglinide
physiologist's viewpoint. Diabetologia 1997; 40: 487-95. taken with food on gastric emptying rates in healthy subjects. Clin
[71] Lang J. Molecular mechanisms and regulation of insulin exocytosis Ther 2003; 25: 1722-38.
as a paradigm of endocrine secretion. Eur J Biochem 1999; 259: 3- [95] Weaver ML, Orwig BA, Rodriguez LC, et al. Pharmacokinetics
17. and metabolism of nateglinide in humans. Drug Metab Dispos
[72] MacDonald PE, Joseph JW, Rorsman P. Glucose-sensing mecha- 2001; 29: 415-21.
nisms in pancreatic beta-cells. Philos Trans R Soc Lond B Biol Sci [96] Gutniak M, Orskov C, Holst JJ, et al. Antidiabetogenic effect of
2005; 360: 2211-25. glucagon-like peptide-1 (7-36)amide in normal subjects and pa-
[73] Domecq JP, Prutsky G, Leppin A, et al. Drugs commonly associ- tients with diabetes mellitus. N Engl J Med 1992; 326: 1316-22.
ated with weight change: a systematic review and meta-analysis. J [97] Drucker DJ. Glucagon-like peptides. Diabetes 1998; 47: 159-69.
Clin Endocrinol Metab 2015; 100: 363-70. [98] Feinle C, Chapman IM, Wishart J, et al. Plasma glucagon-like
[74] Malaisse WJ. Stimulation of insulin release by non-sulfonylurea peptide-1 (GLP-1) responses to duodenal fat and glucose infusions
hypoglycemic agents: the meglitinide family. Horm Metab Res in lean and obese men. Peptides 2002; 23: 1491-5.
1995; 27: 263-6. [99] Kieffer TJ, Francis Habener J. The glucagon-like peptides. Endocr
[75] Horton ES, Clinkingbeard C, Gatlin M, et al. Nateglinide alone and Rev 1999; 20: 876-913.
in combination with metformin improves glycemic control by re- [100] MacDonald PE, El-kholy W, Riedel MJ, et al. The multiple actions
ducing mealtime glucose levels in type 2 diabetes. Diabetes Care of GLP-1 on the process of glucose-stimulated insulin secretion.
2000; 23: 1660-5. Diabetes 2002; 51: S434-42.
[76] Moses R, Slobodniuk R, Boyages S, et al. Effect of repaglinide [101] Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP.
addition to metformin monotherapy on glycemic control in patients Gastroenterology 2007; 132: 2131-57.
with type 2 diabetes. Diabetes Care 1999; 22: 119-24. [102] Imeryuz N, Yegen BC, Bozkurt A, et al. Glucagon-like peptide-1
[77] Sunaga Y, Gonoi T, Shibasaki T, et al. The effects of mitiglinide inhibits gastric emptying via vagal afferent-mediated central
(KAD-1229), a new anti-diabetic drug, on ATP-sensitive K+ chan- mechanisms. Am J Physiol 1997; 273: G920-7.
nels and insulin secretion: comparison with the sulfonylureas and [103] Gutzwiller JP, Drewe J, Goke B, et al. Glucagon-like peptide-1
nateglinide. Eur J Pharmacol 2001; 431: 119-25. promotes satiety and reduces food intake in patients with diabetes
[78] Grell W, Hurnaus R, Griss G, et al. Repaglinide and related hypo- mellitus type 2. Am J Physiol 1999; 276: R1541-4.
glycemic benzoic acid derivatives. J Med Chem 1998; 41: 5219-46. [104] Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like
[79] Gromada J, Dissing S, Kofod H, et al. Effects of the hypoglycae- peptide-1 by human plasma in vitro yields an N-terminally trun-
mic drugs repaglinide and glibenclamide on ATP-sensitive potas- cated peptide that is a major endogenous metabolite in vivo. J Clin
sium-channels and cytosolic calcium levels in  TC3 cells and rat Endocrinol Metab 1995; 80: 952-7.
pancreatic beta cells. Diabetologia 1995; 38: 1025-32. [105] Gallwitz B, Ropeter T, Morys-Wortmann C, et al. GLP-1-
[80] Hansen AMK, Christensen IT, Hansen JB, et al. Differential inter- analogues resistant to degradation by dipeptidyl-peptidase IV in vi-
actions of nateglinide and repaglinide on the human -cell sul- tro. Regul Pept 2000; 86: 103-11.
phonylurea receptor 1. Diabetes 2002; 51: 2789-95. [106] Drucker DJ, Nauck MA. The incretin system: glucagon-like pep-
[81] Dornhorst A. Insulinotropic meglitinide analogues. Lancet 2001; tide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in
358: 1709-16. type 2 diabetes. Lancet 2006; 368: 1696-705.
[82] Marbury T, Huang W-C, Strange P, et al. Repaglinide versus gly- [107] Ristic S, Byiers S, Foley J, et al. Improved glycaemic control with
buride: a one-year comparison trial. Diabetes Res Clin Pract 1999; dipeptidyl peptidase-4 inhibition in patients with type 2 diabetes:
43: 155-66. vildagliptin (LAF237) dose response. Diabetes Obes Metab 2005;
[83] Culy CR, Jarvis B. Repaglinide. Drugs 2001; 61: 1625-60. 7: 692-8.
[84] Van Heiningen PNM, Hatorp V, Nielsen KK, et al. Absorption, [108] Deacon CF, Holst JJ. Dipeptidyl peptidase-4 inhibitors for the
metabolism and excretion of a single oral dose of 14C-repaglinide treatment of type 2 diabetes: comparison, efficacy and safety. Ex-
during repaglinide multiple dosing. Eur J Clin Pharmacol 1999; 55: pert Opin Pharmacother 2013; 14: 2047-58.
521-5. [109] Vardarli I, Nauck MA, Kothe LD, et al. Inhibition of DPP-4 with
[85] Hatorp V, Oliver S, Su CA. Bioavailability of repaglinide, a novel vildagliptin improved insulin secretion in response to oral as well
antidiabetic agent, administered orally in tablet or solution form or as "isoglycemic" intravenous glucose without numerically chang-
intravenously in healthy male volunteers. Int J Clin Pharmacol Ther ing the incretin effect in patients with type 2 diabetes. J Clin Endo-
1998; 36: 636-41. crinol Metab 2011; 96: 945-54.
[86] Kajosaari LI, Laitila J, Neuvonen PJ, et al. Metabolism of repa- [110] Duttaroy A, Voelker F, Merriam K, et al. The DPP-4 inhibitor
glinide by CYP2C8 and CYP3A4 in vitro: effect of fibrates and ri- vildagliptin increases pancreatic beta cell mass in neonatal rats. Eur
fampicin. Basic Clin Pharmacol Toxicol 2005; 97: 249-56. J Pharmacol 2011; 650: 703-7.
[87] Hasslacher C. Safety and efficacy of repaglinide in type 2 diabetic [111] Mu J, Woods J, Zhou YP, et al. Chronic inhibition of dipeptidyl
patients with and without impaired renal function. Diabetes Care peptidase-4 with a sitagliptin analog preserves pancreatic beta-cell
2003; 26: 886-91. mass and function in a rodent model of type 2 diabetes. Diabetes
[88] Hatorp V, Walther KH, Christensen MS, et al. SingleDose Phar- 2006; 55: 1695-704.
macokinetics of Repaglinide in Subjects with Chronic Liver Dis- [112] Gallwitz B. Glucagon-like peptide-1 as a treatment option for type
ease. J Clin Pharmacol 2000; 40: 142-52. 2 diabetes and its role in restoring beta-cell mass. Diabetes Technol
[89] Jovanovic L, Hassman DR, Gooch B, et al. Treatment of type 2 Ther 2005; 7: 651-7.
diabetes with a combination regimen of repaglinide plus pioglita- [113] Vilsbøll T. The effects of glucagon-like peptide-1 on the beta cell.
zone. Diabetes Res Clin Pract 2004; 63: 127-34. Diabetes Obes Metab 2009; 11: 11-18.
[90] Chachin M, Yamada M, Fujita A, et al. Nateglinide, a D- [114] Baetta R, Corsini A. Pharmacology of dipeptidyl peptidase-4 in-
phenylalanine derivative lacking either a sulfonylurea or ben- hibitors: similarities and differences. Drugs 2011; 71: 1441-67.
zamido moiety, specifically inhibits pancreatic beta-cell-type [115] Deacon CF. Dipeptidyl peptidase-4 inhibitors in the treatment of
K(ATP) channels. J Pharmacol Exp Ther 2003; 304: 1025-32. type 2 diabetes: a comparative review. Diabetes Obes Metab 2011;
[91] Hu S, Wang S, Fanelli B, et al. Pancreatic beta-cell K(ATP) chan- 13: 7-18.
nel activity and membrane-binding studies with nateglinide: A
3618 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

[116] Yoshioka T, Fujita T, Kanai T, et al. Studies on hindered phenols [141] Loke YK, Kwok CS, Singh S. Comparative cardiovascular effects
and analogues. 1. Hypolipidemic and hypoglycemic agents with of thiazolidinediones: systematic review and meta-analysis of ob-
ability to inhibit lipid peroxidation. J Med Chem 1989; 32: 421-8. servational studies. Br Med J 2011; 342: d1309.
[117] Fujita T, Sugiyama Y, Taketomi S, et al. Reduction of insulin [142] Wright EM, Loo DDF, Hirayama BA. Biology of human sodium
resistance in obese and/or diabetic animals by 5-[4-(1- glucose transporters. Physiol Rev 2011; 91: 733-94.
methylcyclohexylmethoxy) benzyl]-thiazolidine-2, 4-dione (ADD- [143] Chao EC, Henry RR. SGLT2 inhibition--a novel strategy for diabe-
3878, U-63,287, ciglitazone), a new antidiabetic agent. Diabetes tes treatment. Nat Rev Drug Discov 2010; 9: 551-9.
1983; 32: 804-10. [144] Chen J, Williams S, Ho S, et al. Quantitative PCR tissue expression
[118] Tominaga M, Igarashi M, Daimon M, et al. Thiazolidinediones profiling of the human SGLT2 gene and related family members.
(AD-4833 and CS-045) improve hepatic insulin resistance in strep- Diabetes Ther 2010; 1: 57-92.
tozotocin-induced diabetic rats. Endocr J 1993; 40: 343-9. [145] Idris I, Donnelly R. Sodium-glucose co-transporter-2 inhibitors: an
[119] Isley WL. Hepatotoxicity of thiazolidinediones. Expert Opin Drug emerging new class of oral antidiabetic drug. Diabetes Obes Metab
Saf 2003; 2: 581-6. 2009; 11: 79-88.
[120] Gitlin N, Julie NL, Spurr CL, et al. Two cases of severe clinical [146] Ehrenkranz JR, Lewis NG, Kahn CR, et al. Phlorizin: a review.
and histologic hepatotoxicity associated with troglitazone. Ann In- Diabetes Metab Res Rev 2005; 21: 31-38.
tern Med 1998; 129: 36-38. [147] Marsenic O. Glucose control by the kidney: an emerging target in
[121] US Food and Drug Administration. Pioglitazone HCl (marketed as diabetes. Am J Kidney Dis 2009; 53: 875-83.
Actos, Actoplus Met, and Duetact) Information. 2011. [148] Meng W, Ellsworth BA, Nirschl AA, et al. Discovery of dapagli-
[122] Kung J, Henry RR. Thiazolidinedione safety. Expert Opin Drug Saf flozin: a potent, selective renal sodium-dependent glucose cotrans-
2012; 11: 565-79. porter 2 (SGLT2) inhibitor for the treatment of type 2 diabetes. J
[123] Ahmadian M, Suh JM, Hah N, et al. PPARgamma signaling and Med Chem 2008; 51: 1145-9.
metabolism: the good, the bad and the future. Nat Med 2013; 19: [149] Han S, Hagan DL, Taylor JR, et al. Dapagliflozin, a selective
557-66. SGLT2 inhibitor, improves glucose homeostasis in normal and dia-
[124] Tontonoz P, Spiegelman BM. Fat and beyond: the diverse biology betic rats. Diabetes 2008; 57: 1723-9.
of PPARgamma. Annu Rev Biochem 2008; 77: 289-312. [150] Mather A, Pollock C. Glucose handling by the kidney. Kidney Int
[125] Yamauchi T, Kamon J, Waki H, et al. The mechanisms by which 2011; 79: S1-S6.
both heterozygous peroxisome proliferator-activated receptor [151] Plosker GL. Dapagliflozin: a review of its use in type 2 diabetes
gamma (PPARgamma) deficiency and PPARgamma agonist im- mellitus. Drugs 2012; 72: 2289-312.
prove insulin resistance. J Biol Chem 2001; 276: 41245-54. [152] Schumm-Draeger PM, Burgess L, Koranyi L, et al. Twice-daily
[126] Suzuki S, Arnold LL, Pennington KL, et al. Effects of pioglitazone, dapagliflozin co-administered with metformin in type 2 diabetes: a
a peroxisome proliferator-activated receptor gamma agonist, on the 16-week randomized, placebo-controlled clinical trial. Diabetes
urine and urothelium of the rat. Toxicol Sci 2010; 113: 349-57. Obes Metab 2015; 17: 42-51.
[127] Miyazaki Y, Mahankali A, Matsuda M, et al. Improved glycemic [153] Rosengren AH, Braun M, Mahdi T, et al. Reduced insulin exocyto-
control and enhanced insulin sensitivity in type 2 diabetic subjects sis in human pancreatic beta-cells with gene variants linked to type
treated with pioglitazone. Diabetes Care 2001; 24: 710-9. 2 diabetes. Diabetes 2012; 61: 1726-33.
[128] Qin S, Liu T, Kamanna VS, et al. Pioglitazone stimulates apolipo- [154] Alberti KG, Zimmet PZ. Definition, diagnosis and classification of
protein A-I production without affecting HDL removal in HepG2 diabetes mellitus and its complications. Part 1: diagnosis and classi-
cells: involvement of PPAR-alpha. Arterioscler Thromb Vasc Biol fication of diabetes mellitus provisional report of a WHO consulta-
2007; 27: 2428-34. tion. Diabetes Med 1998; 15: 539-53.
[129] Karim A, Slater M, Bradford D, et al. Oral antidiabetic drugs: [155] Garvey WT, Olefsky J, Griffin J, et al. The effect of insulin treat-
effect of food on absorption of pioglitazone and metformin from a ment on insulin secretion and insulin action in type II diabetes mel-
fixed-dose combination tablet. J Clin Pharmacol 2007; 47: 48-55. litus. Diabetes 1985; 34: 222-34.
[130] Jaakkola T, Backman JT, Neuvonen M, et al. Effect of rifampicin [156] Guerci B, Sauvanet JP. Subcutaneous insulin: pharmacokinetic
on the pharmacokinetics of pioglitazone. Br J Clin Pharmacol variability and glycemic variability. Diabetes Metab 2005; 31: 4S7-
2006; 61: 70-78. 4S24.
[131] Lin ZJ, Ji W, Desai-Krieger D, et al. Simultaneous determination [157] Zinman B. Drug therapy. The physiologic replacement of insulin:
of pioglitazone and its two active metabolites in human plasma by an elusive goal. N Engl J Med 1989; 321: 363-70.
LC–MS/MS. J Pharmaceut Biomed 2003; 33: 101-8. [158] Becker RHA, Frick AD, Burger F, et al. Insulin glulisine, a new
[132] Aronoff S, Rosenblatt S, Braithwaite S, et al. Pioglitazone hydro- rapid-acting insulin analogue, displays a rapid time-action profile
chloride monotherapy improves glycemic control in the treatment in obese non-diabetic subjects. Exp Clin Endocrinol Diabetes 2005;
of patients with type 2 diabetes: a 6-month randomized placebo- 113: 435-43.
controlled dose-response study. The Pioglitazone 001 Study Group. [159] Ahmad K. Insulin sources and types: a review of insulin in terms of
Diabetes Care 2000; 23: 1605-11. its mode on diabetes mellitus. J Tradit Chin Med 2014; 34: 234-7.
[133] Azoulay L, Yin H, Filion KB, et al. The use of pioglitazone and the [160] Siebenhofer A, Plank J, Berghold A, et al. Short acting insulin
risk of bladder cancer in people with type 2 diabetes: nested case- analogues versus regular human insulin in patients with diabetes
control study. Br Med J 2012; 344: e3645. mellitus. Cochrane Database Syst Rev 2006: CD003287.
[134] Wei L, MacDonald TM, Mackenzie IS. Pioglitazone and bladder [161] Francis AJ, Home PD, Hanning I, et al. Intermediate acting insulin
cancer: a propensity score matched cohort study. Br J Clin Pharma- given at bedtime: effect on blood glucose concentrations before and
col 2013; 75: 254-9. after breakfast. Br Med J (Clin Res Ed) 1983; 286: 1173.
[135] Betteridge DJ. Effects of pioglitazone on lipid and lipoprotein [162] Burge MR, Waters DL, Holcombe JH, et al. Prolonged Efficacy of
metabolism. Diabetes Obes Metab 2007; 9: 640-7. Short Acting Insulin Lispro in Combination with Human Ultralente
[136] Marx N, Mach F, Sauty A, et al. Peroxisome proliferator-activated in Insulin-Dependent Diabetes Mellitus 1. J Clin Endocrinol Metab
receptor- activators inhibit IFN--induced expression of the T cell- 1997; 82: 920-4.
active CXC chemokines IP-10, Mig, and I-TAC in human endothe- [163] Rosskamp RH, Park G. Long-acting insulin analogs. Diabetes Care
lial cells. J Immunol 2000; 164: 6503-8. 1999; 22: B109-B13.
[137] Lebovitz HE. Differentiating members of the thiazolidinedione [164] Viollet B, Guigas B, Sanz Garcia N, et al. Cellular and molecular
class: a focus on safety. Diabetes Metab Res Rev 2002; 18 Suppl 2: mechanisms of metformin: an overview. Clin Sci (Lond) 2012;
S23-29. 122: 253-70.
[138] Cox PJ, Ryan DA, Hollis FJ, et al. Absorption, disposition, and [165] Barnett AH. Complementing insulin therapy to achieve glycemic
metabolism of rosiglitazone, a potent thiazolidinedione insulin sen- control. Adv Ther 2013; 30: 557-76.
sitizer, in humans. Drug Metab Dispos 2000; 28: 772-80. [166] Dias TR, Alves MG, Silva BM, et al. Sperm glucose transport and
[139] Wagstaff AJ, Goa KL. Rosiglitazone: a review of its use in the metabolism in diabetic individuals. Mol Cell Endocrinol 2014; 396:
management of type 2 diabetes mellitus. Drugs 2002; 62: 1805-37. 37-45.
[140] Zhu Z, Shen Z, Lu Y, et al. Increased risk of bladder cancer with [167] Scheepers A, Joost HG, Schurmann A. The glucose transporter
pioglitazone therapy in patients with diabetes: a meta-analysis. families SGLT and GLUT: molecular basis of normal and aberrant
Diabetes Res Clin Pract 2012; 98: 159-63. function. (JPEN) J Parenter Enteral Nutr 2004; 28: 364-71.
Antidiabetic Drugs Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3619

[168] Martins AD, Alves MG, Simoes VL, et al. Control of Sertoli cell [194] Hadad SM, Hardie DG, Appleyard V, et al. Effects of metformin
metabolism by sex steroid hormones is mediated through modula- on breast cancer cell proliferation, the AMPK pathway and the cell
tion in glycolysis-related transporters and enzymes. Cell Tissue Res cycle. Clin Transl Oncol 2014; 16: 746-52.
2013; 354: 861-8. [195] Foster DW. Malonyl-CoA: the regulator of fatty acid synthesis and
[169] Chehtane M, Khaled AR. Interleukin-7 mediates glucose utilization oxidation. J Clin Invest 2012; 122: 1958-9.
in lymphocytes through transcriptional regulation of the hexokinase [196] Rebecca JF, Morgan DF, Stephen LP, et al. Metformin and salicy-
II gene. Am J Physiol Cell Physiol 2010; 298: C1560-71. late synergistically activate liver AMPK, inhibit lipogenesis and
[170] Mor I, Cheung EC, Vousden KH. Control of glycolysis through improve insulin sensitivity. Biochem J 2015; 468: 125-32.
regulation of PFK1: old friends and recent additions. Cold Spring [197] Abo Alrob O, Lopaschuk GD. Role of CoA and acetyl-CoA in
Harb Symp Quant Biol 2011; 76: 211-6. regulating cardiac fatty acid and glucose oxidation. Biochem Soc
[171] Rato L, Alves MG, Socorro S, et al. Metabolic regulation is impor- Trans 2014; 42: 1043-51.
tant for spermatogenesis. Nat Rev Urol 2012; 9: 330-8. [198] Shaw RJ, Lamia KA, Vasquez D, et al. The kinase LKB1 mediates
[172] Freedman AD, Graff S. The metabolism of pyruvate in the tricar- glucose homeostasis in liver and therapeutic effects of metformin.
boxylic acid cycle. J Biol Chem 1958; 233: 292-5. Science 2005; 310: 1642-6.
[173] Oliveira PF, Martins AD, Moreira AC, et al. The Warburg Effect [199] Dias TR, Alves MG, Bernardino RL, et al. Dose-dependent effects
Revisited-Lesson from the Sertoli Cell. Med Res Rev 2014. of caffeine in human Sertoli cells metabolism and oxidative profile:
[174] Gladden LB. Lactate metabolism: a new paradigm for the third Relevance for male fertility. Toxicology 2015; 328: 12-20.
millennium. J Physiol 2004; 558: 5-30. [200] Erkkilä K, Aito H, Aalto K, et al. Lactate inhibits germ cell apopto-
[175] Boussouar F, Benahmed M. Lactate and energy metabolism in sis in the human testis. Mol Hum Reprod 2002; 8: 109-17.
male germ cells. Trends Endocrinol Metab 2004; 15: 345-50. [201] Johnson NP. Metformin use in women with polycystic ovary syn-
[176] Herman MA, Kahn BB. Glucose transport and sensing in the main- drome. Ann Transl Med 2014; 2: 56.
tenance of glucose homeostasis and metabolic harmony. J Clin In- [202] Vitek W, Alur S, Hoeger KM. Off-label drug use in the treatment
vest 2006; 116: 1767-75. of polycystic ovary syndrome. Fertil Steril 2015; 103: 605-11.
[177] Mailloux RJ, Beriault R, Lemire J, et al. The tricarboxylic acid [203] Will MA, Palaniappan M, Peegel H, et al. Metformin: direct inhibi-
cycle, an ancient metabolic network with a novel twist. PLoS One tion of rat ovarian theca-interstitial cell proliferation. Fertil Steril
2007; 2: e690. 2012; 98: 207-14.
[178] Wieland OH. The mammalian pyruvate dehydrogenase complex: [204] Shackelford DB, Abt E, Gerken L, et al. LKB1 inactivation dictates
structure and regulation. Rev Physiol Biochem Pharmacol 1983; therapeutic response of non-small cell lung cancer to the metabo-
96: 123-70. lism drug phenformin. Cancer Cell 2013; 23: 143-58.
[179] Colman RF. Mechanisms for the oxidative decarboxylation of [205] Morales DR, Morris AD. Metformin in Cancer Treatment and
isocitrate: implications for control. Adv Enzyme Regul 1975; 13: Prevention. Annu Rev Med 2015; 66: 17-29.
413-33. [206] Kalaany NY, Sabatini DM. Tumours with PI3K activation are
[180] Gnoni GV, Priore P, Geelen MJ, et al. The mitochondrial citrate resistant to dietary restriction. Nature 2009; 458: 725-31.
carrier: metabolic role and regulation of its activity and expression. [207] Rizos CV, Elisaf MS. Metformin and cancer. Eur J Pharmacol
IUBMB Life 2009; 61: 987-94. 2013; 705: 96-108.
[181] Jensen-Urstad AP, Semenkovich CF. Fatty acid synthase and liver [208] Dong L, Zhou Q, Zhang Z, et al. Metformin sensitizes endometrial
triglyceride metabolism: housekeeper or messenger? Biochim Bio- cancer cells to chemotherapy by repressing glyoxalase I expression.
phys Acta 2012; 1821: 747-53. J Obstet Gynaecol Res 2012; 38: 1077-85.
[182] Ameer F, Scandiuzzi L, Hasnain S, et al. De novo lipogenesis in [209] Erices R, Bravo ML, Gonzalez P, et al. Metformin, at concentra-
health and disease. Metabolism 2014; 63: 895-902. tions corresponding to the treatment of diabetes, potentiates the cy-
[183] Kersten S. Mechanisms of nutritional and hormonal regulation of totoxic effects of carboplatin in cultures of ovarian cancer cells.
lipogenesis. EMBO Rep 2001; 2: 282-6. Reprod Sci 2013; 20: 1433-46.
[184] Neely JR, Morgan HE. Relationship between carbohydrate and [210] Sandouk T, Reda D, Hofmann C. The antidiabetic agent pioglita-
lipid metabolism and the energy balance of heart muscle. Annu zone increases expression of glucose transporters in 3T3-F442A
Rev Physiol 1974; 36: 413-59. cells by increasing messenger ribonucleic acid transcript stability.
[185] Eaton S, Bartlett K, Pourfarzam M. Mammalian mitochondrial Endocrinology 1993; 133: 352-9.
beta-oxidation. Biochem J 1996; 320 ( Pt 2): 345-57. [211] Braithwaite SS, Palazuk B, Colca JR, et al. Reduced expression of
[186] Houten SM, Wanders RJ. A general introduction to the biochemis- hexokinase II in insulin-resistant diabetes. Diabetes 1995; 44: 43-
try of mitochondrial fatty acid beta-oxidation. J Inherit Metab Dis 48.
2010; 33: 469-77. [212] Pugazhenthi S, Khandelwal RL. Insulin action on protein phospha-
[187] Moritoh Y, Takeuchi K, Hazama M. Combination treatment with tase-1 activation is enhanced by the antidiabetic agent pioglitazone
alogliptin and voglibose increases active GLP-1 circulation, pre- in cultured diabetic hepatocytes. Mol Cell Biochem 1998; 182:
vents the development of diabetes and preserves pancreatic beta- 185-91.
cells in prediabetic db/db mice. Diabetes Obes Metab 2010; 12: [213] Bodles AM, Banga A, Rasouli N, et al. Pioglitazone increases
224-33. secretion of high-molecular-weight adiponectin from adipocytes.
[188] Bin BH, Seo J, Yang SH, et al. Novel inhibitory effect of the an- Am J Physiol Endocrinol Metab 2006; 291: E1100-5.
tidiabetic drug voglibose on melanogenesis. Exp Dermatol 2013; [214] Gu J, Liu X, Wang QX, et al. Beneficial effects of pioglitazone on
22: 541-6. atrial structural and electrical remodeling in vitro cellular models. J
[189] Nagai K, Matsumaru K, Takahashi Y, et al. Effective therapy using Mol Cell Cardiol 2013; 65: 1-8.
voglibose for nonalcoholic steatohepatitis in a patient with insuffi- [215] Hara S, Takahashi T, Amita M, et al. Pioglitazone counteracts the
cient dietary and exercise therapy: exploring other treatment possi- tumor necrosis factor-alpha inhibition of follicle-stimulating hor-
bilities. Case Rep Gastroenterol 2011; 5: 336-43. mone-induced follicular development and estradiol production in
[190] Shimabukuro M, Higa N, Chinen I, et al. Effects of a single ad- an in vitro mouse preantral follicle culture system. J Ovarian Res
ministration of acarbose on postprandial glucose excursion and en- 2013; 6: 69.
dothelial dysfunction in type 2 diabetic patients: a randomized [216] Stefan N, Fritsche A, Weikert C, et al. Plasma fetuin-A levels and
crossover study. J Clin Endocrinol Metab 2006; 91: 837-42. the risk of type 2 diabetes. Diabetes 2008; 57: 2762-7.
[191] Shimabukuro M, Higa N, Takasu N, et al. A single dose of nateg- [217] Ochi A, Mori K, Emoto M, et al. Direct Inhibitory Effects of Pio-
linide improves post-challenge glucose metabolism and endothelial glitazone on Hepatic Fetuin-A Expression. PLoS One 2014; 9:
dysfunction in Type 2 diabetic patients. Diabet Med 2004; 21: 983- e88704.
6. [218] Sarafidis PA, Stafylas PC, Georgianos PI, et al. Effect of thia-
[192] Kato T, Inoue T, Node K. Postprandial endothelial dysfunction in zolidinediones on albuminuria and proteinuria in diabetes: a meta-
subjects with new-onset type 2 diabetes: an acarbose and nateg- analysis. Am J Kidney Dis 2010; 55: 835-47.
linide comparative study. Cardiovasc Diabetol 2010; 9: 12. [219] Nicholas SB, Kawano Y, Wakino S, et al. Expression and function
[193] Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein of peroxisome proliferator-activated receptor-gamma in mesangial
kinase in mechanism of metformin action. J Clin Invest 2001; 108: cells. Hypertension 2001; 37: 722-7.
1167-74.
3620 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Meneses et al.

[220] Mark M, Grell W. Hypoglycaemic effects of the novel antidiabetic [233] Mason R, Jacob RF, Corbalan J, et al. Saxagliptin reduces lesion
agent repaglinide in rats and dogs. Br J Pharmacol 1997; 121: formation and reverses endothelial dysfunction in CETP-apob100
1597-604. mice with atherosclerosis. Circulation 2013; 128: A15118.
[221] Fuhlendorff J, Rorsman P, Kofod H, et al. Stimulation of insulin [234] Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and Cardio-
release by repaglinide and glibenclamide involves both common vascular Outcomes in Patients with Type 2 Diabetes Mellitus. N
and distinct processes. Diabetes 1998; 47: 345-51. Engl J Med 2013; 369: 1317-26.
[222] Bakkali-Nadi A, Malaisse-Lagae F, Malaisse W. Insulinotropic [235] Nowicki M, Rychlik I, Haller H, et al. Saxagliptin improves gly-
action of meglitinide analogs: concentration-response relationship caemic control and is well tolerated in patients with type 2 diabetes
and nutrient dependency. Diabetes Res 1993; 27: 81-87. mellitus and renal impairment. Diabetes Obes Metab 2011; 13:
[223] Malaisse WJ. Repaglinide, a new oral antidiabetic agent: a review 523-32.
of recent preclinical studies. Eur J Clin Invest 1999; 29: 21-29. [236] Han S, Hagan DL, Taylor JR, et al. Dapagliflozin, a selective
[224] Dunning BE. New non-sulfonylurea insulin secretagogues. Expert SGLT2 inhibitor, improves glucose homeostasis in normal and dia-
Opin Investig Drugs 1997; 6: 1041-8. betic rats. Diabetes 2008; 57: 1723-9.
[225] Viñambres C, Villanueva-Peñacarrillo ML, Valverde I, et al. Repa- [237] Devineni D, Morrow L, Hompesch M, et al. Canagliflozin im-
glinide preserves nutrient-stimulated biosynthetic activity in rat proves glycaemic control over 28 days in subjects with type 2 dia-
pancreatic islets. Pharm Res 1996; 34: 83-85. betes not optimally controlled on insulin. Diabetes Obes Metab
[226] Elliott AD, Ustione A, Piston DW. Somatostatin and insulin medi- 2012; 14: 539-45.
ate glucose-inhibited glucagon secretion in the pancreatic -cell by [238] Nyirjesy P, Sobel JD, Fung A, et al. Genital mycotic infections
lowering cAMP. Am J Physiol Endocrinol Metab 2015; 308: E130- with canagliflozin, a sodium glucose co-transporter 2 inhibitor, in
E43. patients with type 2 diabetes mellitus: a pooled analysis of clinical
[227] Leclercq-Meyer V, Ladriere L, Fuhlendorff J, et al. Stimulation of studies. Curr Med Res Opin 2014; 30: 1109-19.
insulin and somatostatin release by two meglitinide analogs. Endo- [239] Dias TR, Rato L, Martins AD, et al. Insulin deprivation decreases
crine 1997; 7: 311-7. caspase-dependent apoptotic signaling in cultured rat sertoli cells.
[228] Chaudhuri A, Ghanim H, Dhindsa SS, et al., Eds. LBSU-1074: ISRN Urol 2013; 2013: 970370.
Exenatide Induces an Increase in Vasodilatory Mediators. Endo- [240] Oliveira PF, Alves MG, Rato L, et al. Effect of insulin deprivation
crine Society's 96th Annual Meeting and Expo Chicago: Obesity on metabolism and metabolism-associated gene transcript levels of
and Diabetes: From Basic Mechanisms to Treatment 2014. in vitro cultured human Sertoli cells. Biochim Biophys Acta 2012;
[229] Aviles-Olmos I, Dickson J, Kefalopoulou Z, et al. Motor and cog- 1820: 84-89.
nitive advantages persist 12 months after exenatide exposure in [241] Karakelides H, Asmann YW, Bigelow ML, et al. Effect of insulin
Parkinson's disease. J Parkinsons Dis 2014; 4: 337-44. deprivation on muscle mitochondrial ATP production and gene
[230] Aviles-Olmos I, Dickson J, Kefalopoulou Z, et al. Exenatide and transcript levels in type 1 diabetic subjects. Diabetes 2007; 56:
the treatment of patients with Parkinson’s disease. J Clin Invest 2683-9.
2013; 123: 2730-6. [242] Zabielski P, Blachnio-Zabielska A, Lanza IR, et al. Impact of insu-
[231] Ellenbroek JH, Töns HAM, van Meeteren MJAW, et al. Glucagon- lin deprivation and treatment on sphingolipid distribution in differ-
like peptide-1 receptor agonist treatment reduces beta cell mass in ent muscle subcellular compartments of streptozotocin-diabetic
normoglycaemic mice. Diabetologia 2013; 56: 1980-6. C57Bl/6 mice. Am J Physiol Endocrinol Metab 2014; 306: E529-
[232] Cantini G, Di Franco A, Samavat J, et al. Effect of liraglutide on 42.
proliferation and differentiation of human adipose stem cells. Mol
Cell Endocrinol 2015; 402: 43-50.

Received: April 23, 2015 Accepted: July 8, 2015

View publication stats

You might also like