Sie sind auf Seite 1von 19

Diabetes, Obesity and Metabolism 13: 99117, 2011.

2010 Blackwell Publishing Ltd

Pharmacological management of type 2 diabetes:


the potential of incretin-based therapies
B. Charbonnel & B. Cariou
Department of Endocrinology and Diabetes, University Hospital, Nantes, France

Management guidelines recommend metformin as the rst-line therapy for most patients with type 2 diabetes uncontrolled by diet and
exercise. Efcacy with metformin therapy is usually of limited duration, which necessitates the early introduction of one or two additional oral
agents or the initiation of injections, glucagon-like peptide-1 (GLP-1) agonists or insulin. Although safe and effective, metformin monotherapy
has been associated with gastrointestinal side effects (20% of treated patients in randomized studies) and is contraindicated in patients with
renal insufciency or severe liver disease. Patients treated with a sulphonylurea are at increased risk for hypoglycaemia and moderate weight
gain, whereas those receiving a thiazolidinedione are subject to an increased risk of weight gain, oedema, heart failure or fracture. Weight gain
and hypoglycaemia are associated with insulin use. Thus, there is an unmet need for a safe and efcacious add-on agent after initial-therapy
failure. Evidence suggests that incretin-based agents, such as GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, can successfully
achieve glycaemic targets and potentially provide cardiovascular and -cell-function benets. This review will examine current approaches for
treating type 2 diabetes and discuss the place of incretin therapies, mainly GLP-1 agonists, in the type 2 diabetes treatment spectrum.
Keywords: diabetes complications, dipeptidyl peptidase-4 inhibitors, DPP-4 inhibitor, exenatide, GLP-1 analogue, glucagon-like peptide-1
receptor agonists, incretin-based therapies, incretin therapy, liraglutide, metformin, saxagliptin, sitagliptin, type 2 diabetes, type 2 diabetes
management, vildagliptin
Date submitted 30 April 2010; date of first decision 23 June 2010; date of final acceptance 17 September 2010

Introduction

Treatment Intensication in Type 2 Diabetes

Type 2 diabetes is a progressive disease, characterized by a


gradual deterioration in glycaemic control because of the continuing loss of -cell function [1]. Thus, combinations of
glucose-lowering drugs are often needed to maintain blood
glucose at target values: glycated hemoglobin (HbA1c) per
Diabetes Control and Complications Trial (DCCT) <7% or
per International Federation of Clinical Chemistry and Laboratory Medicine <53 mmol/mol [2]. The development and
commercialization of new classes of drugs for diabetes has
increased the treatment options and made the management
of hyperglycaemia more complex. The selection of glucoselowering treatments should take into account patient-specific
factors (e.g. age, duration of diabetes, body weight, waist circumference, associated cardiovascular risk factors), as well as
mechanistic effects of various drugs on the main target organs
involved in the pathogenesis of type 2 diabetes (e.g. liver, skeletal
muscle, adipose tissue, -cell, intestine, kidney). In this review,
we specifically focus on the place of the new incretin therapies,
glucagon-like peptide (GLP)-1 receptor agonists and dipeptidyl
peptidase (DPP)-4 inhibitors, in type 2 diabetes treatment.

Metformin: the First Choice at the First Step

Correspondence to: Prof. Bernard Charbonnel, Clinique dEndocrinologie, Maladies


Metaboliques et Nutrition, Hopital G & R Laennec, Boulevard Jacques Monod, 44093
Nantes Cedex 1, France.
E-mail: bernard.charbonnel@univ-nantes.fr

The choice of initial pharmacotherapy when lifestyle changes


alone do not get patients to HbA1c target is relatively simple.
Treatment guidelines [35] (Figure 1) agree in designating
metformin, a biguanide acting as an insulin-sensitizer on the
liver, as the preferred first therapy for most patients, barring
contraindications such as renal disease or chronic hypoxemic conditions that may lead to lactic acidosis. Metformin
is cost-effective, safe, with an extensive clinical experience
with this drug. In addition, metformin does not cause weight
gain and may even be associated with weight loss. In United
Kingdom Prospective Diabetes Study (UKPDS), intensive treatment with metformin in a relatively small group of overweight
patients with newly diagnosed type 2 diabetes reduced the rate
of macrovascular complications vs. conventional treatment.
A significant benefit vs. sulphonylureas or insulin was seen
for all causes of death [6]. Moreover, observational analyses
have also shown reduced rates of all-cause and cardiovascular mortality among patients with type 2 diabetes newly
treated with metformin monotherapy, relative to sulphonylurea monotherapy [7,8]. Recently, the use of metformin was
found to be significantly associated with a reduced mortality
rate in patients with established atherothrombosis who were
included in the Reduction of Atherothrombosis for Continued
Health registry [9]. Metformin use has also been reported to

review
article

review article

review article

DIABETES, OBESITY AND METABOLISM

Tier 1: Well-validated core therapies

At
diagnosis:

Lifestyle + Metformin
+
Basal Insulin

Lifestyle
+
Metformin

Lifestyle + Metformin
+
Sulfonylurea*

STEP 1

STEP 2

Lifestyle + Metformin
+
Intensive Insulin

STEP 3

Tier 2: Less well-validated therapies


Lifestyle + Metformin
+
Pioglitazone
No hypoglycemia
Weight loss
Nausea/vomiting

Lifestyle + Metformin
+

GLP-1 agonist
No hypoglycemia
Weight loss
Nausea/vomiting

Lifestyle + Metformin
+
Pioglitazone
+
Sulfonylurea*

Lifestyle + Metformin
+
Basal Insulin

Figure 1. ADA/EASD (American Diabetes Association/European Association of the Study of Diabetes) algorithm. (Adapted with permission from
Ref. [2]. Copyright 2009 American Diabetes Association). Sulphonylureas other than glibenclamide (glyburide) or chlorpropamide. Insufficient clinical
use to be confident regarding safety.

be associated with a lower rate of different types of cancer in


patients with type 2 diabetes [10].
In patients with intolerance to metformin (20% experienced gastrointestinal disorders and 5% needed to stop
the treatment), sulphonylurea should be an option, especially
when a rapid drop of blood glucose levels is suitable. Alternatively, thiazolidinedione (TZD) should be considered in
subjects with insulin resistance and/or non-alcoholic fatty liver
disease (NAFLD) [11].

Which Adjunctive Therapy to Metformin? The Debate


is Still Open at Step 2
Longitudinal studiesincluding the A Diabetes Outcome
Progression Trial (ADOPT) [12], UKPDS [13] and an analysis of data from the UK General Practice Research
Database [14]have shown that initial pharmacologic
monotherapy (with metformin, a sulphonylurea, or a TZD)
fails to maintain glycaemic control and/or -cell function
over time. However, the rate of deterioration appears to differ between agents; ADOPT indicated that the incidence of
monotherapy failure [defined as fasting plasma glucose (FPG)
>10 mmol/l] at 5 years was lowest (15%) with the TZD rosiglitazone, greater (21%) with metformin, and highest (34%) with
the sulphonylurea glyburide [12].
The failure of initial pharmacotherapy necessitates the introduction of additional treatment. The American Diabetes Association (ADA)/European Association of the Study of Diabetes
(EASD) consensus statement mandates the introduction of
a sulphonylurea (excluding glibenclamide [i.e. glyburide]) or

100 Charbonnel and Cariou

basal insulin as well-validated adjuncts. Alternatively, this


expert consensus suggests that some less well-validated addon treatments could be used in addition to metformin, such as
the TZD pioglitazone or a GLP-1 receptor agonist, especially
in patients at risk of hypoglycaemia [3]. However, this preference for sulphonylurea as a step-1 therapy has been recently
criticized [15], and alternative algorithms have been suggested.
For instance, the UK National Institute for Health and Clinical Excellence (NICE) guidelines recommend the addition of a
sulphonylurea, TZD or DPP-4 inhibitor after step 1 [16]. Alternatively, the Canadian Diabetes Association (CDA) calls for
augmenting initial pharmacotherapy with a second agent suited
to the individual needs of the patient; drug choice is described
as less important than the need to reach glycaemic target [5].
Oral Agents
Sulphonylureas. Sulphonylureas are commonly added to metformin to improve glucose control and can be expected
to decrease HbA1c by 12% (1122 mmol/mol) [3]. Given
that sulphonylureas stimulate insulin secretion regardless of
blood glucose level, these reductions are accompanied by a
substantial risk of hypoglycaemia. In UKPDS, the mean proportion of patients experiencing one or more hypoglycaemic
episodes per year was 11% for those taking chlorpropamide
monotherapy and 17.7% for those taking glibenclamide
monotherapy [6]. In ADOPT, 38.7% of patients taking
glyburide monotherapy experienced at least one minor hypoglycaemia episode throughout the study, while 0.6% of patients
required a hospitalization for severe hypoglycaemia [12].

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

Clinical studies have suggested that long-acting sulphonylureas (e.g. glibenclamide, glimepiride) are associated with
greater risk of hypoglycaemia than short-acting sulphonylureas
(e.g. gliclazide, glipizide) [17,18]. Therefore, glibenclamide (i.e.
glyburide) and chlorpropamide, which are not specific to the cell receptor and can alter ischemic preconditioning protective
effect at the heart level [19], should be avoided in the management of type 2 diabetes [3]. In parallel to hypoglycaemic
events, the use of sulphonylureas is associated with weight gain.
In ADOPT, a 1.6-kg gain in the first year of sulphonylurea
monotherapy was reported [12].
Another consideration in the use of sulphonylureas is the
hypothesis that, as insulin secretagogues, they may accelerate
disease progression by inducing -cell exhaustion. It should be
noted that some, but not all, relevant data support this hypothesis. Although sulphonylureas were found to induce apoptosis
in pancreatic -cells in some preclinical studies [2022], such
an effect was not observed in a subsequent study [23]. Nevertheless, in all clinical studies, the rate of disease progression
appears to be higher with sulphonylureas. ADOPT showed
a faster annual rate of decline in homeostasis model assessment (HOMA)-B with glibenclamide (i.e. glyburide) (6.1%)
than with metformin (3.1%) or rosiglitazone (2.0%) [12]. This
decrease followed an initial 6-month increase in HOMA-B
with each therapy. As noted above and logical considering
the HOMA-B data, the incidence of monotherapy failure was
highest with glyburide in ADOPT [12]. Similar data were
shown on shorter durations of treatment in the Quartet
studies comparing glicazide, metformin and pioglitazone [24].
UKPDS is difficult to interpret in this regard because of the
small subgroup of patients on metformin. Although the rates
of HOMA-B decline were similar among diet/exercise, metformin and sulphonylurea, the slope of HbA1c deterioration
was higher with sulphonylurea [1]. Finally, the assignment for
5 years to nateglinide, a short-acting insulin secretagogue close
to the sulphonylurea family, fails to reduce the incidence of
diabetes in patients with impaired glucose tolerance [25]. In
summary, sulphonylureas appear to act very quickly, with a
fast initial blood glucose decrease but with a poor durability of
action. In spite of promising preclinical data on -cell function (see Section -Cell Function), no good long-term clinical
studies are yet available concerning the glycaemic durability of
either DPP-4 inhibitors or GLP-1 receptor agonists.
Thiazolidinediones. TZDs, which act as insulin sensitizers at
many different sites, have glucose-lowering potency similar
to metformin or sulphonylureas, with 0.51.4% decrease of
HbA1c (615 mmol/mol) [3]. In contrast to sulphonylureas,
TZDs have been hypothesized to preserve -cell function,
based on an apparent long-term decrease in type 2 diabetes
incidence or progression in patients with prediabetes (Diabetes REduction Assessment with ramipril and rosiglitazone
Medication and Actos Now for the prevention of diabetes
trials) [12,2629]. However, this protective effect was not
maintained after a wash-out period [30]. At step 1, a better
durability of glycaemic control was shown with rosiglitazone
(ADOPT) [12], compared with sulphonylureas or metformin.
A similar good durability profile combined with metformin was

Volume 13 No. 2 February 2011

review article
also shown at step 2 with pioglitazone (Quartet) [31] or rosiglitazone [Rosiglitazone Evaluated for Cardiac Outcomes and
Regulation of glycemia in Diabetes (RECORD)] [32]. From a
clinical viewpoint, the occurrence of weight gain with TZDs is a
significant barrier to their wider use. Weight gain on TZD treatment is due in part to fluid retention but mainly to increased fat
mass [33,34]. In ADOPT, rosiglitazone monotherapy resulted
in an average 4.8-kg increase at 5 years [12].
The TZD-dependent fluid retention can induce or worsen
congestive heart failure [35,36]. This side effect is specifically sustained through the peroxisome proliferator-activated
receptor (PPAR)- stimulation of epithelial sodium channelmediated renal salt absorption [37] and could be counterbalanced by the use of amiloride or spironolactone [38].
Following the publication of several meta-analyses, rosiglitazone labelling was amended to carry a warning on a potential
increased risk of myocardial ischemic events [36]. Data on this
possible association are mixed. The results of the RECORD
study of rosiglitazone found no increase in the primary
endpoint of cardiovascular hospitalization or cardiovascular death [hazard ratio (HR) 0.99; 95% confidence interval
(CI) 0.851.16] or myocardial infarction (MI; HR 1.14; 95%
CI 0.801.63) [32]. More recently, the results of the Bypass
Angioplasty Revascularization Investigation 2 Diabetes study
have yielded a cardiovascular benefit from an insulin-sensitising
therapy (with metformin and/or rosiglitazone) compared with
an insulin-providing strategy (with sulphonylureas and/or
insulin) in patients with type 2 diabetes who underwent coronary artery bypass grafting [39]. By contrast, a meta-analysis of
42 randomized, controlled trials has yielded odds ratios of 1.43
(p = 0.03) for MI and 1.64 (p = 0.06) for death from cardiovascular causes with rosiglitazone-treated patients compared
with controls [40].
These data were recently updated with 56 randomized controlled trials of rosiglitazone at least 24 weeks in duration
that reported cardiovascular events [41], including the largest
RECORD trial (32). This meta-analysis confirmed a relative
risk (RR) of 1.28 (p = 0.04), but no increase in risk for cardiovascular mortality. In addition, a retrospective observational
analysis from a cohort of 227 571 patients older than 65 years
indicates that the prescription of rosiglitazone was associated
with an increased risk of stroke, heart failure and all-cause mortality, when compared with prescription of pioglitazone [42].
In the PROspective PioglitAzone Clinical Trial In macroVascular Events, which evaluated the secondary prevention of
macrovascular events in patients with type 2 diabetes, no significant difference was found between pioglitazone and placebo
in the primary composite endpoint of death from any cause,
non-fatal MI, stroke, acute coronary syndrome, leg amputation and coronary or leg revascularization [43]. Significant
improvement was reported with pioglitazone vs. placebo in
a secondary endpoint of all-cause mortality, MI or stroke,
whereas the rates of oedema and heart failure were increased
in the pioglitazone-treated group.
Of note, several trials have reported that rosiglitazone and
pioglitazone may exert clinically relevant differential effects on
lipids. These studies indicate that pioglitazone treatment results
in increased high-density lipoprotein cholesterol (HDL-C)

doi:10.1111/j.1463-1326.2010.01317.x 101

review article
and reduced levels of total low-density lipoprotein cholesterol
(LDL-C), atherogenic small, dense lipid particles and triglycerides compared with rosiglitazone [4446]. The underlying
molecular mechanism that sustains the clinical differences
between rosiglitazone and pioglitazone is the selective PPAR
modulator concept [47]. Altogether, these data strongly suggest
that the cardiovascular safety profile of pioglitazone is superior
to that of rosiglitazone and that pioglitazone should be the first
choice when prescribing a TZD.
Additionally, data from several studies suggest that women
with diabetes [12,48,49], and probably men [50,51], treated
with a TZD, either rosiglitazone and pioglitazone, may be at
greater risk of fracture and reduced bone mass. This effect is
related to PPAR- activation that inhibits bone formation by
diverting mesenchymal stem cells to the adipocytic rather
than to the osteogenic lineage and by directly increasing
osteoclastic activity [52]. The fracture sites linked to TZDs were
located in the distal upper and lower limb, locations different
from those typically found in postmenopausal osteoporosis.
A clinical study specifically powered to assess fracture risk
in postmenopausal women using pioglitazone is ongoing
(ClinicalTrial.gov identifier NCT00708175).
Given the potential risks associated with this class of drug,
pioglitazone should be used with caution in the treatment of
type 2 diabetes [3], with the greatest expected benefit in patients
with an insulin-resistant profile (severe abdominal obesity, low
HDL-C, liver steatosis and/or NAFLD).
Other Oral Agents (Excluding Incretin-based Therapies). Other
oral agents, such as -glucosidase inhibitors or glinides, have
been available for some time, but their use is limited by
weak glucose-lowering potency and the need for multiple daily
administrations. -Glucosidase inhibitors are associated with
gastrointestinal symptoms that lead to frequent discontinuation (2545% of patients in clinical trials) [3]. Frequent dosing
is necessary with glinides because of short circulating halflives [3], and weight gain and hypoglycaemia are associated
with their use [53].
Insulin and the Need for an Early Intensification of Treatment.
When metformin monotherapy fails, the introduction of
insulin is another option [4,5]. Insulin is the most effective
glucose-lowering medication, given that there is no maximum
dose beyond which reduction in glycaemia will not occur
(except in some very insulin-resistant patients), the reason
why insulin is often recommended when HbA1c values are
very high (>9% or >75 mmol/mol) [3]. Drug intolerance or
existing contraindications for any of the oral glucose-lowering
agents are also indications for insulin therapy. In spite of the
rapid and efficacious action provided by insulin therapy, there
is substantial reluctance to initiate insulin after oral antidiabetic
drug (OAD) failure, at least after monotherapy failure at step 2.
Moreover, the ability of patients to monitor their blood glucose
and adjust the insulin dose must be taken into account.
Weight gain and hypoglycaemia are well-known adverse
effects of exogenous insulin. In UKPDS [6], patients assigned
insulin therapy experienced a significantly greater HbA1c
decrease over 10 years compared with those receiving dietbased therapy [6]. Compared with diet-based therapy, however,

102 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

insulin therapy was associated with a greater weight increase


of 4.0 kg [6]. The mean proportion of patients per year with
hypoglycaemic episodes was 1.2% for diet and 36.5% for
insulin [6]. Newer insulin analogues are associated with less
weight gain and incidence of hypoglycaemia [5456].
Finally, retrospective studies employing flawed statistical
analyses have found an association between insulin and insulin
secretagogues such as sulphonylureas with an increased risk of
various types of cancer [57]. Better-designed studies addressing
this important issue are ongoing.
The natural progression of type 2 diabetes suggests that
insulin should be initiated earlier in many patients. Patients
often cite fear of weight gain and hypoglycaemic episodes
as reasons for reluctance to initiate insulin [58]. Such attitudes may contribute to clinical inertia among providers, in
which treatment intensification is delayed in spite of elevated
HbA1c [59]. The extent of such delay was made apparent in
a retrospective database study, which evaluated time to initiation of insulin among 2501 UK general practice patients with
type 2 diabetes who had failed oral polytherapy [HbA1c
8.0% (64 mmol/mol)] [60]. It was estimated that only 25%
of patients started insulin by 1.8 years after failure of oral therapy, and only 50% started insulin by 4.9 years after failure [60].
The consequences of this postponed treatment intensification are clear. Increased time with elevated HbA1c has been
strongly correlated to the likelihood of developing devastating
microvascular and macrovascular complications [61,62]. Even
if elevated blood glucose levels following loss of treatment
efficacy are subsequently brought down again, vascular stresses
may persist after glucose normalization, a phenomenon known
as metabolic legacy [63].
The concept of metabolic legacy argues strongly for early
aggressive treatment of type 2 diabetes to minimize the longterm effects of uncontrolled glycaemia. Two 10-year observational studies have shown a reduction in risk for macrovascular
and microvascular complications with intensive glycaemic control vs. conventional therapy. The DCCT/Epidemiology of Diabetes Interventions and Complications in type 1 diabetes [64]
and a UKPDS post-trial follow-up in type 2 diabetes [65] found
that an initial intensive therapy was associated with a persistently reduced risk of microvascular and macrovascular complications, even after the cessation of the interventional period.

Insulin Therapy Initiation and/or Intensication at


Step 3
When glucose control with two agents is inadequate, most
patients may benefit from the initiation or intensification of
insulin therapy to achieve target glycaemia [HbA1c < 7% (<53
mmol/mol)].
The addition of a third oral agent may be considered as an
alternative for those with glycaemia not too far from target [3].
For instance, rosiglitazone combined with metformin and a
sulphonylurea resulted in similar HbA1c improvements except
when baseline HbA1c was 9.5% (80 mmol/mol) [66],
compared with insulin glargine patients in whom insulin was
more effective. The NICE guidelines indicate that, at step 3,
insulin, an OAD, or a GLP-1 receptor agonist may be selected,

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

based on individual patient needs [16]. The CDA recommends


the addition of another oral agent, introduction of bedtime
basal insulin, or intensification of existing insulin therapy [5].
However, the application of guideline recommendations is not
always straightforward or simple. Actually, the complexity level
is rapidly increasing as a result of the growing emergence of
new classes of blood glucose-lowering drugs.

GLP-1 Receptor Agonists and DPP-4


Inhibitors in the Management of Type 2
Diabetes
In view of their generally limited efficacy over time, potential
adverse effects associated with established OADs and reluctance
to intensify therapy, prescribers may consider two recently
introduced drug classes, based on the incretin physiology: the
GLP-1 receptor agonists and DPP-4 inhibitors.

Physiology of the Incretins and Implications


for Treatment
The native human GLP-1 hormone plays a key role in glucose
homeostasis. Secreted from L-cells of the intestinal mucosa
following food ingestion [67], GLP-1 decreases postmeal
glycaemia via several pathways. First, GLP-1 stimulates insulin
production and secretion by pancreatic -cells. This effect
is glucose-dependent, that is, occurs only in the presence
of elevated blood glucose levels [68]. The glucose-dependent
character of this mechanism provides a rationale for the low
potential of GLP-1-based therapies to induce hypoglycaemia.
Second, GLP-1 decreases glucagon secretion by pancreatic
-cells, thereby reducing hepatic glucose release. As with the
effect of GLP-1 on -cells, this activity decreases in the presence
of low blood glucose, allowing the normal counter-regulatory
response to hypoglycaemia to occur [69,70]. GLP-1 also slows
gastric emptying, delaying nutrient absorption [71,72], and
promotes the feeling of satiety, thereby decreasing food
intake [73,74]. Animal data suggest that the effects of GLP-1
on satiety may be via direct effects on the brain [75].
It is well established that the incretin effect is diminished
in patients with type 2 diabetes [76]. While initial studies
have suggested that GLP-1 concentrations are reduced in
patients with type 2 diabetes [77], there is actually little evidence
sustaining the hypothesis that impairment in incretin secretion
plays a role in the pathogenesis of type 2 diabetes [78]. The
diminution of incretin effect in patients with type 2 diabetes
is probably as a result of global -cell dysfunction linked
to chronic hyperglycaemia rather than a primary defect in
glucose-dependent insulinotropic polypeptide (GIP) or GLP-1
action [78]. However, it seems that GLP-1 retains significantly
more efficacy than GIP, another incretin hormone, in type 2
diabetes [79]. Unlike GLP-1 [80], raising GIP doses even to
highly supraphysiologic concentrations fails to compensate the
insulinotropic activity in patients with type 2 diabetes [81].
GLP-1, therefore, provided a more logical target than GIP for
incretin-based therapies. The brief half-life of native GLP-1 in
humans (2 min) [82] would necessitate constant exogenous
infusion to be used as a type 2 diabetes therapy; drug

Volume 13 No. 2 February 2011

review article
development has focused on augmenting or extending the
action of native GLP-1.
GLP-1 receptor agonists provide pharmacologic levels of
GLP-1 receptor stimulation beyond those that would occur
from the action of the native hormone alone, and DPP-4
inhibitors preserve endogenous GLP-1 by decreasing its degradation by the DPP-4 enzyme. Indeed, the doses of GLP-1
receptor agonists, like exenatide and liraglutide, result in levels
of the active agent that are fivefold higher compared with
physiologic levels of GLP-1, even when considering the peak
concentrations after meal. By contrast, DPP-4 inhibition only
results in about a twofold increase in GLP-1, within the physiologic range. Both classes of agents have shown glycaemic benefits, with GLP-1 receptor agonists typically associated with more
robust antiglycaemic effects [83]. Clinically, there are other differences as well. DPP-4 inhibitors do not inhibit gastric emptying in humans [84,85], but GLP-1 receptor agonists do [86,87].
GLP-1 agonists, but not DPP-4 inhibitors, have been observed
to increase satiety in animal models. Treatment with the GLP-1
receptor agonist liraglutide was associated with decreased food
intake and a reduced preference for fat/simple carbohydrates in
rats. These effects were not observed with the DPP-4 inhibitor
sitagliptin [88]. As an expected clinical consequence, DPP-4
inhibitors are weight neutral in clinical trials, compared with the
weight loss seen with GLP-1 receptor agonists. Adverse events
profiles also differ, with more frequent gastrointestinal adverse
events in GLP-1 receptor agonist-treated patients [8997].

GLP-1 Receptor Agonists


GLP-1 agonists may represent an option for patients who experience inadequate glycaemic control with metformin alone or
with dual oral therapy. These agents may be preferred for
patients in whom a substantial HbA1c reduction and/or weight
loss is desirable or when hypoglycaemia is a concern [3]. The
NICE guidelines recommend exenatide or liraglutide only at
step 3, in patients who fail two oral drugs and in whom weight
gain or potential drawbacks of insulin are a concern, and propose a pragmatic approach to evaluate the usefulness of GLP-1
agonists as a third-line therapy after metformin and sulphonylurea failure. In this way, continued therapy with GLP-1 agonists is only endorsed in the presence of a beneficial metabolic
response, defined as a 1% (11 mmol/mol) decrease in HbA1c
and 3% reduction in weight from baseline at 6 months [16].

Exenatide and Liraglutide: Blood


Glucose-lowering Efcacy, Weight Loss,
Hypoglycaemia and Tolerability
The following sections review data on available GLP-1 receptor agonists (exenatide twice daily, liraglutide once daily)
when used early in the type 2 diabetes disease course,
whether as an add-on to one oral agent or as monotherapy
(Table 1) [89127].

GLP-1 as Alternative Options After Monotherapy


Failure (at Step 2)
Exenatide. At a dosage of 510 g twice daily, exenatide monotherapy decreased HbA1c by 0.70.9% (810

doi:10.1111/j.1463-1326.2010.01317.x 103

review article

DIABETES, OBESITY AND METABOLISM

Table 1. Changes in glycaemic and weight parameters with incretin therapies in randomized, controlled trials (changes are from baseline).

Regimen
Monotherapy
Exenatide 510 g BID
Liraglutide 1.21.8 mg OD
Sitagliptin 25200 mg/day
Vildagliptin
50100 mg/day

Saxagliptin 2.510 mg/OD


Combined with 1 OAD
Exenatide 510 g BID
Liraglutide 0.61.8 mg OD
Sitagliptin 100 mg OD

Vildagliptin
50100 mg/day
Saxagliptin 2.510 mg OD
Combined with two OADs
Exenatide 510 g BID

Liraglutide 1.21.8 mg OD
Sitagliptin 100 mg OD

Study

 HbA1c, % (mmol/mol)

FPG, mmol/l

Weight, kg

Hypoglycaemia
incidence, %

Moretto et al. [89]


Garber et al. [121]
Aschner et al. [92], Hanefeld
et al. [93], Raz et al. [94]
Foley et al. [102], Pi-Sunyer
et al. [113], Rosenstock
et al. [114], Schweizer
et al. [115]
Rosenstock et al. [105]

0.7 to 0.9 (8 to 10)


0.8 to 1.1 (9 to 12)
0.3 to 0.8 (3 to 9)

1.0
0.8 to 1.4
0.6 to 0.9

2.8 to 3.1
2.0 to 2.4
0.1 to 0.8

4 to 5
8 to 12
0.8 to 1.8

0.5 to 1.1 (5 to 12)

0.2 to 1.3

+0.8 to 0.4

0 to 0.7

0.4 to 0.5 (4 to 5)

0.5 to 0.9

0.1 to 1.2

5.2

DeFronzo et al. [90], Buse


et al. [91]
Marre et al. [103], Nauck
et al. [104]
Charbonnel et al. [95], Raz
et al. [96], Rosenstock
et al. [97]
Ferraninni et al. [101], Garber
et al. [110], Bosi et al. [111],
Garber et al. [112]
Chacra et al. [99], DeFronzo
et al. [100], Allen et al. [108]

0.4 to 0.9 (4 to 10)

0.3 to 0.6

0.9 to 2.8

4.5 to 36

0.6 to 1.1 (7 to 12)

0.7 to 1.7

+0.7 to 2.8

3 to 9.2

0.7 to 1.0 (8 to 11)

0.9 to 1.6

+1.8 to 0.7

1.0 to 1.3

0.4 to 1.0 (4 to 11)

0.1 to 1.1

+2.7 to 0.4

0 to 1.7

0.5 to 0.9 (5 to 10)

0.4 to 1.2

+0.8 to 1.4

0.5 to 14.6

Buse et al. [98], Drucker


et al. [109], Zinman
et al. [116], Kendall
et al. [117], Heine et al. [118]
Buse et al. [98], Russell-Jones
et al. [106], Zinman et al. [107]
Hermansen et al. [127]

0.6 to 1.5 (7 to 16)

0.5 to 1.6

1.6 to 3.6

5.7 to 33.6

1.1 to 1.5 (12 to 16)

1.6 to 2.4

1.0 to 3.2

8 to 27

0.4 (4)

0.2

+0.8

12

BID, twice daily; FPG, fasting plasma glucose; HbA1c, glycated haemoglobin; OAD, oral antidiabetic drugs; OD, once daily.
Included one episode that required the assistance of another person.
Included one severe episode as judged by the study investigator.
Included three episodes that required the assistance of another person. Range does not include hypoglycaemia incidence from the study comparing
exenatide with insulin glargine. In that study, hypoglycaemia was reported as events per patient-year (7.3 for exenatide and 6.3 for insulin glargine), not as
a percentage.
Included five episodes that required the assistance of another person.

mmol/mol) from baseline at 24 weeks in drug-nave patients.


Weight decreased from baseline by 2.83.1 kg, and the incidence of minor hypoglycaemia was 45%, with no severe
episodes, validating glucose dependence when the GLP-1 receptor agonists are used alone. The incidence of gastrointestinal
symptoms was 313% for nausea, 4% for vomiting and 03%
for diarrhoea [89].
Exenatide 510 g twice daily lowered HbA1c from baseline
by 0.40.8% (49 mmol/mol) in combination with metformin and 0.50.9% (510 mmol/mol) in combination with
a sulphonylurea [90,91]. Weight loss in the two studies ranged
from 0.9 to 2.8 kg. The incidence of mild-to-moderate hypoglycaemia was 4.55.3% with exenatide in combination with
metformin and, as would be expected, was higher in the study
with a concomitant sulphonylurea (1436%). However, no
severe episodes were reported. Rates of gastrointestinal adverse
events in both studies were 3651% for nausea. Nausea was
most common in the first 8 weeks of treatment and subsided

104 Charbonnel and Cariou

thereafter [90,91]. The dropout rate linked to nausea was 6%


in randomized studies with exenatide [128].
Patients enrolled in randomized controlled studies of
exenatide added to metformin and/or a sulphonylurea entered
into an open-label extension study of exenatide 10 g twice
daily, yielding data on 3 years of exposure. At 3 years, a 1%
(11 mmol/mol) HbA1c reduction was maintained and weight
loss increased progressively to 5.3 kg from baseline to the end
of the extension period [119].
Liraglutide. Liraglutide was assessed in a program of six phase
III trials [Liraglutide Effect and Action in Diabetes (LEAD)]
designed to validate its efficacy both as monotherapy and
in combination with commonly used OADs [120]. In LEAD
3, a 52-week monotherapy study of liraglutide 1.21.8 mg
once daily, HbA1c decreased by 0.81.1% (912 mmol/mol).
Moreover, in drug-nave patients, a significantly greater reduction in HbA1c was observed with liraglutide 1.8 mg compared

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

with glimepiride at 52 weeks (1.6 vs. 0.88% [17 vs.


10 mmol/mol], p < 0.0001). Weight decreased by 2.02.4 kg
(both from baseline). The incidence of minor hypoglycaemia
was 812%, with no major episodes, and the incidences of
gastrointestinal adverse events ranged from 28 to 29% for
transient nausea, 9 to 12% for vomiting and 16 to 19% for
diarrhoea [121].
Subjects continued to receive liraglutide 1.21.8 mg once
daily during a 1-year extension phase. HbA1c reduction
[0.9 to1.1% (10 to 12 mmol/mol)] and weight loss
(2.12.7 kg) vs. baseline were sustained at the end of year
2 [122].
In addition, two 26-week studies evaluated liraglutide
0.61.8 mg once daily added to metformin (LEAD 2) [104]
or a sulphonylurea (LEAD 1) [103]. HbA1c reductions from
baseline were 0.61.1% (712 mmol/mol) in the two trials. In
a subgroup of patients previously treated with prestudy oral
antihyperglycaemic monotherapy (35% of treated patients),
and thus representing a true monotherapy cohort, the addition of liraglutide to metformin reduced HbA1c as effectively
as glimepiride (1.3 to 1.2% [14 to 13 mmol/mol],
p = NS) [85]. Weight decreased from baseline by 1.82.8 kg
in the metformin combination study, whereas weight change
was negligible (<1 kg) with liraglutide in combination with
sulphonylurea therapy, which is known to increase weight.
Minor hypoglycaemia was somewhat more frequent when
liraglutide was used in combination with sulphonylurea therapy (5.29.2%) than with metformin (3%). To minimize the
risk of hypoglycaemia, it is recommended to reduce by half
the dose of sulphonylurea when initiating a treatment with a
GLP-1 receptor agonist. Across the two studies, the occurrence
of nausea was 1119% and generally subsided after 4 weeks of
treatment [103,104]. The dropout rate because of severe nausea was 2.8% in the LEAD studies [98,103,104,106,107,121],
compared with a 4.1% discontinuation rate because of gastrointestinal side effects with metformin 1500 mg/day in a
24-week trial [129]. It should be stressed that in the LEAD
studies, all patients were subjected to a forced-dose titration to
attain 1.8 mg once daily. In clinical practice, patients experiencing nausea could down-titrate if necessary. One possible way
to reduce the incidence of nausea is to ask for the patients to
reduce their food intake during the initiation of the treatment.

Use of GLP-1 Receptor Agonists in Patients


With Inadequate Response to Two Oral Therapies
(at Step 3)
The treatment of combination failure is even more complex
than addressing inadequate pharmacologic monotherapy. As
the number of co-morbidities and concomitant medications
can be expected to increase in more advanced diabetes,
clinicians need to consider how to address overweight and
obesity, cardiovascular risk and potential drug interactions
most effectively.
Exenatide. Exenatide 10 g twice daily added to a TZD, with or
without metformin, was associated with a 0.9% (10 mmol/mol)
decrease in HbA1c and a 1.75-kg decrease in weight at 16
weeks. Hypoglycaemia occurred in 10.7% of patients, with

Volume 13 No. 2 February 2011

review article
no severe episodes [116]. In a 30-week study of exenatide
510 g twice daily added to metformin and a sulphonylurea,
reductions were 0.60.8% (79 mmol/mol) for HbA1c and
1.6 kg for weight. Hypoglycaemia was reported in 19.027.8%
of patients in the exenatide group (including one episode of
severe hypoglycaemia). Gastrointestinal adverse events were
the most common [117]. A 30-week study evaluated exenatide
10 g twice daily as monotherapy, added to one OAD, or
added to two OADs. HbA1c decreased by 1.5% (16 mmol/mol),
weight loss was 3.6 kg, and hypoglycaemia episodes (all minor)
occurred in 5.7% of patients. Rates of nausea (34%), vomiting
(19%) and diarrhoea (13%) were similar to those seen in
other studies of exenatide [109]. In a 26-week randomized
study, both exenatide and insulin glargine similarly reduced
HbA1c levels by 1.1% (12 mmol/mol) in patients with type 2
diabetes inadequately controlled with combination metformin
and sulphonylurea therapy [118]. However, patients receiving
exenatide had fewer postprandial glucose (PPG) excursions
compared with those taking insulin glargine. Change in weight
was significantly better with exenatide (2.3 vs. +1.8 kg with
insulin glargine). Similar rates of symptomatic hypoglycaemia
were observed between the two groups. Patients in the exenatide
group reported more nausea (57.1 vs. 8.6%), vomiting (17.4
vs. 3.7%) and diarrhoea (8.5 vs. 3.0%) compared with those in
the insulin glargine group.
Liraglutide. Liraglutide 1.21.8 mg once daily, combined with
metformin and a TZD for 26 weeks (LEAD 4), was associated with an HbA1c decrease of 1.5% (16 mmol/mol) and
weight loss of 1.0 to 2.0 kg. Rates of minor hypoglycaemia
were 8.09.0%, with no major episodes. Nausea occurred
in 2940% of patients, and vomiting occurred in 717%.
Most gastrointestinal adverse events were transient, occurring
within the first month of the treatment [107]. Use of liraglutide 1.8 mg once daily, in combination with metformin and
a sulphonylurea, resulted in an HbA1c reduction of 1.33%
(14 mmol/mol) at 26 weeks. Most important, this decrease was
significantly greater than that observed with insulin glargine
(1%; 11 mmol/mol), when used as a trial comparator.
Moreover, the extent of the decrease in FPG was similar between
the two arms (1.6 vs. 1.8 mmol/l, liraglutide 1.8 mg vs.
glargine, respectively). Weight decreased significantly by 1.4 kg
vs. placebo and by 3.4 kg vs. glargine. Minor hypoglycaemia
was reported by 27% of patients, and five patients experienced
a major episode. Transient nausea occurred in 14%, vomiting
in 6% and diarrhoea in 10% [106].
A head-to-head trial compared liraglutide 1.8 mg once daily
and exenatide 10 g twice daily (LEAD 6), each added to metformin and/or a sulphonylurea for 26 weeks. HbA1c decreased
significantly more with liraglutide (1.12%; 11 mmol/mol) compared with exenatide (0.79%; 9 mmol/mol), even though the
incidence of minor hypoglycaemia (blood glucose <3.1 mmol/l
and no necessity for assistance from another person) was significantly lower with liraglutide (26 vs. 34%). As previously
shown in the LEAD 5 study, liraglutide was more effective in
controlling FPG than was exenatide (1.61 vs.0.60 mmol/l,
respectively), because of a more prolonged pharmacokinetics.
By contrast, weight loss was similar with liraglutide (3.2 kg)
and exenatide (2.9 kg) [98].

doi:10.1111/j.1463-1326.2010.01317.x 105

review article
DPP-4 Inhibitors
DPP-4 Inhibitors: Blood Glucose-lowering Efficacy, Weight Loss,
Hypoglycaemia and Tolerability. DPP-4 inhibitors are not a
homogeneous chemical class, but these different chemical products share the same mechanisms of action: DPP-4 inhibition
over 24 h, after oral intake once daily (sitagliptin, saxagliptin),
or twice daily (vildagliptin). While DPP-4 inhibitors are
not included among preferred antidiabetes agents in the
ADA/EASD algorithm [3], the NICE guidelines recommend
the consideration of DPP-4 inhibitors as an add-on therapy
following metformin failure, or as a third-line triple oral therapy option, when sulphonylurea therapy is contraindicated or
not tolerated or when risk of hypoglycaemia is a major concern [12]. Data from trials of DPP-4 inhibitors as monotherapy
or added to one OAD indicate that this class exerts moderate HbA1c reductions, is weight neutral, and does not induce
hypoglycaemia (Table 1).
A DPP-4 inhibitor must be given at a dose which inhibits
DPP-4 >80% for 24 h to get its full efficacy, while increasing
the dose above will do not add more efficacy. It appears that
all the commercially available DPP-4 inhibitors share similar
blood glucose-lowering efficacy. The small differences between
studies came from the differences in the patient populations. To
date, however, it cannot be excluded that each DPP-4 inhibitor
might have different safety profiles, because of differences in
chemistry profile or DPP-4 inhibition selectivity.
Sitagliptin. Twelve to twenty-four weeks of monotherapy
with sitagliptin 25200 mg/day (the recommended dose is
100 mg/day) decreased HbA1c by 0.30.8% (39 mmol/mol)
and decreased weight by 0.10.8 kg from baseline. The incidence of hypoglycaemia was low (0.81.8%), and common
adverse events included nasopharyngitis (2.99.1%) and upper
respiratory tract infection (2.98.8%) [9294], which have
not been confirmed in a larger meta-analysis of the phase
III trials [130]. Sitagliptin 100 mg once daily added to an
OAD (metformin or a TZD) for 2430 weeks resulted in
HbA1c reduction of 0.71.0% (811 mmol/mol) and weight
changes of 0.7 to +1.8 kg from baseline. The incidence of
hypoglycaemia ranged from 1.0 to 1.3%, and incidences were
4.07.3% for nasopharyngitis and 07.3% for respiratory tract
infection [9597].
Longer-term pooled clinical trial data are available for
sitagliptin used as monotherapy or added to metformin. In the
monotherapy pooled data analysis, from which data subsequent
to glycaemic rescue were excluded, the 2-year reduction from
baseline in HbA1c was 1.6% (17 mmol/mol) for sitagliptin
monotherapy. Pooled analysis of two trials in which sitagliptin
was added to metformin showed that the combination reduced
HbA1c by 1.1% (12 mmol/mol) [123].
A 2-week, cross-over study has evaluated the effect of exenatide (510 g BID) vs. sitagliptin (100 mg/day) on 2-h PPG
in type 2 diabetes patients treated with metformin. While the
reduction in FPG was similar between the two groups, 2-h PPG
was lower with exenatide than sitagliptin (133 6 vs. 208
6 mg/dl, p < 0.0001). This effect was associated with slowed
gastric emptying and reduced food intake in the exenatide
group [124]. A liraglutide vs. sitagliptine head-to-head 26-week

106 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

randomized study in patients with type 2 diabetes inadequately


controlled by metformin has been recently published [83].
The HbA1c reduction was significantly greater with liraglutide 1.2 mg (1.24%; 13 mmol/mol) and 1.8 mg (1.50%;
16 mmol/mol) once daily than with sitagliptin [0.90%
(10 mmol/mol), p < 0.0001 for both comparisons], showing
the superiority of GLP-1 agonism (high pharmacologic circulating levels of the drug) vs. DPP-4 inhibition (doubling of
endogenous circulating GLP-1 levels). Mean weight loss was
significantly greater with liraglutide than with sitagliptin after
26 weeks: 3.38 kg for 1.8 mg liraglutide, 2.86 kg for liraglutide 1.2 mg and 0.96 kg for sitagliptin. But nauseas were more
common with liraglutide (21 and 27% patients for 1.2 mg and
1.8 mg, respectively) than with sitagliptin (5% patients).
Vildagliptin. Owing to a shorter half-life than sitagliptin,
vildagliptin has to be given twice a day to fully inhibit DPP-4,
even though the clinical differences in blood glucose-lowering
efficacy between 100 mg once a day or 50 mg twice a day are
small.
Vildagliptin 50100 mg/day as monotherapy for 2452
weeks yielded HbA1c reductions of 0.51.1% (612
mmol/mol) from baseline, weight change of 0.4 to
+0.3 kg from baseline, and hypoglycaemia incidence of
00.6%. Adverse events included headache (4.910.0%) and
nasopharyngitis (3.513.2%) [113115]. A 2-year study of
vildagliptin 100 mg/day in previously drug-nave patients with
type 2 diabetes found a 0.5% (6 mmol/mol) decrease in HbA1c
and a 0.8-kg increase in body weight from baseline, with a 0.7%
incidence of hypoglycaemia. The most common adverse events
were nasopharyngitis (8.8%), influenza (8.4%) and headache
(7.7%) [102].
At the end of a 1-year extension to a 1-year randomized
controlled study [115], vildagliptin 100 mg monotherapy was
associated with a 1.0% (11 mmol/mol) reduction in HbA1c
and a 0.5 kg increase in weight from baseline [125]. Two-year
extension data from patients enrolled in a 24-week, randomized, controlled monotherapy study [114] reported a 0.82%
(9 mmol/mol) reduction in HbA1c and unchanged weight
(0.002 kg) from baseline [126].
Added to an OAD (metformin, a sulphonylurea or a TZD)
for 2452 weeks, vildagliptin 50100 mg/day reduced HbA1c
by 0.41.0% (411 mmol/mol). Change in weight was 0.4
to +2.7 kg. Hypoglycaemia incidence was low (03.6%), and
common adverse events included nasopharyngitis (4.711.3%)
and headache (3.27.6%) [101,110112].
Saxagliptin. Saxagliptin inhibits DPP-4 over 24 h because of a
long-acting active metabolite. The standard dose of saxagliptin
is 5 mg once daily, which inhibits DPP-4 >70% for 24 h.
Saxagliptin 2.510 mg/day monotherapy in previously drugnave patients with type 2 diabetes was associated with
a decrease in HbA1c of 0.40.5% (45 mmol/mol) and a
reduction in weight of 0.11.2 kg at 24 weeks. Hypoglycaemia
occurred in 5.2% of saxagliptin-treated patients, and common
adverse events were dermatologic changes (13.7%), upper
respiratory tract infection (8.8%) and headache (8.2%) [105].
In three 24-week studies of saxagliptin 2.510 mg once
daily added to an OAD (metformin, a sulphonylurea or a

Volume 13 No. 2 February 2011

review article

DIABETES, OBESITY AND METABOLISM

TZD), HbA1c decreased by 0.50.9% (510 mmol/mol) from


baseline. Change in weight from baseline ranged from 1.4 to
+0.8 kg. The incidence of hypoglycaemia was 0.50.6% in the
study of saxagliptin added to metformin, 2.74.1% in the study
of saxagliptin added to a TZD, and 13.314.6% in the study of
saxagliptin added to a sulphonylurea. Common adverse events
included urinary tract infection (5.010.7%), nasopharyngitis
(5.69.9%) and headache (5.89.4%) [99,100,108].

Additional Benecial Effects of Incretin-based


Therapies
Cardiovascular Parameters. Of potential cardioprotective
interest, GLP-1 receptor agonists have been shown to impact
other cardiovascular risk factors such as blood pressure and
plasma lipid profiles. Because the GLP-1 receptor is expressed
in the heart and the vasculature, a direct effect of GLP-1
receptor activation on the vascular tone could be hypothesized. However, the cardiovascular actions of GLP-1 remain
to be further investigated in humans [131]. Alternatively, the
natriuretic effect of GLP-1 could explain the antihypertensive
properties of the incretins [132]. Moreover, promising data
have emerged about a cardioprotective role for GLP-1 agonists
following MI in mice [133] and humans [134].
Exenatide. Although long-term outcomes studies are lacking, exenatide does improve markers of cardiovascular risk
(Table 2) [89101,103,104,106117,121]. Blood pressure and
lipid data were reported in a monotherapy study. A 3.7-mm
Hg decrease in systolic blood pressure (SBP) from baseline occurred with both exenatide 5 and 10 g twice daily.
Decreases from baseline in diastolic blood pressure (DBP)
were 0.8 mm Hg for 5 g and 2.3 mm Hg for 10 g. No
significant changes in lipid parameters occurred [89]. A study
of exenatide added to a sulphonylurea found no significant
change in total cholesterol or triglycerides but showed a significant (but unspecified) reduction in LDL-C from baseline [91].
Cardiovascular parameters improved at 3 years of follow-up

in patients originally enrolled in three 30-week, double-blind


trials. Significant improvements from baseline were observed
in lipids (44.4 mg/dl for triglycerides, 10.8 mg/dl for total
cholesterol, +8.5 mg/dl for HDL-C, and 11.8 mg/dl in LDLC) and blood pressure (3.5 mm Hg for SBP and 3.3 mm
Hg for DBP) [119]. In a study of exenatide as monotherapy
or added to one or two OADs, DBP decreased from baseline (1.7 mm Hg) and no significant effect on SBP was
observed. Triglycerides significantly decreased from baseline
(11%), but beneficial HDL-C also significantly decreased
(0.03 mmol/l) [109]. Exenatide was associated with a reduction in C-reactive protein (CRP) from baseline in an analysis
of extension data [135].
Liraglutide. Liraglutide has also been shown to improve cardiovascular parameters, although long-term outcomes data are
as yet lacking. Across clinical studies, liraglutide 0.61.8 mg
once daily consistently decreased SBP relative to baseline,
by 0.66.7 mm Hg (Table 2) [98,103,104,106,107,121]. Blood
pressure decreased in the first 2 weeks of treatment, prior
to weight loss, suggesting that the improvement in blood
pressure was not dependent on the weight loss [136,137].
Liraglutide significantly improved lipid parameters (total
cholesterol 0.13 mmol/l, triglycerides 0.20 mmol/l, and
LDL-C 0.20 mmol/l) and cardiovascular biomarkers [B-type
natriuretic peptide 11.9%, high-sensitivity CRP (hsCRP)
23.1%] from baseline in a meta-analysis of six clinical
studies [138].
DPP-4 Inhibitors. Data are scarce on the potential impact
of DPP-4 inhibitors on blood pressure or lipid parameters
across clinical studies (Table 2) [9297,101,102,105,110115].
A trial of saxagliptin 2.5 to 5.0 mg/day yielded reductions of
3.23.9 mm Hg in SBP and 1.83.3 mm Hg in DBP [99], but
effects on blood pressure are not reported in other clinical
trials [100,108]. In the recent liraglutide vs. sitagliptin headto-head study, lowering of DBP was significantly greater with
sitagliptin 100 mg/day (1.78 mm Hg) than with liraglutide

Table 2. Incretin therapies and markers of cardiovascular risk: evidence from randomized, controlled trials.

Agent

Study

Reductions in blood pressure


( mm Hg from baseline)

Significantly improved lipids and


other CV markers (vs. comparators)

Exenatide

Moretto [89], DeFronzo [90], Buse [91], Buse et al. [98],


Drucker et al. [109], Zinman [116], Kendall [117]

2.5 to 3.7 SBP


0.8 to 2.3 DBP

LDL-C

Liraglutide

Buse et al. [98], Marre et al. [103], Nauck et al. [104],


Russell-Jones et al. [106], Zinman et al. [107],
Garber et al. [121]

0.6 to 6.7 SBP

LDL-C, VLDL-C, TG, FFA

Sitagliptin

Aschner et al. [92], Hanefeld et al. [93], Raz et al. [94],


Charbonnel et al. [95], Raz et al. [96], Rosenstock
et al. [97], Foley [102], Hermansen et al. [127]
Ferrannini et al. [101], Garber et al. [110], Bosi et al. [111],
Garber et al. [112], Pi-Sunyer et al. [113],
Rosenstock et al. [114], Schweizer et al. [115]

Not reported

TG, TC, LDL-C, non-HDL-C,


HDL-C, TG-to-HDL-C

No major changes
in vital signs
across studies

TG, TC, VLDL-C, non-HDL-C

Chacra et al. [99], DeFronzo et al. [100], Allen et al. [108]

3.2 to 3.9 SBP


1.8 to 3.3 DBP

None reported

Vildagliptin

Saxagliptin

CV, cardiovascular; DBP, diastolic blood pressure; FFA, free fatty acids; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein
cholesterol; SBP, systolic blood pressure; TC, total cholesterol; TG, triglycerides; VLDL-C, very low-density lipoprotein cholesterol.
The listed improvements occurred in at least one of the studies cited.

Volume 13 No. 2 February 2011

doi:10.1111/j.1463-1326.2010.01317.x 107

review article
1.8 mg/day (+0.07 mg Hg) [83]. However, the reduction of
SBP and DBP was similar between sitagliptin 100 mm Hg
and liraglutide 1.2 mg/day. Of note, heart rate increased with
liraglutide and decreased slightly with sitagliptin. Concerning
the other cardiovascular biomarkers, saxagliptin did not affect
the levels of hsCRP, platelet activation factor-1, fibrinogen or
interleukin-6 in one trial [105].
Large, long-term cardiovascular outcome studies are
planned with liraglutide (LEAD: Evaluation of Cardiovascular Outcome Results; ClinicalTrial.gov identifier NCT01179048)
and with DPP-4 inhibitors. The most advanced of the DPP4 studies is the Trial to Evaluate Cardiovascular Outcomes
after treatment with Sitagliptin (ClinicalTrial.gov identifier
NCT00790205). The primary end point of these studies is
to validate the cardiovascular safety of these drugs, but cardiovascular benefit may be prespecified as a secondary end point.
-Cell Function. Preclinical studies are encouraging, but clinical data are limited to surrogate parameters of -cell function,
the most controversial being HOMA-B which may simply
reflect the ability of a drug to stimulate insulin. Clearly, a good
glycaemic clinical durability study (like ADOPT for the TZDs)
is lacking in this field.
Exenatide. Exenatide appears to exert a beneficial effect on
-cells, which could have implications for disease progression,
although long-term outcomes research is needed. -cell mass
increased in insulin-resistant obese fa/fa Zucker rats treated
with exenatide [139]. In vitro and in mice, exenatide inhibited
-cell apoptosis [140], and exposure to exenatide promoted
replication and neogenesis of -cells in a rat model of type
2 diabetes [141]. Exenatide-protected islet allografts against
rejection and increased insulin secretion post-transplant in
diabetic non-human primates (Macaca fascicularis) [142].
Although changes in -cell mass cannot be evaluated
in humans, exenatide achieved significant improvements
vs. placebo in indices of -cell function: HOMA-B (1932%) [89,116] and proinsulin:insulin ratio [90,91]. An analysis
of pooled data indicated that exenatide improved the postprandial -cell response [143]. Patients with type 2 diabetes
who received exenatide for 1 year experienced improvement
in indices of -cell function, vs. baseline and compared with
insulin glargine (for a similar HbA1c reduction), although it
should be noted that these benefits were not sustained after a
12-week washout period [144]. Exenatide has been observed
to increase insulin secretion following islet transplantation in
type 1 diabetes [145].
Liraglutide. Liraglutide has also been shown to increase
-cell mass in diabetic mice [146] and Zucker diabetic fatty
rats [147], to inhibit -cell apoptosis by cytokines and free
fatty acids in rat islet cells [148], and to improve engraftment and function of islet transplants in diabetic mice [149]
and in a porcine marginal mass islet autograft transplant
model [150]. In humans, liraglutide exerted beneficial effects
vs. comparators on measures of -cell function, including HOMA-B (2743%) [87,103,107,151], proinsulin:insulin
ratio [103,104,107,151] and proinsulin : C-peptide ratio [106].
Other improvements with liraglutide therapy included

108 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

increased first-phase insulin secretion and maximum -cell


insulin secretory capacity [152]. Liraglutide was associated with
significant improvement in -cell function head to head compared with sitagliptin, with no treatment-related differences
recorded for markers of insulin resistance (i.e. HOMA-IR) [83].
DPP-4 Inhibitors. Similar to the GLP-1 receptor agonists, the
DPP-4 inhibitors exert beneficial effects on -cells in preclinical and clinical studies. A sitagliptin analogue increased -cell
mass and function in a murine model of type 2 diabetes [153].
Sitagliptin achieved improvements in HOMA-B (1020%) and
proinsulin:insulin ratio compared with placebo in clinical trials [9297,127,154]. A meta-analysis of data from randomized,
placebo- or active-controlled trials found that sitagliptin significantly improved HOMA-B and proinsulin:insulin ratio relative
to placebo. Proinsulin:insulin ratio did not differ between
sitagliptin-treated patients and patients receiving active control
medication, however, and sitagliptin was significantly inferior
to active control on HOMA-B [155], which may simply indicate a difference in the ability to secrete insulin more than a
true effect on -cell health, when the active control is sulphonylurea. Saxagliptin also significantly improved HOMA-2 vs.
placebo in several trials [100,105,108].
Vildagliptin was associated with improvements in proinsulin:insulin ratio [102,110] and insulin secretory rate area
under the curve (AUC) : glucose AUC ratio vs. placebo
in clinical studies [110112]. A 52-week study found that
vildagliptin increased meal-related insulin secretion independently of changes in insulin sensitivity [156]. Improvements in
-cell function with vildagliptin have not been sustained after
washout [157,158].

Safety of Incretin-based Therapies


Tolerability issues (e.g. gastrointestinal disorders, hypoglycemia) with GLP-1 receptor agonists and DPP-4 inhibitors
had been reviewed in previous sections.
Acute Pancreatitis. Acute pancreatitis has been reported in
patients treated with both GLP-1 receptor agonists and DPP-4
inhibitors; exenatide and sitagliptin labels recommend vigilance
for signs and symptoms of acute pancreatitis [159,160]. A
claims-based safety surveillance system that assessed risk
for acute pancreatitis with either exenatide or sitagliptin
found no difference in risk between those patients receiving
either of the incretin-based therapies vs. those who were on
metformin or glyburide [161]. Incidence of acute pancreatitis
among patients using either liraglutide or a comparator
in clinical trials is similar to that expected in a type 2
diabetes population [98,103,104,107,121], but the incidence
of pancreatitis was found to be increased in the liraglutide
group in LEAD-6 study [98]. One potential explanation is
that patients with type 2 diabetes have a threefold greater
risk of developing pancreatitis compared with the general
population [162]. Thus, the increasing number of cases of
pancreatitis following incretin-based therapies published or
reported to the US Food and Drug Administration (FDA) could
be a consequence of increased awareness of the potential link.
Alternatively, some animal studies suggest that treatments with

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

GLP-1 agonist or DPP-4 inhibitors may favour the development


of histologically proven low-grade pancreatitis [163,164].
Exenatide [163] and sitagliptin [164] seem to induce pancreatic
ductal cell replication, a critical step in the pathogenesis
of low-grade pancreatitis and also a well-established factor
for pancreatic adenocarcinoma [165]. Interestingly, metformin
counterbalances the positive effect of sitagliptin on pancreatic
ductal replication [164], suggesting that GLP-1-based therapies
should be preferentially used in combination with metformin in
clinical practice. Thus, an accurate postmarketing surveillance
is needed to further assess the effect of GLP-1-based therapies
on exocrine pancreas function.
Antibodies to GLP-1 Agonists. One safety consideration with
GLP-1 agonists is the potential to induce antibodies, as has
been reported for other protein-based therapies. Antiexenatide antibody formation was common in clinical studies,
with a 2748% incidence [8991,109,116,117]. The presence of antibodies had little apparent effect on efficacy or
safety [90,91,116,117], although the prescribing information
for exenatide notes that, among a small proportion (6%)
of patients who developed high-titre blocking antibody levels in clinical trials, approximately half had no glycaemic
response [159]. Liraglutide exhibited a low potential to induce
antibodies (413%) in clinical trials, compared with the incidence reported for exenatide [103,106,107]. This is probably
because of the greater amino acid sequence homology of
liraglutide (97%) as compared with exenatide (50%) to
native human GLP-1 [166,167]. Unfortunately, no data are
available concerning the prevalence of antibodies following
either exenatide or liraglutide treatments in the LEAD-6
head-to-head study [98]. It should be mentioned here, that
during phase III studies with the not yet approved long-acting
GLP-1 receptor agonist taspoglutide, a potential association
between hypersensitivity reactions (mainly skin reactions and
gastrointestinal symptoms) and antitaspoglutide antibodies
was identified [168]. Similar hypersensitivity reactions were
not reported with either exenatide or liraglutide in clinical
trials. However, postmarketing hypersensitivity reactions were
reported at very low rate for exenatide [159].
Given that DPP-4 inhibitors, unlike GLP-1 receptor agonists, are not protein-based therapies, no antibody formation
is expected. Nonetheless, the expression of DPP-4 on lymphocytes [169] suggests the need for vigilance regarding possible
immune system effects. A first meta-analysis of clinical studies
data found a significantly increased risk of all-cause infection
(e.g. nasopharyngitis, upper respiratory tract infection, urinary
tract infection) associated with sitagliptin (RR 1.15; 95% CI
1.021.31, p = 0.03), but not with vildagliptin (RR 1.04; 95%
CI 0.871.24, p = 0.05) [169]. This finding was not confirmed
for sitagliptin in a subsequent meta-analysis of a larger pool of
data [130].
Kidney Function and Pharmacological Interactions. Kidney
function is frequently compromised in patients with
long-standing type 2 diabetes. Pharmacokinetics study indicates that exenatide is eliminated renally, with a reduction
of mean clearance of the compound and a poor tolerance in
subjects with end-stage renal disease (ESRD) [170,171]. Thus,

Volume 13 No. 2 February 2011

review article
use of exenatide is not recommended in patients with severe
renal impairment [creatinine clearance (CrCl) <30 ml/min]
or ESRD. Dose escalation from 5 to 10 g should proceed conservatively in patients with moderate renal impairment (CrCl:
3050 ml/min) [158]. Exenatide has not exhibited clinically
significant interactions with other medications (e.g. statins,
warfarin, digoxin, paracetamol) in clinical studies [172175],
although postmarketing reports have been made of increased
international normalized ratio in patients taking exenatide and
warfarin [158]. Caution is accordingly warranted in patients
taking this combination.
Liraglutide half-life was not found to be increased, and
clearance was not found to be decreased in subjects with
renal impairment, including ESRD, supporting the observation
that the kidneys are not a major site for the elimination
and degradation of liraglutide [176,177]. However, the
liraglutide-prescribing information does not recommend use
in patients with moderate (CrCl 3059 ml/min) or worse renal
dysfunction because of limited therapeutic experience [178].
Clinical data indicate only minor pharmacokinetic interactions
between liraglutide and a range of drugs (e.g. atorvastatin,
lisinopril, digoxin, griseofulvin, paracetamol or an oral
contraceptive) and no need for dosage adjustment [179181].
Elimination of sitagliptin occurs primarily via renal excretion. Given the limited experience of sitagliptin use in moderate
or greater renal impairment (i.e. CrCl <50 ml/min), sitagliptin
is not recommended for use in such patients. In the United
States, but not in Europe, sitagliptin at dose of 50 mg once
daily is recommended instead of 100 mg in such patients. No
clinically meaningful pharmacokinetic interaction has been
found between sitagliptin and metformin, glyburide, simvastatin, rosiglitazone, warfarin or oral contraceptives [182].
Vildagliptin should not be used in patients with moderate or
greater renal impairment or in those with hepatic impairment,
including those with alanine aminotransferase or aspartate
aminotransferase threefold greater than the upper limit of
normal. The increase of liver enzymes in some patients on
vildagliptin 100 mg once daily has resulted in approval of
vildagliptin 50 mg once or twice daily in Europe (vildagliptin
is not approved in the United States). No clinically significant interactions have been found with a range of medications,
including metformin, glyburide, digoxin, amlodipine, ramipril,
valsartan or simvastatin [183].
The US product information for saxagliptin recommends
dosage reduction in patients with moderate or greater renal
impairment; the EU summary of product characteristics warns
against use in these patients owing to the lack of clinical data [184,185]. Saxagliptin has exhibited pharmacokinetic
interactions with strong inhibitors of hepatic cytochrome P450
3A4/5 enzymes. US labelling recommends dosage reduction
of saxagliptin when used concomitantly with such inhibitors,
such as ketoconazole, atazanavir, telithromycin or nefazodone;
the EU label states that clinical data suggest a low risk of interaction with these drugs and recommends careful monitoring of
glucose control when saxagliptin is used with them concomitantly [184,185].
Calcitonin-producing C-cell Hyperplasia. The GLP-1 receptor
was found to be expressed in thyroid C-cell lines of several

doi:10.1111/j.1463-1326.2010.01317.x 109

review article
species, including mice, rats, monkeys and humans, with no
expression in other thyroid cell types [186]. However, the
expression level of GLP-1 receptor was 22-fold higher in mouse
cell line than in human line, and in situ hybridization experiments failed to detect GLP-1 receptor mRNA in sections
from human thyroid gland [186]. Moreover, GLP-1 agonists
increase cyclic adenosine monophosphate (cAMP) and calcitonin release in rat, but not in human, C-cell lines. Liraglutide
induced C-cell hyperplasia, C-cell adenoma, and, at the highest
doses, C-cell carcinoma in preclinical rodent studies. Similar
findings did not occur in non-human primates (who express
similar levels of GLP-1 receptor to human) exposed to 60 times
the human dose of 1.8 mg, confirming that rodent C-cells are
sensitive to GLP-1-agonist activation, yet human and nonhuman primate C-cells are not [186]. More important, there
were no cases of medullary thyroid carcinoma in the LEAD
studies, and calcitonin levels, the biomarker of medullary thyroid carcinoma, were not increased after 2 years of clinical exposure to liraglutide. FDA requires the establishment of a cancer
registry to monitor the annual incidence of medullary thyroid
carcinoma over the next 15 years but does not encourage checking calctinonin levels for patients receiving liraglutide [187].

The Place of Incretin-based Therapies in Clinical


Practice
In view of the data presented here, some practical principles can
be formulated for the clinical use of GLP-1 receptor agonists
and DPP-4 inhibitors (Figure 2 [2]).
1. At step 1, most newly diagnosed patients with type 2 diabetes
will be initiated on metformin pharmacotherapy. Longterm clinical studies are lacking to promote the utilization
of incretin-based therapy at this initial step, and metformin
remains the most cost-effective drug.
2. At step 2 of treatment intensification, incretin-based therapies are currently recommended as add-on therapy, when
metformin alone is not adequate to reach the HbA1c target,
by both the ADA/EASD (which suggest GLP-1 receptor
agonists) and the NICE (which suggests DPP-4 inhibitors)
guidelines, mainly in patients at risk for hypoglycaemia
or who are obese [3,16]. Early DPP-4 inhibitor initiation
appears to be a good option, facilitating early intensification
and reducing clinical inertia. The stopping rules proposed
in the NICE guidelines [to switch to another option if HbA1c
has not decreased by 0.5% (5 mmol/mol)] could allow differentiation between good and bad responders, because
no baseline characteristics are predictive of response.
Beyond the lack of hypoglycaemia and weight gain, there are
other benefits in prescribing incretin-based therapies over
other available antidiabetic agents. Poor durability over time
of sulphonylureas makes incretin-based therapies theoretically preferable, even though the precise durability of DPP-4
inhibitors or GLP-1 agonists is not known. Adverse effects
associated with TZDs also make incretin-based therapies
preferable, although the long-term safety of the newer drugs
remains to be confirmed. Finally, the only disadvantage to
prescribing these drugs at step 2, compared with the other
available options, is their cost.

110 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

The choice of incretin therapy has to be individualized:


DPP-4 inhibitors are easy to use and well tolerated, although
this advantage must be weighed against the expectations
about HbA1c reductions. Indeed, compared with TZDs,
HbA1c decrease with DDP-4 inhibitors is on an average
similar (in non-inferiority trials) or slightly less (in clinical practice). DPP-4 inhibitors have a slower onset of
action compared with sulphonylureas, but HbA1c decrease
with DPP-4 inhibitors is similar (in non-inferiority trials)
or slightly less (in clinical practice) after 1 year of treatment. Compared with the GLP-1 agonist liraglutide, DPP-4
inhibitors are less potent, do not result in weight loss and
are not associated with nausea. GLP-1 receptor agonists
are injectable drugs and therefore perhaps less easily taken
than DPP-4 inhibitors. Nausea, at least when starting treatment, also has to be considered. Weight loss, however, at
least in those patients who lose >5 kg, is of considerable
interest.
3. At step 3 of treatment intensification, NICE Guidelines [16]
recommend GLP-1 agonists in very obese patients with body
mass index (BMI) >35 kg/m2 . Alternatives to liraglutide at
step 3 are insulin, which is the preferred option by NICE, or
adding a third agent. In the case of failure of a dual oral treatment, adding a third oral drug may provide a good choice,
if the prospect of injection with insulin results in a delay in
intensification of treatment or clinical inertia. Pioglitazone
is primarily indicated for initiation after dual therapy failure.
It may also be a good indication for DPP-4 inhibitors, especially in patients on a metforminTZD combination. DPP-4
inhibitorsulfonylurea combination is not optimal, even
though efficacy has beenshown. Combining two insulin secretagogues, even though acting through different pathways,
is not ideal, and the benefits of DPP-4 inhibitors (e.g. weight
neutral, no hypogycaemia) are lost in this combination with
durability of effect which appears to be uncertain.
Basal insulin is generally recommended at step 3, for
robust reasons. Insulin is the most effective glucose-lowering
medication, given that there is no maximum dose beyond
which reduction in glycaemia will not occur, it is a wellvalidated option (ADA/EASD), and the natural progressive
history of the disease suggests that most patients will eventually require it.
GLP-1 receptor agonists appear to be an alternative to
insulin: a single injection without need for titration, weight
loss instead of weight gain, no hypoglycemia (if sulphonylureas are reduced or stopped), a similar or even better blood
glucose-lowering efficacy, at least in the clinical studies.
It may be anticipated, if the cost is not the priority, that
this option will become the recommended intensification of
treatment at step 3; insulin may then become step 4 in the
case of GLP-1 receptor agonist failure. In this scenario, the
change from GLP-1 receptor agonists to insulin may occur
early, if HbA1c does not decrease by 1% (11 mmol/mol) in
the stopping rules proposed in the NICE guidelines [16].
If the choice of a GLP-1 receptor agonist is made at step 3 in
patients on a combined DPP-4 inhibitormetformin treatment not adequately controlling glycemia, it seems logical
to stop the DPP-4 inhibitor.

Volume 13 No. 2 February 2011

Volume 13 No. 2 February 2011

OAD tritherapy
Met/SU/PIO
Met/DPP-4 Inh/PIO
MET/SU/DPP-4 Inh

Metformin

Metformin +
DPP-4
DPP 4 iinhibitors
hibit
++ lack of hypoglycaemia
+ weight neutral
+ tolerability
-- cost
? durability
? pancreatitis

++ cost efficacy
++ long-term safety
- GI disorders

Metformin/OAD +
Insulin (bed-time)
Met/SU/insulin
Met/PIO/insulin
++ cost efficacy
+ no upper limit dose
-- hypoglycaemia
-weight gain (PIO>SU)
- fluid retention (PIO)

et o
Metformin
+
GLP-1 agonists
++ lack of hypoglycaemia
++ weight loss
-- cost
--nausea
? durability
? pancreatitis
titi

y
++ cost efficacy
++ no upper limit dose
-hypoglycaemia
- weight gain

Metformin +
Insulin multi-injections
(premix twice daily,
basal-bolus regimens)

Metformin +
Insulin
I
li
(bed-time regimen)
++ cost efficacy
+ no upper limit dose
-- hypoglycaemia
-weight gain

Figure 2. Pharmacological management of type 2 diabetes. Therapeutic options at each of three steps of treatment intensification are shown. + and indicate the benefits and disadvantages of each options,
respectively. For more details, see the text. DPP-4 inh, DPP-4 inhibitors; GI, gastrointestinal; GLP-1, GLP-1 receptor agonists; MET, metformin; NAFLD, non-alcoholic fatty liver disease; OAD, oral antidiabetic
drug; PIO, pioglitazone; SU, sulphonylureas.

OAD bitherapy + GLP-1


GLP-1/Met
GLP-1/MET/SU
GLP-1/Met/PIO
++ weight loss
+lack of hypoglycaemia (PIO)
-hypoglycaemia (SU)
-- nauseas
-- cost

Metformin +
Pioglitazone
++ durability
++ insulin-sensitization
+ NAFLD
-- weigth gain
-- fluid retention
-- fractures

+ efficacy for HbA1C 9%


-- clinical inertia for HbA1c > 9%

Step 3

Metformin
M
tf
i +
Sulphonylureas
++ cost efficacy
+ long-term safety
-- durability
-- hypoglycaemia
- weight gain

Step 2

Step 1

HbA1C target < 7% (53 mmol/mol)


Life style changes associated at each step
Maximum 6 months delay between each step

DIABETES, OBESITY AND METABOLISM

review article

doi:10.1111/j.1463-1326.2010.01317.x 111

review article
Conclusions
Disease progression in patients with type 2 diabetes inevitably
necessitates treatment intensification to cope with declining
-cell function. Although guidelines differ on when to
consider the introduction of incretin therapies, clinical data
support the glycaemic efficacy and extraglycaemic benefits
of the GLP-1 receptor agonists and DPP-4 inhibitors. The
differing mechanisms of action of GLP-1 receptor agonists
and DPP-4 inhibitors appear to result in differing therapeutic
profiles. Both agents significantly reduce HbA1c, although the
effect is consistently more pronounced with GLP-1 receptor
agonists. The low risk of hypoglycaemia associated with these
agents provides an advantage relative to the introduction of
sulphonylurea or insulin therapy. Weight loss (GLP-1 receptor
agonists) or weight neutrality (DPP-4 inhibitors) support the
use of incretin therapies in patients for whom overweight
or obesity is a concern. The lack of clinically significant
drug interactions suggests the utility of incretin therapies
in patients receiving treatment for co-morbid conditions.
Incretin therapies improve -cell function, but there is
currently no evidence of a long-term, disease-modifying
effect. GLP-1 receptor agonists merit further investigation
to determine if improvements in markers of cardiovascular
function (e.g. blood pressure, lipids) correlate with better
long-term cardiovascular outcomes.

Acknowledgements
The authors thank Andrew Horgan, PhD, and Patricia Abramo
of AdelphiEden Health Communications for providing medical
writing and editorial services. Funding for the preparation of
this review was provided by Novo Nordisk, Inc.

Conict of Interest
B. Charbonnel has received fees for consultancy, speaking,
travel or accommodation from Takeda, GlaxoSmithKline,
Merck Sharpe & Dohme, AstraZeneca, Bristol Myers Squibb,
Boehringer Ingelheim, Novo Nordisk, Roche, Sanofi-Aventis
and Novartis.
B. Cariou has acted as an investigator for Roche, Boehringer
Ingelheim, Novo Nordisk, Eli Lilly & Co, Genfit, Johnson
& Johnson and has received fees for consultancy, speaking,
travel or accommodation from Novo Nordisk, Eli Lilly & Co,
Sanofi-Aventis, Merck & Co Inc, Pierre Fabre, Takeda, Genfit,
GlaxoSmithKline, Novartis and Servier.
Prof. B. Charbonnel and Prof. B. Cariou received no
honorarium or payment for authorship of this review.
The authors report that they have made substantial
contributions to the conception and design of the study,
acquisition of data, analysis and interpretation of data, drafting
the article and final approval of the version to be submitted.

References
1. UK Prospective Diabetes Study Group. UK Prospective Diabetes Study 16.
Overview of 6 years therapy of type II diabetes: a progressive disease.
Diabetes 1995; 44: 12491258.

112 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

2. American Diabetes Association. Standard of Medical Care in Diabetes2010. Diabetes Care 2010; 33: S1161.
3. Nathan DM, Buse JB, Davidson MB et al., American Diabetes Association
& European Association for Study of Diabetes. Medical management of
hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation
and adjustment of therapy: a consensus statement of the American
Diabetes Association and the European Association for the Study of
Diabetes. Diabetes Care 2009; 32: 193203.
4. National Institute for Health and Clinical Excellence. NICE Clinical Guideline
87. Type 2 Diabetes: The Management of Type 2 Diabetes. London,
England: National Institute for Health and Clinical Excellence, 2009.
5. Canadian Diabetes Association Clinical Practice Guidelines Expert
Committee. Canadian Diabetes Association 2008 clinical practice
guidelines for the prevention and management of diabetes in Canada.
Can J Diabetes 2008; 32(Suppl.1): S1201.
6. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose
control with sulphonylureas or insulin compared with conventional
treatment and risk of complications in patients with type 2 diabetes
(UKPDS 33). Lancet 1998; 352: 837853.
7. Evans JM, Ogston SA, Emslie-Smith A, Morris AD. Risk of mortality and
adverse cardiovascular outcomes in type 2 diabetes: a comparison of
patients treated with sulfonylureas and metformin. Diabetologia 2006;
49: 930936.
8. Johnson JA, Majumdar SR, Simpson SH, Toth EL. Decreased mortality
associated with the use of metformin compared with sulfonylurea
monotherapy in type 2 diabetes. Diabetes Care 2002; 25: 22442248.
9. Roussel R, Bernard C. Metformin use and mortality among diabetic
patients with established atherothrombosis (Abstract). 70th American
Diabetes Association Scientic Sessions. Orlando, Florida, June 2529
2010.
10. Bowker SL, Yasui Y, Veugelers P, Johnson JA. Glucose-lowering agents
and cancer mortality rates in type 2 diabetes: assessing effects of
time-varying exposure. Diabetologia 2010; 53: 16311637.
11. Belfort R, Harrison SA, Brown K et al. A placebo-controlled trial of
pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med
2006; 355: 22972307.
12. Kahn SE, Haffner SM, Heise MA et al. & ADOPT Study Group. Glycemic
durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl
J Med 2006; 355: 24272443.
13. Turner RC, Cull CA, Frighi V, Holman RR & UK Prospective Diabetes Study
(UKPDS) Group. Glycemic control with diet, sulfonylurea, metformin, or
insulin in patients with type 2 diabetes mellitus: progressive requirement
for multiple therapies (UKPDS 49). JAMA 1999; 281: 20052012.
14. Cook MN, Girman CJ, Stein PP, Alexander CM. Initial monotherapy with
either metformin or sulphonylureas often fails to achieve or maintain
current glycaemic goals in patients with type 2 diabetes in UK primary
care. Diabet Med, 2007; 24: 350358.
15. Schernthaner G, Barnett AH, Betteridge DJ et al. Is the ADA/EASD
algorithm for the management of type 2 diabetes (January 2009)
based on evidence or opinion? A critical analysis. Diabetologia 2010; 53:
12581269.
16. National Institute for Health and Clinical Excellence. NICE Short Clinical
Guideline 87. Type 2 Diabetes: Newer Agents for Blood Glucose Control
in Type 2 Diabetes. London, England: National Institute for Health and
Clinical Excellence, 2009.
17. Shorr RI, Ray WA, Daugherty JR, Grifn MR. Individual sulfonylureas and
serious hypoglycemia in older people. J Am Geriatr Soc 1996; 44:
751755.
18. Stahl M, Berger W. Higher incidence of severe hypoglycaemia leading to
hospital admission in type 2 diabetic patients treated with long-acting
versus short-acting sulphonylureas. Diabet Med 1999; 16: 586590.

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

19. Lee TM, Chou TF. Impairment of myocardial protection in type 2 diabetic
patients. J Clin Endocrinol Metab 2003; 88: 531537.
20. Maedler K, Carr RD, Bosco D, Zuellig RA, Berney T, Donath MY. Sulfonylurea induced -cell apoptosis in cultured human islets. J Clin Endocrinol
Metab 2005; 90: 501506.
21. Iwakura T, Fujimoto S, Kagimoto S et al. Sustained enhancement of
Ca(2+) inux by glibenclamide induces apoptosis in RINm5F cells.
Biochem Biophys Res Commun 2000; 271: 422428.
22. Efanova IB, Zaitsev SV, Zhivotovsky B et al. Glucose and tolbutamide
induce apoptosis in pancreatic -cells. A process dependent on
intracellular Ca2+ concentration. J Biol Chem 1998; 273: 3350133507.
23. Del Guerra S, Marselli L, Lupi R et al. Effects of prolonged in vitro exposure
to sulphonylureas on the function and survival of human islets. J Diabetes
Complications 2005; 19: 6064.
24. Charbonnel B, Schernthaner G, Brunetti P et al. Long-term efcacy and
tolerability of add-on pioglitazone therapy to failing monotherapy
compared with addition of gliclazide or metformin in patients with
type 2 diabetes. Diabetologia 2005; 48: 10931104.
25. Holman RR, Haffner SM, McMurray JJ et al., for the Navigator Study Group.
Effect of nateglinide on the incidence of diabetes and cardiovascular
events. N Engl J Med 2010; 362: 14631476.
26. DeFronzo RA, Banerji MA, Bray GA et al., for the ACT NOW Study Group.
Determinants of glucose tolerance in impaired glucose tolerance at
baseline in the Actos Now for Prevention of Diabetes (ACT NOW) Study.
Diabetologia 2010; 53: 435445.
27. DeFronzo RA, Banerji M, Bray GA et al. Actos Now for the Prevention of
Diabetes (ACT NOW) Study. BMC Endocr Disord 2009; 9: 17.
28. Gerstein HC, Yusuf S, Bosch J et al. & DREAM (Diabetes REduction Assessment with ramipril and rosiglitazone Medication) Trial Investigators.
Effect of rosiglitazone on the frequency of diabetes in patients with
impaired glucose tolerance or impaired fasting glucose: a randomised
controlled trial. Lancet 2006; 368: 10961105.
29. Buchanan TA, Xiang AH, Peters RK et al. Preservation of pancreatic -cell
function and prevention of type 2 diabetes by pharmacological treatment
of insulin resistance in high-risk Hispanic women. Diabetes 2002; 51:
27962803.
30. Roussel R. Three studies in type 2 diabetes: DREAM the wash-out results.
Ann Endocrinol (Paris) [French] 2008; 69: 459460.
31. Hanefeld M, Brunetti P, Schernthaner GH, Matthews DR, Charbonnel BH
& QUARTET Study Group. One-year glycemic control with a sulfonylurea
plus pioglitazone versus a sulfonylurea plus metformin in patients with
type 2 diabetes. Diabetes Care 2004; 27: 141147.

review article
37. Guan Y, Hao C, Cha DR et al. Thiazolidinediones expand body uid
volume through PPAR gamma stimulation of ENaC-mediated renal salt
absorption. Nat Med 2005; 11: 861866.
38. Karalliedde J, Buckingham R, Starkie M, Lorand D, Stewart M, Viberti G,
for the Rosiglitazone Fluid Retention Study Group. Effect of various
diuretic treatments on rosiglitazone-induced uid retention. J Am Soc
Nephrol 2006; 17: 34823490.
39. Chaitman BR, Hardison RM, Adler D et al., for the Bypass Angioplasty
Revascularization Investigation 2 Diabetes (BARI 2D) Study Group. The
bypass angioplasty revascularisation investigation 2 diabetes randomized
trial of different treatment strategies in type 2 diabetes mellitus with
stable ischemic heart disease: impact of treatment strategy on cardiac
mortality and myocardial infarction. Circulation 2009; 120: 25292540.
40. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial
infarction and death from cardiovascular causes. N Engl J Med 2007;
356: 24572471.
41. Nissen SE, Wolski K. Rosiglitazone revisited: an updated meta-analysis of
risk for myocardial infarction and cardiovascular mortality. Arch Intern
Med 2010 170: 11911201.
42. Graham DJ, Ouellet-Hellstrom R, MaCurdy TE et al. Risk of acute myocardial infarction, stroke, heart failure, and death in elderly Medicare patients
treated with rosiglitazone or pioglitazone. JAMA 2010; 304: 411418.
43. Dormandy JA, Charbonnel B, Eckland DJ et al., on behalf of the PROactive
Investigators. Secondary prevention of macrovascular events in patients
with type 2 diabetes in the PROactive Study (PROspective pioglitAzone
Clinical Trial In macroVascular Events): a randomised controlled trial.
Lancet 2005; 366: 12791289.
44. Berneis K, Rizzo M, Stettler C et al. Comparative effects of rosiglitazone
and pioglitazone on fasting and postprandial low-density lipoprotein
size and subclasses in patients with Type 2 diabetes. Expert Opin
Pharmacother 2008; 9: 343349.
45. Davidson M, Meyer PM, Haffner S et al. Increased high-density lipoprotein cholesterol predicts the pioglitazone-mediated reduction of carotid
intima-media thickness progression in patients with type 2 diabetes
mellitus. Circulation 2008; 117: 21232130.
46. Deeg MA, Buse JB, Goldberg RB et al., on behalf of the GLAI Study
Investigators. Pioglitazone and rosiglitazone have different effects on
serum lipoprotein particle concentrations and sizes in patients with type
2 diabetes and dyslipidemia. Diabetes Care 2007; 30: 24582464.
47. Olefsky JM. Treatment of insulin resistance with peroxisome proliferatoractivated receptor gamma agonists. J Clin Invest 2000; 106: 467472.
48. Schwartz AV, Sellmeyer DE, Vittinghoff E et al. Thiazolidinedione use and
bone loss in older diabetic adults. J Clin Endocrinol Metab 2006; 91:
33493354.

32. Home PD, Pocock SJ, Beck-Nielsen H et al. & RECORD Study Team.
Rosiglitazone evaluated for cardiovascular outcomes in oral agent
combination therapy for type 2 diabetes (RECORD): a multicentre,
randomised, open-label trial. Lancet 2009; 373: 21252135.

49. Loke YK, Singh S, Furberg CD. Long-term use of thiazolidinediones and
fractures in type 2 diabetes: a meta-analysis. CMAJ 2009; 180: 3239.

33. Smith SR, De Jonge L, Volaufova J, Li Y, Xie H, Bray GA. Effect of


pioglitazone on body composition and energy expenditure: a randomized
controlled trial. Metabolism 2005; 54: 2432.

50. Douglas IJ, Evans SJ, Pocock S, Smeeth L. The risk of fractures associated
with thiazolidinediones: a self-controlled case-series study. PLoS Med
2009; 6: e1000154.

34. Carey DG, Cowin GJ, Galloway GJ et al. Effect of rosiglitazone on insulin
sensitivity and body composition in type 2 diabetic patients [corrected].
Obes Res 2002; 10: 10081015.

51. Dormuth CR, Carney G, Carleton B, Bassett K, Wright JM. Thiazolidinediones and fractures in men and women. Arch Intern Med 2009; 169:
13951402.

35. Takeda Global Research and Development Centre (Europe) Ltd. Actos
(pioglitazone) summary of product information. London, England: Takeda
Global Research and Development Centre (Europe) Ltd, 2009. Available from URL: http://www.emea.europa.eu/humandocs/PDFs/EPAR/
Actos/emea-combined-h285en.pdf. Accessed 30 April 2010.

52. Wan Y, Chong LW, Evans RM. PPAR-gamma regulates osteoclastogenesis


in mice. Nat Med 2007; 13: 14961503.
53. Black C, Donnelly P, McIntyre L, Royle PL, Shepherd JP, Thomas S.
Meglitinide analogues for type 2 diabetes mellitus. Cochrane Database
Syst Rev 2007; 18: CD004654.

36. SmithKline Beecham Ltd. Avandia (rosiglitazone) product information.


Middlesex, England: SmithKline Beecham Ltd, 2008. Available from
URL: http://www.emea.europa.eu/humandocs/PDFs/EPAR/Avandia/
emea-combined-h268en.pdf. Accessed 30 April 2010.

54. Raslova K, Tamer SC, Clauson P, Karl D. Insulin detemir results in less
weight gain than NPH insulin when used in basal-bolus therapy for type
2 diabetes mellitus, and this advantage increases with baseline body
mass index. Clin Drug Investig 2007; 27: 279285.

Volume 13 No. 2 February 2011

doi:10.1111/j.1463-1326.2010.01317.x 113

review article

DIABETES, OBESITY AND METABOLISM

55. Hermansen K, Davies M, Derezinski T, Martinez Ravn G, Clauson P,


Home P & Sitagliptin Study 035 Group. A 26-week, randomized, parallel,
treat-to-target trial comparing insulin detemir with NPH insulin as add-on
therapy to oral glucose-lowering drugs in insulin-naive people with type
2 diabetes. Diabetes Care 2006; 29: 12691274.

73. Gutzwiller JP, Drewe J, Goke B et al. Glucagon-like peptide-1 promotes


satiety and reduces food intake in patients with diabetes mellitus type
2. Am J Physiol 1999; 276(Pt 2): R1541R1544.

56. Rosenstock J, Schwartz SL, Clark CM Jr., Park GD, Donley DW, Edwards MB.
Basal insulin therapy in type 2 diabetes: 28-week comparison of insulin
glargine (HOE 901) and NPH insulin. Diabetes Care 2001; 24: 631636.

74. Naslund E, Gutniak M, Skogar S, Rossner S, Hellstrom PM. Glucagon-like


peptide 1 increases the period of postprandial satiety and slows gastric
emptying in obese men. Am J Clin Nutr 1998; 68: 525530.

57. Smith U, Gale EA. Does diabetes therapy inuence the risk of cancer?
Diabetologia 2009; 52: 16991708.

75. Turton MD, OShea D, Gunn I et al. A role for glucagon-like peptide-1 in
the central regulation of feeding. Nature 1996; 379 :6972.

58. Leslie CA, Satin-Rapaport W, Matheson D, Stone R, Eneld G. Psychological insulin resistance: a missed diagnosis? Diab Spect 1994; 7: 5257.

76. Nauck M, Stockmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in


type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29: 4652.

59. Ziemer DC, Miller CD, Rhee MK et al. Clinical inertia contributes to poor
diabetes control in a primary care setting. Diabetes Educ 2005; 31:
564571.

77. Vilsbll T, Krarup T, Deacon CF, Madsbad S, Holst JJ. Reduced postprandial
concentrations of intact biologically active glucagon-like peptide 1 in
type 2 diabetic patients. Diabetes 2001; 50: 609613.

60. Rubino A, McQuay LJ, Gough SC, Kvasz M, Tennis P. Delayed initiation
of subcutaneous insulin therapy after failure of oral glucose-lowering
agents in patients with Type 2 diabetes: a population-based analysis in
the UK. Diabet Med 2007; 24: 14121418.

78. Meier JJ, Nauck MA. Is the diminished incretin effect in type 2 diabetes
just an epi-phenomenon of impaired beta-cell function? Diabetes 2010;
59: 11171125.

61. Stratton IM, Adler AI, Neil HA et al, on behalf of the UKPDS. Association
of glycaemia with macrovascular and microvascular complications of
type 2 diabetes (UKPDS 35): prospective observational study. BMJ 2000;
321: 405412.
62. Klein R. Hyperglycemia and microvascular and macrovascular disease in
diabetes. Diabetes Care 1995; 18: 258268.
63. Ceriello A, Ihnat MA, Thorpe JE. The metabolic memory: is more than
just tight glucose control necessary to prevent diabetic complications?
J Clin Endocrinol Metab 2009; 94: 410415.
64. Nathan DM, Cleary PA, Backlund JY et al., Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications
(DCCT/EDIC) Study Research Group. Intensive diabetes treatment and
cardiovascular disease in patients with type 1 diabetes. N Engl J Med
2005; 353: 26432653.
65. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year followup of intensive glucose control in type 2 diabetes. N Engl J Med 2008;
359: 15771589.
66. Rosenstock J, Sugimoto D, Strange P, Stewart J, Soltes-Rak E, Dailey G,
for the Insulin Glargine 4014 Study Investigators. Triple therapy in type 2
diabetes: insulin glargine or rosiglitazone added to combination therapy
of sulfonylurea plus metformin in insulin-nave patients. Diabetes Care
2006; 29: 554559.
67. Theodorakis MJ, Carlson O, Michopoulos S et al. Human duodenal
enteroendocrine cells: source of both incretin peptides, GLP-1 and GIP.
Am J Physiol Endocrinol Metab 2006; 290: E550559.
68. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide
1 (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients.
Diabetologia 1993; 36: 741744.
69. Gutniak M, Orskov C, Holst JJ, Ahren B, Efendic S. Antidiabetogenic effect
of glucagon-like peptide-1 (7-36)amide in normal subjects and patients
with diabetes mellitus. N Engl J Med 1992; 326: 13161322.
70. Nauck MA, Heimesaat MM, Behle K et al. Effects of glucagon-like peptide
1 on counterregulatory hormone responses, cognitive functions, and
insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp
experiments in healthy volunteers. J Clin Endocrinol Metab 2002; 87:
12391246.
71. Nauck MA, Niedereichholz U, Ettler R et al. Glucagon-like peptide 1
inhibition of gastric emptying outweighs its insulinotropic effects in
healthy humans. Am J Physiol 1997; 273(Pt 1): E981988.
72. Meier JJ, Gallwitz B, Salmen S et al. Normalization of glucose concentrations and deceleration of gastric emptying after solid meals during

114 Charbonnel and Cariou

intravenous glucagon-like peptide 1 in patients with type 2 diabetes.


J Clin Endocrinol Metab 2003; 88: 27192725.

79. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W.
Preserved incretin activity of glucagon-like peptide 1 [7-36 amide]
but not of synthetic human gastric inhibitory polypeptide in patients with
type-2 diabetes mellitus. J Clin Invest 1993; 91: 301307.
80. Kjems LL, Holst JJ, Vlund A, Madsbad S. The inuence of GLP-1 on
glucose-stimulated insulin secretion: effects on beta-cell sensitivity in
type 2 and nondiabetic subjects. Diabetes 2003; 52: 380386.
81. Meier JJ, Gallwitz B, Kask B et al. Stimulation of insulin secretion by
intravenous bolus injection and continuous infusion of gastric inhibitory
polypeptide in patients with type 2 diabetes and healthy control subjects.
Diabetes 2004; 53(Suppl.2): S220224.
82. Vilsbll T, Agers H, Krarup T, Holst JJ. Similar elimination rates of
glucagon-like peptide-1 in obese type 2 diabetic patients and healthy
subjects. J Clin Endocrinol Metab 2003; 88: 220224.
83. Pratley RE, Nauck M, Bailey T et al., for the 1860-LIRA-DPP-4 Study
Group. Liraglutide versus sitagliptin for patients with type 2 diabetes
who did not have adequate glycaemic control with metformin: a 26week, randomised, parallel-group, open-label trial. Lancet 2010; 375:
14471456.
84. Nauck MA, El-Ouaghlidi A. The therapeutic actions of DPP-IV inhibition
are not mediated by glucagon-like peptide-1. Diabetologia 2005; 48:
608611.
85. Vella A, Bock G, Giesler PD et al. Effects of dipeptidyl peptidase-4
inhibition on gastrointestinal function, meal appearance, and glucose
metabolism in type 2 diabetes. Diabetes 2007; 56: 14751480.
86. Edwards CM, Stanley SA, Davis R et al. Exendin-4 reduces fasting and
postprandial glucose and decreases energy intake in healthy volunteers.
Am J Physiol Endocrinol Metab 2001; 281: E155161.
87. Juhl CB, Hollingdal M, Sturis J et al. Bedtime administration of NN2211,
a long-acting GLP-1 derivative, substantially reduces fasting and
postprandial glycemia in type 2 diabetes. Diabetes 2002; 51: 424429.
88. Raun K, von Voss P, Gotfredsen CF, Golozoubova V, Rolin B, Knudsen LB.
Liraglutide, a long-acting glucagon-like peptide-1 analog, reduces body
weight and food intake in obese candy-fed rats, whereas a dipeptidyl
peptidase-IV inhibitor, vildagliptin, does not. Diabetes 2007; 56: 815.
89. Moretto TJ, Milton DR, Ridge TD et al. Efcacy and tolerability of exenatide
monotherapy over 24 weeks in antidiabetic drug-naive patients with type
2 diabetes: a randomized, double-blind, placebo-controlled, parallelgroup study. Clin Ther 2008; 30: 14481460.
90. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects
of exenatide (exendin-4) on glycemic control and weight over 30 weeks
in metformin-treated patients with type 2 diabetes. Diabetes Care 2005;
28: 10921100.

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

review article

91. Buse JB, Henry RR, Han J, Kim DD, Fineman MS, Baron AD. Effects
of exenatide (exendin-4) on glycemic control over 30 weeks in
sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004;
27: 26282635.

106. Russell-Jones D, Vaag A, Schmitz O et al. Liraglutide versus insulin


glargine in combination with metformin and sulfonylurea therapy in type
2 diabetes: a randomized controlled trial (LEAD-5 met+SU). Diabetologia
2009; 52: 20462055.

92. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, WilliamsHerman DE, for the Sitagliptin Study 021 Group. Effect of the dipeptidyl
peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in
patients with type 2 diabetes. Diabetes Care 2006; 29: 26322637.

107. Zinman B, Gerich J, Buse JB et al. Efcacy and safety of the human GLP-1
analog liraglutide in combination with metformin and TZD in patients
with type 2 diabetes mellitus (LEAD-4 Met+TZD). Diabetes Care 2009;
32: 12241230.

93. Hanefeld M, Herman GA, Wu M, Mickel C, Sanchez M, Stein PP &


Sitagliptin Study 014 Investigators. Once-daily sitagliptin, a dipeptidyl
peptidase-4 inhibitor, for the treatment of patients with type 2 diabetes.
Curr Med Res Opin 2007; 23: 13291339.

108. Allen E, Hollander P, Li J, Chen R. Saxagliptin added to a thiazolidinedione


improves glycaemic control in patients with inadequately controlled type
2 diabetes. 44th Annual Meeting of the European Association for the
Study of Diabetes. Rome, Italy, 711 September 2008.

94. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H &


Sitagliptin Study 023 Group. Efcacy and safety of the dipeptidyl
peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2
diabetes mellitus. Diabetologia 2006; 49: 25642571.

109. Drucker DJ, Buse JB, Taylor K et al. & DURATION-1 Study Group. Exenatide
once weekly versus twice daily for the treatment of type 2 diabetes:
a randomised, open-label, non-inferiority study. Lancet 2008; 372:
12401250.

95. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G. Efcacy and safety


of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing
metformin therapy in patients with type 2 diabetes inadequately
controlled with metformin alone. Diabetes Care 2006; 29: 26382643.

110. Garber AJ, Foley JE, Banerji MA et al. Effects of vildagliptin on glucose
control in patients with type 2 diabetes inadequately controlled with a
sulphonylurea. Diabetes Obes Metab 2008; 10: 10471056.

96. Raz I, Chen Y, Wu M et al. Efcacy and safety of sitagliptin added to


ongoing metformin therapy in patients with type 2 diabetes. Curr Med
Res Opin 2008; 24: 537550.

111. Bosi E, Camisasca RP, Collober C, Rochotte E, Garber AJ. Effects of


vildagliptin on glucose control over 24 weeks in patients with type
2 diabetes inadequately controlled with metformin. Diabetes Care 2007;
30: 890895.

97. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P. Efcacy and safety of the
dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone
therapy in patients with type 2 diabetes: a 24-week, multicenter,
randomized, double-blind, placebo-controlled, parallel-group study. Clin
Ther 2006; 28: 15561568.
98. Buse JB, Rosenstock J, Sesti G et al. & LEAD-6 Study Group. Liraglutide
once a day versus exenatide twice a day for type 2 diabetes: a 26-week
randomised, parallel-group, multinational, open-label trial (LEAD-6).
Lancet 2009; 374: 3947.
99. Chacra AR, Tan GH, Apanovitch A, Ravichandran S, List J, Chen R, CV181040 Investigators. Saxagliptin added to a submaximal dose of
sulphonylurea improves glycaemic control compared with uptitration of
sulphonylurea in patients with type 2 diabetes: a randomised controlled
trial. Int J Clin Pract 2009; 63: 13951406.
100. DeFronzo RA, Hissa MN, Garber AJ et al. & Saxagliptin 014 Study Group.
The efcacy and safety of saxagliptin when added to metformin therapy
in patients with inadequately controlled type 2 diabetes with metformin
alone. Diabetes Care 2009; 32: 16491655.
101. Ferrannini E, Fonseca V, Zinman B et al. Fifty-two-week efcacy and
safety of vildagliptin vs. glimepiride in patients with type 2 diabetes
mellitus inadequately controlled on metformin monotherapy. Diabetes
Obes Metab 2009; 11: 157166.
102. Foley JE, Sreenan S. Efcacy and safety comparison between the DPP-4
inhibitor vildagliptin and the sulfonylurea gliclazide after two years of
monotherapy in drug-nave patients with type 2 diabetes. Horm Metab
Res 2009; 41: 905909.
103. Marre M, Shaw J, Brandle M et al. & LEAD-1 SU Study Group. Liraglutide,
a once-daily human GLP-1 analogue, added to a sulphonylurea over 26
weeks produces greater improvements in glycaemic and weight control
compared with adding rosiglitazone or placebo in subjects with Type 2
diabetes (LEAD-1 SU). Diabet Med 2009; 26: 268278.
104. Nauck M, Frid A, Hermansen K et al. Efcacy and safety comparison of
liraglutide, glimepiride, and placebo, all in combination with metformin,
in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2
study. Diabetes Care 2009; 32: 8490.
105. Rosenstock J, Aguilar-Salinas C, Klein E, Nepal S, List J, Chen R, for the
CV181-011 Study Investigators. Effect of saxagliptin monotherapy in
treatment-nave patients with type 2 diabetes. Curr Med Res Opin 2009;
25: 24012411.

Volume 13 No. 2 February 2011

112. Garber AJ, Schweizer A, Baron MA, Rochotte E, Dejager S. Vildagliptin in


combination with pioglitazone improves glycaemic control in patients
with type 2 diabetes failing thiazolidinedione monotherapy: a
randomized, placebo-controlled study. Diabetes Obes Metab 2007; 9:
166174.
113. Pi-Sunyer FX, Schweizer A, Mills D, Dejager S. Efcacy and tolerability of
vildagliptin monotherapy in drug-nave patients with type 2 diabetes.
Diabetes Res Clin Pract 2007; 76: 132138.
114. Rosenstock J, Baron MA, Dejager S, Mills D, Schweizer A. Comparison
of vildagliptin and rosiglitazone monotherapy in patients with type 2
diabetes: a 24-week, double-blind, randomized trial. Diabetes Care 2007;
30: 217223.
115. Schweizer A, Couturier A, Foley JE, Dejager S. Comparison between
vildagliptin and metformin to sustain reductions in HbA(1c ) over 1 year in
drug-naive patients with type 2 diabetes. Diabet Med 2007; 24: 955961.
116. Zinman B, Hoogwerf BJ, Duran Garca S et al. The effect of adding
exenatide to a thiazolidinedione in suboptimally controlled type 2
diabetes: a randomized trial. Ann Intern Med 2007; 146: 477485.
117. Kendall DM, Riddle MC, Rosenstock J et al. Effects of exenatide (exendin4) on glycemic control over 30 weeks in patients with type 2 diabetes
treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:
10831091.
118. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG &
GWAA Study Group. Exenatide versus insulin glargine in patients with
suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern
Med 2005; 143: 559569.
119. Klonoff DC, Buse JB, Nielsen LL et al. Exenatide effects on diabetes,
obesity, cardiovascular risk factors and hepatic biomarkers in patients
with type 2 diabetes treated for at least 3 years. Curr Med Res Opin
2008; 24: 275286.
120. Blonde L, Russell-Jones D. The safety and efcacy of liraglutide with or
without oral antidiabetic drug therapy in type 2 diabetes: an overview of
the LEAD 15 studies. Diabetes Obes Metabol 2009; 11(Suppl.3): 2634.
121. Garber A, Henry R, Ratner R et al. & LEAD-3 (Mono) Study Group.
Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD3 Mono): a randomised, 52-week, phase III, double-blind, paralleltreatment trial. Lancet 2009; 373: 473481.

doi:10.1111/j.1463-1326.2010.01317.x 115

review article
122. Garber A, Henry R, Ratner R, Hale P, Chang CT, Bode B. Monotherapy
with liraglutide, a once-daily human GLP-1 analog, provides sustained
reductions in A1c , FPG, and weight compared with glimepiride in type
2 diabetes: LEAD-3 mono 2-year results. Program and abstracts of
the American Diabetes Association (ADA) 69th Scientic Sessions. New
Orleans, Louisiana, 59 June 2009.
123. Williams-Herman D, Seck TL, Golm G et al. Long-term efcacy with
sitagliptin as monotherapy or add-on therapy to metformin: improvement in glycemic control over 2 years in patients with type 2 diabetes.
Diabetes 2009; 58(Suppl.1): A144145.
124. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell
L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin
and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24: 29432952.
125. Goke B, Hershon K, Kerr D et al. Efcacy and safety of vildagliptin
monotherapy during 2-year treatment of drug-nave patients with type
2 diabetes: comparison with metformin. Horm Metab Res 2008; 40:
892895.
126. Rosenstock J, Niggli M, Maldonado-Lutomirsky M. Long-term 2-year
safety and efcacy of vildagliptin compared with rosiglitazone in drugnave patients with type 2 diabetes mellitus. Diabetes Obes Metab 2009;
11: 571578.
127. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P. Efcacy and
safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with
type 2 diabetes mellitus inadequately controlled on glimepiride alone or
on glimepiride and metformin. Diabetes Obes Metab 2007; 9: 733745.
128. Gallwitz B. Benet-risk assessment of exenatide in the therapy of type 2
diabetes mellitus. Drug Saf 2010; 33: 87100.
129. Schwartz S, Fonseca V, Berner B, Cramer M, Chiang YK, Lewin A. Efcacy,
tolerability, and safety of a novel once-daily extended-release metformin
in patients with type 2 diabetes. Diabetes Care 2006; 29: 759764.
130. Williams-Herman D, Engel SS, Round E et al. Safety and tolerability of
sitagliptin in clinical studies: a pooled analysis of data from 10,246
patients with type 2 diabetes. BMC Endocr Dis 2010; 10: 7.
131. Grieve DJ, Cassidy RS, Green BD. Emerging cardiovascular actions of the
incretin hormone glucagon-like peptide-1: potential therapeutic benets
beyond glycaemic control? Br J Pharmacol 2009; 157: 13401351.
132. Gutzwiller JP, Tschopp S, Bock A et al. Glucagon-like peptide 1 induces
natriuresis in healthy subjects and in insulin-resistant obese men. J Clin
Endocrinol Metab 2004; 89: 30553061.
133. Noyan-Ashraf MH, Momen MA, Ban K et al. GLP-1R agonist liraglutide
activates cytoprotective pathways and improves outcomes after
experimental myocardial infarction in mice. Diabetes 2009; 58: 975983.
134. Nikolaidis LA, Mankad S, Sokos GG et al. Effects of glucagon-like
peptide-1 in patients with acute myocardial infarction and left ventricular
dysfunction after successful reperfusion. Circulation 2004; 109: 962965.
135. Kendall DM, Bhole D, Guan X et al. Exenatide treatment for 82 weeks
reduced C-reactive protein, HbA1C, and body weight in patients with type
2 diabetes mellitus (Abstract 784). Diabetologia 2006; 49(Suppl.1): 475.
136. Gallwitz B, Vaag A, Falahati A, Madsbad S. Adding liraglutide to oral
antidiabetic drug therapy: onset of treatment effects over. Int J Clin Pract
2010; 64: 267276.
137. Fonseca V, Madsbad S, Falahati A, Zychma M, Plutzky J. Once-daily
human GLP-1 analog liraglutide reduces systolic BPa meta-analysis of 6
clinical trials. Program and abstracts of the American Diabetes Association
(ADA) 69th Scientic Sessions. New Orleans, Louisiana, 59 June 2009.
138. Plutzky J, Garber AJ, Falahati A, Toft AD, Poulter NR. The once-daily
human GLP-1 analogue, liraglutide, signicantly reduces markers of
cardiovascular risk in type 2 diabetes: a meta-analysis of six clinical
trials. European Society of Cardiology Congress 2009. Barcelona, Spain,
29 August2 September 2009.

116 Charbonnel and Cariou

DIABETES, OBESITY AND METABOLISM

139. Gedulin BR, Nikoulina SE, Smith PA et al. Exenatide (exendin-4) improves
insulin sensitivity and -cell mass in insulin-resistant obese fa/fa Zucker
rats independent of glycemia and body weight. Endocrinology 2005;
146: 20692076.
140. Chen J, Couto FM, Minn AH, Shalev A. Exenatide inhibits -cell apoptosis by decreasing thioredoxin-interacting protein. Biochem Biophys Res
Commun 2006; 346: 10671074.
141. Xu G, Stoffers DA, Habener JF, Bonner-Weir S. Exendin-4 stimulates both
-cell replication and neogenesis, resulting in increased -cell mass and
improved glucose tolerance in diabetic rats. Diabetes 1999; 48:
22702276.
142. Rajab A, Buss J, Diakoff E, Hadley GA, Osei K, Ferguson RM. Comparison
of the portal vein and kidney subcapsule as sites for primate islet
autotransplantation. Cell Transplant 2008; 17: 10151023.
143. Mari A, Nielsen LL, Nanayakkara N, DeFronzo RA, Ferrannini E, Halseth A.
Mathematical modeling shows exenatide improved -cell function in
patients with type 2 diabetes treated with metformin or metformin and
a sulfonylurea. Horm Metab Res 2006; 38: 838844.
144. Corner A, Bunck MC, Diamant M et al. Beta cell function and glycaemic
control following one year exenatide therapy, and after 12 week washout, in patients with type 2 diabetes. 44th Annual Meeting of the
European Association for the Study of Diabetes. Rome, Italy, 711
September 2008.
145. Ghofaili KA, Fung M, Ao Z et al. Effect of exenatide on cell function after
islet transplantation in type 1 diabetes. Transplantation 2007; 83: 2428.
146. Rolin B, Larsen MO, Gotfredsen CF et al. The long-acting GLP-1 derivative
NN2211 ameliorates glycemia and increases -cell mass in diabetic
mice. Am J Physiol Endocrinol Metab 2002; 283: E745752.
147. Sturis J, Gotfredsen CF, Rmer J et al. GLP-1 derivative liraglutide in rats
with -cell deciencies: inuence of metabolic state on -cell mass
dynamics. Br J Pharmacol 2003; 140: 123132.
148. Bregenholt S, Mldrup A, Blume N et al. The long-acting glucagon-like
peptide-1 analogue, liraglutide, inhibits -cell apoptosis in vitro. Biochem
Biophys Res Commun 2005; 330: 577584.
149. Merani S, Truong W, Emamaullee JA, Toso C, Knudsen LB, Shapiro AM.
Liraglutide, a long-acting human glucagon-like peptide 1 analog,
improves glucose homeostasis in marginal mass islet transplantation
in mice. Endocrinology 2008; 149: 43224328.
150. Emamaullee JA, Merani S, Toso C et al. Porcine marginal mass islet autografts resist metabolic failure over time and are enhanced by early
treatment with liraglutide. Endocrinology 2009; 150: 21452152.
151. Matthews D, Marre M, Le-Thi TD, Zdravkovic M, Simo R. Liraglutide, a
once-daily human GLP-1 analog, signicantly improves -cell function in
subjects with type 2 diabetes. Program and abstracts of the American
Diabetes Association (ADA) 68th Sessions. San Francisco, California, 610
June 2008.
152. Vilsbll T, Brock B, Perrild H et al. Liraglutide, a once-daily human GLP-1
analogue, improves pancreatic B-cell function and arginine-stimulated
insulin secretion during hyperglycaemia in patients with Type 2 diabetes
mellitus. Diabet Med 2008; 25: 152156.
153. Mu J, Woods J, Zhou YP et al. Chronic inhibition of dipeptidyl peptidase-4
with a sitagliptin analog preserves pancreatic -cell mass and function
in a rodent model of type 2 diabetes. Diabetes 2006; 55: 16951704.
154. Goldstein BJ, Feinglos MN, Lunceford JK, Johnson J, Williams-Herman DE
& Sitagliptin 036 Study Group. Effect of initial combination therapy
with sitagliptin, a dipeptidyl peptidase-4 inhibitor, and metformin on
glycemic control in patients with type 2 diabetes. Diabetes Care 2007;
30: 19791987.
155. Riche DM, East HE, Riche KD. Impact of sitagliptin on markers of -cell
function: a meta-analysis. Am J Med Sci 2009; 337: 321328.

Volume 13 No. 2 February 2011

DIABETES, OBESITY AND METABOLISM

156. Ahren B, Pacini G, Foley JE, Schweizer A. Improved meal-related -cell


function and insulin sensitivity by the dipeptidyl peptidase-IV inhibitor
vildagliptin in metformin-treated patients with type 2 diabetes over 1
year. Diabetes Care 2005; 28: 19361940.
157. Utzschneider KM, Tong J, Montgomery B et al. The dipeptidyl peptidase-4
inhibitor vildagliptin improves -cell function and insulin sensitivity in
subjects with impaired fasting glucose. Diabetes Care 2008; 31: 108113.
158. Mari A, Scherbaum WA, Nilsson PM et al. Characterization of the inuence of vildagliptin on model-assessed -cell function in patients with
type 2 diabetes and mild hyperglycemia. J Clin Endocrinol Metab 2008;
93: 103109.
159. Byetta (exenatide) product information. Hampshire, England: Eli Lilly and
Company Limited, 2009. Available from URL: http://www.emea.europa.
eu/humandocs/PDFs/EPAR/byetta/H-698-PI-en.pdf. Accessed 30 April
2010.
160. US Food and Drug Administration. Information for healthcare professionalsacute pancreatitis and sitagliptin (marketed as Januvia and
Janumet). Available from URL: http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyIn
formationforHeathcareProfessionals/ucm183764.htm. Accessed 30 April
2010.
161. Dore DD, Seeger JD, Arnold Chan K. Use of a claims-based active drug
safety surveillance system to assess the risk of acute pancreatitis with
exenatide or sitagliptin compared to metformin or glyburide. Curr Med
Res Opin 2009; 25: 10191027.
162. Noel RA, Braun DK, Patterson R, Bloomgren G. Increased risk of acute
pancreatitis observed in patients with type 2 diabetes. Pancreas 2008;
37: 487.
163. Nachnani JS, Bulchandani DG, Nookala A et al. Biochemical and histological effects of exendin-4 (exenatide) on the rat pancreas. Diabetologia
2010; 53: 153159.
164. Matveyenko AV, Dry S, Cox HI et al. Benecial endocrine but adverse
exocrine effects of sitagliptin in the human islet amyloid polypeptide
transgenic rat model of type 2 diabetes: interactions with metformin.
Diabetes 2009; 28: 16041615.
165. Jura N, Archer H, Bar-Sagi D. Chronic pancreatitis, pancreatic adenocarcinoma and the black box in-between. Cell Res 2005; 15: 7277.

review article
172. Kothare PA, Linnebjerg H, Skrivanek Z et al. Exenatide effects on statin
pharmacokinetics and lipid response. Int J Clin Pharmacol Ther 2007; 45:
114120.
173. Soon D, Kothare PA, Linnebjerg H et al. Effect of exenatide on the pharmacokinetics and pharmacodynamics of warfarin in healthy Asian men.
J Clin Pharmacol 2006; 46: 11791187.
174. Kothare PA, Soon DK, Linnebjerg H et al. Effect of exenatide on the
steady-state pharmacokinetics of digoxin. J Clin Pharmacol 2005; 45:
10321037.
175. Blase E, Taylor K, Gao HY, Wintle M, Fineman M. Pharmacokinetics of an
oral drug (acetaminophen) administered at various times in relation to
subcutaneous injection of exenatide (exendin-4) in healthy subjects. J
Clin Pharmacol 2005; 45: 570577.
176. Jacobsen LV, Hindsberger C, Robson R, Zdravkovic M. Effect of renal
impairment on the pharmacokinetics of the GLP-1 analogue liraglutide.
Br J Clin Pharmacol 2009; 68: 898905.
177. Helleberg H, Malm-Erjefalt M, Bjrnsdottir I et al. Metabolism and excretion of [[Pal-3H]]-liraglutide in human healthy subjects. Program and
Abstracts of the American Diabetes Association (ADA) 68th Scientic
Sessions. San Francisco, California, 610 June 2008.
178. European Medicines Agency. Victoza (liraglutide) product information.
2009. Available from URL: http://www.emea.europa.eu/humandocs/
PDFs/EPAR/victoza/H-1026-PI-en.pdf. Accessed 30 April 2010.
179. Malm-Erjefalt M, Ekblom M, Brndsted L, Vouis J, Lennernas H, Zdravkovic
M. A randomised, double-blind, cross-over trial investigating the effect of
liraglutide on the absorption pharmacokinetics of concomitantly administered oral drugs in healthy subjects. Diabetes 2008; 57(Suppl.1):
A130.
180. Jacobsen LV, Brndsted L, Vouis J, Zdravkovic M. A randomized, doubleblind, cross-over trial investigating the effect of liraglutide on the
absorption of an oral contraceptive drug. Program and Abstracts of
the American Diabetes Association (ADA) 68th Scientic Sessions. San
Francisco, California, 610 June 2008.
181. Kapitza C, Flint A, Hindsberger C, Zdravkovic M. The effect of the oncedaily human GLP-1 analogue liraglutide on the pharmacokinetics of
paracetamol. Diabetes 2008; 57(Suppl.1): A593.

166. Knudsen LB, Nielsen PF, Huusfeldt PO et al. Potent derivatives of


glucagon-like peptide-1 with pharmacokinetic properties suitable for
once daily administration. J Med Chem 2000; 43: 16641669.

182. Merck Sharp & Dohme Ltd. Januvia (sitagliptin) product information. Hertfordshire, England: Merck Sharp & Dohme Ltd., 2008. Available from URL:
http://www.emea.europa.eu/humandocs/PDFs/EPAR/januvia/ emeacombined-h722en.pdf. Accessed 30 April 2010.

167. Eng J, Kleinman WA, Singh L, Singh G, Raufman JP. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma
suspectum venom. Further evidence for an exendin receptor on dispersed
acini from guinea pig pancreas. J Biol Chem 1992; 267: 74027405.

183. Novartis Europharm Limited. Jalra (vildagliptin) product information. Horsham, England: Novartis Europharm Limited, 2009. Available from URL:
http:// www.emea.europa.eu/humandocs/PDFs/EPAR/jalra/emeacombined-h1048en.pdf. Accessed 30 April 2010.

168. F. Hoffmann-La Roche Ltd. Roche announces amendment of the


trial protocols for the taspoglutide Phase III programme [Investor
Update]. Basel, Switzerland: F. Hoffmann-La Roche Ltd. Available from
URL: http://www.roche.com/investors/ir update/inv-update-2010-0618b.htm. Accessed 9 August 2010.

184. Bristol-Myers Squibb. Onglyza [package insert]. Princeton, NJ: BristolMyers Squibb, 2009. Available from URL: http://www.medicines.org.uk/
emc/medicine/22315. Accessed 30 April 2010.

169. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1
receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes.
Lancet 2006; 368: 16961705.
170. Richter B, Bandeira-Echtler E, Bergerhoff K, Lerch CL. Dipeptidyl peptidase- 4 (DPP-4) inhibitors for type 2 diabetes mellitus. Cochrane Database
Syst Rev 2008; 16: CD006739.
171. Linnebjerg H, Kothare PA, Park S et al. Effect of renal impairment on the
pharmacokinetics of exenatide. Br J Clin Pharmacol 2007; 64: 317327.

Volume 13 No. 2 February 2011

185. Bristol-Myers Squibb. Onglyza [Summary of Product Characteristics]. Middlesex, England: Bristol-Myers Squibb/AstraZeneca EEIG, 2009. Available
from URL: http://packageinserts.bms.com/pi/pi onglyza.pdf. Accessed
30 April 2010.
186. Knudsen LB, Madsen LW, Andersen S et al. Glucagon-like peptide-1
receptor agonists activate rodent thyroid c-cells causing calcitonin release
and c-cell proliferation. Endocrinology 2010; 151: 14731486.
187. Parks M, Rosebraugh C. Weighing risks and benets of liraglutidethe
FDAs review of a new antidiabetic therapy. N Engl J Med 2010; 362:
774777.

doi:10.1111/j.1463-1326.2010.01317.x 117

Das könnte Ihnen auch gefallen