Defining the role of GLP-1 receptor agonists for individualized treatment of type 2 diabetes

12
Defining the role of GLP-1 receptor agonists for individualized treatment of type 2 diabetes Expert Rev. Endocrinol. Metab. Early online, 1–12 (2014) Daisuke Yabe* 1–3 and Yutaka Seino 1 1 Center for Diabetes, Endocrinology and Metabolism, Kansai Electric Power Hospital, Osaka, Japan 2 Center for Metabolism and Clinical Nutrition, Kansai Electric Power Hospital, Osaka, Japan 3 Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan *Author for correspondence: Tel.: +81 664 585 821 Fax: +81 664 586 994 [email protected] With the advent of dipeptidyl peptidase (DPP)-4 inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1 RAs) over the past decade, incretin therapy has become established as an important treatment strategy for type 2 diabetes mellitus (T2DM), with an efficacy and safety profile distinct from that of other anti-hyperglycemic agents. However, our understanding of the optimal clinical use of incretins remains incomplete. This review focuses on the use of GLP-1 RAs in the treatment of T2DM, with reference to the differing dominant mechanisms of action between short- and long-acting GLP-1 RAs and the clinical implications of this difference. The role of GLP-1 and the effects of GLP-1 RAs in various organs other than the pancreas will also be discussed. KEYWORDS: albiglutide • DPP-4 inhibitor • dulaglutide • exenatide • exenatide LAR • GLP-1 receptor agonist • incretin • liraglutide • lixisenatide • type 2 diabetes Type 2 diabetes mellitus (T2DM) is a progres- sive metabolic disorder characterized by deteri- oration of pancreatic b-cell function and insulin resistance in the liver and peripheral tissues. These factors combine and result in overproduction of hepatic blood glucose and elevations in plasma glucose (FPG) in the fast- ing state, and insufficient glucose uptake by muscle cells and elevated postprandial plasma glucose (PPG) following a meal [1]. In addition to the b-cell, liver and muscle, dysfunction of the pancreatic a-cell (hyperglucagonemia), the fat cell (accelerated lipolysis), kidney (increased glucose reabsorption), brain (insulin resistance) and gastrointestinal tract (incretin system) can all play an important role in the development and progression of T2DM [1]. The incretin system consists of two gut hor- mones, glucose-dependent insulinotropic poly- peptide and glucagon-like peptide-1 (GLP-1), which are released by gastrointestinal endocrine cells following food intake to stimulate insulin secretion from b-cells glucose dependently [2,3]. GLP-1 is also associated with suppression of glucagon secretion from a-cells and a delaying effect of gastric emptying that reduces the rate of glucose absorption from the small intestine [4], and has an inhibitory neurological influence on appetite and glucose ingestion [2,3,57]. Together, the incretin effects in response to glucose intake ensure that PPG excursions are limited, regardless of carbohydrate load from a meal [8]. Patients with T2DM demonstrate an impaired capability to regulate their incretin effect, which may contribute to the exagger- ated PPG excursions [9]. Efforts to correct this dysfunction with exogenous GLP-1 have been hampered by the short half-life of the peptide in the body [10]. Human GLP-1 is rapidly bro- ken down by the DPP-4 enzyme and metabo- lized in the liver in a matter of minutes, with only a small percentage of the intact hormone circulating systemically [11]. The advent of DPP-4 inhibitors and GLP-1 receptor agonists (GLP-1 RAs) over the past decade has seen incretin therapy become established as an important treatment strategy for T2DM, with an efficacy and safety profile distinct from other anti-diabetic agents [12,13]. However, our understanding of the optimal clinical use of incretins remains incomplete. This review will focus on the GLP-1 RAs and their use in the treatment of T2DM, with reference to the informahealthcare.com 10.1586/17446651.2014.949672 Ó 2014 Informa UK Ltd ISSN 1744-6651 1 Review Expert Review of Endocrinology & Metabolism Downloaded from informahealthcare.com by 182.249.247.14 on 09/03/14 For personal use only.

Transcript of Defining the role of GLP-1 receptor agonists for individualized treatment of type 2 diabetes

Defining the role ofGLP-1 receptor agonists forindividualized treatment oftype 2 diabetesExpert Rev. Endocrinol. Metab. Early online, 1–12 (2014)

Daisuke Yabe*1–3

and Yutaka Seino1

1Center for Diabetes, Endocrinology

and Metabolism, Kansai Electric Power

Hospital, Osaka, Japan2Center for Metabolism and Clinical

Nutrition, Kansai Electric Power

Hospital, Osaka, Japan3Division of Molecular and Metabolic

Medicine, Kobe University Graduate

School of Medicine, Kobe, Japan

*Author for correspondence:

Tel.: +81 664 585 821

Fax: +81 664 586 994

[email protected]

With the advent of dipeptidyl peptidase (DPP)-4 inhibitors and glucagon-like peptide-1 receptoragonists (GLP-1 RAs) over the past decade, incretin therapy has become established as animportant treatment strategy for type 2 diabetes mellitus (T2DM), with an efficacy and safetyprofile distinct from that of other anti-hyperglycemic agents. However, our understanding ofthe optimal clinical use of incretins remains incomplete. This review focuses on the use ofGLP-1 RAs in the treatment of T2DM, with reference to the differing dominant mechanismsof action between short- and long-acting GLP-1 RAs and the clinical implications of thisdifference. The role of GLP-1 and the effects of GLP-1 RAs in various organs other than thepancreas will also be discussed.

KEYWORDS: albiglutide • DPP-4 inhibitor • dulaglutide • exenatide • exenatide LAR • GLP-1 receptor agonist • incretin

• liraglutide • lixisenatide • type 2 diabetes

Type 2 diabetes mellitus (T2DM) is a progres-sive metabolic disorder characterized by deteri-oration of pancreatic b-cell function andinsulin resistance in the liver and peripheraltissues. These factors combine and result inoverproduction of hepatic blood glucose andelevations in plasma glucose (FPG) in the fast-ing state, and insufficient glucose uptake bymuscle cells and elevated postprandial plasmaglucose (PPG) following a meal [1]. In additionto the b-cell, liver and muscle, dysfunction ofthe pancreatic a-cell (hyperglucagonemia), thefat cell (accelerated lipolysis), kidney (increasedglucose reabsorption), brain (insulin resistance)and gastrointestinal tract (incretin system) canall play an important role in the developmentand progression of T2DM [1].

The incretin system consists of two gut hor-mones, glucose-dependent insulinotropic poly-peptide and glucagon-like peptide-1 (GLP-1),which are released by gastrointestinal endocrinecells following food intake to stimulate insulinsecretion from b-cells glucose dependently [2,3].GLP-1 is also associated with suppression ofglucagon secretion from a-cells and a delayingeffect of gastric emptying that reduces therate of glucose absorption from the small

intestine [4], and has an inhibitory neurologicalinfluence on appetite and glucose ingestion[2,3,5–7]. Together, the incretin effects in responseto glucose intake ensure that PPG excursionsare limited, regardless of carbohydrate loadfrom a meal [8].

Patients with T2DM demonstrate animpaired capability to regulate their incretineffect, which may contribute to the exagger-ated PPG excursions [9]. Efforts to correct thisdysfunction with exogenous GLP-1 have beenhampered by the short half-life of the peptidein the body [10]. Human GLP-1 is rapidly bro-ken down by the DPP-4 enzyme and metabo-lized in the liver in a matter of minutes, withonly a small percentage of the intact hormonecirculating systemically [11]. The advent ofDPP-4 inhibitors and GLP-1 receptor agonists(GLP-1 RAs) over the past decade has seenincretin therapy become established as animportant treatment strategy for T2DM, withan efficacy and safety profile distinct fromother anti-diabetic agents [12,13]. However, ourunderstanding of the optimal clinical use ofincretins remains incomplete. This review willfocus on the GLP-1 RAs and their use in thetreatment of T2DM, with reference to the

informahealthcare.com 10.1586/17446651.2014.949672 � 2014 Informa UK Ltd ISSN 1744-6651 1

Review

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

differing dominant mechanisms of action between GLP-1 RAs.The role played by GLP-1 and the effects of GLP-1 RAs invarious organs other than the pancreas will also be discussed.

Classification & comparison of GLP-1 RAs in bothclinical & basic researchThere are currently five different GLP-1 RAs approved for usein patients with poorly controlled T2DM (TABLE 1). From a clin-ical perspective, these five agents can be divided into two broadcategories according to their relative half-lives: twice-daily exe-natide and once-daily lixisenatide are short-acting GLP-1 RAs,while once-daily liraglutide, once-weekly exenatide long-actingrelease (LAR) and albiglutide are long-acting GLP-1 RAs. Thedifference in the half-lives of these two classes appears to affectthe mechanistic action by which they exert their glucose-lowering effects. Short-acting GLP-1 RAs have limited effectson insulin secretion but strongly inhibit postprandial glucagonsecretion [14], which may aid PPG control [15]. They also have adelaying effect on gastric emptying, which results in pro-nounced reductions in PPG levels by reducing the rate of glu-cose absorption from the small intestine [5,16,17]. In contrast,long-acting GLP-1 RAs have a limited gastric emptying effect,and a consequently more modest effect on PPG excur-sions [18,19]. Instead, long-acting GLP-1 RAs provide reductionsin both FPG and PPG through stimulation of insulin secretionand inhibition of glucagon secretion [20].

Short-acting GLP-1 RAs

The first GLP-1 RA to be developed was exenatide, a syntheticversion of the 39-residue exendin-4 peptide originally isolatedfrom Gila monster lizard salivary secretions [21]. Exendin-4shares approximately 50% sequence homology with mammalianGLP-1 and is a potent agonist of the GLP-1 receptor [22]. Cru-cially, however, exendin-4 is resistant to proteolytic cleavage bythe DPP-4 enzyme, and so has a half-life of approximately 2.5 hin plasma, making it clinically viable for therapeutic use [21].Exenatide 10 mg is administered twice daily (b.i.d.) within60 min prior to the breakfast and evening meal [23,24]. In the piv-otal clinical studies of exenatide, glycated hemoglobin (HbA1c)levels were reduced by 0.8–0.9% over a period of 30 weeks,with relatively modest reductions in FPG (-0.6 mmol/l[-10.1 mg/dl]) indicating that the predominant glucose-loweringeffect was via reductions in PPG levels [25–27]. Blood glucosemeasurements confirmed that exenatide provides significantreductions in PPG after breakfast and after the evening meal [28].

The structure of the once-daily short-acting GLP-1 RA lixi-senatide is also based on the exendin-4 peptide, with modifica-tions to the C-terminal end. The binding affinity oflixisenatide for the human GLP-1 receptor is approximatelyfourfold greater than that of human GLP-1 in vitro [29,30].Like exenatide, the structure of lixisenatide is resistantto DPP-4 metabolism, and it has a half-life in plasma of2–4 h [20,31]. A dose-ranging study comparing once- versustwice-daily lixisenatide administration showed comparablereductions in HbA1c over 13 weeks, with a dose of 20 mg once

daily (q.d.) selected for Phase III studies on the basis that itprovided the best tolerability profile while conferring glycemiccontrol [32]. In the Phase III clinical study program, addition oflixisenatide to existing oral therapy resulted in significant reduc-tion in HbA1c of 0.7–0.9% over 24 weeks [33–36]. As with exe-natide, reductions in FPG were modest, although statisticallysignificant; however, reductions in PPG of 4.5–6.2 mmol/l(82–113 mg/dl) were much more marked [37]. Although thelargest reductions in PPG are generally seen post-breakfast, anunsurprising observation given the half-life of lixisenatide andit’s usual timing of administration, a pharmacologic study ofonce-daily lixisenatide administered before breakfast revealed asignificant reduction in PPG versus placebo after breakfast(p < 0.0001), lunch (p < 0.001) and dinner (p < 0.05) [16],demonstrating a significant effect on plasma glucose levelsthroughout the day. Furthermore, the effect of lixisenatide onPPG was correlated with a reduction in the rate of gastric emp-tying [16]. A recent study demonstrated that lixisenatide wasequally effective and well tolerated when administered q.d.before the main meal of the day versus before-breakfastadministration [38].

A head-to-head study of once-daily lixisenatide versus twice-daily exenatide showed statistically non-inferior reductions inHbA1c over 24 weeks (-0.8 vs -1.0%; treatment difference0.17% [95% CI: 0.033–0.297]) despite the different dosingregimens [36]. Although PPG levels were not recorded in thisstudy, both lixisenatide and exenatide showed moderate reduc-tions in FPG (-1.22 vs -1.45 mmol/l [-22 vs -26.1 mg/dl]), inline with the predominant PPG-lowering effects associated withshort-acting GLP-1 RAs.

Long-acting GLP-1 RAs

A LAR formulation of exenatide was subsequently developed toprovide consistent availability in plasma with once-weekly dos-ing. The LAR formulation consists of injectable microspheres ofexenatide and a biodegradable polymer that provides sustainedexenatide release into plasma with a half-life of approximately2 weeks [39]. In Phase III clinical studies, once-weekly exenatideLAR was associated with reduction in HbA1c of 1.3–1.9% over30 weeks of treatment [40,41]. In the DURATION-1 study,which compared once-weekly exenatide LAR with twice-dailyexenatide, HbA1c reductions were significantly greater withthe long-acting versus regular formulation after 30 weeksof treatment (-1.9 vs -1.5%; treatment difference -0.33 [95%CI: -0.54 to -0.12]; p = 0.0023) [41]. Reductions in FPG weresignificantly greater with exenatide LAR versus exenatide(-2.3 vs -1.4 [-42 vs -25 mg/dl]; p < 0.0001), while changes in2-h PPG were significantly lower in patients treated with exena-tide b.i.d. (-5.3 vs -6.9 mmol/l [-96 vs -126 mg/dl];p = 0.0124) [41]. Consistent with these observations, delay ingastric emptying was markedly less pronounced with exenatideLAR than with exenatide [41].

Liraglutide is a second long-acting GLP-1 RA and the firstanalog of human GLP-1 RA, which is administered q.d. Theliraglutide structure includes modifications for attachment of a

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

16-carbon free fatty acid chain that promotes reversible bindingwith plasma proteins, increasing resistance to DPP-4 cleavage,inhibiting renal clearance and extending its half-life to approxi-mately 12 h [42]. The Phase III clinical studies showed a reduc-tion in HbA1c levels of 1.1–1.8% [18,43–46]. Similar to exenatideLAR, head-to-head comparison of once-daily liraglutide versustwice-daily regular exenatide over 26 weeks revealed

significantly greater reductions in HbA1c (-1.12 vs -0.79%;treatment difference -0.33 [95% CI: -0.47 to -0.18]) and FPG(-1.61 vs -0.60 mmol/l [29 vs 11 mg/dl]; p < 0.0001), but sig-nificantly lower reductions in PPG after breakfast and din-ner [18]. Two recent meta-analyses revealed that exenatide LARand liraglutide showed similar reductions in HbA1c levels, butthat these reductions were superior to those seen with twice-

Table 1. Summary of approved short- and long-acting glucagon-like peptide-1 receptor agonists for thetreatment of type 2 diabetes.

Peptideorigin

Modifications Half-life Dosingregimen

Glucose-loweringeffect and changein body weight

Ref.

Short-acting GLP-1 receptor agonists

Exenatide Exendin-4 Synthetic Exendin-4:

39-amino acids with

53% homology to

human GLP-1. An

Ala2Gly substitution

(relative to human

GLP-1) gives resistance

to DPP-mediated

inactivation

~2.5 h b.i.d. HbA1c

reduction: 0.8–0.9%,

body weight

reduction:

1.6–2.8 kg, over

30 weeks

[25–27]

Lixisenatide Exendin-4 Pro38 removed and

C-terminus modified

with six additional Lys

residues

2–4 h q.d. HbA1c

reduction: 0.7–0.9%,

body weight

reduction:

1.6–3.0 kg, over

24 weeks

[37]

Long-acting GLP-1 receptor agonists

Exenatide

long-acting release

Exendin-4 Exendin-4

administered in a

matrix of

biodegradable polymer

(poly-D,L-lactide-co-

glycolide), resulting in

a release over time

after administration

~2 weeks Once weekly HbA1c

reduction: 1.3–1.9%,

body weight

reduction:

2.3–3.7 kg, over

30 weeks

[40,41,118,119]

Liraglutide GLP-1 Lys34Arg substitution,

and addition of a

C-16 fatty acid

(palmitoyl) at Lys26,

attached by a

glutamate linker

12 h q.d. HbA1c reduction:

1.1–1.8%, body

weight reduction:

0.2–3.2 kg, over

26 week

[18,43–46]

Albiglutide GLP-1 Dimer of a 30 amino

acid peptide, with an

Ala2Glu substitution to

increase resistance to

DPP-4, and fused to

human albumin

6–7 days Once weekly HbA1c

reduction:0.8%,

body weight

reduction: 0.6 kg,

over 32 weeks

[51]

Dulaglutide GLP-1 Covalently linked to an

Fc fragment of human

IgG4

90 h Once weekly HbA1c reduction:

0.8–1.1%, body

weight reduction:

2.6–3.0 kg, over

52 weeks

[120]

b.i.d.: Twice daily; DDP-4: Dipeptidyl peptidase 4; FPG: Fasting plasma glucose; GLP-1: Glucagon-like peptide-1; PPG: Postprandial plasma glucose; q.d.: Once daily.

Defining the role of GLP-1 RAs for individualized treatment of type 2 diabetes Review

informahealthcare.com doi: 10.1586/17446651.2014.949672

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

daily exenatide [47,48]. However, a head-to-head trial of liraglu-tide and exenatide LAR found that while both resulted inimprovements in glycemic control, greater reductions inHbA1c, reduced persistence of nausea and fewer occurrences ofhypoglycemia were seen with once-daily liraglutide [49].

The most recent GLP-RA to be approved is albiglutide.Albiglutide is a fusion peptide composed of two DPP-4-resist-ant GLP-1 analogs non-reversibly bound to human serumalbumin, resulting in a half-life of 6–7 days [50]. Data wererecently published from a 32-week Phase III study comparingonce-weekly albiglutide with once-daily liraglutide, whichshowed a reduction in HbA1c of -0.78 versus -0.99%, respec-tively (treatment difference 0.21% [95% CI: 0.08–0.34];p = 0.0846 for non-inferiority) [51]. However, while patientswho received once-daily liraglutide had greater reductions inHbA1c, they also experienced more gastrointestinal side effects,though fewer injection site reactions, than patients whoreceived once-weekly albiglutide [51].

Safety issues associated with the use of GLP-1 RAsOne of the principal attractive features of GLP-1 RAs, in addi-tion to their glucose-lowering benefits, is the relatively low riskof hypoglycemia associated with these agents. GLP-1-inducedstimulation of insulin is glucose-dependent and is negligibleunder conditions of hypoglycemia [52]. Consequently, hypogly-cemia associated with GLP-1 RAs is markedly lower than withsome other anti-diabetic therapies [53,54]. A meta-analysis of27 randomized controlled studies of non-insulin anti-diabeticagents added to metformin therapy showed that GLP-1 RAswere associated with levels of hypoglycemia comparable withplacebo [53]. In contrast, treatment with thiazolidinediones, sul-fonylureas (SUs) and glinides resulted in an increase in hypo-glycemia 2, 2.6- and 7-fold greater than placebo, respectively.As GLP-1 receptor activation by the GLP-1 RA exenatide hasbeen shown to ameliorate ATP production in pancreatic b-cellsand enhance sensitivity to SU [55,56], caution needs to takenwith respect to the potential for hypoglycemia whenGLP-1 RAs are used in combination with SU.

The most common class of adverse events (AEs) experiencedby patients receiving GLP-1 RAs are gastrointestinal AEs. In ameta-analysis of clinical studies of GLP-1 RAs versus insulin, asignificantly greater proportion of patients treated withGLP-1 RAs experienced gastrointestinal AEs compared withthose treated with insulin (Mantel–Haenszel odds ratio 15.00[5.44, 41.35]; p < 0.01) [57]. A review and meta-analysis ofGLP-1 RAs and DPP-4 inhibitors as an add-on to metforminby Deacon et al. found that while AEs were rare overall, theincidence of nausea and vomiting increased with GLP-1 RAtreatment [48]. However, it should also be noted that these gas-trointestinal side effects were mild to moderate in intensity,rarely resulting in treatment withdrawal, were transient in themajority of cases [37,58–60] and the frequency of events appearedto decline over the first few weeks of treatment [61]. The risk ofgastrointestinal side effects may also be influenced by dosingtitration, meal size, meal frequency and BMI.

While short-acting GLP-1 RAs, such as exenatide and lixise-natide, have been associated with a small or non-significanteffect, or even a reduction in resting heart rate, several long-acting GLP-1 RAs, including dulaglutide, liraglutide and LARexenatide, have been shown to be associated with a significantincrease in resting heart rate [62,63]. A significant effect ofGLP-1 RAs on electrocardiogram QT intervals has not beendemonstrated [64–66]. Administration of liraglutide 0.6, 1.2 or1.8 mg q.d. over 7 days found no concentration–exposuredependency on QTc intervals compared with placebo, and noQTc thresholds 500 ms or increases over >60 ms [64], andsingle-dose exenatide 10 mg was not found to be associatedwith clinically meaningful prolongation of the QTc interval,compared with placebo, in healthy subjects [66]. Analysis ofcardiovascular events from Phase II and Phase III liraglutidestudies demonstrated that the overall risk of cardiovascularevents was low, meeting the US FDA criteria for ruling outunacceptable increases in cardiovascular risk [67]. Data fromongoing long-term large-scale cardiovascular outcomes studiesare needed to confirm the lack of cardiovascular risk withGLP-1 RAs [68,69].

Acute pancreatitis is another rare but potentially serioussafety issue with regard to use of GLP-RAs and DPP-4 inhibi-tors. Concerns were raised initially on the basis of individualcase reports in exenatide- and liraglutide-treated patients [70],and retrospective analyses of medical databases [71]. However,both of these sources of information can be unrepresentative ofthe clinical reality and subject to potential bias. Evidence fromrandomized clinical studies of GLP-1 RAs provides a more rig-orous assessment and has not identified a link, although thestatistical power of individual studies is limited for detectingsuch a rare event [72]. More recently, a meta-analysis of 41 ran-domized clinical studies with GLP-1 RAs showed no increasein the risk of pancreatitis with the use of GLP-1 RAs [73], andrecent incretin pancreatic safety reviews by both the FDA andEMA found no evidence of a causal relationship [74]. Concernshave also been raised regarding a potential increased risk ofmedullary thyroid cancer with GLP-1 RAs based on evidenceof thyroid C-cell hyperplasia and tumors associated with long-term liraglutide exposure in rodents [75]. However, large-scalemonitoring during clinical studies has not identified any indica-tors of cancer development, and no increased incidence of C-cell cancer was identified during the clinical studies of otherGLP-1 RAs [76]. An analysis of data reported to the FDAadverse event reporting system did seem to show an increasedrisk of pancreatic and thyroid cancer with incretin therapies,although the data were not consistent and have been discred-ited on the basis of a bias in reporting of events [77,78].

Predictors of efficacy for GLP-1 RAsBaseline HbA1c

A meta-analysis that included seven studies relating to short-acting GLP-1 RAs and seven studies relating to long-actingGLP-1 RAs identified a linear significant negative correlationbetween baseline HbA1c and reduction in HbA1c in both

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

groups over the course of the studies, with a significantlygreater reduction seen in the long-acting GLP-1 RA group [48].Similarly, Kawata et al. identified HbA1c at baseline as the sin-gle largest predictive factor for reduction in HbA1c with liraglu-tide, with significant associations seen at 12 weeks after thestart of liraglutide (b = -0.854, p = 0.001) and at 24 weeks(b = -0.602, p = 0.013) [79]. The authors concluded that base-line HbA1c may be a predictive factor for HbA1c reduction forall incretin therapies, and this could explain why increasedbaseline HbA1c is associated with a greater degree of HbA1c

reduction on treatment with liraglutide.

Ethnicity

Accumulating evidence suggests that GLP-1RAs exert a greatereffect on glycemic control in Asian patients than in non-Asianpatients [80]. In a study comparing the efficacy of lixisenatide inJapanese and Caucasian patients with T2DM insufficiently con-trolled with SU with or without metformin, Japanese patientsexperienced a greater benefit in terms of reduction in PPG excur-sions compared with Caucasian patients [81]. In Japanese patients,the LS mean difference versus placebo in PPG AUC[0:29–4:30 h]

after a standardized breakfast was -406.7 (36.7) h•mg/dl for thelixisenatide q.d. group and -346.3 (35.1) h•mg/dl for the lixise-natide b.i.d. group compared with -260.1 (39.5) h•mg/dl and-231.3 (38.6) h•mg/dl, respectively, in Caucasian patients [81].The authors suggest that greater efficacy of GLP-1 RAs in Asianpatients may be attributable to lower insulin secretory capacity [82]

along with little enhancement of meal-induced GLP-1 secretionin Japanese subjects [83,84].

Residual b-cell function

Pancreatic b-cells are critical for regulation of glucose homeosta-sis, and their progressive loss is a cardinal symptom of T2DMthat underlies the progression of deteriorating glucose control.Long-acting GLP-RAs significantly improve glycemic controlpartly by stimulating insulin release from pancreatic b-cells,unlike short-acting GLP-RAs, which act primarily througheffects on gastric motility. This difference in mechanism ofaction suggests that a certain amount of residual b-cell functionis required for optimal efficacy of long-acting GLP-1 RAs, whileresidual b-cell function is less important for the efficacy ofshort-acting GLP-1 RAs, such as exenatide and lixisenatide. Evi-dence in support of this hypothesis comes from an analysis ofdata from the Phase III studies of liraglutide, which showed asignificantly greater beneficial effect on HbA1c, with liraglutideversus exenatide in patients with high-estimated b-cell mass butno difference in patients with low-estimated b-cell mass [85]. Incontrast, a similar pooled analysis of data from two Phase IIIstudies of short-acting lixisenatide showed that HbA1c reduc-tions were consistent regardless of patients’ b-cell function, andthat PPG reductions were actually significantly greater inpatients with low- versus high-residual b-cell function [17].

The clinical importance of b-cell function for the efficacy oflong-acting GLP-1 RAs has been shown in a recent analysis ofpatients with T2DM switching from insulin to liraglutide

therapy [86]. Patients whose hyperglycemia was uncontrolledafter 12 weeks of liraglutide therapy had a longer duration ofdiabetes, higher baseline insulin doses and lower b-cell functioncompared with responders, indicating a more advanced stageof disease.

Gender

Treatment of T2DM with twice-daily exenatide 10 mg as anadd-on to oral therapy (metformin or metformin ± SU) hasbeen shown to result in better glycemic control in male patientsthan in female patients after 12 months of treatment, withHbA1c £7% achieved in a significantly greater proportion ofmales than females (38 vs 27%). With regard to safety, nosignificant gender difference regarding gastrointestinal AEs hasbeen reported; for example, 60 and 63 cases of nausea werereported with exenatide therapy in males and females,respectively [87].

Predictors of achieving therapeutic glycemic response (HbA1c

£7%) at 1 year among males were found to be inversely relatedto a longer duration of disease and baseline HcA1c. In females,baseline therapy with metformin was related to a positive glyce-mic outcome at 1 year, and baseline SU treatment or combina-tion treatment was associated with a negative effect onachievement of glycemic targets. In females only, an increase inbaseline homeostatic model assessment-b was also associatedwith a positive glycemic response [87].

Expectations beyond glycemic controlThe benefit of improved glycemic control with a low risk ofhypoglycemia makes the GLP-1 RAs an attractive treatmentoption in T2DM. However, the multiple physiologicaleffects of GLP-1 indicate that the clinical benefits ofGLP-1 RAs are likely to extend further (FIGURE 1). Preclinicalstudies of animal models indicate that incretins play a role ina range of diverse biological activities, with potential implica-tions for improvements in diabetes-related microvascularcomplications (e.g., retinopathy, nephropathy and neuropa-thy) and macrovascular complications (e.g., coronary arterydisease, peripheral artery disease and cerebrovascular disease),as well as diabetes-related comorbidities (e.g., obesity,non-alcoholic fatty liver disease, bone fracture and cognitivedysfunction) [88–90].

GLP-1 RAs have a clinically demonstrable benefit in termsof weight, which can be helpful in ameliorating early T2DMand the effects of weight gain from other anti-diabetictherapies [91–93]. A meta-analysis of 25 randomized controlledstudies of GLP-1 RAs of at least 20 weeks’ duration showedthat weight decrease was a consistent phenomenon, with areduction in body weight of approximately 2.9 kg per study [94].Another meta-analysis that included 29 randomized controlledstudies of GLP-1 RAs of at least 24 weeks’ duration reported areduction in BMI at 6 months of -1.0 kg/m2

[95]. Comparablereductions in weight were seen with short- and long-actingGLP-1 RAs. Weight decrease with GLP-1 RAs is thought tooccur through delayed gastric emptying and neurological

Defining the role of GLP-1 RAs for individualized treatment of type 2 diabetes Review

informahealthcare.com doi: 10.1586/17446651.2014.949672

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

suppression of appetite by increasing the perception of fullnessand satiety [96]. As the effects of long-acting GLP-1 RAs ongastric emptying could be lost with sustained dosing, as dis-cussed above, the fact that weight reduction with GLP-1 RAsis independent of the duration of action highlights the impor-tance of GLP-1 RA-mediated suppression of appetite via thebrain [20]. The potential for weight decrease with GLP-1 RAs isan attractive feature, particularly for patients with considerableobesity and for those needing intensified therapy with a treat-ment option that may contribute to an increase body weight,such as insulin.

Studies in animals show that GLP-1 RAs may have potentialcardioprotective effects [88,90,97]. In patients with acute myocar-dial infarction and left ventricular ejection fraction dysfunction,GLP-1 infused over 72 h significantly improved left ventricularejection fraction, and other measures of cardiac function,including global wall motion indices and regional wallmotion [98]. More recently, it was reported that GLP-1 infusionprotects the heart from ischemic left ventricle dysfunctioninduced by dobutamine stress in patients with coronary arterydisease [99]. Exenatide has also been shown to reduce reperfu-sion injury and final infarct size in patients with ST-segmentelevated myocardial infarction [100,101].

GLP-1 RAs also have well-documented benefits against riskfactors for cardiovascular disease, including hypertension anddyslipidemia. The anti-hypertensive effects of GLP-1 have consis-tently been shown in several clinical studies. A meta-analysis of

16 randomized controlled studies of GLP-1 RAs in patients withT2DM revealed that GLP-1 RAs reduced systolic blood pressureand diastolic blood pressure by 1–5 mmHg compared with someother anti-diabetic drugs including insulin, glimepiride and pla-cebo [102]. A pooled analysis of six Phase III clinical trials with lir-aglutide also found significantly greater systolic blood pressurereductions from baseline with liraglutide 1.2 mg q.d. (2.7 [0.8]mmHg) and 1.8 mg (2.9 [0.7] mmHg) than with placebo (0.5[0.9] mmHg) (p = 0.0029 and p = 0.0004, respectively), after26 weeks [103]. Measurement of flow-mediated vasodilatation ofthe brachial artery in patients with T2DM has shown a signifi-cantly greater increase in flow-mediated vasodilatation with exe-natide versus glimepiride [104]. In mice, activation of GLP-1receptors expressed in cardiac atria has been shown to stimulateatrial natriuretic peptide production, a powerful vasodilator [105],indicating that GLP-1 RAs exert their anti-hypertensive effectsthrough a vasodilatory action.

GLP-1 infusion inhibits postprandial elevation of triglyceridesand free fatty acids in healthy individuals [106]. A single subcuta-neous injection of exenatide in patients with newly diagnosedT2DM was also associated with a marked reduction in postpran-dial triacylglycerol, as well as in apolipoprotein B48 [107].Whether these effects on triglycerides and free fatty acids are aresult of delayed gastric emptying or some other mechanism hasyet to be determined. Nevertheless, the data show thatGLP-1 receptor activation ameliorates postprandial lipidemia,which is likely to be beneficial for patients with T2DM.

Extrapancreatic actions Macrovascular Microvascular Co-morbidities

Cerebrovasculardisease

GLP-1: Prevent

Cardiovasculardisease

GLP-1: Prevent

RetinopathyGLP-1: Prevent

DementiaGLP-1: Improve

Fatty liverGLP-1: Improve

ObesityGLP-1: Improve

Bone fractureGLP-1: Prevent

NephropathyGLP-1: Prevent

NeuropathyGLP-1: Prevent

HypertensionGLP-1: Improve

DyslipidemiaGLP-1: Improve

InflammationGLP-1: Prevent

Peripheral arterydisease

GLP-1: Prevent

Intrapancreatic actions

Insulin secretionGLP-1: Stimulate

Glucagon secretionGLP-1: Suppress

β cell massGLP-1: Increase

Figure 1. Intra- and extrapancreatic function of glucagon-like peptide-1 and expected effects on diabetic complications andco-morbidities.GLP-1: Glucagon-like peptide-1.Adapted with permission from [90] � Asian Association for the Study of Diabetes and Wiley Publishing Asia Pty Ltd (2013).

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

GLP-1 RAs as add-on therapies in clinical useMeta-analyses comparing multiple different add-on therapies tometformin for treatment of T2DM have reported that treat-ment with add-on GLP-1 RAs was as effective as basal orbiphasic insulin in terms of glycemic control [54] and resultedin a greater decrease in HbA1c compared with adjunctive SU,glinides, thiazolidinediones, a-glucosidase inhibitors andDPP-4 inhibitors [48,54]. In a meta-analysis comparing metfor-min in combination with either DPP-4 inhibitors orGLP-1 RAs, both were found to significantly reduce HbA1c,but long-term concomitant treatment with metformin andlong-acting GLP-1 RAs (liraglutide or exenatide once weekly)were associated with significantly greater reductions in FPGthan add-on treatment with short-acting exenatide (b.i.d.) orDPP-4 inhibitors [48]. The addition of short-acting agents tobasal insulin has been shown to be effective in achievingHbA1c targets in patients with T2DM insufficiently controlledwith basal insulin alone [91,108–111]. In particular, the additionof short-acting GLP-1 RAs to basal insulin is a favorableoption for treatment intensification in patients with T2DMdue to complementary effects on PPG and FPG, respectively,and can result in improved overall glycemic control versus basalinsulin alone [112–114]. The efficacy of long-acting GLP-1 RAsto basal insulin still needs to be compared with that of short-acting GLP-1 RAs in clinical trials in the future.

Some T2DM therapy combinations are associated withweight increase. While add-on treatment with DPP-4 inhibitorsis reported to be weight-neutral, [48,53,54,115], SU, glinides, thiazo-lidinediones, basal insulin and biphasic insulin add-on therapiesare associated with weight increase. By contrast, botha-glucosidase inhibitors [53,54] and GLP-1 RAs as add-on thera-pies have been associated with weight decrease [48,53,54,115].The addition of GLP-1 RAs to basal insulin is associated withsignificant weight reduction compared with placebo plus basalinsulin [114,116], which may be of particular clinical importanceand relevance to patients.

A final important consideration for add-on therapy regimensis the risk of hypoglycemia. Neither DPP-4 inhibitors norGLP-1 RAs in combination with metformin were associatedwith an increase in the risk of hypoglycemia [54,117]. Additionof GLP-1 RAs to basal insulin was not associated withincreased risk of hypoglycemia compared with basal insulinalone [114,116].

ConclusionIncretin-based therapy, specifically GLP-1 RAs, represent avaluable addition to treatment options for T2DM. A variety ofGLP-1 RAs are available which, due to differing mechanismsof action, provide different clinical profiles. Long-actingGLP-1 RAs help patients reach glycemic targets through FPGand PPG control as a result of insulinotropic effects, whereasshort-acting GLP-1 RAs confer overall glycemic controlthrough pronounced effects on PPG, mainly as a result ofdelayed gastric emptying. Safety and tolerability profiles alsodiffer somewhat between agents, although gastrointestinal

events remain the most common type of AE with this class ofdrug. Beyond the glycemic control actions of GLP-1 RAs, thesedrugs are also associated with multiple physiological effects,with potential implications for improvements in diabetes-related microvascular and macrovascular complications, as wellas diabetes-related comorbidities. GLP-1 RAs are associatedwith reduced weight increase or even weight decrease. Finally,GLP-1 RAs represent an attractive option as add-on therapyfor patients who may not have adequate glycemic control withtheir existing treatment regimen. In particular, the addition ofshort-acting GLP-1 RAs to basal insulin represents a clinicallyuseful treatment option for patients with inadequate PPG con-trol despite well-controlled FPG.

In summary, GLP-1 RAs represent an effective and well-tolerated treatment option for many different types of patientswith T2DM.

Expert commentaryIt has been thought that GLP-1 RAs improve glycemia by bothameliorating impaired secretion of insulin and glucagon, andby delaying gastric emptying. Accumulating evidence showsthat the primary mode of action is different among differentGLP-1 RAs; long-acting GLP-1 RAs acting preferentially viapancreatic b-cells, while short-acting GLP-1 RAs do so mainlyvia delayed gastric emptying. Such differences are reflected bydifferential effects of long- and short-acting GLP-1 RAs onfasting and postprandial glucose levels, although effects arecomparable on body weight changes. Although it remains to betested in clinical trials, our current knowledge strongly suggeststhat long-acting GLP-1 RAs ameliorate glycemia more effi-ciently in patients with sufficient b-cell function, while short-acting GLP-1 RAs, possibly combined with basal insulin, couldimprove postprandial glucose excursions even in patients withlow b-cell function.

Five-year viewWhile long-term efficacy of GLP-1 RAs in glucose loweringand body weight reduction have been documented over theyears, beneficial effects on diabetes complications (e.g., micro-and macroangiopathies) need to be assessed through large-scalerandomized clinical trials, along with any potential AE risks (e.g., pancreatic diseases). Meanwhile, development of once-weekly or once-monthly GLP-1 RAs, as well as oral or nasalGLP-1 RAs, should be continued to improve the quality of lifeof patients with T2DM.

Financial & competing interests disclosure

D Yabe has taken part in advisory panels for Sanofi, Novo Nordisk, Inc.

and Novartis Pharma AG, and has taken part in speaker’ s bureaux for

Sanofi, Novo Nordisk Inc., Merck Sharp & Dohme Limited, Takeda

Pharmaceuticals USA, Inc., Boehringer Ingelheim Pharmaceuticals, Inc.,

Eli Lilly and Company and Bristol-Myers Squibb Company. Y Seino

has acted as medical advisor for Eli Lilly, Sanoi, Novo Nordisk,

GlaxoSmithKline, Taisho Pharmaceuticals, Astellas Pharmaceuticals,

Becton, Dickinson & Company, Boerhringer Ingelheim, Johnson &

Defining the role of GLP-1 RAs for individualized treatment of type 2 diabetes Review

informahealthcare.com doi: 10.1586/17446651.2014.949672

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

Johnson, Takeda Pharmaceuticals and Otsuka Pharmaceuticals. The

authors have no other relevant affiliations or financial involvement with

any organization or entity with a financial interest in or financial conflict

with the subject matter or materials discussed in the manuscript apart

from those disclosed. Writing and editorial assistance was provided by

A Shepherd of Caudex Medical, funded by Sanofi.

Key issues

• Patients with type 2 diabetes mellitus (T2DM) demonstrate an impaired capability to regulate their incretin effect, and correction of this

dysfunction with exogenous glucagon-like peptide-1, GLP-1 receptor agonists (GLP-1 RAs), has become established as an important

treatment strategy for T2DM, with an efficacy and safety profile distinct from that of other anti-diabetic agents.

• The different GLP-1 RAs approved for use in patients with poorly controlled T2DM can be divided into two broad categories according

to their relative half-lives as short- and long-acting GLP-1 RAs, which exert their glucose-lowering effects through different dominant

mechanistic actions: delayed gastric emptying and enhanced insulin secretion.

• Short-acting GLP-1 RAs have limited effects on insulin and glucagon secretion, but have a delaying effect on gastric emptying, which

results in pronounced reductions in postprandial plasma glucose (PPG) levels by reducing the rate of glucose absorption from the

small intestine.

• Long-acting GLP-1 RAs have a limited effect on gastric emptying, but provide reductions in fasting plasma glucose (FPG) and PPG – the

latter less robustly than short-acting GLP-1RAs – through amelioration of insulin and glucagon secretion.

• The clinical implication of these mechanistic differences is that short-acting GLP-RAs are better suited to improve control of PPG

excursions, even in patients with limited residual b-cell function, while long-acting GLP-1 RAs are more appropriate for patients needing

control of FPG, in addition to PPG, but require a certain amount of residual b-cell function.• Compared with thiazolidinediones, sulfonylureas and glinides, GLP-1 RAs are associated with a relatively low risk of hypoglycemia, as

GLP-1-induced stimulation of insulin is glucose-dependent and is negligible under conditions of hypoglycemia, although caution is

advised if GLP-1 RAs are used in combination with sulfonylureas, as they may enhance b-cell sensitivity to this agent.

• Both short- and long-acting GLP-1 RAs have a clinically demonstrable benefit in terms of weight, which can be helpful in ameliorating

early T2DM and the effects of weight gain from other anti-diabetic therapies, such as insulin.

• GLP-1 RAs also have well-documented benefits against risk factors for cardiovascular disease, including hypertension and dyslipidemia.

• The most common class of adverse events experienced by patients receiving GLP-1 RAs are gastrointestinal adverse events, and

although concerns have been raised about a potential increased risk of acute pancreatitis, thyroid C-cell hyperplasia and tumors, no

increased risk has been identified.

• GLP-1 RAs represent an attractive option as add-on therapy for patients who may not have adequate glycemic control with their

existing treatment regimen, and in particular, the addition of GLP-1 RAs to basal insulin represents a clinically useful treatment option

for patients with inadequate PPG control and body weight gain despite well-controlled FPG.

References

Papers of special note have been highlighted as:

• of interest

1. Defronzo RA. Banting Lecture. From the

triumvirate to the ominous octet: a new

paradigm for the treatment of type 2

diabetes mellitus. Diabetes 2009;58(4):

773-95

2. Seino Y, Fukushima M, Yabe D. GIP and

GLP-1, the two incretin hormones:

similarities and differences. J Diabetes

Investig 2010;1(1-2):8-23

3. Campbell JE, Drucker DJ. Pharmacology,

physiology, and mechanisms of incretin

hormone action. Cell Metab 2013;17(6):

819-37

4. Deane AM, Nguyen NQ, Stevens JE, et al.

Endogenous glucagon-like peptide-1 slows

gastric emptying in healthy subjects,

attenuating postprandial glycemia. J Clin

Endocrinol Metab 2010;95(1):215-21

5. 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(1):E155-61

6. Flint A, Raben A, Ersboll AK, et al. The

effect of physiological levels of glucagon-like

peptide-1 on appetite, gastric emptying,

energy and substrate metabolism in obesity.

Int J Obes Relat Metab Disord 2001;25(6):

781-92

7. Meier JJ, Gallwitz B, Salmen S, et al.

Normalization of glucose concentrations and

deceleration of gastric emptying after solid

meals during intravenous glucagon-like

peptide 1 in patients with type 2 diabetes.

J Clin Endocrinol Metab 2003;88(6):

2719-25

8. Holst JJ. The physiology of glucagon-like

peptide 1. Physiol Rev 2007;87(4):1409-39

9. Bagger JI, Knop FK, Lund A, et al.

Impaired regulation of the incretin effect in

patients with type 2 diabetes. J Clin

Endocrinol Metab 2011;96(3):737-45

10. Deacon CF, Nauck MA, Toft-Nielsen M,

et al. Both subcutaneously and intravenously

administered glucagon-like peptide I are

rapidly degraded from the NH2-terminus in

type II diabetic patients and in healthy

subjects. Diabetes 1995;44(9):1126-31

11. Meier JJ, Nauck MA, Kranz D, et al.

Secretion, degradation, and elimination of

glucagon-like peptide 1 and gastric

inhibitory polypeptide in patients with

chronic renal insufficiency and healthy

control subjects. Diabetes 2004;53(3):

654-62

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

12. Ahren B, Schmitz O. GLP-1 receptor

agonists and DPP-4 inhibitors in the

treatment of type 2 diabetes. Horm Metab

Res 2004;36(11-12):867-76

13. Garber AJ. Incretin-based therapies in the

management of type 2 diabetes: rationale

and reality in a managed care setting. Am J

Manag Care 2010;16(Suppl 7):S187-94

14. Ahren B, Gautier JF, Berria R, et al.

Pronounced reduction of post-prandial

glucagon by lixisenatide: a meta-analysis of

randomized clinical trials. Diabetes Obes

Metab 2014. [Epub ahead of print]

15. Shah P, Vella A, Basu A, et al. Lack of

suppression of glucagon contributes to

postprandial hyperglycemia in subjects with

type 2 diabetes mellitus. J Clin Endocrinol

Metab 2000;85(11):4053-9

16. Lorenz M, Pfeiffer C, Steinstrasser A, et al.

Effects of lixisenatide once daily on gastric

emptying in type 2 diabetes - Relationship

to postprandial glycemia. Regul Pept 2013;

185(1):8

17. Meier JJ, Yabe D, Wang E, et al. Efficacy

of lixisenatide in patients with different

levels of beta cell function as assessed by C-

peptide/glucose ratio. Diabetologia 2013;

56(Suppl 1):S1-S566; Abstract 896

• A study that demonstrates the significant

role of gastric emptying and glucagon

inhibition in the mechanism of glycemic

control with the short-acting glucagon-

like peptide-1 receptor agonist (GLP-1

RA), lixisenatide.

18. Buse JB, Rosenstock J, Sesti G, et al.

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(9683):39-47

19. Degn KB, Juhl CB, Sturis J, et al. One

week’s treatment with the long-acting

glucagon-like peptide 1 derivative liraglutide

(NN2211) markedly improves 24-h

glycemia and alpha- and beta-cell function

and reduces endogenous glucose release in

patients with type 2 diabetes. Diabetes

2004;53(5):1187-94

20. Meier JJ. GLP-1 receptor agonists for

individualized treatment of type 2 diabetes

mellitus. Nat Rev Endocrinol 2012;8(12):

728-42

21. Kolterman OG, Buse JB, Fineman MS,

et al. Synthetic exendin-4 (exenatide)

significantly reduces postprandial and fasting

plasma glucose in subjects with type 2

diabetes. J Clin Endocrinol Metab 2003;

88(7):3082-9

22. Goke R, Fehmann HC, Linn T, et al.

Exendin-4 is a high potency agonist and

truncated exendin-(9-39)-amide an

antagonist at the glucagon-like peptide 1-(7-

36)-amide receptor of insulin-secreting beta-

cells. J Biol Chem 1993;268(26):19650-5

23. FDA. Amylin Pharmaceuticals Inc. Byetta

prescribing information. 2009

24. Bristol-Myers Squibb and AstraZeneca

EEIG. Byetta: Summary of product

characteristics. EMA. 2012

25. Defronzo RA, Ratner RE, Han J, et al.

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(5):

1092-100

26. Kendall DM, Riddle MC, Rosenstock J,

et al. Effects of exenatide (exendin-4) on

glycemic control over 30 weeks in patients

with type 2 diabetes treated with metformin

and a sulfonylurea. Diabetes Care 2005;

28(5):1083-91

27. Buse JB, Henry RR, Han J, et al. Effects of

exenatide (exendin-4) on glycemic control

over 30 weeks in sulfonylurea-treated

patients with type 2 diabetes. Diabetes Care

2004;27(11):2628-35

28. Heine RJ, Van Gaal LF, Johns D, et al.

Exenatide versus insulin glargine in patients

with suboptimally controlled type 2

diabetes: a randomized trial. Ann Intern

Med 2005;143(8):559-69

29. Thorkildsen C, Neve S, Larsen BD, et al.

Glucagon-like peptide 1 receptor agonist

ZP10A increases insulin mRNA expression

and prevents diabetic progression in db/db

mice. J Pharmacol Exp Ther 2003;307(2):

490-6

30. Werner U, Haschke G, Herling AW,

Kramer W. Pharmacological profile of

lixisenatide: a new GLP-1 receptor agonist

for the treatment of type 2 diabetes. Regul

Pept 2010;164(2-3):58-64

31. Distiller RA, Ruus P; on behalf of the

ACT6011 Study Group. Pharmacokinetics

and pharmacodynamics of a new GLP-1

agonist AVE0010 in type 2 diabetes

patients. Diabetes 2008;58(Suppl 1):A154-5

32. Ratner RE, Rosenstock J, Boka G. Dose-

dependent effects of the once-daily GLP-1

receptor agonist lixisenatide in patients with

Type 2 diabetes inadequately controlled

with metformin: a randomized, double-

blind, placebo-controlled trial. Diabet Med

2010;27(9):1024-32

33. Ahren B, Leguizamo DA, Miossec P, et al.

Efficacy and safety of lixisenatide once-daily

morning or evening injections in type 2

diabetes inadequately controlled on

metformin (GetGoal-M). Diabetes Care

2013;36(9):2543-50

34. Bolli GB, Munteanu M, Dotsenko S, et al.

Efficacy and safety of lixisenatide once daily

versus placebo in patients with type 2

diabetes insufficiently controlled on

metformin (GetGoal-F1). Diabet Med

2014;31:176-84

35. Pinget M, Goldenberg R, Niemoeller E,

et al. Efficacy and safety of lixisenatide once

daily versus placebo in type 2 diabetes

insufficiently controlled on pioglitazone

(GetGoal-P). Diabetes Obes Metab 2013;

15(11):1000-7

36. Rosenstock J, Raccah D, Koranyi L, et al.

Efficacy and safety of lixisenatide once daily

versus exenatide twice daily in type 2

diabetes inadequately controlled on

metformin: a 24-week, randomized, open-

label, active-controlled study (GetGoal-X).

Diabetes Care 2013;36(10):2945-51

37. Bolli GB, Owens DR. Lixisenatide, a novel

GLP-1 receptor agonist: efficacy, safety and

clinical implications for type 2 diabetes

mellitus. Diabetes Obes Metab 2014;16(7):

588-601

38. Ahren B. Equal improvement in glycemia

with lixisenatide given before breakfast or

the main meal of the day. J Diabetes

Complications 2014. [Epub ahead of print]

39. Fineman M, Flanagan S, Taylor K, et al.

Pharmacokinetics and pharmacodynamics of

exenatide extended-release after single and

multiple dosing. Clin Pharmacokinet 2011;

50(1):65-74

40. Bergenstal RM, Wysham C, Macconell L,

et al. Efficacy and safety of exenatide once

weekly versus sitagliptin or pioglitazone as

an adjunct to metformin for treatment of

type 2 diabetes (DURATION-2): a

randomised trial. Lancet 2010;376(9739):

431-9

41. Drucker DJ, Buse JB, Taylor K, et al.

Exenatide once weekly versus twice daily for

the treatment of type 2 diabetes: a

randomised, open-label, non-inferiority

study. Lancet 2008;372(9645):1240-50

42. Agerso H, Jensen LB, Elbrond B, et al. The

pharmacokinetics, pharmacodynamics, safety

and tolerability of NN2211, a new long-

acting GLP-1 derivative, in healthy men.

Diabetologia 2002;45(2):195-202

43. Garber A, Henry R, Ratner R, et al.

Liraglutide versus glimepiride monotherapy

for type 2 diabetes (LEAD-3 Mono): a

randomised, 52-week, phase III, double-

blind, parallel-treatment trial. Lancet 2009;

373(9662):473-81

Defining the role of GLP-1 RAs for individualized treatment of type 2 diabetes Review

informahealthcare.com doi: 10.1586/17446651.2014.949672

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

44. Marre M, Shaw J, Brandle M, et al.

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(3):268-78

45. Nauck M, Frid A, Hermansen K, et al.

Efficacy 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(1):84-90

46. Zinman B, Gerich J, Buse JB, et al. Efficacy

and safety of the human glucagon-like

peptide-1 analog liraglutide in combination

with metformin and thiazolidinedione in

patients with type 2 diabetes (LEAD-4 Met

+TZD). Diabetes Care 2009;32(7):1224-30

47. Scott DA, Boye KS, Timlin L, et al. A

network meta-analysis to compare glycaemic

control in patients with type 2 diabetes

treated with exenatide once weekly or

liraglutide once daily in comparison with

insulin glargine, exenatide twice daily or

placebo. Diabetes Obes Metab 2013;15(3):

213-23

48. Deacon CF, Mannucci E, Ahren B.

Glycaemic efficacy of glucagon-like peptide-

1 receptor agonists and dipeptidyl peptidase-

4 inhibitors as add-on therapy to metformin

in subjects with type 2 diabetes-a review

and meta analysis. Diabetes Obes Metab

2012;14(8):762-7

49. Buse JB, Nauck M, Forst T, et al.

Exenatide once weekly versus liraglutide

once daily in patients with type 2 diabetes

(DURATION-6): a randomised, open-label

study. Lancet 2013;381(9861):117-24

50. Matthews JE, Stewart MW, De Boever EH,

et al. Pharmacodynamics, pharmacokinetics,

safety, and tolerability of albiglutide, a long-

acting glucagon-like peptide-1 mimetic, in

patients with type 2 diabetes. J Clin

Endocrinol Metab 2008;93(12):4810-17

51. Pratley RE, Nauck MA, Barnett AH, et al.

Once-weekly albiglutide versus once-daily

liraglutide in patients with type 2 diabetes

inadequately controlled on oral drugs

(HARMONY 7): a randomised, open-label,

multicentre, non-inferiority phase 3 study.

Lancet Diabetes Endocrinol 2014;2(4):

289-97

52. 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(3):1239-46

53. Phung OJ, Scholle JM, Talwar M,

Coleman CI. Effect of noninsulin

antidiabetic drugs added to metformin

therapy on glycemic control, weight gain,

and hypoglycemia in type 2 diabetes. JAMA

2010;303(14):1410-18

54. Liu SC, Tu YK, Chien MN, Chien KL.

Effect of antidiabetic agents added to

metformin on glycaemic control,

hypoglycaemia and weight change in

patients with type 2 diabetes: a network

meta-analysis. Diabetes Obes Metab 2012;

14(9):810-20

55. Mukai E, Fujimoto S, Sato H, et al.

Exendin-4 suppresses SRC activation and

reactive oxygen species production in

diabetic Goto-Kakizaki rat islets in an Epac-

dependent manner. Diabetes 2011;60(1):

218-26

56. Yabe D, Seino S. Dipeptidyl peptidase-4

inhibitors and sulfonylureas for type 2

diabetes: Friend or Foe J Diabetes Investig

2014. [Epub ahead of print]

57. Wang Y, Li L, Yang M, et al. Glucagon-like

peptide-1 receptor agonists versus insulin in

inadequately controlled patients with type 2

diabetes mellitus: a meta-analysis of clinical

trials. Diabetes Obes Metab 2011;13(11):

972-81

58. Mulligan CM, Harper R, Harding J, et al.

A retrospective audit of type 2 diabetes

patients prescribed liraglutide in real-life

clinical practice. Diabetes Ther 2013;4(1):

147-51

59. Wysham C, Blevins T, Arakaki R, et al.

Efficacy and safety of dulaglutide added

onto pioglitazone and metformin versus

exenatide in type 2 diabetes in a

randomized controlled trial (AWARD-1).

Diabetes Care 2014;37(8):2159-67

60. Grimm M, Han J, Weaver C, et al.

Efficacy, safety, and tolerability of exenatide

once weekly in patients with type 2 diabetes

mellitus: an integrated analysis of the

DURATION trials. Postgrad Med 2013;

125(3):47-57

61. Rosenstock J, Forst T, Aronson R, et al.

Efficacy and safety of once-daily lixisenatide

added on to titrated glargine plus oral

agents in type 2 diabetes: getgoal-duo 1

study. Abstract #62-OR. Presented at:

American Diabetes Association, 2012

62. Ferdinand KC, White WB, Calhoun DA,

et al. Effects of the once-weekly glucagon-

like peptide-1 receptor agonist dulaglutide

on ambulatory blood pressure and heart rate

in patients with type 2 diabetes mellitus.

Hypertension 2014. [Epub ahead of print]

63. Robinson LE, Holt TA, Rees K, et al.

Effects of exenatide and liraglutide on heart

rate, blood pressure and body weight:

systematic review and meta-analysis. BMJ

Open 2013;3(1):e001986

64. Chatterjee DJ, Khutoryansky N,

Zdravkovic M, et al. Absence of QTc

prolongation in a thorough QT study with

subcutaneous liraglutide, a once-daily

human GLP-1 analog for treatment of type

2 diabetes. J Clin Pharmacol 2009;49(11):

1353-62

65. Darpo B, Zhou M, Matthews J, et al.

Albiglutide does not prolong QTc interval

in healthy subjects: a thorough ECG study.

Diabetes Ther 2014;5(1):141-53

66. Linnebjerg H, Seger M, Kothare PA, et al.

A thorough QT study to evaluate the effects

of singledose exenatide 10 mug on cardiac

repolarization in healthy subjects. Int J Clin

Pharmacol Ther 2011;49(10):594-604

67. Parks M, Rosebraugh C. Weighing risks

and benefits of liraglutide - the FDA’sreview of a new antidiabetic therapy. N

Engl J Med 2010;362(9):774-7

68. Marso SP, Lindsey JB, Stolker JM, et al.

Cardiovascular safety of liraglutide assessed

in a patient-level pooled analysis of phase 2:

3 liraglutide clinical development studies.

Diab Vasc Dis Res 2011;8(3):237-40

69. Monami M, Dicembrini I, Nardini C, et al.

Effects of glucagon-like peptide-1 receptor

agonists on cardiovascular risk: a meta-

analysis of randomized clinical trials.

Diabetes Obes Metab 2014;16(1):38-47

70. Anderson SL, Trujillo JM. Association of

pancreatitis with glucagon-like peptide-1

agonist use. Ann Pharmacother 2010;44(5):

904-9

71. Singh S, Chang HY, Richards TM, et al.

Glucagon-like peptide 1-based therapies and

risk of hospitalization for acute pancreatitis

in type 2 diabetes mellitus: a population-

based matched case-control study. JAMA

Intern Med 2013;173(7):534-9

72. Garg R, Chen W, Pendergrass M. Acute

pancreatitis in type 2 diabetes treated with

exenatide or sitagliptin: a retrospective

observational pharmacy claims analysis.

Diabetes Care 2010;33(11):2349-54

• This large observational study identified

an increased risk of acute pancreatitis in

diabetic versus non-diabetic patients, but

did not see an association between

exenatide use and pancreatitis.

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

73. Monami M, Dicembrini I, Nardini C, et al.

Glucagon-like peptide-1 receptor agonists

and pancreatitis: a meta-analysis of

randomized clinical trials. Diabetes Res Clin

Pract 2014;103(2):269-75

74. Egan AG, Blind E, Dunder K, et al.

Pancreatic safety of incretin-based drugs –

FDA and EMA assessment. N Engl J Med

2014;370(9):794-7

• This study reported comprehensive

independent evaluations by both the

European and US regulatory bodies that

found that assertions of a causal

relationship between GLP-1 RAs and

pacreatitis or pancreatic cancer were not

consistent with the currently available

data.

75. Bjerre KL, 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(4):1473-86

76. Hegedus L, Moses AC, Zdravkovic M, et al.

GLP-1 and calcitonin concentration in

humans: lack of evidence of calcitonin

release from sequential screening in over

5000 subjects with type 2 diabetes or

nondiabetic obese subjects treated with the

human GLP-1 analog, liraglutide. J Clin

Endocrinol Metab 2011;96(3):853-60

77. Elashoff M, Matveyenko AV, Gier B, et al.

Pancreatitis, pancreatic, and thyroid cancer

with glucagon-like peptide-1-based

therapies. Gastroenterology 2011;141(1):

150-6

78. Nauck MA, Friedrich N. Do GLP-1-based

therapies increase cancer risk? Diabetes Care

2013;36(Suppl 2):S245-52

79. Kawata T, Kanamori A, Kubota A, et al. Is

a switch from insulin therapy to liraglutide

possible in Japanese type 2 diabetes mellitus

patients? J Clin Med Res 2014;6(2):138-44

80. Kim YG, Hahn S, Oh TJ, et al. Differences

in the HbA1c-lowering efficacy of glucagon-

like peptide-1 analogues between Asians and

non-Asians: a systematic review and meta-

analysis. Diabetes Obes Metab 2014. [Epub

ahead of print]

81. Seino Y, Takami A, Boka G, et al.

Pharmacodynamics of the glucagon-like

peptide-1 receptor agonist lixisenatide in

Japanese and Caucasian patients with type 2

diabetes mellitus poorly controlled on

sulphonylureas with/without metformin.

Diabetes Obes Metab 2014;16(8):739-47

82. Fukushima M, Suzuki H, Seino Y. Insulin

secretion capacity in the development from

normal glucose tolerance to type 2 diabetes.

Diabetes Res Clin Pract 2004;66(Suppl 1):

S37-43

83. Yabe D, Watanabe K, Sugawara K, et al.

Comparison of incretin immunoassays with

or without plasma extraction: incretin

secretion in Japanese patients with type 2

diabetes. J Diabetes Investig 2012;3(1):70-9

84. Yabe D, Kuroe A, Watanabe K, et al. Little

enhancement of meal-induced GLP-1

secretion in Japanese: comparison of type 2

diabetes and healthy controls. J Diabetes

Investig 2010;1(1-2):56-9

85. Meier JJ, Vilsboll T, Donsmark M, et al.

Greater glycaemic control is achieved with

the once-daily human GLP-1 analogue

liraglutide vs comparators across the

continuum of estimated beta cell mass.

Diabetologia 2011;54(Suppl 1):1-542

• A study that demonstrated the potential

for GLP-1 RAs in aiding glycemic

control, even in patients with advanced

diabetes (low b-cell mass).

86. Usui R, Yabe D, Kuwata H, et al.

Retrospective analysis of safety and efficacy

of insulin-to-liraglutide switch in Japanese

type 2 diabetes: a caution against

inappropriate use in patients with reduced

I2-cell function. J Diabetes Invest 2001;4(6):

585-94

87. Anichini R, Cosimi S, Di CA, et al. Gender

difference in response predictors after 1-year

exenatide therapy twice daily in type 2

diabetic patients: a real world experience.

Diabetes Metab Syndr Obes 2013;6:123-9

88. Yabe D, Seino Y. Incretin actions beyond

the pancreas: lessons from knockout mice.

Curr Opin Pharmacol 2013;13(6):946-53

89. Fujita H, Morii T, Fujishima H, et al. The

protective roles of GLP-1R signaling in

diabetic nephropathy: possible mechanism

and therapeutic potential. Kidney Int 2014;

85(3):579-89

• This study in a mouse model identified a

protective role for GLP-1 against

oxidative stress in the kidney.

90. Seino Y, Yabe D. GIP and GLP-1: incretin

actions beyond the pancreas. J Diabetes

Investig 2013;4(2):108-30

91. Buse JB, Bergenstal RM, Glass LC, et al.

Use of twice-daily exenatide in Basal

insulin-treated patients with type 2 diabetes:

a randomized, controlled trial. Ann Intern

Med 2011;154(2):103-12

92. DeVries JH, Bain SC, Rodbard HW, et al.

Sequential intensification of metformin

treatment in type 2 diabetes with liraglutide

followed by randomized addition of basal

insulin prompted by A1C targets. Diabetes

Care 2012;35(7):1446-54

93. Riddle MC, Aronson R, Home P, et al.

Adding once-daily lixisenatide for type 2

diabetes inadequately controlled by

established basal insulin: a 24-week,

randomized, placebo-controlled comparison

(GetGoal-L). Diabetes Care 2013;36(9):

2489-96

94. Vilsboll T, Christensen M, Junker AE, et al.

Effects of glucagon-like peptide-1 receptor

agonists on weight loss: systematic review

and meta-analyses of randomised controlled

trials. BMJ 2012;344:d7771

95. Monami M, Dicembrini I, Marchionni N,

et al. Effects of glucagon-like peptide-1

receptor agonists on body weight: a meta-

analysis. Exp Diabetes Res

2012;2012:672658

96. Flint A, Raben A, Astrup A, Holst JJ.

Glucagon-like peptide 1 promotes satiety

and suppresses energy intake in humans. J

Clin Invest 1998;101(3):515-20

97. Ussher JR, Drucker DJ. Cardiovascular

biology of the incretin system. Endocr Rev

2012;33(2):187-215

98. 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(8):962-5

99. Read PA, Khan FZ, Dutka DP.

Cardioprotection against ischaemia induced

by dobutamine stress using glucagon-like

peptide-1 in patients with coronary artery

disease. Heart 2012;98(5):408-13

100. Lonborg J, Kelbaek H, Vejlstrup N, et al.

Exenatide reduces final infarct size in

patients with ST-segment-elevation

myocardial infarction and short-duration of

ischemia. Circ Cardiovasc Interv 2012;5(2):

288-95

101. Lonborg J, Vejlstrup N, Kelbaek H, et al.

Exenatide reduces reperfusion injury in

patients with ST-segment elevation

myocardial infarction. Eur Heart J 2012;

33(12):1491-9

102. Wang B, Zhong J, Lin H, et al. Blood

pressure-lowering effects of GLP-1 receptor

agonists exenatide and liraglutide: a meta-

analysis of clinical trials. Diabetes Obes

Metab 2013;15(8):737-49

103. Fonseca VA, Devries JH, Henry RR, et al.

Reductions in systolic blood pressure with

liraglutide in patients with type 2 diabetes:

insights from a patient-level pooled

analysis of six randomized clinical trials. J

Diabetes Complications 2014;28(3):

399-405

Defining the role of GLP-1 RAs for individualized treatment of type 2 diabetes Review

informahealthcare.com doi: 10.1586/17446651.2014.949672

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.

104. Irace C, De LS, Shehaj E, et al. Exenatide

improves endothelial function assessed by

flow mediated dilation technique in subjects

with type 2 diabetes: results from an

observational research. Diab Vasc Dis Res

2013;10(1):72-7

105. Kim M, Platt MJ, Shibasaki T, et al. GLP-

1 receptor activation and Epac2 link atrial

natriuretic peptide secretion to control of

blood pressure. Nat Med 2013;19(5):

567-75

• This study identified a novel mechanism

by which GLP-1 plays a role in

regulation of blood pressure.

106. Meier JJ, Gethmann A, Gotze O, et al.

Glucagon-like peptide 1 abolishes the

postprandial rise in triglyceride

concentrations and lowers levels of non-

esterified fatty acids in humans.

Diabetologia 2006;49(3):452-8

107. Schwartz EA, Koska J, Mullin MP, et al.

Exenatide suppresses postprandial elevations

in lipids and lipoproteins in individuals

with impaired glucose tolerance and recent

onset type 2 diabetes mellitus.

Atherosclerosis 2010;212(1):217-22

108. Arnolds S, Dellweg S, Clair J, et al. Further

improvement in postprandial glucose

control with addition of exenatide or

sitagliptin to combination therapy with

insulin glargine and metformin: a proof-of-

concept study. Diabetes Care 2010;33(7):

1509-15

109. Leahy JL. Insulin therapy in type 2 diabetes

mellitus. Endocrinol Metab Clin North Am

2012;41(1):119-44

110. Nayak UA, Govindan J, Baskar V, et al.

Exenatide therapy in insulin-treated type 2

diabetes and obesity. QJM 2010;103(9):

687-94

111. Owens DR, Luzio SD, Sert-Langeron C,

Riddle MC. Effects of initiation and

titration of a single pre-prandial dose of

insulin glulisine while continuing titrated

insulin glargine in type 2 diabetes: a

6-month ‘proof-of-concept’ study. Diabetes

Obes Metab 2011;13(11):1020-7

112. Riddle MC, Aronson R, Home P, et al.

Adding once-daily lixisenatide for type 2

diabetes inadequately controlled by

established basal insulin: a 24-week,

randomized, placebo-controlled comparison

(GetGoal-L). Diabetes Care 2013;36(9):

2489-96

113. Riddle MC, Forst T, Aronson R, et al.

Adding once-daily lixisenatide for type 2

diabetes inadequately controlled with newly

initiated and continuously titrated basal

insulin glargine: a 24-Week, randomized,

placebo-controlled study (GetGoal-Duo 1).

Diabetes Care 2013;36(9):2497-503

114. Tobin GS, Cavaghan MK, Hoogwerf BJ,

McGill JB. Addition of exenatide twice

daily to basal insulin for the treatment of

type 2 diabetes: clinical studies and practical

approaches to therapy. Int J Clin Pract

2012;66(12):1147-57

115. Gross JL, Kramer CK, Leitao CB, et al.

Effect of antihyperglycemic agents added to

metformin and a sulfonylurea on glycemic

control and weight gain in type 2 diabetes:

a network meta-analysis. Ann Intern Med

2011;154(10):672-9

116. Mathieu C, Rodbard HW, Cariou B, et al.

A comparison of adding liraglutide versus a

single daily dose of insulin aspart to insulin

degludec in subjects with type 2 diabetes

(BEGIN: VICTOZA ADD-ON). Diabetes

Obes Metab 2014;16(7):636-44

117. Wu D, Li L, Liu C. Efficacy and safety of

dipeptidyl peptidase-4 inhibitors and

metformin as initial combination therapy

and as monotherapy in patients with type 2

diabetes mellitus: a meta-analysis. Diabetes

Obes Metab 2013;16(1):30-7

118. Diamant M, Van GL, Stranks S, et al.

Once weekly exenatide compared with

insulin glargine titrated to target in patients

with type 2 diabetes (DURATION-3): an

open-label randomised trial. Lancet 2010;

375(9733):2234-43

119. Blevins T, Pullman J, Malloy J, et al.

DURATION-5: exenatide once weekly

resulted in greater improvements in

glycemic control compared with exenatide

twice daily in patients with type 2 diabetes.

J Clin Endocrinol Metab 2011;96(5):

1301-10

120. Umpierrez G, Tofe Povedano S,

Perez Manghi F, et al. Efficacy and Safety

of Dulaglutide Monotherapy Versus

Metformin in Type 2 Diabetes in a

Randomized Controlled Trial (AWARD-3).

Diabetes Care 2014;37(8):2168-76

Review Yabe & Seino

doi: 10.1586/17446651.2014.949672 Expert Rev. Endocrinol. Metab.

Exp

ert R

evie

w o

f E

ndoc

rino

logy

& M

etab

olis

m D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y 18

2.24

9.24

7.14

on

09/0

3/14

For

pers

onal

use

onl

y.