You are on page 1of 9

CE: Tripti; MED/270108; Total nos of Pages: 9;

MED 270108

REVIEW

CURRENT
OPINION Incretin therapy for diabetes mellitus type 2
Jens Juul Holst

Purpose of review
Among the gastrointestinal hormones, the incretins: glucose-dependent insulinotropic polypeptide and
glucagon-like peptide-1 have attracted interest because of their importance for the development and
therapy of type 2 diabetes and obesity. New agonists and formulations of particularly the GLP-1 receptor
have been developed recently showing great therapeutic efficacy for both diseases.
Recent findings
The status of the currently available GLP-1 receptor agonists (GLP-1RAs) is described, and their strengths
and weaknesses analyzed. Their ability to also reduce cardiovascular and renal risk is described and
analysed. The most recent development of orally available agonists and of very potent monomolecular
co-agonists for both the GLP-1 and GIP receptor is also discussed.
Summary
The GLP-1RAs are currently the most efficacious agents for weight loss, and show potential for further
efficacy in combination with other food-intake-regulating peptides. Because of their glycemic efficacy and
cardiorenal protection, the GLP-1 RAs will be prominent elements in future diabetes therapy.
Keywords
dulaglutide, glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide, semaglutide,
weight-losing therapy

INTRODUCTION THE PHYSIOLOGICAL BACKGROUND FOR


According to the newest guidelines and recommen- THE USE OF GLP-1 RECEPTOR AGONISTS
dations, GLP-1 receptor agonists and DPP-4 inhib-
itors (which act by increasing levels of active Action on insulin secretion and food intake
incretin hormones) are now recommended as at The GLP-1 RAs act via activation of the GLP-1 recep-
least second-line treatment for type 2 diabetes [1]. tor [4] – there are reports of actions that are thought
The GLP-1 receptor agonists are particularly recom- to be independent of the receptor, but the existence
mended for patients with evidence of cardiovascular of such effects needs better experimental support.
disease complicating their diabetes. These recom- The GLP-1 receptor is expressed in numerous tissues,
mendations are mainly based on the results of very but even today – about 18 years after the cloning of
large outcome trials, in which the GLP-1 receptor the receptor – its precise localization is not known
agonists on top of their well established antidiabetic in detail [5]. A site of very dense expression of the
activity have proven superior to placebo in reducing receptor is the pancreatic beta cells, where activa-
&
the risk of cardiovascular events [2 ] (whereas the tion by the ligands leads to cAMP formation
DPP-4 inhibitors have proven neutral in this respect and eventually insulin secretion via PKA and
[3]). Another important finding is the risk of devel- Epac activation [6]. The insulinotropic action is
oping renal adverse events, but in this case, both the best described as potentiation of glucose-induced
GLP-1 receptor agonists (GLP-1RAs) and DPP-4
inhibitors have shown reduced risk compared with
NNF Center for Basic Metabolic Research and Department of Biomedi-
placebo. The purpose of this brief overview is to
cal Sciences, The Panum Institute, Faculty of Health Sciences, University
discuss the background for this remarkable develop- of Copenhagen, Blegdamsvej 3, Copenhagen N, Denmark
ment, and to discuss briefly the role of the incretin- Correspondence to Jens Juul Holst, NNF Center for Basic Metabolic
based therapies in the modern/future therapy of Research and Department of Biomedical Sciences, The Panum Institute,
type 2 diabetes. The focus will be on GLP-1 receptor Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3,
agonists, but the possible role of glucose-dependent DK-2200 Copenhagen N, Denmark. E-mail: jjholst@sund.ku.dk
insulinotropic polypeptide (GIP) will also be briefly Curr Opin Endocrinol Diabetes Obes 2019, 26:000–000
discussed. DOI:10.1097/MED.0000000000000516

1752-296X Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-endocrinology.com

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Gastrointestinal hormones

food intake is dependent on this uptake [13]. The


KEY POINTS GLP-1 receptor is barely expressed on the pancreatic
 Agonists of the receptor for the incretin hormone, GLP-1 alpha cells (if at all) [14], but abundantly on the
(GLP-1RAs) have proven successful for the therapy of somatostatin-producing delta cells, and it is gener-
both obesity and diabetes and currently range among ally thought that the inhibition of glucagon secre-
the most efficacious therapies for these diseases. tion caused by the GLP-1 RAs is exerted via
stimulation of islet somatostatin, inhibiting gluca-
 Newer GLP-1 RAs suitable for once weekly
administration have been developed and an orally gon secretion in a paracrine manner [15]. The GLP-1
active GLP-1 RA has recently been approved. induced inhibition of glucagon secretion may be of
considerable importance for the antidiabetic effects
 In large cardiovascular outcome trials, the GLP-1RAs of the GLP-1 RAs [16], and from pancreatic clamping
are not only well tolerated, but reduce risk of
experiments in humans, it has been calculated that
cardiovascular and renal adverse events, extending
their applicability for metabolic therapy. around 50% of the glucose-lowering effect of GLP-1
in type 2 diabetes is because of inhibition of gluca-
 The other incretin hormone, glucose-dependent gon secretion whereas the remaining 50% is because
insulinotropic polypeptide (GIP), has little acute effect of stimulation of insulin secretion [17]. Although
on insulin secretion in patients with type 2 diabetes,
there are clearly GLP-1 receptors in the mentioned
and no effect on food intake, but may show additional
effects when combined with GLP-1 in monomolecular circumventricular organs [18], there are many more
co-agonists. GLP-1 receptors in the brain, which do not seem to
be accessible for peripheral GLP-1 or GP-1RAs [19].
These receptors are probably targets for GLP-1
released from nerve fibers projecting from a small
secretion [7], and this is important, because it group of proglucagon-producing neurons in the
explains that GLP1RAs in general do not produce nucleus of the solitary tract [20]. These neurons
hypoglycemia (unless they are combined with sul- may be activated by a number of peripheral mainly
fonylurea compounds, which uncouple this glucose vagally transmitted stimuli, and in this way signal to
dependency [8]) and also explains that insulin secre- parts of the brain that are involved in regulation of
tion usually is not increased during therapy, but appetite, food intake, and reward [21]. The relation-
now occurs at a lower ambient glucose concentra- ship between peripheral GLP-1 and the GLP-1 neu-
tion [9]. Being peptide-based, all GLP-1 RAs require rons in the brain stem is not clear, but gastric
subcutaneous injection [note, however, that the emptying, for instance, has been reported to activate
first oral GLP1RA has just been approved by the the GLP-1 neurons [22]. It cannot be excluded that,
Food and Drug Administration (FDA) [10]], and this physiologically, the GLP-1 system mainly functions
means that their plasma concentration will increase as a part of the so-called ileal brake, endocrine
after the injection, and that increased concentra- mechanisms evoked by the presence of nutrients
tions may reach the beta cell and activate the recep- in the distal small intestine (where GLP-1 expression
tors. Most likely, this scenario differs from the is greatest), which cause inhibition of appetite and
actions of the endogenous hormone, GLP-1, which food intake as well as inhibition of upper gastroin-
is degraded extensively and rapidly by the dipep- testinal secretion and motility [23]. Only with large
tidyl peptidase-4 in the intestinal capillaries of the meals would the GLP-1 system be sufficiently acti-
gut and the liver after its release from the gut, vated (the secretory responses are related to meal
leaving only a very small fraction to reach the size [24]) to allow endocrine interactions with the
pancreas in the intact form [11]. Instead, the endog- pancreatic islets.
enous peptide is thought to activate enteric neurons
and sensory afferent fibers of the vagus (there is a
dense expression of the receptors in the enteric Other actions
plexuses), with impulses reaching the brain stem Other sites of GLP-1 receptor expression include the
and hypothalamic nuclei involved in regulation of Brunner’s glands of the duodenum [25], where GLP-
appetite and gastrointestinal secretions and motility 1 may induce mucus secretion and secretion of
[7]. To what extent exogenous GLP-1 receptor ago- antibacterial agents, and smooth muscles of the
nists are able to activate this neural system is not afferent arterioles in the kidney [26], suggesting
known. For them, an important site of GLP-1 recep- involvement in the regulation of renal hemody-
tor activation may be the circumventricular organs namics and perhaps consistent with the proposal
of the brain [12], and it appears that the agonists of an intestinorenal axis regulating sodium excre-
may access the area postrema, the median eminence tion [27]. Finally, a dense receptor expression
and the arcuate nucleus and that their effects on of receptors has been found on intestinal

2 www.co-endocrinology.com Volume 26  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Incretin therapy for diabetes mellitus type 2 Holst

intraepithelial lymphocytes, which may represent evidence for this probably derives from the pro-
an entry for GLP-1 to the immune systems, perhaps longed effect on glycated hemoglobin (hemoglobin
consistent with the many reports of anti-inflamma- A1c, a measure of average glucose concentrations
tory actions of GLP-1 and the GLP-1RAs [28]. Local- over 3 months) upon continued therapy with a
ization of GLP-1 receptors that can explain the GLP1 RA (for instance up to 5 years with liraglutide
antiatherosclerotic and cardiovascular effects [29] in the LEADER trial [39]). Signs of beta cell regener-
of the GLP-1RAs is currently lacking, possibly ation or proliferation, originally proposed to occur
because of insufficient sensitivity of the applied [40], have not been observed, however. In addition
methods. to the effects on the islets, GLP-1RAs inhibit appetite
and food intake [41] leading to weight losses.
Weight loss per se has incredibly important conse-
The antidiabetic effect of the incretin quences for metabolic disease and diabetes with
hormones improvement of insulin resistance being one of
So being normally secreted, why is it that the incre- the most important elements [42]. On top of that
tins do not prevent type 2 diabetes from occurring? comes the cardiovascular and renal benefit.
It turns out that the incretin effect, the amplifica-
tion of insulin secretion exerted by hormones from
the gut, is severely impaired in patients with T2DM WHERE ARE WE TODAY WITH RESPECT
[30]. The main reason is that the two hormones, in TO INCRETIN-BASED THERAPIES?
physiological concentrations, have lost most of The first problem to solve was the apparent instabil-
their insulinotropic effects in these patients [31]. ity of the GLP-1 molecule which, as mentioned, is
But whereas GIP is ineffective regardless of dose [32], degraded by DPP-4, leaving the peptide with a half-
higher doses of exogenous GLP-1 are able to restore life in the circulation of less than 2 min [43]. The
beta cell responses to glucose to completely normal discovery of this inactivation [44] lead to the pro-
values [33] (although the response is lower than that posal that inhibition of the DPP-4 enzyme might be
observed in healthy individuals after combined glu- useful in diabetes therapy [45], and indeed this
cose and incretins). This explains the clinical effec- turned out to be the case, as first demonstrated
tiveness of the GLP-1 RAs and also explains that experimentally [46] and subsequently in clinical
GLP-1s have little effect on beta cells with poor studies [47]. Given the extensive degradation of
residual secretory capacity [34]. There are also endogenous GLP-1 (80–90%), it was expected that
abnormalities of glucagon secretion in T2DM with levels of intact GLP-1 might increase by a factor of
delayed and reduced suppression by glucose, and it more than 5 after inhibition, but clinically the levels
appears that the higher doses of GLP-1 can also only rise by a factor of 2–3 [48], apparently because
restore alpha cell sensitivity to glucose [32], of a negative feed-back cycle involving intestinal
although in general, the inhibitory effect of glucose somatostatin restraining on GLP-1 secretion [49].
itself is retained in the diabetic alpha cell [35], as However, levels of active GLP-1 are apparently ele-
opposed to the beta cells. vated sufficiently to influence pancreatic islet secre-
tion. Also the other incretin hormone, GIP, is
protected from degradation by DPP-4 [48], and
The therapeutic potential of glucagon-like although its contribution to the antidiabetic action
peptide-1 of the inhibitors is uncertain, there are reports indi-
All together the therapeutic potential of GLP-1 in cating that the improved glycemic control may
T2DM is surprising. It acts on the diabetic beta cells restore some of the lost insulinotropic actions of
to stimulate insulin secretion, and it inhibits gluca- GIP [50].
gon secretion, which is generally increased in T2DM Another way to exploit the GLP-1 system was of
(presumably because of hepatic glucagon resistance course to develop DPP-4 resistant analogs, and this
and stimulation of glucagon secretion by elevated turned out to be rather simple, a substitution of the
aminoacid levels [36]) and is known to contribute amino acid residue in position 2 was sufficient [51].
importantly to the diabetic hyperglycemia in the This, however, as it turned out, only prolonged the
fasting state (as demonstrated by the actions of half-life to 4–5 min because of extensive renal elim-
glucagon receptor antagonists [37]). GLP-1 has also ination of the peptide. The discovery of a resistant
been proposed to protect beta cells against progres- GLP-1 RA, exendin-4, a peptide from the saliva of
sion of disease. Thus GLP-1 appears to protect beta the Gila Monster, a poisonous lizard from Arizona
cells, also human, from apoptosis induced by cyto- [52] was a solution to this problem. This peptide is
kines [38], but whether protection may actually stable and exclusively cleared by glomerular filtra-
occur clinically in humans is uncertain. The best tion in the kidneys, giving it an intravenous half-life

1752-296X Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-endocrinology.com 3

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Gastrointestinal hormones

of about 30 min (2–3 h after subcutaneous injec- commercial grounds, although in a large recent
tion) [53]. A synthetic product, exenatide, came to cardiovascular outcome trial (the Harmony trial
the market in 2005 [54] and is still available as the [63]), it performed surprisingly well.
prototype of the so-called short-acting agonists. The Lilly company introduced dulaglutide, a
Their duration of action is brief, covering only long-acting fusion product of two stabilized GLP-1
one or two meals. This problem has been partly moieties with an fc immunoglobulin fragment, in
addressed by giving two daily injections (before 2015 [64], and immediately this seemed comparable
meals) or increasing the dose (as is done with a with liraglutide, but with the benefit of once weekly
modified analogue, lixisenatide). The short-acting administration. Also the cardiovascular outcomes
agonists very potently reduce postprandial glucose were positive with dulaglutide (the REWIND trial
&
excursions because of the pronounced effects on [65 ], see below). NovoNordisk subsequently intro-
gastric emptying [54]), but they have, as expected duced another acylated but stabilized and optimized
because of their short duration of action, poor version of liragutide, semaglutide, suitable for weekly
effects on fasting glycemia, and in a cardiovascular administration and selected for greater efficacy with
outcome study (CVOT, see below), lixisenatide (a respect to both glycemic control and weight loss [66].
slightly modified analogue of exendin-4) did not The general features of the GLP-1RAs are thus
show cardiovascular benefits [55], which does not their ability to improve glycemic control in patients
support the use of short-acting agonists. A formula- with T2DM and to cause weight loss. It is outside the
tion of exenatide (Bydureon) making it suitable for scope of this short overview to discuss the importance
once weekly administration was introduced [56] of improved glycemic control in T2DM, but this
from 2008. The subcutaneous administration of this target is still considered key to diabetes therapy.
agonist is not without problems and it has a ten- Losing overweight, on the other hand, is extremely
dency to generate antibodies and nodules at the important: because of the involvement of overweight
injection site. In addition, its efficacy is weaker in in the diabetes pathogenesis, because of the meta-
head-to-head comparisons with other weekly ago- bolic and cardiovascular improvements following
nists (possibly a dosing problem), and this com- weight loss, and because of the congruence with most
bined with somewhat discouraging results of a patients’ desire [42]. When choosing among the vari-
very large outcome study (EXSCEL [57]) suggests ous GLP-1 receptor agonists, it is, therefore, relevant
that in its current dosing scheme, it is less competi- to compare their effects on these targets. In addition
tive. A modified long-acting (weekly) and poten- to that comes convenience, which, regarding the
tially more potent exendin-derived agonist, injectables, would provide the weekly agonists with
efpeglenatide, is currently under development [58]. a considerable advantage; and equally importantly,
The remaining agonists are derived from the the side effects, which although the frequency of
backbone of the human/mammalian GLP-1 mole- severe side effects is low, still remains a problem
cule. The first was liraglutide, consisting of the GLP- for the GLP-1RAs, which almost inevitably cause
1 molecule with a fatty acid chain (palmitate) nausea, vomiting, diarrhea and stomach pain in
attached via a linker, whereby it binds to albumin some patients in the initial phase of the therapy.
in the circulation [59]. This provides it with a half- The importance the patients (and the doctor) may
life in the circulation (12 h) sufficient to cover the attribute to each of these features varies a lot (gener-
24 h of the day, ensuring a stable exposure in steady ally weight loss is the most valued property [67]), so
state. Liraglutide has been extensively evaluated, the choice is not easy. It is important to emphasize
particularly in the previously mentioned 3.5 years that the magnitude of the effects of the compounds
(up to 5 years) cardiovascular outcome study, varies considerably from person to person and from
LEADER [39]. A discussion of its cardiovascular study to study, which means that direct comparisons
effect will follow below. As already mentioned, this in controlled clinical trials are absolutely essential.
agonist provides lasting weight loss and improve- On the basis of such considerations, the currently
ments of HbA1c. A fusion molecule between two most attractive compounds would be the weekly
GLP-1 sequences and human albumin, designed for agonists, semaglutide, and dulaglutide, which are
at least weekly use, albiglutide, was introduced in quite comparable, although with a slightly greater
2012 [60]. This molecule was the first of the large efficacy for semaglutide, particularly with respect to
molecular weight agonists and was suspected to weight loss (see also below) [68].
have lesser effect on body weight [61] (cf. the dis-
cussion on the possible mechanism of action above),
and in comparative studies, it is clearly weaker THE CARDIOVASCULAR OUTCOME TRIALS
in this respect [62]. The company behind this When it comes to the cardiovascular benefits of the
compound has withdrawn the compound on GLP-1 RAs, the picture is less clear, because the

4 www.co-endocrinology.com Volume 26  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Incretin therapy for diabetes mellitus type 2 Holst

outcome trials differ so much. In general, they have comparison of MACE and its individual compo-
been designed to be event driven, in order to fulfill nents in the major cardiovascular outcome studies
requirements regarding cardiovascular safety completed so far. Judging from meta-analyses of
demanded by the FDA in 2010. The events typically these data, it may be concluded that the GLP-1
constitute MACE: major cardiovascular adverse receptor agonists are not only well tolerated, but
events, including nonfatal myocardial infarction, also that they are in general superior to placebo,
nonfatal stroke, and cardiovascular death. The sta- showing an average 13% relative risk reduction of
tistical strategy of the studies has been to secure MACE. Inconsistences between the performances of
significance for cardiovascular safety, that is, non- the drugs in the various trials regarding the individ-
inferiority compared with placebo, but this design ual components of MACE as well as all-cause mor-
unfortunately also implies that observed additional tality may, therefore, reflect differences between the
effects rarely reach statistical significance. There- trials more than differences between the agonists.
fore, one should not put too much weight on the For instance, in the SUSTAIN 6 trial of semaglutide
results of the secondary analyses. Figure 1 shows a [69], there was a significant reduction of nonfatal

FIGURE 1. Risk of major cardiovascular adverse events and each of its components in the GLP-1 receptor agonist
cardiovascular outcome trials (CVOTs) published so far. Data from Kristensen et al. [2 ].
&

1752-296X Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-endocrinology.com 5

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Gastrointestinal hormones

stroke and near-significant effects on nonfatal myo- pressure in agreement with the increase in heart
cardial infarction, but no effect whatsoever on rate and output). Also, in the kidneys, the beneficial
cardiovascular death. This should be contrasted effect on adverse events is mainly restricted to a
with the LEADER trial [39], where liraglutide had reduced risk of albuminuria, whereas a clear effect
a marked beneficial effects on precisely cardiovas- on glomerular filtration rate is not seen (again, in
cular death. Does this mean that the two very related contrast to the SGLT2 inhibitors) [72]. The most
compounds, liraglutide and semaglutide, differ with recent research has pointed to a lowering of angio-
respect to protection against cardiovascular death? tensin 2 levels [27], but this needs further investiga-
This is unlikely; rather, the explanation is that the tion. Thus, choosing among the different agonists
SUSTAIN 6 trial was a short, preregistration trial in a on the basis of their cardiovascular effects may be
limited number of patients, in which the number of difficult (the ‘weakest’ antidiabetic, albiglutide, had
cardiovascular deaths were too few to reveal a sig- a very significant effects on MACE in the HARMONY
nificant difference against placebo. Indeed, in this trial), although of course, a documented effect,
trial, the risk reduction regarding overall MACE would weigh more than no significant effect.
(26%) was the greatest among all the trials com-
pleted so far, consistent with the considerable effi-
cacy of this agonist. WHAT ABOUT THE FUTURE?
A major problem regarding the evaluation of the The GLP-1 receptor agonists are potent antidiabetic
cardiovascular effects of the agonists in these trials is agents, and when it comes to T2DM, they often
that we really do not know the mechanisms that are perform significantly better than insulin. Thus, after
responsible for the effects. The only established failure of oral therapy (with metformin and SU) the
cardiovascular effect of the agonists is an increase GLP-1 agonists may perform better than basal insu-
in heart rate, which is generally considered harmful lin [73], and in more advanced cases, better than
by cardiologists. The CVOTs are in general designed basal-bolus insulin therapy [74], both in terms of
for glycemic equipoise between placebo and study glycemic control and with respect to body weight
compound – thus, the studies are blinded and the control and absence of hypoglycemia. In addition,
clinicians instructed to do their best to keep hemo- the agonists may be associated with reduced athero-
globin A1C levels below the recommended target of sclerotic progression and improved cardiovascular
7% in all patients. As the GLP-1RAs agonists are risk, as discussed above.
efficient glucose-lowering agents, this means that The SGLT-2 inhibitors represent an extremely
the placebo groups receive much larger amounts of valuable addition to the diabetes therapeutics, but
additional antidiabetic medications, in particular are in general less effective with respect to glucose
more insulin. As a result, hypoglycemia is much control and weight loss [75], and they are not
more frequent in the placebo groups [39]. Could thought to be associated with slowing of atheroscle-
the seemingly beneficial effects of the GLP-1 ago- rotic progression. Their beneficial effects on cardio-
nists in reality be because of harmful effects of the vascular (in particular, heart failure) and renal
additional therapeutics given to the placebo groups? complications are so clear that their use in patients
This cannot be excluded at present. Regarding heart with nondiabetic cardiovascular and renal disease is
failure, it is a general finding that the GLP-1RAs do now being implemented [76]. Whether the GLP-
not have a beneficial effect (but, importantly, they 1RAs may also be accepted for nondiabetic patients
also do not cause or aggravate heart failure [70]). is more uncertain. At least the REWIND study with
This is in stark contrasts to the SGLT2 inhibitors [71] dulaglutide suggested that beneficial cardiovascular
showing a marked effect on exactly heart failure, effects could also be expected in diabetic individuals
perhaps consistent with their ability to cause extra- without established cardiovascular disease at the
&
cellular volume constriction. Although still incom- start of trial [65 ]. A number of studies are currently
pletely founded in experimental research, this has being performed looking at combinations of SGLT2-
led many to conclude that the GLP-1RAs act to inhibitors and GLP-1 receptor agonists, which from
slow down atherosclerotic disease progression [70] a theoretical point of view, would be very attractive.
(as illustrated by their effects to reduce the risk of Indeed, there seems to be additive effects of com-
myocardial infarction, incidence of stroke, or revas- bining the two therapies [77].
cularization procedures by 35%) as observed in the As mentioned above, the GLP-1 RAs may even-
SUSTAIN 6 trial [69]. tually be associated with secondary failure because
Other beneficial effects include a significant of progression of beta cell failure [34]. One obvious
lowering of blood pressure in most trials, but again way of solving this problem is to treat the patients
the mechanism behind this is completely unknown with combinations of basal insulin and a GLP-1 RA,
(acute administration of GLP-1 increases blood and such combinations have shown remarkable

6 www.co-endocrinology.com Volume 26  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Incretin therapy for diabetes mellitus type 2 Holst

efficacy, and the fixed combinations currently rep- effects of food intake and no effects on insulin
resent the most efficacious diabetes therapy at all. secretion and glycemia in patients with T2DM.
Thus, the introduction of the GLP-1RAs to diabetes In conclusion, it seems safe to conclude, on the
therapy must be considered a very significant basis of these recent developments that the GLP-1
improvement RAs have a bright future ahead of them and that
As discussed above, some of the agonists have diabetes and obesity therapy is likely to include
shown efficacy as weight-losing agents. By slow and some form of GLP-1 receptor agonism for many
careful up-titration of the agonists, it may be possi- years ahead.
ble to reach higher doses than those recommended
for diabetes therapy without creating intolerable Acknowledgements
side effects whereby their food-intake inhibiting None.
effects can be exploited better. Thus, liraglutide at
3 mg (as opposed to 1.8 mg for diabetes therapy) has Financial support and sponsorship
been approved for nondiabetic (and diabetic) obe- None.
sity. It is moderately effective [78], but has the
benefit, compared with other weight-losing agents, Conflicts of interest
that it also counteracts the metabolic complications
JJH has received lecture fees and participated in advisory
of obesity, above all the development of diabetes,
boards for MSD, NovoNordisk, Lilly, GSK, Sanofi,
which may be prevented or delayed to a consider-
Hamni, and AstraZeneca.
able extent [79]. The more effective agonist, sema-
glutide, has also been investigated in higher doses,
for example, using lower but daily injections
amounting to a several fold higher total dose per REFERENCES AND RECOMMENDED
week than the currently approved 1 mg dose, and in READING
Papers of particular interest, published within the annual period of review, have
these studies up to 15% weight losses have been been highlighted as:
&
observed [80 ]. This makes semaglutide the most & of special interest
&& of outstanding interest
efficacious weight-losing agents of all, illustrating
the huge future potential of the GLP-1RAs in 1. Davies MJ, D’Alessio DA, Fradkin J, et al. Management of hyperglycemia in
type 2 diabetes, 2018. A consensus report by the American Diabetes
this field. Association (ADA) and the European Association for the Study of Diabetes
GLP-1 is not the only appetite-inhibiting and (EASD). Diabetes Care 2018; 41:2669–2701.
2. Kristensen SL, Rorth R, Jhund PS, et al. Cardiovascular, mortality, and kidney
food intake-inhibiting hormone from the gastroin- & outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a
testinal tract, and studies of patients after bariatric systematic review and meta-analysis of cardiovascular outcome trials. Lancet
Diabetes Endocrinol 2019; 7:776–785.
surgery have taught us that the weight loss associ- Very useful updated analysis of all the published cardiovascular outcome trials for
ated with these operations is probably because of the GLP-1RAs.
3. Alfayez OM, Almutairi AR, Aldosari A, Al Yami MS. Update on cardiovascular
exaggerated secretion of gut hormones, perhaps in safety of incretin-based therapy in adults with type 2 diabetes mellitus: a
particular the hormones GLP-1 and PYY [81]. Long- meta-analysis of cardiovascular outcome trials. Can J Diabetes 2019;
43:538.e2–545.e2.
acting agonists of PYY are, therefore, being devel- 4. Thorens B. Expression cloning of the pancreatic beta cell receptor for the
oped for combinations with GLP-1 RAs for obesity gluco- incretin hormone glucagon-like peptide 1. Proc Natl Acad Sci U S A
1992; 89:8641–8645.
therapy. Another combination that deserves men- 5. Pujadas G, Drucker DJ. Vascular biology of glucagon receptor superfamily
tioning is the GLP-1-amylin combination. Amylin peptides: mechanistic and clinical relevance. Endocr Rev 2016;
37:554–583.
has been around as diabetes therapy for decades in 6. Holz GG. Epac: A new cAMP-binding protein in support of glucagon-like
the form of pramlintide, a modified and soluble peptide-1 receptor-mediated signal transduction in the pancreatic beta-cell.
Diabetes 2004; 53:5–13.
form of the human beta cell product. It was observed 7. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev 2007;
that treating T2DM patients with large doses of 87:1409–1439.
8. de Heer J, Holst JJ. Sulfonylurea compounds uncouple the glucose depen-
pramlintide resulted in weight loss, and therefore, dence of the insulinotropic effect of glucagon-like peptide 1. Diabetes 2007;
long-acting amylin receptor agonists are being 56:438–443.
9. Holst JJ, Deacon CF, Vilsboll T, et al. Glucagon-like peptide-1, glucose
developed, again for combination with GLP-1 recep- homeostasis and diabetes. Trends Mol Med 2008; 14:161–168.
tor agonists [82]. A final, unexpected development, 10. Rosenstock J, Allison D, Birkenfeld AL, et al., PIONEER 3 Investigators. Effect
of additional oral semaglutide vs sitagliptin on glycated hemoglobin in adults
was the generation of monomolecular GLP-1–GIP with type 2 diabetes uncontrolled with metformin alone or with sulfonylurea:
dual agonists, among which tirzepatide from Eli the PIONEER 3 randomized clinical trial. JAMA 2019; 321:1466–1480.
11. Deacon CF, Holst JJ. Dipeptidyl peptidase-4 inhibitors for the treatment of
Lilly is the most interesting. In phase two studies, type 2 diabetes: comparison, efficacy and safety. Expert Opin Pharmacother
this compound had antidiabetic and weight-losing 2013; 14:2047–2058.
12. Sisley S, Gutierrez-Aguilar R, Scott M, et al. Neuronal GLP1R mediates
effects that appeared (at least) comparable with liraglutide’s anorectic but not glucose-lowering effect. J Clin Invest 2014;
&&
those of semaglutide [83 ]. The contribution of 124:2456–2463.
13. Secher A, Jelsing J, Baquero AF, et al. The arcuate nucleus mediates GLP-1
GIP to the effects of tirzepatide is unclear at present, receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014;
given the general understanding that GIP has no 124:4473–4488.

1752-296X Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-endocrinology.com 7

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Gastrointestinal hormones

14. Adriaenssens AE, Svendsen B, Lam BY, et al. Transcriptomic profiling of 42. Taylor R, Al-Mrabeh A, Sattar N. Understanding the mechanisms of reversal of
pancreatic alpha, beta and delta cell populations identifies delta cells type 2 diabetes. Lancet Diabetes Endocrinol 2019; 7:726–736.
as a principal target for ghrelin in mouse islets. Diabetologia 2016; 43. Vilsboll T, Agerso H, Krarup T, Holst JJ. Similar elimination rates of glucagon-
59:2156–2165. like peptide-1 in obese type 2 diabetic patients and healthy subjects. J Clin
15. Orgaard A, Holst JJ. The role of somatostatin in GLP-1-induced inhibition of Endocrinol Metab 2003; 88:220–224.
glucagon secretion in mice. Diabetologia 2017; 60:1731–1739. 44. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by
16. Nauck MA, Kleine N, Orskov C, et al. Normalization of fasting hyperglycaemia human plasma in vitro yields an N-terminally truncated peptide that is a major
by exogenous glucagon- like peptide 1 (7–36 amide) in type 2 (noninsulin- endogenous metabolite in vivo. J Clin Endocrinol Metab 1995; 80:952–957.
dependent) diabetic patients. Diabetologia 1993; 36:741–744. 45. Deacon CF, Nauck MA, Toft-Nielsen M, et al. Both subcutaneously and
17. Hare KJ, Vilsboll T, Asmar M, et al. The glucagonostatic and insulinotropic intravenously administered glucagon-like peptide I are rapidly degraded from
effects of glucagon-like peptide-1 contribute equally to its glucose-lowering the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes
action. Diabetes 2010; 59:1765–1770. 1995; 44:1126–1131.
18. Orskov C, Poulsen SS, Moller M, Holst JJ. Glucagon-like peptide I receptors in 46. Deacon CF, Hughes TE, Holst JJ. Dipeptidyl peptidase IV inhibition potenti-
the subfornical organ and the area postrema are accessible to circulating ates the insulinotropic effect of glucagon-like peptide 1 in the anesthetized
glucagon-like peptide I. Diabetes 1996; 45:832–835. pig. Diabetes 1998; 47:764–769.
19. Jensen CB, Pyke C, Rasch MG, et al. Characterization of the glucagonlike 47. Ahren B, Gomis R, Standl E, et al. Twelve- and 52-week efficacy of the
peptide-1 receptor in male mouse brain using a novel antibody and in situ dipeptidyl peptidase IV inhibitor LAF237 in metformin-treated patients with
hybridization. Endocrinology 2018; 159:665–675. type 2 diabetes. Diabetes Care 2004; 27:2874–2880.
20. Larsen PJ, Tang-Christensen M, Holst JJ, Orskov C. Distribution of glucagon- 48. Mari A, Sallas WM, He YL, et al. Vildagliptin, a dipeptidyl peptidase-IV
like peptide-1 and other preproglucagon- derived peptides in the rat hypotha- inhibitor, improves model-assessed {beta}-cell function in patients with type
lamus and brainstem. Neuroscience 1997; 77:257–270. 2 diabetes. J Clin Endocrinol Metab 2005; 90:4888–4894.
21. Langhans W, Holst JJ. Afferent endocrine control of eating. In: Dixon S, 49. Hansen L, Hartmann B, Bisgaard T, et al. Somatostatin restrains the secretion
Mercer JG, editors. Neuroendocrinology of appetite. Wiley-Blackwell; 2016. of glucagon-like peptide-1 and 2 from isolated perfused porcine ileum. Am J
pp. 24–54. Physiol 2000; 278:E1010–E1018.
22. Vrang N, Phifer CB, Corkern MM, Berthoud HR. Gastric distension induces c- 50. Aaboe K, Knop FK, Vilsboll T, et al. Twelve weeks treatment with the DPP-4
Fos in medullary GLP-1/2-containing neurons. Am J Physiol Regul Integr inhibitor, sitagliptin, prevents degradation of peptide YY and improves glu-
Comp Physiol 2003; 285:R470–R478. cose and nonglucose induced insulin secretion in patients with type 2
23. Holst JJ. Glucagon-like peptide 1 (GLP-1): an intestinal hormone signalling diabetes mellitus. Diabetes Obes Metab 2010; 12:323–333.
nutritional abundance, with an unusual therapeutic potential. Trends Endo- 51. Deacon CF, Knudsen LB, Madsen K, et al. Dipeptidyl peptidase IV resistant
crinol Metab 1999; 10:229–234. analogues of glucagon-like peptide-1 which have extended metabolic stability
24. Vilsboll T, Krarup T, Sonne J, et al. Incretin secretion in relation to meal size and and improved biological activity. Diabetologia 1998; 41:271–278.
body weight in healthy subjects and people with type 1 and type 2 diabetes 52. Eng J, Kleinman WA, Singh L, et al. Isolation and characterization of exendin-4,
mellitus. J Clin Endocrinol Metab 2003; 88:2706–2713. an exendin-3 analogue, from Heloderma suspectum venom. Further evidence
25. Richards P, Parker HE, Adriaenssens AE, et al. Identification and character- for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol
ization of GLP-1 receptor-expressing cells using a new transgenic mouse Chem 1992; 267:7402–7405.
model. Diabetes 2014; 63:1224–1233. 53. Edwards CM, Stanley SA, Davis R, et al. Exendin-4 reduces fasting and
26. Jensen EP, Poulsen SS, Kissow H, et al. Activation of GLP-1 receptors on postprandial glucose and decreases energy intake in healthy volunteers. Am J
vascular smooth muscle cells reduces the autoregulatory response in afferent Physiol Endocrinol Metab 2001; 281:E155–E161.
arterioles and increases renal blood flow. Am J Physiol Renal Physiol 2015; 54. Kolterman OG, Kim DD, Shen L, et al. Pharmacokinetics, pharmacodynamics,
308:F867–F877. and safety of exenatide in patients with type 2 diabetes mellitus. Am J Health
27. Asmar A, Cramon PK, Simonsen L, et al. Extracellular fluid volume expansion Syst Pharm 2005; 62:173–181.
uncovers a natriuretic action of GLP-1: a functional GLP-1-renal axis in man. J 55. Pfeffer MA, Claggett B, Diaz R, et al. Lixisenatide in patients with type 2
Clin Endocrinol Metab 2019; 104:2509–2519. diabetes and acute coronary syndrome. N Engl J Med 2015;
28. Yusta B, Baggio LL, Koehler J, et al. GLP-1R agonists modulate enteric 373:2247–2257.
immune responses through the intestinal intraepithelial lymphocyte GLP-1R. 56. Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily
Diabetes 2015; 64:2537–2549. for the treatment of type 2 diabetes: a randomised, open-label, noninferiority
29. Drucker DJ. The cardiovascular biology of glucagon-like peptide-1. Cell Metab study. Lancet 2008; 372:1240–1250.
2016; 24:15–30. 57. Holman RR, Bethel MA, Mentz RJ, et al. Effects of once-weekly exenatide on
30. Nauck M, Stockmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017;
type 2 (noninsulin-dependent) diabetes. Diabetologia 1986; 29:46–52. 377:1228–1239.
31. Hojberg PV, Zander M, Vilsboll T, et al. Near normalisation of blood glucose 58. Rosenstock J, Sorli CH, Trautmann ME, et al. Once-weekly efpeglenatide
improves the potentiating effect of GLP-1 on glucose-induced insulin secre- dose-range effects on glycemic control and body weight in patients with type
tion in patients with type 2 diabetes. Diabetologia 2008; 51:632–640. 2 diabetes on metformin or drug naive, referenced to liraglutide. Diabetes
32. Vilsboll T, Knop FK, Krarup T, et al. The pathophysiology of diabetes involves a Care 2019; 42:1733–1741.
defective amplification of the late-phase insulin response to glucose by 59. Russell-Jones D. Molecular, pharmacological and clinical aspects of liraglu-
glucose-dependent insulinotropic polypeptide-regardless of etiology and tide, a once-daily human GLP-1 analogue. Mol Cell Endocrinol 2009; 297(1–
phenotype. J Clin Endocrinol Metab 2003; 88:4897–4903. 2):137–140.
33. Nauck MA, Heimesaat MM, Orskov C, et al. Preserved incretin activity of 60. Madsbad S, Kielgast U, Asmar M, et al. An overview of once-weekly glucagon-
glucagon-like peptide 1 [7–36 amide] but not of synthetic human gastric like peptide-1 receptor agonists–available efficacy and safety data and
inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest perspectives for the future. Diabetes Obes Metab 2011; 13:394–407.
1993; 91:301–307. 61. Baggio LL, Huang Q, Brown TJ, Drucker DJ. A Recombinant human glucagon-
34. Jones AG, McDonald TJ, Shields BM, et al. Markers of beta-cell failure predict like peptide (GLP)-1-albumin protein (albugon) mimics peptidergic activation
poor glycemic response to GLP-1 receptor agonist therapy in type 2 diabetes. of GLP-1 receptor-dependent pathways coupled with satiety, gastrointestinal
Diabetes Care 2016; 39:250–257. motility, and glucose homeostasis. Diabetes 2004; 53:2492–2500.
35. Hare KJ, Knop FK, Asmar M, et al. Preserved inhibitory potency of GLP-1 on 62. Madsbad S, Kielgast U, Asmar M, et al. An overview of once-weekly GLP-1
glucagon secretion in type 2 diabetes mellitus. J Clin Endocrinol Metab 2009; receptor agonists - available efficacy and safety data and perspectives for the
94:4679–4687. future. Diabetes Obes Metab 2011; 13:394–407.
36. Wewer Albrechtsen NJ, Pedersen J, Galsgaard KD, et al. The liver-alpha-cell 63. Hernandez AF, Green JB, Janmohamed S, et al., Harmony Outcomes Com-
axis and type 2 diabetes. Endocr Rev 2019; 40:1353–1366. mittees and Investigators. Albiglutide and cardiovascular outcomes in pa-
37. Kazda CM, Ding Y, Kelly RP, et al. Evaluation of efficacy and safety of the tients with type 2 diabetes and cardiovascular disease (Harmony Outcomes):
glucagon receptor antagonist LY2409021 in patients with type 2 diabetes: a double-blind, randomised placebo-controlled trial. Lancet 2018;
12- and 24-week phase 2 studies. Diabetes Care 2016; 39:1241–1249. 392:1519–1529.
38. Buteau J, El-Assaad W, Rhodes CJ, et al. Glucagon-like peptide-1 prevents 64. Thompson AM, Trujillo JM. Dulaglutide: the newest GLP-1 receptor agonist
beta cell glucolipotoxicity. Diabetologia 2004; 47:806–815. for the management of type 2 diabetes. Ann Pharmacother 2015;
39. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and 49:351–359.
cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375: 65. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovas-
311–322. & cular outcomes in type 2 diabetes (REWIND): a double-blind, randomised
40. Xu G, Stoffers DA, Habener JF, Bonner-Weir S. Exendin-4 stimulates both placebo-controlled trial. Lancet 2019; 394:121–130.
beta-cell replication and neogenesis, resulting in increased beta-cell Cardiovascular outcome trial of the first once-weekly GLP-1 agonist in the market.
mass and improved glucose tolerance in diabetic rats. Diabetes 1999; 66. Nauck MA, Petrie JR, Sesti G, et al., Study 1821 Investigators. A phase 2,
48:2270–2276. randomized, dose-finding study of the novel once-weekly human GLP-1
41. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety analog, semaglutide, compared with placebo and open-label liraglutide in
and suppresses energy intake in humans. J Clin Invest 1998; 101:515–520. patients with type 2 diabetes. Diabetes Care 2016; 39:231–241.

8 www.co-endocrinology.com Volume 26  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; MED/270108; Total nos of Pages: 9;
MED 270108

Incretin therapy for diabetes mellitus type 2 Holst

67. Jendle J, Torffvit O, Ridderstrale M, et al. Willingness to pay for diabetes drug 76. Lan NSR, Fegan PG, Yeap BB, Dwivedi G. The effects of sodium-glucose
therapy in type 2 diabetes patients: based on LEAD clinical programme cotransporter 2 inhibitors on left ventricular function: current evidence and
results. J Med Econ 2012; 15(Suppl 2):1–5. future directions. ESC Heart Fail 2019; 6:927–935.
68. Pratley RE, Aroda VR, Lingvay I, et al., SUSTAIN 7 investigators. Semaglutide 77. Frias JP, Guja C, Hardy E, et al. Exenatide once weekly plus dapagliflozin once
versus dulaglutide once weekly in patients with type 2 diabetes (SUSTAIN 7): daily versus exenatide or dapagliflozin alone in patients with type 2 diabetes
a randomised, open-label, phase 3b trial. Lancet Diabetes Endocrinol 2018; inadequately controlled with metformin monotherapy (DURATION-8): a 28
6:275–286. week, multicentre, double-blind, phase 3, randomised controlled trial. Lancet
69. Marso SP, Bain SC, Consoli A, et al., SUSTAIN-6 Investigators. Semaglutide Diabetes Endocrinol 2016; 4:1004–1016.
and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 78. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg
2016; 375:1834–1844. of liraglutide in weight management. N Engl J Med 2015; 373:11–22.
70. Caruso I, Cignarelli A, Giorgino F. Heterogeneity and similarities in GLP-1 79. Le Roux CW, Astrup A, Fujioka K, et al., SCALE Obesity Prediabetes
receptor agonist cardiovascular outcomes trials. Trends Endocrinol Metab NN8022-1839 Study Group. 3 years of liraglutide versus placebo for
2019; 30:578–589. type 2 diabetes risk reduction and weight management in individuals with
71. North EJ, Newman JD. Review of cardiovascular outcomes trials of sodium- prediabetes: a randomised, double-blind trial. Lancet 2017; 389:
glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor 1399–1409.
agonists. Curr Opin Cardiol 2019; 34:687–692. 80. O’Neil PM, Birkenfeld AL, McGowan B, et al. Efficacy and safety of semaglu-
72. Nagahisa T, Saisho Y. Cardiorenal protection: potential of SGLT2 inhibitors & tide compared with liraglutide and placebo for weight loss in patients with
and GLP-1 receptor agonists in the treatment of type 2 diabetes. Diabetes obesity: a randomised, double-blind, placebo and active controlled, dose-
Ther 2019; 10:1733–1752. ranging, phase 2 trial. Lancet 2018; 392:637–649.
73. Aroda VR, Bain SC, Cariou B, et al. Efficacy and safety of once-weekly The first demonstration of two-digit percent weight losses with a GLP-1 agonist.
semaglutide versus once-daily insulin glargine as add-on to metformin (with or 81. Svane MS, Jorgensen NB, Bojsen-Moller KN, et al. Peptide YY and glucagon-
without sulfonylureas) in insulin-naive patients with type 2 diabetes (SUSTAIN like peptide-1 contribute to decreased food intake after Roux-en-Y gastric
4): a randomised, open-label, parallel-group, multicentre, multinational, phase bypass surgery. Int J Obes (Lond) 2016; 40:1699–1706.
3a trial. Lancet Diabetes Endocrinol 2017; 5:355–366. 82. Boyle CN, Lutz TA, Le FC. Amylin - its role in the homeostatic and hedonic
74. Castellana M, Cignarelli A, Brescia F, et al. GLP-1 receptor agonist added to control of eating and recent developments of amylin analogs to treat obesity.
insulin versus basal-plus or basal-bolus insulin therapy in type 2 diabetes: A Mol Metab 2018; 8:203–210.
systematic review and meta-analysis. Diabetes Metab Res Rev 2019; 83. Frias JP, Nauck MA, Van J, et al. Efficacy and safety of LY3298176, a novel
35:e3082. && dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a
75. Lingvay I, Catarig AM, Frias JP, et al. Efficacy and safety of once-weekly randomised, placebo-controlled and active comparator-controlled phase 2
semaglutide versus daily canagliflozin as add-on to metformin in patients with trial. Lancet 2018.
type 2 diabetes (SUSTAIN 8): a double-blind, phase 3b, randomised con- A phase two trial demonstrating seemingly unsurpassed glycemic and and weight-
trolled trial. Lancet Diabetes Endocrinol 2019; 7:834–844. loosing efficacy of a long-acting GLP-1-GIP co-agonist.

1752-296X Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-endocrinology.com 9

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

You might also like