throbber
REVIEW ARTICLES
`
`Glucagon-like peptide-1 and diabetes treatment
`
`Tina Vilsbøll, Kristine J. Hare, Jonatan O. Bagger and Filip K. Knop
`Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen,
`Hellerup, Denmark (t.vilsboll@dadlnet.dk)
`
`Abstract
`
`Type 2 diabetes is characterized by insulin resis-
`tance, impaired glucose-induced insulin secre-
`tion and inappropriately regulated glucagon
`secretion, which in combination eventually result
`in hyperglycemia and in the longer term in
`microvascular and macrovascular complications
`affecting multiple organ systems. Traditional
`treatment modalities — even multidrug
`approaches — for type 2 diabetes are often
`unsatisfactory at achieving glycemic goals as the
`disease progresses due to a steady, relentless
`decline in pancreatic β-cell function. Further-
`more, current treatment modalities are often
`limited by inconvenient dosing regimens, safety
`and tolerability issues, the latter including hypo-
`glycemia, body weight gain, edema and gastro-
`intestinal side effects.
`The incretin hormones glucagon-like peptide
`(GLP)-1 and glucose-dependent insulinotropic
`polypeptide (GIP) are intestinal hormones that
`augment insulin secretion in response to the
`ingestion of nutrients. The actions of GLP-1 and
`GIP, which also include trophic effects on the β-
`cells, have attracted a lot of interest. GLP-1 also
`inhibits glucagon secretion and suppresses food
`intake and appetite. Recently, an entirely new
`therapeutic modality for the treatment of type 2
`diabetes based on the effect of GLP-1 was intro-
`duced onto the market.
`Incretin-based therapies fall into two groups:
`(1) GLP-1 receptor agonists, i.e. injectable pep-
`tide preparations with actions similar to the nat-
`ural incretin hormone GLP-1; and (2) the
`incretin enhancers, which are orally available
`agents that inhibit the degradation of the
`incretin hormones and thereby increase their
`plasma concentrations and biological actions.
`This review outlines the scientific basis for the
`development of GLP-1 receptor agonists and
`incretin enhancers, assesses the clinical experi-
`ence gathered so far and discusses future expec-
`tations for incretin-based therapy.
`
`Key words:
`Incretin hormones, dipeptidyl peptidase-4 (DPP-4),
`glucagon-like peptide-1 (GLP-1), glucose-dependent
`insulinotropic polypeptide (GIP), incretin mimetics,
`GLP-1 receptor agonists, type 2 diabetes
`
`Incretin hormones: secretion, effect and
`degradation
`
`The ‘incretin effect’ refers to the amplification of
`glucose-stimulated insulin secretion elicited by
`hormones secreted from the gastrointestinal
`tract. In the strictest sense, it is quantified by
`comparing insulin responses to oral and intra-
`venous glucose administration where the intra-
`venous infusion is adjusted so as to result in the
`same (isoglycemic) plasma glucose concentra-
`tions as the oral stimulus [1, 2]. In healthy sub-
`jects, oral administration causes a two- to
`threefold larger insulin response compared with
`the isoglycemic intravenous stimulus.
`
`The incretin hormones GLP-1 and GIP are
`intestinal hormones that augment
`insulin secretion in response to the
`ingestion of nutrients
`
`This discrepancy in insulin secretion between
`the two stimuli is due to the actions of incretin
`hormones, glucagon-like peptide (GLP)-1 and
`glucose-dependent insulinotropic polypeptide
`(GIP). GLP-1 is a 30-amino acid polypeptide
`produced in the endocrine L-cells of the intesti-
`nal epithelium as a product of glucagon gene
`expression. The GLP-1 moiety is liberated from
`proglucagon by the action of prohormone
`convertase 1/3 (PC1/3). This is in contrast to
`proglucagon processing in the pancreatic
`α-cells, where prohormone convertase 2 cleaves
`out glucagon but leaves the GLP-1 molecule
`embedded in a large inactive fragment [3].
`The L-cells are found throughout the intestinal
`
`Volume 21, Number 1, 2009
`
`International Diabetes Monitor
`
`1
`
`MPI EXHIBIT 1040 PAGE 1
`
`MPI EXHIBIT 1040 PAGE 1
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 1 of 7
`
`

`

`Review articles
`
`tract, but their density is highest in the ileum
`and parts of the colon.
`GIP is produced in the endocrine K-cells,
`which are more frequent in the proximal small
`intestine [3]. As in the L-cells, PC1/3 is respon-
`sible for the processing of proGIP in endocrine
`K-cells. After the secretion of GIP and GLP-1,
`both hormones are degraded by the enzyme
`dipeptidyl peptidase-4 (DPP-4). This enzyme
`cleaves off the two N-terminal amino acids of
`the incretin hormones, which abolishes their
`insulinotropic activity.
`GLP-1 has an apparent half-life of 1–2 min
`[4], whereas GIP is degraded more slowly, with a
`half-life of 7 min [5]. The truncated metabolites
`are eliminated through the kidneys. The intact
`and active forms of GLP-1 and GIP elicit signifi-
`cant insulin responses at plasma glucose levels
`above 4 mM. At higher levels (e.g. in the post-
`prandial state) this insulinotropic power
`increases dramatically [6]. These insulinotropic
`actions are exerted by activation of specific
`GLP-1 and GIP receptors, respectively, on the
`pancreatic β-cells and consist of potentiation of
`glucose-induced insulin secretion [7]. At lower
`plasma glucose concentrations (4 mM and
`below) both hormones lose their insulinotropic
`activity completely. Therefore the incretin hor-
`mones seem to play a very important role in the
`regulation of, particularly, postprandial glucose
`clearance from the bloodstream.
`
`GLP-1 has an apparent half-life of 1–2
`min, whereas GIP is degraded more
`slowly, with a half-life of 7 min
`
`The incretin hormones have several additional
`actions besides being ‘incretins’. In addition to
`its insulinotropic action, GLP-1 enhances
`insulin biosynthesis and insulin gene expression,
`it has trophic and protective effects on the β-
`cells (stimulating β-cell growth, proliferation and
`neogenesis, and reducing β-cell apoptosis) [8].
`Furthermore, GLP-1 strongly inhibits glucagon
`secretion, which, combined with its effect on
`insulin secretion, results in an inhibition of
`hepatic glucose production that contributes to
`the glucose-lowering effect of the hormone [9].
`GLP-1 also functions as an inhibitor of upper
`gastrointestinal motility and secretion (reducing
`postprandial glucose excursions) [10], suppress-
`ing appetite and food intake. Chronic adminis-
`tration of GLP-1 has been shown to lead to
`weight loss [11]. Finally, GLP-1 appears to have
`beneficial actions on the cardiovascular system:
`
`enhancing myocardial performance in experi-
`mental and clinical cardiac insufficiency, reduc-
`ing infarct size in experimental myocardial
`infarction and improving endothelial dysfunction
`in patients with type 2 diabetes [3]. GIP appears
`to have similar actions to GLP-1 on the β-cells,
`but its effects with regard to glucagon secretion
`remain relatively unclear.
`
`Incretin hormones and type 2 diabetes
`mellitus
`
`When patients with type 2 diabetes are subjected
`to isoglycemic oral and intravenous glucose chal-
`lenges, the amplification of insulin secretion dur-
`ing the oral stimulus is markedly reduced unlike
`in healthy subjects [12]. Considering the power
`of the incretin effect to maintain postprandial
`glucose levels in healthy subjects, there can be
`little doubt that the attenuated incretin effect in
`type 2 diabetes contributes to the glucose intol-
`erance of these patients.
`When trying to explain why the incretin effect
`is lost, one may ask whether there is something
`wrong with the secretion or actions of GIP and
`GLP-1. Mixed-meal stimulation tests have
`revealed that postprandial GIP secretion is near
`normal or slightly impaired, whereas particularly
`the late phase of the GLP-1 response is signifi-
`cantly reduced in patients with type 2 diabetes
`[13]. Furthermore, in early studies of the actions
`of the two peptides, it was clearly demonstrated
`that the insulinotropic effect of GLP-1 was
`retained, whereas that of GIP was almost com-
`pletely lost [14].
`Subsequent studies of the effects of GLP-1 on
`glucose-stimulated insulin secretion (β-cell
`responsiveness to glucose) revealed that it was
`possible to completely normalize the glucose
`responsiveness with GLP-1, but also that the
`potency of GLP-1 in this respect was reduced in
`type 2 diabetic patients [15]. Further clamp
`studies revealed that particularly the late-phase
`insulin response to GIP was completely lost in
`type 2 diabetes [16]. Thus the loss of incretin
`effect in type 2 diabetes seems to be due to an
`impaired secretion of GLP-1 in particular and,
`perhaps more importantly, a loss of the
`insulinotropic effect of GIP and reduced
`insulinotropic potency of GLP-1. However, since
`the insulinotropic effect of high doses of GLP-1 is
`preserved, it should be possible to restore incretin
`action in patients with type 2 diabetes with supra-
`physiological doses of GLP-1.
`
`2
`
`International Diabetes Monitor
`
`Volume 21, Number 1, 2009
`
`MPI EXHIBIT 1040 PAGE 2
`
`MPI EXHIBIT 1040 PAGE 2
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 2 of 7
`
`

`

`GLP-1 receptor agonists
`
`It is fairly easy to stabilize the GLP-1 molecule
`against DPP-4: a substitution of alanine in posi-
`tion 2 with, for example, valine is sufficient and
`does not change the biological activity of the
`peptide [17]. However, the stabilized molecule
`is still eliminated extremely rapidly in the
`kidneys (with a half-life of 4–5 min), which
`leaves such analogues unsuitable for prolonged
`drug exposure.
`However, exendin-4, a peptide with about
`50% sequence homology to GLP-1, which was
`isolated from the saliva of the Gila monster
`(Heloderma suspectum) during a search for bio-
`logically active peptides, turned out to be a full
`agonist for the GLP-1 receptor, to be stable
`against DPP-4, and to be eliminated through the
`kidneys exclusively by glomerular filtration [18].
`After subcutaneous injection of exenatide (a syn-
`thetic replica of exendin-4) in the dose selected
`for clinical use (10 µg), the plasma concentration
`is elevated into the insulinotropic range for
`about 5–6 h. Exenatide is therefore administered
`twice daily [19]. Exenatide has been developed
`by Amylin and Eli Lilly for the treatment of type
`2 diabetes under the trade name Byetta®.
`Another GLP-1 receptor agonist under clinical
`development is liraglutide (Novo Nordisk),
`which is based on the structure of human GLP-
`1 (97% homology with the native peptide) but
`modified to include an amino acid substitution
`and an attachment of a C16 acyl chain [20],
`enabling the molecule to bind to albumin, thus
`preventing renal elimination and degradation by
`DPP-4. Following subcutaneous administration,
`liraglutide is slowly absorbed into the blood-
`stream and has a plasma half-life of approxi-
`mately 11–13 h [21], making it suitable for
`once-daily injection. Clinically, the molecule has
`similar actions to continuously infused GLP-1
`and appears to have a similar clinical potential to
`that of exendin-4 [22].
`Clinical studies using exenatide have demon-
`strated sustained beneficial effects on HbA1c,
`body weight and β-cell function in patients with
`type 2 diabetes. Controlled studies comparing
`exenatide and placebo injections as add-on ther-
`apy to already instituted antidiabetic treatment
`have revealed a statistically significant decline in
`HbA1c of approximately 1% from baseline (base-
`line HbA1c 8.2%) [23] and significant weight
`loss in favour of exenatide. The weight loss was
`progressive, dose-dependent and with no appar-
`ent plateau by week 30 (−2.3 kg); however, it
`appeared to plateau after 2–3 years of treatment
`(with a weight loss of 5.3 kg) in completers par-
`
`Review articles
`
`ticipating in an open-label extension of the trials
`[24, 25]. Side effects were primarily dose-depen-
`dent nausea and vomiting, occurring in as many
`as 57% and 17% of participants, respectively,
`although nausea was generally mild to moderate
`and declined with time.
`The clinical efficacy of exenatide in patients
`with type 2 diabetes over a period of 6 months
`has been evaluated by extractions of data from a
`primary care electronic medical record database
`[26]. In this study, weight loss among the 1785
`patients was >3 kg (baseline weight 121 kg) and
`as many as 70% of the patients lost weight. Low-
`ering of HbA1c ranged from 0.7% to 0.9%
`regardless of weight loss. It was concluded that
`the effectiveness of exenatide in a primary care
`setting is similar to that observed in controlled
`clinical trials.
`Exenatide given as twice-daily injections may
`not provide complete 24-h coverage, especially
`after midday meals, and overnight plasma con-
`centrations seem inadequate to obtain optimal
`glycemic control. Therefore a long-acting release
`(LAR) formulation of exenatide for subcuta-
`neous injection in patients with type 2 diabetes
`has recently been developed. Data from a phase-
`II trial (n = 295) in which exenatide LAR 2.0
`mg once weekly was compared with exenatide
`10 µg twice daily for 30 weeks were recently
`published [27]. These data showed significant
`reductions in HbA1c (1.9% and 1.5%, respec-
`tively) and body weight (average decrease of 4 kg
`in both groups). The trial was extended for 22
`weeks with all subjects being switched to
`exenatide LAR in the extension period. A sus-
`tained effect of exenatide LAR was seen after 52
`weeks with improvements in both HbA1c (−2%)
`and fasting plasma glucose (−2.6 mmol/l). Three
`out of four patients achieved an HbA1c of 7.0%
`or below. The change in body weight was −4 kg
`by week 52 compared with baseline. Exenatide
`LAR was well tolerated. The most common side
`effect, nausea, was predominantly mild and
`transient during the study; during the 22-week
`follow-up the incidence of nausea was 7% [28].
`Exenatide LAR is currently in phase-III clinical
`development and is expected to reach the market
`in 2010 (Table I).
`The next GLP-1 receptor agonist for the treat-
`ment of type 2 diabetes to reach the market is
`expected to be liraglutide. Published data have
`demonstrated that liraglutide as monotherapy
`(1.9 mg once daily) is capable of decreasing
`fasting plasma glucose levels by 3.4 mmol/l on
`average in patients with type 2 diabetes when
`compared with placebo [22]. In the same study,
`a decrease in HbA1c of up to 1.7% (baseline
`
`Volume 21, Number 1, 2009
`
`International Diabetes Monitor
`
`3
`
`MPI EXHIBIT 1040 PAGE 3
`
`MPI EXHIBIT 1040 PAGE 3
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 3 of 7
`
`

`

`Review articles
`
`Table I: The GLP-1 receptor agonists currently on the market or in clinical development.
`
`Company
`
`Compound
`
`Status of development
`
`Formulation
`
`Eli Lilly/Amylin
`
`Byetta® (exenatide)
`
`Launched
`
`Novo Nordisk
`
`Liraglutide
`
`Filed USA/EU May 2008
`
`Eli Lilly/Amylin
`
`Exenatide LAR
`
`Phase III Expected 2010
`
`Sanofi-Aventis
`
`AVE0010(ZP10)
`
`Phase III Expected 2011
`
`Roche/Ipsen
`
`Taspoglutide (R1583)
`
`Phase IIb–III Expected 2011
`
`GlaxoSmithKline
`
`Syncria® (albiglutide)
`
`Phase IIb
`
`ConjuChem
`
`Eli Lilly
`
`Sanofi-Aventis
`
`Novo Nordisk
`
`CJC-1134
`
`LY2405319
`
`AVE0010(ZP10)
`
`NN9535
`
`Phase II
`
`Phase II
`
`Phase II
`
`Phase II
`
`Twice daily
`
`Once daily
`
`Once weekly
`
`Once daily
`
`Once weekly
`
`Once weekly
`
`Once weekly
`
`Once weekly
`
`Once weekly
`
`Once weekly
`
`HbA1c 8.0%) was observed, and almost 50% of
`the patients managed to reach the HbA1c goal of
`<7%. In the highest liraglutide dose group
`(1.9 mg/day) the change from baseline in body
`weight was −3 kg (−1.2 kg compared with
`placebo). As for exenatide, transient and mild
`nausea was reported in the liraglutide-treated
`subjects (liraglutide 10%, placebo 3%). Data
`from a number of phase-III trials with liraglutide
`were presented at the 2008 annual scientific
`meetings of the American Diabetes Association
`(ADA) and the European Association for the
`Study of Diabetes (EASD). These trials, called
`the LEAD (Liraglutide Effect and Action in Dia-
`betes) trials, demonstrated that liraglutide given
`as a once-daily injection, as monotherapy and in
`combination with a range of antidiabetic drugs,
`is associated with significant improvements in
`HbA1c (sustained reductions of up to 1.6%),
`fasting plasma glucose, postprandial glucose and
`β-cell function [29, 30]. Furthermore, in trials of
`up to 1 year liraglutide showed maintained
`weight reduction (up to 4 kg in subjects with a
`high BMI) [29], minimal risk of hypoglycemia,
`reductions of up to 3.6 mmHg in systolic blood
`pressure [31], low and transient incidence of
`nausea and negligible antibody formation.
`
`Many GLP-1 receptor agonists that
`are in their late clinical development
`are anticipated to have optimized
`pharmacokinetic profiles, fewer
`gastrointestinal side effects and
`are expected to reach the market
`from 2010 onwards
`
`Very recently, liraglutide was compared with
`exenatide twice daily (for 26 weeks) and signifi-
`
`cant differences in glycemic control were
`observed (HbA1c −1.1% and −0.8%, respec-
`tively), a tendency towards a more pronounced
`weight loss with liraglutide (−3 kg vs. −2 kg)
`and less nausea in favour of liraglutide (Blonde
`L et al. Can J Diabetes 2008; 32 suppl: A107).
`Applications for liraglutide as a new drug were
`filed with the authorities in both the United
`States and the European Union in May 2008.
`Regarding adverse events of GLP-1 receptor
`agonists, attention has recently been drawn
`towards a few cases of acute pancreatitis in
`patients treated with exenatide. These cases have
`been reviewed by the US Food and Drug
`Administration during post-marketing. A very
`few cases of acute pancreatitis have also been
`diagnosed during treatment with liraglutide in
`the LEAD programme. However, it is not clear
`whether the incidence of acute pancreatitis in
`patients with type 2 diabetes treated with GLP-1
`receptor agonists is higher than in a type 2 dia-
`betic population not treated with GLP-1 recep-
`tor agonists, and, so far, the reports are few and
`seem to be correlated to other causes of pancre-
`atitis (hypertriglyceridemia, alcohol abuse, gall
`bladder stones/operation).
`Many GLP-1 receptor agonists that are in
`their late clinical development are anticipated to
`have optimized pharmacokinetic profiles and
`presumably fewer gastrointestinal side effects.
`Many of these are expected to reach the market
`from 2010 onwards (Table I).
`
`DPP-4 inhibitors
`
`The extremely rapid and extensive degradation
`of GLP-1 by DPP-4 has given rise to the
`proposal that inhibitors of the enzyme could
`be used as a therapy for patients with type 2
`
`4
`
`International Diabetes Monitor
`
`Volume 21, Number 1, 2009
`
`MPI EXHIBIT 1040 PAGE 4
`
`MPI EXHIBIT 1040 PAGE 4
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 4 of 7
`
`

`

`Table II: The leading DPP-4 inhibitors — all orally available — in clinical development or on the market.
`
`Review articles
`
`Compound
`
`Status of development
`
`Januvia® (sitagliptin, MK-0431)
`
`Launched 2006
`
`Galvus® (vildagliptin, LAF-237)
`
`Launched 2008
`
`Filed USA 2008
`
`Filed USA/EU 2008
`
`Phase III
`
`Phase IIb
`
`Phase IIb
`
`Phase II
`
`Phase II
`
`Phase II
`
`Phase II
`
`Phase II
`
`Phase II
`
`Company
`
`Merck
`
`Novartis
`
`Takeda
`
`Alogliptin (SYR-322)
`
`Bristol-Myers Squibb
`
`Onglyza™ (saxagliptin)
`
`Boehringer-Ingelheim
`
`BI-1356
`
`Glenmark
`
`Phenomix
`
`Melogliptin (GRC 8200)
`
`Dutogliptin (PHX1149)
`
`Mitsubishi Tanabe Pharma
`
`Mitsubishi Tanabe Pharma
`
`TA-6666
`
`MP-513
`
`Amgen/Servier
`
`AMG 222/ALS 2-0426
`
`Pfizer
`
`Takeda
`
`Abbott Laboratories
`
`PF 00734200
`
`SYR-472
`
`ABT-279
`
`diabetes by protecting and thereby enhancing
`the circulating levels of endogenous GLP-1 [32].
`Early experiments documented that administra-
`tion of an inhibitor of DPP-4 to pigs completely
`protected both endogenous and exogenous
`GLP-1 and, furthermore, greatly enhanced
`insulin responses to glucose [33]. In a subse-
`quent study, DPP-4 inhibition was demonstrated
`also to protect GIP from degradation, again
`resulting in enhanced insulinotropic activity of
`infused GIP [34]. The idea was quickly accepted
`by the pharmaceutical industry and numerous
`companies embarked on the development of
`DPP-4 inhibitors for clinical use in the treat-
`ment of type 2 diabetes (Table II).
`
`The rapid and extensive degradation of
`GLP-1 by DPP-4 suggests that inhibitors
`of the enzyme could be used as a
`therapy for type 2 diabetes by
`enhancing the circulating levels of
`endogenous GLP-1, resulting in
`enhanced insulinotropic activity of
`infused GIP
`
`The first DPP-4 inhibitor to reach the market
`in 2007 was sitagliptin (Merck) [35]. Vildagliptin
`(Novartis) [36] was launched in the European
`Union in spring 2008. Both inhibitors have
`good oral bioavailability and a relatively
`long duration of action, such that once-daily
`(sitagliptin) or twice-daily (vildagliptin) dosing
`gives 70–90% inhibition of plasma DPP-4 activ-
`ity over a 24-h period [37], which is sufficient to
`fully protect the endogenous incretin hormones
`from degradation. Sitagliptin and vildagliptin
`
`have significant antidiabetic effects when given
`in monotherapy and result in further improve-
`ments in glycemic control when given in combi-
`nation with other antidiabetic agents including
`metformin, sulfonylurea and thiazolidinediones
`[37]. More than 25 studies have been published,
`comparing treatment with sitagliptin and
`vildagliptin with placebo, and in a few studies
`with other oral agents [23]. Most studies have
`been of a relatively short duration (<30 weeks)
`and the observed reductions in HbA1c approxi-
`mated 0.6–0.8% compared with placebo during
`the first 6 months of treatment [23].
`In a recent study sitagliptin was given for 26
`weeks in combination with various doses of
`metformin after a washout of previous medica-
`tion. The combination of the highest doses (100
`mg sitagliptin + 2000 mg metformin) resulted
`in large reductions in HbA1c with 66% of the
`patients reaching values below 7%, considered
`a therapeutic target by the ADA [38]. Data
`from the same subjects completing a 2-year
`extension trial demonstrated a sustained effect
`on glycemic control and were recently presented
`at the annual meeting of the EASD [39]. The
`combination with metformin is of particular
`interest, because recent studies have indicated
`that metformin may increase GLP-1 biosynthe-
`sis and secretion, so that a larger increase in the
`concentration of active GLP-1 may be obtained
`with the combination compared with either
`agent alone [40]. If the combination treatment
`can be demonstrated to prevent deterioration
`of β-cell function with time better than the
`currently recommended initial treatment
`(metformin) it may be recommended as an
`initial treatment for newly diagnosed patients
`with type 2 diabetes.
`
`Volume 21, Number 1, 2009
`
`International Diabetes Monitor
`
`5
`
`MPI EXHIBIT 1040 PAGE 5
`
`MPI EXHIBIT 1040 PAGE 5
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 5 of 7
`
`

`

`Review articles
`
`DPP-4 inhibition may also be combined with
`insulin treatment. Vildagliptin (50 mg b.i.d.)
`added to insulin treatment (80 U/day) reduced
`HbA1c levels by 0.5% (baseline 8.5%) vs. a
`reduction of 0.2% with placebo, and, despite
`the improvement in glycemic control, signifi-
`cantly fewer hypoglycemic events occurred
`in the patients who received the combination
`therapy [41].
`The safety profiles of the DPP-4 inhibitors
`were recently reviewed by scientists from the
`Cochrane Institute, who concluded that both
`vildagliptin and sitagliptin were well tolerated
`[42]. As opposed to the GLP-1 agonists, the
`inhibitors do not cause weight reduction but
`appear to be weight neutral. This is in itself of
`interest, since the significant reduction in blood
`glucose they provide per se would be expected to
`result in weight gain. The simplest explanation
`for the lack of effect on body weight is the fact
`that the effect of the endogenous concentrations
`of intact GLP-1 obtained with the inhibitors is
`limited compared with the effect that can be
`obtained with the GLP-1 agonists. Several
`DPP-4 inhibitors are currently undergoing clini-
`cal development and many of these are expected
`to reach the market within the next few years
`(Table II).
`
`Conclusion
`
`As the treatment efficacy of GLP-1 receptor
`agonists and incretin enhancers has been firmly
`established in respect to lowering HbA1c and
`improving β-cell function during treatment,
`these treatment modalities are expected to be
`incorporated as standard treatments and be
`included in the recommendations for the treat-
`ment of type 2 diabetes in the coming years.
`Incretin-based approaches are pleiotropic and,
`unlike existing therapies, target both α- and
`β-cell dysfunction. Studies indicate that incretin-
`based therapies — perhaps especially because of
`their trophic effects on the pancreatic β-cells —
`may halt the progression of disease that
`inevitably seems to accompany conventional
`treatment. So far this has not been established in
`clinical trials, but animal studies show that
`administration of GLP-1 receptor agonists or
`DPP-4 inhibitors is associated with β-cell prolif-
`eration and β-cell protection. If the β-cell-
`preserving potential of these drug classes can be
`demonstrated in humans, incretin-based thera-
`pies may be able to address one of the under-
`lying causes of progression of type 2 diabetes —
`the gradual loss of β-cell function and mass.
`
`Within the next year many new GLP-1 receptor
`agonists and DPP-4 inhibitors will be intro-
`duced to the market.
`
`Animal studies show that administration
`of GLP-1 receptor agonists or DPP-4
`inhibitors is associated with (cid:2)-cell
`proliferation and (cid:2)-cell protection
`
`References
`
`1. McIntyre N, Holdsworth CD, Turner DS. New inter-
`pretation of oral glucose tolerance. Lancet 1964; II:
`20–1.
`2. Perley M, Kipnis DM. Plasma insulin responses to oral
`and intravenous glucose: studies in normal and diabetic
`subjects. J Clin Invest 1967; 46: 1954–62.
`3. Holst JJ. The physiology of glucagon-like peptide 1.
`Physiol Rev 2007; 87: 1409–39.
`4. Vilsbøll T, Agersø H, Krarup T, Holst JJ. Similar elimi-
`nation rates of glucagon-like peptide-1 in obese type 2
`diabetic patients and healthy subjects. J Clin Endocrinol
`Metab 2003; 88: 220–4.
`5. Vilsbøll T, Agersø H, Lauritsen T et al. The elimination
`rates of intact GIP as well as its primary metabolite,
`GIP 3–42, are similar in type 2 diabetic patients and
`healthy subjects. Regul Pept 2006; 137: 168–72.
`6. Vilsbøll T, Krarup T, Madsbad S, Holst JJ. Both GLP-1
`and GIP are insulinotropic at basal and postprandial
`glucose levels and contribute nearly equally to the
`incretin effect of a meal in healthy subjects. Regul Pept
`2003; 114: 115–21.
`7. Fehmann HC, Goke R, Goke B. Cell and molecular
`biology of the incretin hormones glucagon-like peptide-
`I and glucose-dependent insulin releasing polypeptide.
`Endocr Rev 1995; 16: 390–410.
`8. Drucker DJ. The biology of incretin hormones. Cell
`Metab 2006; 3: 153–65.
`9. Hvidberg A, Nielsen MT, Hilsted J et al. Effect of
`glucagon-like peptide-1 (proglucagon 78–107 amide)
`on hepatic glucose production in healthy man.
`Metabolism 1994; 43: 104–8.
`10. Holst JJ. Glucagon-like peptide 1 (GLP-1): an intestinal
`hormone signalling nutritional abundance, with an
`unusual therapeutic potential. Trends Endocrinol Metab
`1999; 10: 229–34.
`11. Zander M, Madsbad S, Madsen JL, Holst JJ. Effect of
`6-week course of glucagon-like peptide 1 on glycaemic
`control, insulin sensitivity, and beta-cell function in type
`2 diabetes: a parallel-group study. Lancet 2002; 359:
`824–30.
`12. Knop FK, Vilsbøll T, Hojberg PV et al. Reduced
`incretin effect in type 2 diabetes: cause or consequence
`of the diabetic state? Diabetes 2007; 56: 1951–9.
`13. Toft-Nielsen MB, Damholt MB, Madsbad S et al.
`Determinants of the impaired secretion of glucagon-like
`peptide-1 in type 2 diabetic patients. J Clin Endocrinol
`Metab 2001; 86: 3717–23.
`14. Nauck MA, Heimesaat MM, Ørskov C et al. Preserved
`incretin activity of glucagon-like peptide 1 [7–36 amide]
`but not of synthetic human gastric inhibitory poly-
`peptide in patients with type-2 diabetes mellitus. J Clin
`Invest 1993; 91: 301–7.
`15. Kjems LL, Holst JJ, Volund A, Madsbad S. The influ-
`ence of GLP-1 on glucose-stimulated insulin secretion:
`effects on β-cell sensitivity in type 2 and nondiabetic
`subjects. Diabetes 2003; 52: 380–6.
`
`6
`
`International Diabetes Monitor
`
`Volume 21, Number 1, 2009
`
`MPI EXHIBIT 1040 PAGE 6
`
`MPI EXHIBIT 1040 PAGE 6
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1040, p. 6 of 7
`
`

`

`16. Vilsbøll T, Krarup T, Madsbad S, Holst JJ. Defective
`amplification of the late phase insulin response to
`glucose by GIP in obese Type II diabetic patients.
`Diabetologia 2002; 45: 1111–19.
`17. Deacon CF, Knudsen LB, Madsen K et al. Dipeptidyl
`peptidase IV resistant analogues of glucagon-like
`peptide-1 which have extended metabolic stability and
`improved biological activity. Diabetologia 1998; 41:
`271–8.
`18. Simonsen L, Holst JJ, Deacon CF. Exendin-4, but not
`glucagon-like peptide-1, is cleared exclusively by
`glomerular filtration in anaesthetised pigs. Diabetologia
`2006; 49: 706–12.
`19. Kolterman OG, Kim DD, Shen L et al. Pharmaco-
`kinetics, pharmacodynamics, and safety of exenatide in
`patients with type 2 diabetes mellitus. Am J Health Syst
`Pharm 2005; 62: 173–81.
`20. Knudsen LB, Nielsen PF, Huusfeldt PO et al. Potent
`derivatives of glucagon-like peptide-1 with pharmaco-
`kinetic properties suitable for once daily administration.
`J Med Chem 2000; 43: 1664–9.
`21. Agersø H, Jensen LB, Elbrond B et al. The pharmaco-
`kinetics, pharmacodynamics, safety and tolerability of
`NN2211, a new long-acting GLP-1 derivative, in
`healthy men. Diabetologia 2002; 45: 195–202.
`22. Vilsbøll T, Zdravkovic M, Le-Thi T et al. Liraglutide, a
`long-acting human glucagon-like peptide-1 analog,
`given as monotherapy significantly improves glycemic
`control and lowers body weight without risk of hypo-
`glycemia in patients with type 2 diabetes. Diabetes Care
`2007; 30: 1608–10.
`23. Amori RE, Lau J, Pittas AG. Efficacy and safety of
`incretin therapy in type 2 diabetes. Systematic review
`and meta-analysis. JAMA 2007; 298: 194–206.
`24. Klonoff DC, Buse JB, Nielsen LL et al. Exenatide
`effects on diabetes, obesity, cardiovascular risk factors
`and hepatic biomarkers in patients with type 2 diabetes
`treated for at least 3 years. Curr Med Res Opin 2008; 24:
`275–86.
`25. Blonde L, Klein EJ, Han J et al. Interim analysis of the
`effects of exenatide treatment on A1c, weight and car-
`diovascular risk factors over 82 weeks in 314 overweight
`patients with type 2 diabetes. Diabetes Obes Metab 2006;
`8: 436–47.
`26. Brixner D, Oderda G, Xiangyang Y et al. Clinical effec-
`tiveness of exenatide in patients with type 2 diabetes in
`a primary care electronic medical record database
`(abstract). Diabetes 2008; 57 (suppl 1).
`27. 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: 1240–50.
`28. Buse JB, Drucker DJ, Taylor K et al. Exenatide once
`weekly elicits sustained glycaemic control and weight loss
`over 52 weeks (abstract). Diabetologia 2008; suppl: A146.
`29. 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 2008; Sep 24 [Epub
`ahead of print].
`
`Review articles
`
`30. Vaag AA, Nauck MA, Brandle M et al. Liraglutide, a
`human GLP-1 analogue, substantially reduces HbA1c in
`subjects with type 2 diabetes, irrespective of HbA1c at
`baseline (abstract). Diabetologia. In press.
`31. Colagiuri S, Frid A, Zdravkovic M et al. Liraglutide, a
`human GLP-1 analogue, reduces systolic blood
`pressure in subjects with type 2 diabetes (abstract).
`Diabetologia. In press.
`32. 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: 1126–31.
`33. Deacon CF, Hughes TE, Holst JJ. Dipeptidyl peptidase
`IV inhibition potentiates the insulinotropic effect of
`glucagon-like peptide 1 in the anesthetized pig. Diabetes
`1998; 47: 764–9.
`34. Deacon CF, Danielsen P, Klarskov L et al. Dipeptidyl
`peptidase IV inhibition reduces the degradation and
`clearance of GIP and potentiates its insulinotropic and
`antihyperglycemic effects in anesthetized pigs. Diabetes
`2001; 50: 1588–97.
`35. Kim D, Kowalchick JE, Edmondson SD et al.
`Triazolopiperazine-amides as dipeptidyl peptidase IV
`inhibitors: close analogs of JANUVIA® (sitagliptin
`phosphate). Bioorg Med Chem Lett 2007; 17: 3373–7.
`36. Villhauer EB, Brinkman JA, Naderi GB et al. 1-
`[(3-hydroxy-1-adamantyl)amino]acetyl]-2-cyano-(S)-
`pyrrolidine, a potent, selective, and orally bioavailable
`dipeptidyl peptidase IV inhibitor with antihyper-
`glycemic properties. J Med Chem 2003; 46: 2774–89.
`37. Deacon CF, Holst JJ. Dipeptidyl peptidase IV
`inhibitors, a promising new therapeutic approach for
`the management of type 2 diabete

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket