throbber
Best Practice & Research Clinical Endocrinology & Metabolism
`Vol. 18, No. 4, pp. 531–554, 2004
`doi:10.1016/j.beem.2004.08.001
`available online at http://www.sciencedirect.com
`
`6 G
`
`lucagon-like peptide-1 and glucagon-like
`peptide-2
`
`Laurie L. Baggio PhD
`Research Associate
`
`Daniel J. Drucker* MD
`Director
`
`Department of Medicine, The Banting and Best Diabetes Centre, University of Toronto, Toronto General Hospital,
`200 Elizabeth Street, MBRW 4R-402, Toronto, Ont., Canada M5G 2C4
`
`The glucagon-like peptides (glucagon-like peptide-1 (GLP-1) and glucagon-like peptide-2 (GLP-2))
`are released from enteroendocrine cells in response to nutrient ingestion. GLP-1 enhances
`glucose-stimulated insulin secretion and inhibits glucagon secretion, gastric emptying and feeding.
`GLP-1 also has proliferative, neogenic and antiapoptotic effects on pancreatic b-cells. More
`recent studies illustrate a potential protective role for GLP-1 in the cardiovascular and central
`nervous systems. GLP-2 is an intestinal trophic peptide that stimulates cell proliferation and
`inhibits apoptosis in the intestinal crypt compartment. GLP-2 also regulates intestinal glucose
`transport, food intake and gastric acid secretion and emptying, and improves intestinal barrier
`function. Thus, GLP-1 and GLP-2 exhibit a diverse array of metabolic, proliferative and
`cytoprotective actions with important clinical implications for the treatment of diabetes and
`gastrointestinal disease, respectively. This review will highlight our current understanding of the
`biology of GLP-1 and GLP-2, with an emphasis on both well-characterized and more novel
`therapeutic applications of these peptides.
`
`Key words: diabetes; obesity; food intake; intestinal disease; cell proliferation; apoptosis; insulin
`secretion.
`
`Glucagon-like peptide-1 (GLP-1) is an incretin hormone that regulates blood glucose
`level through its combined actions on the stimulation of glucose-dependent insulin
`secretion and the inhibition of glucagon secretion, gastric emptying and food intake.1
`GLP-1 also increases b-cell mass via a stimulation of b-cell proliferation and neogenesis,
`and an inhibition of b-cell apoptosis.2 The observation that the pharmacological
`
`* Corresponding author. Tel.: C1 416 340 4125; Fax: C1 416 978 4108.
`E-mail address: d.drucker@utoronto.ca (D.J. Drucker).
`
`1521-690X/$ - see front matter Q 2004 Elsevier Ltd. All rights reserved.
`
`MPI EXHIBIT 1069 PAGE 1
`
`MPI EXHIBIT 1069 PAGE 1
`
`

`

`532 L. L. Baggio and D. J. Drucker
`
`administration of GLP-1 and related analogues can reduce elevated fasting and
`postprandial blood glucose levels in diabetic human subjects has generated intense
`interest in the development of GLP-1R agonist-based therapies for the treatment of
`diabetes mellitus.
`GLP-2 is a 33 amino acid peptide that regulates energy homeostasis via acute and
`chronic effects on gut motility, and nutrient ingestion and absorption. In rodents,
`exogenous GLP-2 administration promotes the growth and survival of epithelial cells
`within the small and large bowel mucosa via an inhibition of apoptotic cell death and a
`stimulation of cellular proliferation.1 GLP-2 also enhances barrier function and
`increases the resistance to and recovery from a variety of experimental models of gut
`injury.1 The reported actions of GLP-2 have fostered the initiation of clinical studies to
`assess the ability of GLP-2 to improve nutrient absorption and epithelial integrity and
`restore functional bowel mass in human patients with intestinal disease.
`The actions of GLP-1 and GLP-2 are mediated via unique G protein-coupled
`receptors. The GLP-2 receptor (GLP-2R) is expressed in a highly tissue-specific
`manner, predominantly in the gastrointestinal tract and brain.3 In contrast, the GLP-1
`receptor (GLP-1R) has a more widespread distribution and is expressed in a number of
`tissues,
`including the pancreas,
`intestine, stomach, central nervous system (CNS),
`heart, pituitary, lung and kidney.4–6
`Despite the potential therapeutic benefits of GLP-1 and GLP-2, the durations of
`their action are limited owing to a rapid inactivation of these peptides by the ubiquitous
`protease dipeptidyl peptidase-IV (DPP-IV). Consequently, the inhibition of DPP-IV
`activity or the development of DPP-IV-resistant glucagon-like peptide analogues offers
`additional therapeutic options for treating human disease.
`
`SYNTHESIS, SECRETION AND METABOLISM OF GLP-1 AND GLP-2
`
`GLP-1 and GLP-2 are co-encoded within the proglucagon gene, which, in mammals,
`gives rise to a single mRNA transcript that is expressed in the a-cells of the endocrine
`pancreas, in the enteroendocrine L-cells of the intestine and in the hypothalamus and
`brainstem in the CNS.7,8 The proglucagon mRNA is translated into a single 160 amino
`acid precursor protein that undergoes tissue-specific post-translational processing to
`produce several biologically active proglucagon-derived peptides (PGDPs), including
`glucagon in the pancreatic a-cells and glicentin, oxyntomodulin, GLP-1 and GLP-2 in the
`intestine and brain (Figure 1). Glucagon is the major counter-regulatory hormone to
`insulin and is essential for maintaining blood glucose levels in the physiological range
`during the post-absorptive state. Oxyntomodulin has inhibitory effects on gastroin-
`testinal secretion and motility, and stimulatory effects on pancreatic enzyme secretion
`and intestinal glucose uptake.9 More recently,
`it has been demonstrated that
`oxyntomodulin can reduce food intake in both rodents and humans.10,11 In contrast,
`the physiological actions of glicentin are poorly defined. GLP-1 and GLP-2 exhibit
`trophic effects in the pancreas and intestine, respectively, and play important roles in
`the regulation of nutrient assimilation and energy homeostasis (see below).
`The PGDP sequences within the proglucagon precursor are flanked by pairs of basic
`amino acids, and the post-translational processing of proglucagon is carried out by
`prohormone convertases, which are endoproteolytic enzymes that cleave C-terminal
`to paired basic amino acid residues.12 Although the prohormone convertase enzymes
`responsible for the production of GLP-1 and GLP-2 in the CNS have not been
`
`MPI EXHIBIT 1069 PAGE 2
`
`MPI EXHIBIT 1069 PAGE 2
`
`

`

`Glucagon-like peptide-1 and glucagon-like peptide-2 533
`
`GRPP
`
`Glucagon
`
`IP-1
`
`GLP-1
`
`IP-2
`
`GLP-2
`
`Glicentin
`
`MPGF
`
`Oxyntomodulin
`
`Pancreas
`Glucagon
`MPGF
`
`Intestine & Brain
`Glicentin
`Oxyntomodulin
`GLP-1
`GLP-2
`IP-1
`
`Figure 1. Mammalian proglucagon structure and tissue-specific post-translational processing of the
`proglucagon-derived peptides. GRPP, glicentin-related polypeptide; IP-1 and IP-2, intervening peptide-1 and -2;
`MPGF, major proglucagon fragment; GLP-1 and GLP-2, glucagon-like peptide-1 and -2.
`
`definitively established, prohormone convertase 1/3 has been localized to intestinal
`L-cells and is both necessary and sufficient for the post-translational processing of
`proglucagon to GLP-1 and GLP-2.13 In contrast, prohormone convertase 2 is expressed
`in the islet a-cell and is essential for the generation of 29 amino acid pancreatic
`glucagon.14
`GLP-1 and GLP-2 are secreted in a 1:1 ratio from intestinal L-cells15, the majority of
`which are located in the distal ileum and colon.16 The major stimulus for GLP-1 and
`GLP-2 secretion is the ingestion of nutrients, including glucose, fatty acids and dietary
`fibre.17 Fasting plasma levels of the biologically active forms of GLP-1 and GLP-2 in
`healthy humans are 5–10 and 15–20 pM, respectively, and increase 2–5-fold following
`food ingestion, the absolute peak level being dependent on the size and nutrient
`composition of the meal.18,19 When nutrients are ingested, the release of GLP-1
`and GLP-2 into the circulation occurs in a bi-phasic manner, consisting of a rapid (within
`10–15 minutes) early phase followed by a more prolonged (30–60 minutes) second
`phase.20 The distal location of most L-cells that produce GLP-1 and GLP-2 makes it
`unlikely that the rapid nutrient-stimulated increase in plasma levels of these peptides is
`due to a direct effect of nutrients on the L-cell. Indeed, studies in rodents and humans
`clearly indicate that the vagus nerve, the neurotransmitter gastrin-releasing peptide and
`the hormone glucose-dependent insulinotropic peptide all contribute to the rapid
`release of GLP-1 and GLP-2 from distal L-cells in response to nutritional stimuli.17 In
`contrast, the second phase of peptide secretion probably results from a direct
`stimulation of the L-cell by digested nutrients.21 Thus, nutrient-induced stimulatory
`signals are transmitted to intestinal L-cells indirectly, via neural and endocrine effectors,
`and also by direct interaction with these cells, to mediate the first and second phase,
`respectively, of GLP-1 and GLP-2 secretion.
`The half-life of circulating biologically active GLP-1 is less than 2 minutes22, whereas
`GLP-2 is more stable, with a half-life of approximately 5–7 minutes.23,24 The relatively
`short circulating half-lives of the bioactive forms of these peptides can be attributed to
`renal clearance and enzymatic inactivation. DPP-IV, a serine protease that cleaves
`dipeptides from the amino terminus of oligopeptides or proteins that have a proline or
`
`MPI EXHIBIT 1069 PAGE 3
`
`MPI EXHIBIT 1069 PAGE 3
`
`

`

`534 L. L. Baggio and D. J. Drucker
`
`alanine residue in the penultimate position25, is a critical determinant of GLP-1/GLP-2
`degradation. DPP-IV cleaves GLP-1 and GLP-2 at the alanine residue in position 2,
`yielding the inactive peptides GLP-1 (9–37/36NH2) and GLP-2 (3–33). DPP-IV
`expression is fairly widespread and is found on the surface of circulating white blood
`cells and in cells constituting the vascular endothelium of the small intestine, adjacent to
`the sites of GLP-1 and GLP-2 secretion.25,26 Thus, the majority of GLP-1 and GLP-2
`entering the portal circulation has already been inactivated by DPP-IV prior to entry
`into the systemic circulation. The kidney provides the major route of clearance for both
`GLP-1 and GLP-227, and patients with uraemia or chronic renal
`insufficiency have
`elevated levels of circulating GLP-1 relative to healthy control individuals.28,29
`
`GLP-1 AND GLUCOSE HOMEOSTASIS
`
`GLP-1 elicits multiple actions in the pancreas and in extra-pancreatic tissues that lead to
`the reduction of blood glucose (Figure 2). The first physiological action to be described
`for GLP-1 was the augmentation of glucose-stimulated insulin secretion.30–32 GLP-1
`binds to its specific receptor on the pancreatic b-cell and stimulates insulin secretion
`C
`through mechanisms that involve an inhibition of ATP-sensitive K
`channels (KATP) and
`subsequent b-cell depolarization, elevations in intracellular Ca2C
`level, an inhibition of
`channels and direct effects on the b-cell exocytotic
`voltage-dependent K
`machinery.33,34 The intracellular signalling events that modulate GLP-1-regulated
`insulin release include activation of the cAMP/protein kinase A (PKA), cAMP/guanine-
`nucleotide exchange factor and phosphatidylinositol-3 kinase (PI-3K)/protein kinase C
`(PKC)z pathways.34–36 Unlike other insulin secretagogues, GLP-1 also promotes insulin
`gene transcription, mRNA stability and biosynthesis, and thus has the capacity to
`replenish depleted b-cell insulin stores.37,38
`
`C
`
`GLP-1 Actions
`
`Pancreas
`
`CNS
`
`↑ Insulin synthesis & secretion
`↓ Glucagon secretion
`↑ Somatostatin secretion
`↑ Expression of genes that
`modify β-cell function
`↑ β-cell proliferation & neogenesis
`↑ β-cell survival
`
`↓ Food intake
`↑ Satiety
`↑ Neuronal cell proliferation &
`neogenesis
`↑ Neuronal cell survival
`↑ Learning & memory
`
`Stomach & Intestine
`
`Liver/Fat/Muscle
`
`Heart
`
`↓ Gastric emptying
`↓ Bowel motility
`
`↑ Glycogen synthesis
`↑ Lipogenesis
`
`Structure/ function
`Cardioprotection
`
`Figure 2. Pancreatic and extra-pancreatic glucagon-like peptide-1 receptor agonist-dependent actions. CNS,
`central nervous system.
`
`MPI EXHIBIT 1069 PAGE 4
`
`MPI EXHIBIT 1069 PAGE 4
`
`

`

`Glucagon-like peptide-1 and glucagon-like peptide-2 535
`
`In addition to its ability to stimulate glucose-dependent insulin secretion by direct
`interaction with the pancreatic b-cell GLP-1R, GLP-1 may also stimulate insulin
`secretion indirectly via neural mechanisms. It has been estimated that more than half of
`the GLP-1 secreted from intestinal L-cells is inactivated by DPP-IV, while the majority of
`the remaining intact peptide is inactivated as it passes through the liver.26,39 It is thus
`likely that only small amounts of bioactive GLP-1 actually reach the pancreas intact.
`Studies using ganglionic blockers in rats or chemical denervation experiments in mice
`following capsaicin treatment suggest that endogenously released GLP-1 can stimulate
`insulin secretion in part by a sensory neural reflex that probably initiates in the
`hepatoportal system.40,41
`GLP-1 can also increase the steady-state levels of mRNA transcripts for key
`components of the molecular machinery involved in b-cell exocytosis, such as the
`sulphonylurea receptor and the inwardly rectifying potassium channel (Kir 6.2),
`molecular subunits of the b-cell KATP channel. Similarly, GLP-1 also regulates b-cell KATP
`channel function via the prevention of glucose-dependent inhibition of KATP channel
`activity.42
`Studies in both rodents and humans reveal that GLP-1 can improve the ability of the
`b-cell to sense and respond to glucose, thereby conferring glucose sensitivity to
`previously resistant b-cells.43,44 A potential mechanism whereby GLP-1 could restore
`b-cell glucose responsivity is suggested by studies demonstrating that GLP-1 can
`upregulate the expression of glucose transporters and glucokinases, components of the
`b-cell glucose sensor.45,46
`GLP-1 also lowers blood glucose levels by inhibiting the secretion of glucagon.47 The
`inhibitory effects of GLP-1 on glucagon secretion may occur through direct interaction
`with GLP-1 receptors on pancreatic a-cells48 or indirectly through GLP-1-mediated
`stimulation of insulin and/or somatostatin secretion.49
`Both animal and human studies demonstrate that GLP-1 delays gastric emptying
`and intestinal motility, thereby slowing the transit of nutrients from the stomach to
`the small
`intestine and attenuating meal-associated elevations in plasma glucose
`levels.50,51 The inhibitory effects of GLP-1 on the gut are likely to involve both CNS-
`and intestinal-derived GLP-1.52 The mechanism whereby peripheral GLP-1 inhibits
`gastrointestinal motility appears to involve either direct interaction with CNS
`centres that regulate visceral motility or an indirect mechanism via vagal afferent
`pathways.52
`GLP-1 may also regulate glucose disposal through peripheral actions on liver, skeletal
`muscle and adipose tissue. GLP-1 has been shown to increase glucose incorporation
`into glycogen in isolated rat hepatocytes and skeletal muscle53, and enhance insulin-
`stimulated glucose metabolism in 3T3 L1 adipocyte cultures and isolated rat
`adipocytes.54,55 However, subsequent studies have failed to support a direct extra-
`pancreatic role for GLP-1 in liver, muscle or adipose tissue.56,57 Whether GLP-1 has
`direct effects on glucose disposal independent of changes in the levels of islet hormones
`in humans remains unclear. A number of studies in healthy and diabetic humans suggest
`that GLP-1 can increase glucose disappearance,
`independent of
`insulin and
`glucagon58–60; in contrast, other studies indicate that GLP-1 has no direct influence
`on glucose disposition.61–65 Recent evidence suggests that the effects of GLP-1 may
`involve a suppression of endogenous glucose production rather than an increase in
`peripheral glucose disposal.66 The mechanism whereby GLP-1 could mediate these
`extra-pancreatic effects in humans, independent of changes in the insulin:glucagon ratio,
`remains uncertain. There is no consistent evidence from rodent or human studies that
`GLP-1 receptors are present in liver, fat or muscle tissues.67–71 Since GLP-1R binding,
`
`MPI EXHIBIT 1069 PAGE 5
`
`MPI EXHIBIT 1069 PAGE 5
`
`

`

`536 L. L. Baggio and D. J. Drucker
`
`signal transduction pathways and the effects of GLP-1R agonists and antagonists in
`these non-pancreatic cell types differ from those observed with the known pancreatic
`GLP-1R, it seems likely that GLP-1 may mediate its effects in extra-pancreatic tissues via
`a ‘second’ or related GLP-1 receptor.72–76
`The importance of GLP-1 as a physiological regulator of glucose homeostasis is
`illustrated by studies in which GLP-1 action has been reduced or eliminated. Inhibition
`of GLP-1 activity with exendin (9–39), a specific GLP-1R antagonist, leads to impaired
`glucose tolerance and diminished levels of glucose-stimulated insulin in rodent, baboon
`and human studies.77–79 Similarly, mice with a targeted genetic disruption of the GLP-1
`K/K
`) are characterized by mild fasting hyperglycaemia, glucose
`receptor (GLP-1R
`intolerance in the face of an oral or intraperitoneal glucose challenge, and decreased
`circulating levels of insulin following glycaemic stimulation.80 Moreover, the relative
`importance of basal GLP-1 levels for glucoregulation is demonstrated in human studies
`in which the administration of exendin (9–39) produces significant elevations in the
`levels of fasting glucose and glucagon, suggesting that GLP-1 may have a tonic inhibitory
`effect on the a-cell.81
`More recent experiments have demonstrated that GLP-1 has proliferative and
`neogenic effects on pancreatic b-cells. Studies with the INS-1 b-cell line demonstrate
`that GLP-1 can activate the expression of immediate early genes, whose products are
`transcription factors that play a role in genetic programmes regulating islet cell
`proliferation and differentiation.82,83 The treatment of islet hormone-negative AR42J
`pancreatic exocrine cells with GLP-1 promotes their conversion into cells that produce
`and secrete insulin in a glucose-dependent manner.84 Moreover, the treatment of rat
`and human pancreatic ductal cell lines with GLP-1 causes them to differentiate into cells
`exhibiting endocrine properties.85,86 GLP-1 has also been shown to accelerate both the
`differentiation and maturation of human and porcine fetal islet cells.87,88 In both normal
`and diabetic rodents, short-term treatment with GLP-1R agonists leads to improved
`glucose tolerance, enhanced b-cell proliferation and neogenesis, leading to increased
`b-cell mass.89–91 In db/db mice and Goto-Kakizaki rats, rodent models of type 2
`diabetes, GLP-1R agonist administration is associated with increased b-cell mass and a
`delayed onset of diabetes.92,93 Moreover, GLP-1R agonist administration during the
`prediabetic neonatal period prevented the development of adult-onset diabetes in rats
`following experimentally induced intrauterine growth retardation.94 A key feature
`revealed by these animal studies is that the beneficial effects of GLP-1R agonists on
`glucose homeostasis may be sustained for prolonged periods following the suspension
`of GLP-1R agonist treatment.
`In addition to stimulating cell proliferation and differentiation, GLP-1 has also been
`shown to have cytoprotective effects. GLP-1 reduces apoptosis in rodent islets and islet
`lines exposed to cytotoxic agents95,96 and preserves morphology,
`cell
`improves
`glucose-stimulated insulin secretion and inhibits apoptosis in freshly isolated human
`islets.97 Similarly, GLP-1 reduces the lipotoxic effects of fatty acids in both human
`lines.98 GLP-1R agonist administration was associated with
`islets and rodent cell
`proliferative and anti-apoptotic effects in both the endocrine and exocrine
`compartments of the pancreas in Zucker diabetic rats99 and significantly reduced the
`number of apoptotic b-cells in db/db mice as well as following streptozotocin
`administration to wildtype mice.92,96
`A direct role for the GLP-1R in the proliferative, neogenic and anti-apoptotic actions
`of GLP-1 is supported by studies examining GLP-1-dependent effects in the presence of
`the GLP-1R antagonist exendin (9–39) in normal cells and rodents, or in separate
`K/K
`studies of GLP-1R
`mice. Exendin (9–39) blocks the GLP-1R agonist-mediated
`
`MPI EXHIBIT 1069 PAGE 6
`
`MPI EXHIBIT 1069 PAGE 6
`
`

`

`Glucagon-like peptide-1 and glucagon-like peptide-2 537
`
`differentiation of human pancreatic ductal cells86 and inhibits the anti-apoptotic effects
`K/K
`of GLP-1 in the MIN6 b-cell line.95 GLP-1R
`mice have fewer large b-cell clusters,
`K/K
`islets exhibit an altered a-cell topography100, display an impaired ability
`and GLP-1R
`to regenerate their b-cell mass following partial pancreatectomy101 and manifest an
`increased susceptibility to streptozotocin-induced apoptosis.96
`The signal-transduction pathways downstream of GLP-1R activation coupled to
`stimulation of cell proliferation have been examined in islets and islet cell lines and
`appear to include trans-activation of the epidermal growth factor receptor, which leads
`to increased PI-3K activity and a subsequent activation of PKCz.102 The precise
`mechanisms involved in GLP-1-dependent b-cell differentiation/neogenesis are poorly
`defined but may involve the activation of PKC and mitogen-activated protein kinase.84
`Notably, a common observation following the GLP-1R agonist treatment of b-cells is an
`increase in levels of PDX-1, a transcription factor essential for pancreatic development
`function.84–87,103,104 The molecular mechanisms implicated in GLP-1-
`and b-cell
`dependent anti-apoptotic pathways appear to be diverse and involve multiple signalling
`pathways. Studies in rodents and experiments with isolated islets or b-cell
`lines
`demonstrate that the anti-apoptotic effects of GLP-1 are associated with reduced levels
`of the pro-apoptotic markers active caspase-3 and poly-ADP-ribose polymerase
`cleavage, and an upregulation of anti-apoptotic factors including Bcl-2, Bcl-xL and
`inhibitor of apoptosis protein-2.92,95,98,99,105 The cumulative experimental evidence
`indicates that the intracellular signalling pathways that mediate GLP-1-dependent
`cytoprotective effects include:
`
`1. cAMP/PKA;
`2. PI-3K-dependent activation of the prosurvival kinase Akt/protein kinase B;
`3. activation of nuclear factor-kappa B activity;
`4. cAMP/PKA-mediated phosphorylation and activation of cAMP response element
`binding protein (CREB), subsequent CREB-dependent activation of insulin receptor
`substrate-2 and induction of Akt/protein kinase B.92,98,106,107
`
`As b-cell mass is reduced and b-cell apoptosis is increased in autopsy studies of
`pancreata from patients with type 2 diabetes108, the ability of GLP-1 to promote b-cell
`proliferation and neogenesis, and inhibit b-cell apoptosis, raises the possibility that
`GLP-1 therapy has the potential to preserve or even restore functional b-cell mass in
`diabetic individuals.
`
`GLP-1 AND FEEDING BEHAVIOR
`
`Numerous studies in rodents have demonstrated that the central (intracerebroven-
`tricular) or peripheral administration of GLP-1R agonists leads to the inhibition of
`food intake and reductions in body weight.109–111 Moreover, GLP-1 also inhibits food
`intake and promotes satiety in normal, obese and diabetic humans112–114, suggesting
`that GLP-1 could play an important role in controlling appetite and body weight.
`K/K
`However, GLP-1R
`mice exhibit normal
`feeding behaviour and body weight,
`indicating that GLP-1R signalling may not be essential for the regulation of satiety and
`maintenance of body weight.80
`There is considerable interest in how GLP-1 mediates its effects on ingestive
`behaviour, and experimental evidence indicates that GLP-1 probably modifies food
`
`MPI EXHIBIT 1069 PAGE 7
`
`MPI EXHIBIT 1069 PAGE 7
`
`

`

`538 L. L. Baggio and D. J. Drucker
`
`It has been proposed that the
`intake via a number of different mechanisms.
`inhibitory effects of GLP-1 on food intake can be mediated indirectly, by its ability to
`slow gastric emptying, thereby promoting gastric distension and a sensation of
`satiety. In addition, GLP-1 receptors are expressed in the hypothalamus and nucleus
`of the solitary tract (NTS), CNS regions that are thought to be important for
`regulating appetite and satiety.5,115 Thus, GLP-1 could also modify feeding by direct
`interaction with these CNS centres. Alternatively,
`it has been suggested that the
`inhibitory effect on food intake could reflect a secondary physiological response to
`the GLP-1-dependent activation of aversive signalling pathways that produce visceral
`illness.116–118
`Binding sites or receptors for GLP-1 have been detected in numerous CNS
`regions, including the hypothalamus, NTS, subfornical organ (SFO) and area postrema
`(AP).5,115,119,120 The SFO and AP represent blood–brain barrier-free CNS locations and
`have also been shown to play a role in the regulation of feeding.121 Although GLP-1 is
`produced in CNS neurons located in the NTS, studies in rats have demonstrated that
`peripherally administered GLP-1 can also gain access to the CNS through interaction
`with GLP-1 receptors in the SFO and AP.122 However, the relative importance of
`peripheral versus central GLP-1 for appetite regulation remains to be elucidated.
`
`GLP-1 AND THE CARDIOVASCULAR SYSTEM
`
`Studies using anaesthetized rats or conscious calves and central or intravenous GLP-
`1 administration are associated with increases in heart rate and systolic, diastolic and
`mean arterial blood pressure.123,124 Consistent with these effects, GLP-1 receptors
`are expressed in the heart and in the NTS and AP, CNS regions known to regulate
`cardiovascular function.125–127 The stimulatory effects of GLP-1 on the rat
`cardiovascular system are independent of catecholamine action, are blocked by
`administration of the GLP-1R antagonist exendin (9–39) and appear to be mediated
`by both direct and indirect GLP-1R-dependent effects on the nervous system and
`heart.124 More recent studies using telemetric monitoring to measure heart rate and
`blood pressure in freely moving rats demonstrated that GLP-1R-dependent increases
`in heart rate and blood pressure are associated with the activation of (i) neuronal
`activity in several autonomic control regions of the rat CNS, (ii) GLP-1-sensitive
`hypothalamic and medullary catecholamine neurons that
`innervate sympathetic
`preganglionic neurons, and (iii) neuronal activity in the adrenal medulla, suggesting
`that central GLP-1 regulates cardiovascular function by activating the sympathetic
`nervous system.128 An essential role for GLP-1R signalling in the maintenance of
`K/K
`normal cardiac structure and function is demonstrated in studies using GLP-1R
`K/K
`mice. GLP-1R
`hearts are characterized by increased septal and posterolateral
`K/K
`myocardial wall
`thickness, and GLP-1R
`mice display abnormal cardiac
`haemodynamic responses to external stress.129
`In contrast to its stimulatory effects on the cardiovascular system in animals, limited
`studies in humans indicate that GLP-1R agonist administration has no significant effects
`on heart rate or blood pressure.130–132 Moreover, GLP-1 may also have cardiopro-
`tective effects under certain conditions. A 14 day infusion of GLP-1 prevents the
`development of hypertension, improves endothelial function and reduces renal and
`cardiac damage in Dahl salt-sensitive rats maintained on a high-salt diet.133
`The antihypertensive effect of GLP-1 in these hypertension-prone rats is attributed
`
`MPI EXHIBIT 1069 PAGE 8
`
`MPI EXHIBIT 1069 PAGE 8
`
`

`

`Glucagon-like peptide-1 and glucagon-like peptide-2 539
`
`to a GLP-1-dependent increase in salt and water excretion133, actions recently
`demonstrated in human subjects.134 Additionally, a 72 hour infusion of GLP-1 in
`patients with acute myocardial infarction and severe left ventricular systolic dysfunction
`following successful reperfusion with primary angioplasty led to improved regional and
`global left ventricular function and was associated with reduced in-hospital mortality
`rate and length of hospitalization.135
`
`GLP-1 AND NEUROPROTECTION
`
`Comparable to actions observed in studies of pancreatic b-cells, GLP-1 also exerts
`proliferative, neogenic and anti-apoptotic effects on neuronal cells. GLP-1R agonist
`treatment of PC12 cells stimulates neurite outgrowth, enhances nerve growth factor-
`induced differentiation and improves cell survival after nerve growth factor with-
`drawal.136 GLP-1R agonists prevent glutamate-induced apoptosis in cultured rat
`hippocampal neurons and restore cholinergic marker activity in the basal forebrain of
`ibotenic acid-treated rats, a rodent model of neurodegeneration.137 Furthermore,
`GLP-1R-activated pathways seem to be important for learning and memory. GLP-1R
`agonist administration is associated with enhanced learning in rats, an effect that can be
`K/K
`blocked by the co-administration of exendin (9–39).138 In contrast, GLP-1R
`mice
`exhibit deficits in learning that can be overcome by hippocampal Glp1r gene transfer.138
`K/K
`GLP-1R
`mice are also more susceptible to kainate-induced seizures and
`hippocampal neuronal degeneration, whereas GLP-1R agonist treatment prevents
`kainate-induced apoptosis in wildtype animals.138 These observations have led to the
`suggestion that GLP-1R agonists may potentially be useful
`for the treatment of
`neurological disorders,
`including the neuropathy that results as a secondary
`complication of diabetes.139 In contrast, endogenous GLP-1 has also been implicated
`in the pathogenesis of b-amyloid protein-induced neurotoxicity as a continuous co-
`infusion of a GLP-1R antagonist with b-amyloid protein prevented memory impairment
`and hippocampal apoptosis in rats.140
`
`GLP-1 AND THE TREATMENT OF TYPE 2 DIABETES
`
`Numerous studies have demonstrated that GLP-1 can enhance glucose-stimulated
`insulin secretion and lower fasting and postprandial blood glucose levels in individuals
`with type 2 diabetes. The administration of GLP-1 by continuous subcutaneous infusion
`for 6 weeks increased insulin secretion, reduced fasting and postprandial glucose levels,
`lowered haemoglobin A1c (HbA1c) values, and decreased food intake and body weight
`in patients with type 2 diabetes141, indicating that GLP-1 retains its effectiveness, even
`with continuous long-term treatment. As obesity can be a contributing factor to the
`pathogenesis of diabetes, the ability of GLP-1 to reduce food intake and promote satiety
`and weight loss provides an additional means for improving glycaemia in these
`individuals. GLP-1 also lowers fasting and postprandial glucose and reduces the meal-
`associated insulin requirement in human patients with type 1 diabetes, probably via its
`inhibitory effects on glucagon secretion and/or gastric emptying.142–144 Moreover, the
`insulinotropic and glucagonostatic effects of GLP-1 are glucose dependent145–147; thus
`under conditions of normoglycaemia, GLP-1 has no effect on the levels of plasma insulin
`or glucagon.
`
`MPI EXHIBIT 1069 PAGE 9
`
`MPI EXHIBIT 1069 PAGE 9
`
`

`

`540 L. L. Baggio and D. J. Drucker
`
`The ability of GLP-1 to lower blood glucose levels and promote weight loss,
`combined with its potential to preserve or restore functional b-cell mass, has generated
`considerable interest in the use of GLP-1 as a therapeutic agent for the treatment of
`diabetes. Moreover, the observation that GLP-1 secretion is deficient in type 2 diabetics
`suggests that the restoration of normal GLP-1 concentrations in these patients may be
`beneficial.19,148 However, because the plasma half-life of native GLP-1 is very short,
`owing to its rapid inactivation by DPP-IV, continuous infusion or multiple injections of
`GLP-1 are required to attain adequate glycaemic control. Consequently, alternative
`therapeutic strategies have been devised focused on the generation of GLP-1 analogues
`that are resistant to DPP-IV and the development of compounds that inhibit DPP-IV
`activity.
`
`DPP-IV-RESISTANT GLP-1R AGONISTS AND DPP-IV INHIBITION
`AS ALTERNATIVE THERAPEUTIC STRATEGIES
`
`A number of structurally unique GLP-1R agonists have been developed with
`prolonged activity such that once- or twice-daily injections of these molecules are
`potentially as efficacious as continuous GLP-1 infusion for the treatment of
`experimental or clinical diabetes. Exendin-4 is a naturally occurring, DPP-IV-
`resistant GLP-1R agonist originally isolated from the venom of the Heloderma
`suspectum lizard149, and exenatide is a synthetic version of exendin-4. A single
`subcutaneous injection of exenatide in patients with poorly controlled type 2
`diabetes significantly reduced fasting and postprandial glucose concentrations,
`in
`association with increased levels of plasma insulin and decreased plasma
`glucagon.150 Four weeks of twice-daily exendin-4/exenatide treatment significantly
`reduced postprandial glucose and HbA1c levels151, and, when used in combination
`with metformin and/or sulphonylurea treatment, also lowered postprandial plasma
`glucose and HbA1c in diabetic patients with poor glycaemic control.132 The results
`of recently completed phase 3 clinical trials demonstrated that the exenatide
`treatment

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