throbber
Perspectives in Diabetes
`Glucagon-Like Peptides
`
`Daniel J. Drucker
`
`Proglucagon contains the sequence of two glucagon-
`like peptides, GLP-1 and GLP-2, secreted
`from
`enteroendocrine cells of the small and large intestine.
`GLP-1 lowers blood glucose in both NIDDM and IDDM
`patients and may be therapeutically useful for treat-
`ment of patients with diabetes. GLP-1 regulates blood
`glucose via stimulation of glucose-dependent insulin
`secretion, inhibition of gastric emptying, and inhibi-
`tion of glucagon secretion. GLP-1 may also regulate
`glycogen synthesis in adipose tissue and muscle; how-
`ever, the mechanism for these peripheral effects
`remains unclear. GLP-1 is produced in the brain, and
`intracerebroventricular GLP-1 in rodents is a potent
`inhibitor of food and water intake. The short duration
`of action of GLP-1 may be accounted for in part by the
`enzyme dipeptidyl peptidase 4 (DPP-IV), which cleaves
`GLP-1 at the NH 2-terminus; hence GLP-1 analogs or the
`lizard peptide exendin-4 that are resistant to DPP-IV
`cleavage may be more potent GLP-1 molecules in vivo.
`GLP-2 has recently been shown to display intestinal
`growth factor activity in rodents, raising the possibility
`that GLP-2 may be therapeutically useful for enhance-
`ment of mucosal regeneration in patients with intesti-
`nal disease. This review discusses recent advances in
`our understanding of the biological activity of the
`glucagon-like peptides. Diabetes 47:159-169, 1998
`
`THE INCRETIN CONCEPT AND p-CELL FUNCTION
`Enhancement of insulin secretion from the islet |3 cell is a prin-
`cipal goal for treatment of patients with NIDDM. The obser-
`vation that sulfonylureas stimulate insulin secretion has pro-
`vided the rationale for the therapeutic use of these agents in
`the treatment of NIDDM. Nevertheless, the sulfonylurea
`stimulation of insulin secretion is not strictly glucose depen-
`dent, and hence hypoglycemia is an undesirable side effect of
`sulfonylurea treatment, particularly in elderly patients.
`The observation that glucose administered via the gas-
`trointestinal tract is associated with a greater stimulation of
`insulin release compared with a comparable glucose chal-
`lenge given intravenously (1,2) prompted a search for the
`responsible "incretins," gut-derived factors that increase glu-
`cose-stimulated insulin secretion (3). The concept of the
`
`From the Department of Medicine, the Toronto Hospital; and the Banting
`and Best Diabetes Centre, University of Toronto, Ontario, Canada.
`Address correspondence and reprint requests to Dr. Daniel J. Drucker,
`The Toronto Hospital, 200 Elizabeth St., CCRW3-838, Toronto, Canada M5G
`2C4. E-mail: d.drucker@utoronto.ca.
`Received for publication 9 September 1997 and accepted in revised
`form 8 October 1997.
`CNS, central nervous system; DPP-IV, dipeptidyl peptidase 4; GIP, glu-
`cose-dependent insulinotropic peptide; GLP, glucagon-like peptide; GLP-1R,
`GLP-1 receptor; GRP, gastrin-releasing peptide; ICV, intracerebroventricu-
`lar; PC, prohormone convertase; PGDP, proglucagon-derived peptide; RT-
`PCR, reverse transcription-polymerase chain reaction.
`
`DIABETES, VOL. 47, FEBRUARY 1998
`
`enteroinsular axis suggested that insulin secretagogues were
`synthesized in and released from the intestinal endocrine sys-
`tem after nutrient ingestion. The isolation and characterization
`of glucose-dependent insulinotropic peptide (GIP) repre-
`sented an important advance in the identification of intestinal
`incretin hormones. GIP is released from enteroendocrine cells
`in the duodenum and proximal jejunum after nutrient intake
`and stimulates insulin secretion in a glucose-dependent man-
`ner (3,4). Nevertheless, immunoneutralization of GIP or
`removal of GIP from intestinal extracts does not result in
`complete elimination of incretin activity, consistent with the
`presence of additional gut-derived factors with insulinotropic
`activity (4).
`After the isolation of the cDNAs and genes encoding
`proglucagon approximately 15 years ago (5-7), two novel
`glucagon-like peptides, GLP-1 and GLP-2, were identified
`COOH-terminal to the glucagon sequence in mammalian
`proglucagon (Figs. 1 and 2). Initial characterization of GLP-
`1 bioactivity using NH2-terminally extended GLP-l(l-37)
`failed to demonstrate effects on blood glucose or insulin
`secretion; however, subsequent experiments using the NH2-
`terminally truncated GLP-l(7-36) amide or GLP-l(7-37) pep-
`tides demonstrated potent effects on glucose-dependent
`insulin secretion, islet cell cAMP formation, and insulin gene
`expression (8-12). The principal aim of this review is to high-
`light recent advances in our understanding of the biology of
`the GLPs. Previous reviews of the biology of GLP-1 and GLP-
`2 offer a detailed introduction to the subject (4,13-15).
`
`GLP-1 BIOSYNTHESIS AND SECRETION
`A single proglucagon gene in mammals gives rise to an iden-
`tical proglucagon RNA transcript that is translated and
`processed differently in brain, pancreatic islets, and intestine
`(Fig. 1) (16). In contrast, vertebrates such as the chicken, fish,
`frog, and lizard may contain two proglucagon genes, and use
`alternative RNA splicing for the generation of proglucagon
`mRNA transcripts that encode for GLP-1 but not GLP-2 in the
`pancreas and both GLP-1 and GLP-2 in the intestine (17,18).
`Although considerable progress has been made in elucidat-
`ing the factors that control proglucagon gene expression in
`islet cells, much less is known about the regulation of
`proglucagon gene expression and, hence, the control of GLP-
`1 biosynthesis, in enteroendocrine cells. Transgenic experi-
`ments have demonstrated that different
`tissue-specific
`enhancers specify islet versus intestinal proglucagon gene
`expression (19,20); however, the intestine-specific pro-
`glucagon gene enhancer remains poorly defined (21). The pan-
`creatic A-cell and enteroendocrine L-cell both express the
`transcription factor cdx-2/3, which regulates proglucagon
`gene expression in pancreas and intestine (22,23); however,
`transcription factors important for regulation of the
`proglucagon promoter specifically in the enteroendocrine
`
`159
`
`AstraZeneca Exhibit 2069
`Mylan v. AstraZeneca
`IPR2015-01340
`
`Page 1 of 11
`
`

`

`GLUCAGON-LIKE PEPTIDES
`
`Proglucagon
`
`GRPP
`
`30
`
`33
`
`Glucagon
`
`64
`IP-1
`
`69
`
`78
`
`107/8
`
`126
`
`GLP-1
`
`IP-2
`
`GLP-2
`
`61
`
`72
`
`111
`
`123
`
`158
`
`158
`
`Glicentin
`
`Oxyntomodulin
`
`MPGF
`
`Pancreas I ™ g on
`
`Oxyntomodulin
`Intestine G UM
`GLP-2
`LIP-2
`FIG. 1. Structural organization of mammalian proglucagon. The numbers refer to amino acid sequences in proglucagon. The peptides released
`by posttranslational processing in the pancreas and intestine are shown. GRPP, glicentin-related pancreatic polypeptide; IP, intervening pep-
`tide; MPGF, major proglucagon fragment.
`
`cell have not been extensively characterized. Furthermore,
`despite interest in potential new strategies for increasing
`GLP-1 synthesis and secretion in diabetic patients, the factors
`important for the regulation of human intestinal proglucagon
`biosynthesis remain unknown.
`The liberation of GLP-1 in the intestine but not the pancreas
`appears to be due to the tissue-specific expression of pro-
`hormone convertases (PCs) in the enteroendocrine cells of the
`small and large bowel. Whereas both PCI and PC2 cleave
`proglucagon to generate the major proglucagon fragment and
`glicentin and oxyntomodulin (24,25), PCI expressed in
`enteroendocrine cells appears to be the enzyme responsible
`for the liberation of both GLP-1 and GLP-2 (25). Although
`multiple immunoreactive forms of GLP-1 are liberated in vivo,
`including GLP-l(7-36) amide and GLP-l(7-37), the majority of
`circulating GLP-1 in humans appears to be GLP-l(7-36) amide
`(26). Nevertheless, in vivo studies have shown that both
`molecular forms of NH2-terminaUy truncated GLP-1 are
`equipotent with regard to their insulin-stimulating properties;
`in addition, both appear to exhibit similar half-lives in vivo (27).
`
`Whether control of PC activity in the intestine is an important
`regulator of GLP-1 synthesis remains to be determined.
`Because the enteroendocrine cell is exposed to both cir-
`culating humoral factors and luminal intestinal contents,
`intestinal proglucagon-derived peptide (PGDP) biosynthesis
`and secretion are subject to regulation by both hormonal
`and nutritional factors. Nutrient intake stimulates the syn-
`thesis and secretion of PGDPs from the enteroendocrine cell
`in rodents (28). In one study, proglucagon mRNA abundance
`decreased with fasting and increased with refeeding in rat
`jejunum, and the profile of circulating enteroglucagon and
`GLP-1 paralleled changes observed in jejunal proglucagon
`mRNA (29). The rapid rise in plasma GLP-1 levels after nutri-
`ent ingestion and the distal location of the majority of GLP-
`1-containing enteroendocrine cells in the ileum and colon has
`led to the suggestion that one component of the nutrient-
`induced secretory signal may be indirect, perhaps via GIP or
`gastrin-releasing peptide (GRP) release from the proximal
`jejunum. GLP-1 secretion from the distal ileum was abol-
`ished when intervening segments of intestine were resected,
`
`DPP-IVI
`(7-37)
`Human GLP-1 HAEGT FTSDV SSYLE GQAAK EFIAW LVKGR G
`(7-36)Mdde
`Human GLP-1 HAEGT FTSDV SSYLE GQAAK EFIAW LVKGR
`Exendin-4
`HGEGT FTSDL SKQME EEAVR LFIEW LKNGG PSSGA PPPSG
`YAEGT FISDY SIAMD KIHQQ DFVNW LLAQK GKKND WKHNI TQ
`Human GIP
`
`Human GLP2 HADGS FSDEM NTILD NLAAR DFINW LIQTK ITD
`Rat GLP2
`HADGS FSDEM NTILD NLATR DFINW LIQTK ITD
`Mouse GLP2 HADGS FSDEM STILD NLATR DFINW LIQTK ITD
`
`FIG. 2. Amino acid sequences of GLP-1, exendin-4, GIP, and GLP-2. The arrow designates the recognition site for DPP-IV enzymatic cleavage.
`Residues in rat or mouse GLP-2 that differ in sequence from human GLP-2 are underlined.
`
`160
`
`DIABETES, VOL. 47, FEBRUARY 1998
`
`Page 2 of 11
`
`

`

`Biological activities of GLP-1
`
`Adipose
`tissue
`
`and water intake
`
`7 Glycogen synthesis
`Glycogenolysis
`
`JGastric emptying
`Islets of Langerhans
`f Glucagon
`•Insulin
`ASomatostatin
`
`Nutrients
`L
`
`[ Intestine"!
`FIG. 3. Schematic representation of GLP-1 action.
`
`and infusion of a GRP antagonist inhibited the L-cell
`response to nutrient ingestion, observations consistent with
`GRP having an important role in the humoral regulation of
`GLP-1 secretion (30).
`In isolated perfused rat ileum preparations, GLP-1 secretion
`was stimulated by cholinergic agonists, bombesin, calci-
`tonin-gene-related peptide, and GIP (31,32), and in the iso-
`lated perfused rat colon, it was stimulated by pj-adrenergic and
`cholinergic agonists, bombesin, and calcitonin-gene-related
`peptide, and GIP (33). Somatostatin directly inhibits L-cell and
`GLP-1 secretion, and galanin antagonizes the stimulatory
`effect of GIP on GLP-1 release in rats (34). Luminal perfusion
`with glucose, pectin, or the bile acid hodeoxycholate stimu-
`lated GLP-1 secretion from the rat colon, suggesting that
`enteroendocrine cells in the large bowel are also sensitive to
`luminal contents in vivo (35).
`GLP-1 secretion is also stimulated by nutrient ingestion in
`humans. Basal circulating levels of human GLP-1 (7-36)
`amide range from 0.4 to 7.0 pmol/1, depending on the assay,
`and are stimulated after oral but not intravenous glucose
`administration (26,36). Secretion of GLP-1 throughout the
`day increases after meal ingestion, in parallel with meal-
`related increases in insulin secretion (37,38). GLP-1 release
`was also stimulated after oral administration of galactose,
`amino acids, and corn oil (36). GLP-1 is secreted in a pulsatile
`manner in humans; glucose ingestion increases the amplitude,
`but not the frequency, of GLP-1 secretion (39). The integrated
`GLP-1 pulse amplitude was reduced by atropine, consistent
`with the importance of cholinergic mechanisms in the con-
`trol of GLP-1 secretion.
`
`GLP-1 DEGRADATION
`An important determinant of the circulating levels of bioactive
`GLP-1 appears to be the NHg-terminal degradation of the pep-
`tide by the enzyme DPP-TV (40). Cleavage of GLP-1 at the
`penultimate alanine residue to generate GLP-1 (9-36)amide
`occurs rapidly in plasma (40), and the half-life of intact GLP-
`1 in vivo appears to be less than 2 min (41). GLP-l(9-36)amide
`constitutes 53.5% of the concentration of intact GLP-l(7-36)
`amide in the fasted state; however, after nutrient ingestion,
`human GLP-l(9-36)amide is relatively more abundant than the
`intact 7-36(amide) molecule (42). GLP-1 (9-36)amide also
`
`DIABETES, VOL. 47, FEBRUARY 1998
`
`D.J. DRUCKER
`
`binds to the GLP-1 receptor, albeit with lower affinity than the
`(7-36)amide form, and may function as a competitive antago-
`nist of the GLP-1 receptor in vivo (43). Radioimmunoassays
`that do not distinguish between intact GLP-l(7-36) amide and
`the NH2-terminally deleted GLP-1 (9-36)amide may overesti-
`mate the actual concentration of circulating bioactive GLP-1
`(42); experiments that measure total immunoreactive circu-
`lating GLP-1 need to be interpreted with caution in light of this
`new information. DPP-IV activity is inhibited by low temper-
`ature and diprotin A (41,42), hence the importance of col-
`lecting blood samples for measurements of GLP-1 immunore-
`activity on ice in the presence of appropriate protease
`inhibitors. Intact GLP-1 appears to be cleared predominantly
`through renal extraction; the contribution of extrarenal tissues
`to clearance of GLP-1 under normal physiological conditions
`remains to be determined (44).
`
`GLP-1 ACTION
`The GLP-1 receptor. GLP-1 exerts its actions via binding to
`a G-protein-linked receptor expressed on islet (3-cells (45).
`The human GLP-1 receptor (46,47) is 90% homologous to the
`rat receptor, and the gene has been localized to 6p21 (48). No
`GLP-1 receptor mutations have been reported in NIDDM
`patients, and genetic analysis has failed to demonstrate link-
`age between the GLP-1 receptor gene and populations with
`maturity-onset diabetes of the young or NIDDM (49). GLP-1
`receptor mRNA transcripts have been detected by Northern
`blotting in rodent tissues such as the islets, lung, kidney,
`stomach, and brain (45,50). GLP-1 receptor mRNA tran-
`scripts have been more difficult to detect by Northern blot-
`ting in human tissues (46,47), but have been identified in
`human pancreas, lung, kidney, stomach, heart, and brain by
`RNAse protection analyses (51).
`Some controversy remains with regard to the expression
`of GLP-1 receptor mRNA transcripts in peripheral tissues.
`Although low levels of GLP-1 receptor mRNA transcripts
`and GLP-1 binding have been reported in rat muscle and
`liver (50), these findings have not been universally con-
`firmed (51,52). Furthermore, discrepancies among results
`obtained using ligand binding, in situ hybridization, RNAse
`protection, and reverse transcription-polymerase chain
`reaction (RT-PCR) for characterization of GLP-1 receptor
`expression have led to the suggestion that structural variants
`of the GLP-1 receptor, or a second closely related receptor,
`may be expressed in different tissues (52-54); however,
`cDNAs encoding variant GLP-1 receptors have not yet been
`identified. Experiments using primary islet cultures, (J-cell
`lines, and cells transfected with the GLP-1 receptor cDNA
`have shown that GLP-1 signaling is coupled to both activa-
`tion of adenylate cyclase and phospholipase C pathways
`(12,45,50,55,56). GLP-1 binding is associated with an
`increase in cytosolic-free calcium (50,56,57). GLP-1 may
`increase intracellular [Ca2+] via activation of a prolonged
`cAMP-sensitive inward current leading to membrane depo-
`larization and increases in intracellular calcium (58). GLP-1
`receptor responsivity may be desensitized in vitro after
`exposure to agonist or activation of protein kinase C (59,60),
`and receptor desensitization appears to correlate with
`receptor phosphorylation (59,61).
`A number of distinct yet complementary actions con-
`tribute to the glucose-lowering properties of GLP-1 (Fig. 3).
`Binding of GLP-1 to its p-cell receptor stimulates insulin
`
`161
`
`Page 3 of 11
`
`

`

`GLUCAGON-LIKE PEPTIDES
`
`secretion in a glucose-dependent manner, and GLP-1
`increases insulin mRNA (12), likely via induction of insulin
`gene transcription through a cAMP-dependent mechanism
`(62,63). GLP-1 also confers glucose sensitivity to glucose-
`resistant (3-cells (55), thereby enhancing the ability of p-cells
`to secrete insulin in a glucose-dependent manner. Consis-
`tent with this hypothesis, GLP-1 increased the insulinotropic
`effect of glibenclamide in the perfused rat pancreas (64),
`and GLP-1 administration to glucose-intolerant aging Wistar
`rats lowered plasma glucose and increased circulating
`insulin and insulin RNA, in keeping with a role for GLP-1 in
`the restoration of normal islet function and control of insulin
`biosynthesis (65).
`GLP-1 also lowers blood glucose via inhibition of
`glucagon secretion (66,67). GLP-1 likely acts directly on the
`pancreatic A-cell and via indirect mechanisms through stim-
`ulation of somatostatin and insulin secretion. GLP-1 infusion
`in C-peptide-negative diabetic dogs lowered circulating
`plasma glucagon, suggesting that the glucagonostatic effects
`of GLP-1 are at least partially independent of circulating
`insulin (68). Consistent with a direct effect of GLP-1 on
`glucagon and somatostatin secretion, GLP-1 receptors have
`been localized to the a- and 8-cells of the islets (69). Para-
`doxically, GLP-1 stimulated glucagon secretion from iso-
`lated rat a cells, and this stimulatory effect was inhibited by
`somatostatin, raising the possibility that the glucagonostatic
`effects of GLP-1 are partly indirect through a paracrine
`effect on somatostatin secretion (70). In contrast, GLP-1
`directly inhibited glucagon secretion and intracellular cAMP
`in the glucagon-producing InRl-G9 cell line in the absence
`of somatostatin (71); the relative contributions of different
`mechanism(s) underlying the inhibitory effect of GLP-1 on
`the A-cell remain unclear.
`Extrapancreatic effects of GLP-1. GLP-1 attenuates
`meal-associated glucose excursion by directly inhibiting gas-
`tric emptying (72). GLP-1 also inhibits postprandial acid
`secretion, and GLP-1 receptors have been demonstrated in
`the stomach (45,73). The GLP-1 receptor expressed in heart
`is structurally identical to the pancreatic islet receptor (51),
`and GLP-1 increased systolic and diastolic pressure and
`heart rate in rats (74). The highest levels of GLP-1 receptor
`mRNA transcripts are found in lung (45), consistent with
`identification of GLP-1 binding sites in lung membrane
`preparations (75,76). Although GLP-1 stimulated macro-
`molecule secretion from tracheal ring preparations (77), a
`physiological role, if any, for GLP-1 in pulmonary physiology
`in vivo remains to be determined.
`The mechanism of action of GLP-1 in peripheral tissues
`such as liver, muscle, and adipose tissue remains unclear
`(Fig. 3). GLP-1-stimulated glycogen synthesis in isolated
`hepatocytes from normal and diabetic rats (78) and GLP-1
`binding in rat hepatocyte and liver membrane preparations
`(79) has been demonstrated. Both fish and human GLP-1
`stimulated glycogenolysis in fish hepatocyte preparations
`(80). In contrast, other investigators failed to demonstrate an
`effect of GLP-1 on hepatic glycogenolysis or glycogen syn-
`thesis in rat liver (81). Similarly, GLP-1 binding activity has
`been observed in rat skeletal muscle membranes (54), and
`GLP-1 effects on glucose incorporation into glycogen have
`been demonstrated by some investigators (82,83), but not by
`others (84). GLP-1 binding has also been demonstrated in rat
`and human adipose tissue membranes (53,85), and GLP-
`
`162
`
`1-enhanced insulin-stimulated glucose uptake in 3T3-L1
`adipocytes (86) and isolated rat adipocytes (87). Intriguingly,
`GLP-1 decreased intracellular cAMP in 3T3-L1 adipocytes,
`thereby providing indirect evidence for the presence of a
`second receptor with signaling properties distinct from those
`described for the pancreatic GLP-1 receptor (88). Although
`GLP-1 receptor expression has been demonstrated by RT-PCR
`in RNA from rat muscle and fat pad (86), other investigators,
`using a combination of RT-PCR, RNAse-protection, and in situ
`hybridization experiments, failed to detect GLP-1 receptor
`mRNA transcripts in adipose tissue, liver, and muscle (52).
`Given the lack of conclusive evidence for the expression of
`the pancreatic GLP-1 receptor in muscle, liver, and adipose
`tissue, the mechanisms and receptor(s) mediating these
`peripheral effects of GLP-1 remain unclear.
`PGDPs and GLP-1 are synthesized in the central nervous
`system (CNS), and GLP-1 receptors have been localized
`through a combination of in situ autoradiography and
`hybridization studies to different regions of the CNS (89,90).
`Although GLP-1 immunoreactivity is widely distributed in
`many regions of the brain, GLP-1 mRNA transcripts are local-
`ized predominantly to the brain stem and, to a lesser extent,
`the hypothalamus, thereby supporting a role for peptidergic
`transport from brain stem neurons in the regulation of GLP-
`1 CNS distribution (91-93). Consistent with these findings,
`GLP-1 binding sites and GLP-1 receptor RNA transcripts have
`been identified throughout the CNS (89,90,94,95) and in the
`pituitary (96,97). GLP-1 may also play a role in the peripheral
`nervous system, as intraportal GLP-1 activates vagal nerve
`activity in rats (98).
`A potential role for GLP-1 in the central control of feeding
`behavior was suggested by studies demonstrating that intra-
`cerebroventricular (ICV) administration of GLP-1 in rats
`inhibited food intake and induced c-fos immunoreactivity in
`the paraventricular nucleus and amygdala (99). Although
`ICV GLP-1 and leptin both inhibit food intake, leptin acti-
`vated c-/os-like immunoreactivity in regions of the rat brain
`different from those activated by GLP-1 (100). Furthermore,
`the inhibitory effects of leptin were of comparatively longer
`duration, and GLP-1, but not leptin, produced conditioned
`taste aversion, implying distinct roles for these peptides in the
`central regulation of feeding (101). ICV GLP-1 inhibited basal
`water intake (102) and stimulated urinary excretion of water
`and sodium, and both ICV and intraperitoneal GLP-1 inhibited
`basal and ANG II-induced drinking behavior (103) and
`reduced body temperature in rats. Whether the GLP-1 effects
`on water regulation are related to or distinct from the periph-
`eral effects of GLP-1 on heart rate and blood pressure (74)
`remains uncertain.
`Studies using GLP-1 receptor antagonists. The obser-
`vation that a truncated lizard GLP-1-related peptide,
`exendin(9-39), binds to the mammalian GLP-1 receptor and
`functions as a GLP-1 antagonist (46,104) has provided the
`opportunity to carry out studies examining the transient
`reduction or loss of GLP-1 action both in vitro and in vivo.
`Exendin(9-39) administered to rats reduced postprandial
`insulin levels (105), reduced insulin secretory response, and
`increased blood glucose after intraduodenal glucose infu-
`sion (106), thereby providing important evidence that GLP-1
`is a physiologically relevant incretin in vivo. Infusion of
`exendin(9-39) in baboons increased fasting levels of glucose
`and glucagon and increased postprandial glycemic excur-
`
`DIABETES, VOL. 47, FEBRUARY 1998
`
`Page 4 of 11
`
`

`

`D.J. DRUCKER
`
`sion with reduction of postprandial insulin secretion (107).
`The postprandial glycemic excursion was also increased
`after infusion of a GLP-1-specific monoclonal antibody (107).
`Exendin(9-39) blocks the extrapancreatic effects of GLP-1 in
`the cardiovascular system, liver, and muscle (108,109), and
`functions as an antagonist of the brain GLP-1 receptor,
`inhibiting the effects of ICV GLP-1 on food and water intake
`(99,103), and potentiating the stimulatory actions of neu-
`ropeptide Y on food intake (99).
`Despite experimental evidence that exendin(9-39) may be
`a relatively specific GLP-1 receptor antagonist, experiments
`with the cloned rat and human GIP receptors have demon-
`strated that relatively high concentrations of exendin(9-39)
`may also function as a GIP receptor antagonist (110,111).
`The results of recent experiments suggest that truncation of
`the first 3-7 amino acids of exendin-4 may produce exendin
`analogs that are up to 10-fold more potent as GLP-1 antago-
`nists than exendin(9-39) (112); however, the relative specificity
`of these analogs for the GLP-1 versus the GIP receptor has not
`yet been reported.
`The GLP-1 receptor - /- mouse. Targeted disruption of the
`gene encoding the pancreatic islet GLP-1 receptor (GLP-1R) in
`embryonic stem cells followed by the derivation oftransgenic
`GLP-1R -/- mice has permitted the analysis of the role of GLP-
`1 in both glucose control and appetite regulation in vivo. GLP-
`1R -/- mice exhibit mild fasting hyperglycemia and glucose
`intolerance after oral glucose challenge (113). The abnormal
`glycemic excursion after oral glucose loading was associated
`with a reduction in glucose-stimulated insulin secretion, con-
`sistent with an essential role for GLP-1 signaling in the regu-
`lation of glucose-dependent insulin secretion (113). Remark-
`ably, GLP-1R -/- mice also exhibit abnormal glycemic excur-
`sion after intraperitoneal glucose challenge, suggesting that
`intact GLP-1 signaling is important for the handling of a glucose
`load, independent of the site of glucose entry.
`Despite evidence that ICV GLP-1 is a potent inhibitor of
`food intake, analysis of body weight in GLP-1R -/- mice (up
`to age 18 months; D.J.D., unpublished observations) did not
`demonstrate any significant changes in body mass com-
`pared with age- and sex-matched control mice (113). Dis-
`ruption of GLP-1 signaling in the brain does not appear to
`be associated with chronically increased food intake, and
`GLP-1 receptor -/- mice do not eat more than control mice
`in short-term feeding studies (113). Furthermore, despite
`evidence for significantly increased leptin sensitivity in the
`GLP-1R -/- islet, the inhibition of food intake after ICV
`leptin appears relatively normal in the GLP-1 R -/- mouse
`(114). No GLP-1 binding sites are detectable in the CNS of
`GLP-1R -/- mice, consistent with the presence of a single
`brain GLP-1 receptor. Taken together, studies with the
`GLP-1R -/- mouse support an essential role for GLP-1 in the
`regulation of glycemia and glucose-stimulated insulin
`secretion; however, the available data suggest that GLP-1
`signaling may not be essential for regulation of satiety or
`body weight. The CNS results may be explained by genetic
`redundancy, in that multiple compensatory mechanisms
`likely exist for central regulation of food intake and body
`weight (115). Furthermore, the possibility that disruption
`of GLP-1 signaling from birth may be associated with sub-
`tle developmental abnormalities in the CNS that may influ-
`ence the regulation of feeding and body weight cannot be
`excluded.
`
`DIABETES, VOL. 47, FEBRUAEY 1998
`
`GLP-1 IN HUMAN STUDIES
`Normal human subjects. GLP-1, either as the (7-36)amide
`or in the 7-37 forms (27), stimulates insulin secretion,
`inhibits glucagon secretion, and lowers blood glucose in
`humans in the fasting or postprandial state (116,117). Infu-
`sion of GLP-1 in normal human volunteers delays gastric
`emptying (72), although nausea and vomiting, likely due to
`inhibition of gastric emptying, have been observed with
`higher dosages of GLP-1 (117). GLP-1 may also regulate gly-
`cemia by modulating hepatic glucose production, predomi-
`nantly through its effects on levels of circulating insulin and
`glucagon (118,119). The mechanisms underlying the insulin-
`independent effects of GLP-1 facilitating glucose disposal
`remain unclear. GLP-1 infusion increased glucose disposal
`and glucose effectiveness in short-term (4-h) studies in nor-
`mal subjects (120,121), but had no effect on glucose dis-
`posal (independent from the effect of insulin) after intra-
`venous glucose loading (119). Furthermore, no effect of
`GLP-1 on insulin sensitivity was observed during a 3-h
`hyperinsulinemic, euglycemic clamp (122) or after oral fat
`ingestion or intravenous glucose loading (121). Although
`the majority of human studies deliver GLP-1 by intravenous
`or subcutaneous injection, a recent promising study demon-
`strated that formulation of GLP-1 as a buccal tablet pro-
`motes transmucosal absorption, resulting in increased lev-
`els of insulin and decreased glucagon and glucose in healthy
`human volunteers (123).
`NIDDM patients. The demonstration that GLP-1 exhibited
`considerably greater potency compared with that of GIP as
`a glucose-dependent stimulator of insulin secretion in diabetic
`subjects has stimulated considerable interest in the use of
`GLP-1 for the treatment of NIDDM (124). Although both GLP-
`1 and GIP stimulate insulin secretion, GLP-1, but not GIP,
`inhibits gastric emptying and lowers circulating glucagon in
`NIDDM patients (124,125). GLP-1 infusion normalized fasting
`hyperglycemia in NIDDM patients with poor glycemic control
`(126) and improved basal and glucose- and arginine-stimu-
`lated insulin secretion in NIDDM subjects (127). Several
`short-term studies in NIDDM patients have demonstrated
`that GLP-1, whether administered by intravenous infusion or
`subcutaneous injection, normalizes both fasting and post-
`prandial glycemia (128), predominantly by enhancing fJ-cell
`function and inhibiting both gastric emptying and glucagon
`secretion (129-131). Additional evidence for a beneficial
`effect of GLP-1 on islet function in NIDDM patients derives
`from studies demonstrating that the glucose-lowering effect
`of GLP-1 is enhanced by the sulfonylurea glibenclamide in
`patients previously resistant to glibenclamide alone (64).
`A recent study has examined the effect of more prolonged
`GLP-1 treatment on glucose control in NIDDM patients
`receiving intensive insulin therapy for 1 week followed by
`either an additional 7 days on insulin alone or insulin plus
`GLP-1 at meals. The GLP-1-treated group required less
`exogenous insulin and exhibited a reduction in postprandial
`hyperglycemia but increased preprandial glycemia, possibly
`due to the short duration of action of GLP-1 (132). GLP-1 treat-
`ment also increased LDL particle diameter and reduced both
`lipoprotein lipase and hepatic lipase activity. A second study
`reported the results of a 3-week, double-blind crossover trial
`of GLP-1 or saline three times a day before meals in five
`NIDDM patients with poor glycemic control. GLP-1 treat-
`ment lowered postprandial glucagon levels and improved
`
`163
`
`Page 5 of 11
`
`

`

`GLUCAGON-LIKE PEPTIDES
`
`postprandial glycemic control, despite no significant increase
`in postprandial insulin levels in these patients (133).
`IDDM patients. GLP-1 lowered postprandial blood glucose
`and the meal-related insulin requirement in IDDM patients in
`association with a reduction in circulating glucagon and
`somatostatin (116). These observations emphasized the
`potential importance of GLP-1 for lowering blood glucose
`independent of its actions on the pancreatic (3-cell. The glu-
`cose-lowering properties of GLP-1 in IDDM patients after
`meal ingestion are likely due in large part to a delay of gas-
`tric emptying and inhibition of glucagon secretion (134).
`Administration of lower dosages of GLP-1 to IDDM patients
`decreased postprandial glycemic excursion but not glucagon
`levels, suggesting that delayed gastric emptying is a major con-
`tributor to the decreased blood glucose observed in these
`studies (135). In contrast, inhibition of glucagon secretion is
`the primary determinant of the reduction in fasting glycemia
`observed in IDDM patients infused with GLP-1 (136).
`Novel glucagon-like peptides. Peptides originally isolated
`from the venom of the Gila monster lizard H. suspectum or
`H. horridum increased cyclic AMP and amylase secretion
`from dispersed pancreatic acini in vitro. Screening of lizard
`venom for the presence of peptides with amino terminal his-
`tidine residues culminated in the isolation of two peptides,
`designated exendin-3 (137) and exendin-4 (138). Both peptides
`exhibit approximately 50% amino acid identity to mammalian
`GLP-1 (Fig. 2), but are encoded by unique exendin genes
`with different patterns of tissue-specific expression in the
`lizard (18). Both exendin peptides increase cAMP in dis-
`persed pancreatic acini, but only exendin-3 increases amylase
`secretion; exendin-4 does not most likely because exendin-4
`does not bind to the pancreatic vasoactive intestinal peptide
`(VIP) receptor (138). Exendin-4 binds to the GLP-1 receptor
`and stimulates glucose-dependent insulin secretion in islet
`cells in vitro (46,104) and in animal studies in vivo (106).
`Exendin-4 also mimics the majority of peripheral actions of
`GLP-1 in the cardiovascular system, stomach, and brain
`(139). Intriguingly, GLP-1 also binds to the putative exendin
`receptor and increases acinar cAMP; however, the identity of
`the exendin receptor expressed on pancreatic acinar cells
`remains unclear (140).
`The first three amino acids at the NH2-terminus of

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