throbber
R e v i e w s / C o m m e n t a r i e s / P o s i t i o n S t a t e m e n t s
`R E V I E W A R T I C L E
`
`Enhancing Incretin Action for the
`Treatment of Type 2 Diabetes
`
`DANIEL J. DRUCKER, MD
`
`OBJECTIVE — To examine the mechanisms of action, therapeutic potential, and challenges
`inherent in the use of incretin peptides and dipeptidyl peptidase-IV (DPP-IV) inhibitors for the
`treatment of type 2 diabetes.
`
`RESEARCH DESIGN AND METHODS — The scientific literature describing the bio-
`logical importance of incretin peptides and DPP-IV inhibitors in the control of glucose homeosta-
`sis has been reviewed, with an emphasis on mechanisms of action, experimental diabetes, human
`physiological experiments, and short-term clinical studies in normal and diabetic human sub-
`jects.
`
`RESULTS — Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide
`(GIP) exert important effects on ␤-cells to stimulate glucose-dependent insulin secretion. Both
`peptides also regulate ␤-cell proliferation and cytoprotection. GLP-1, but not GIP, inhibits
`gastric emptying, glucagon secretion, and food intake. The glucose-lowering actions of GLP-1,
`but not GIP, are preserved in subjects with type 2 diabetes. However, native GLP-1 is rapidly
`degraded by DPP-IV after parenteral administration; hence, degradation-resistant, long-acting
`GLP-1 receptor (GLP-1R) agonists are preferable agents for the chronic treatment of human
`diabetes. Alternatively, inhibition of DPP-IV–mediated incretin degradation represents a com-
`plementary therapeutic approach, as orally available DPP-IV inhibitors have been shown to
`lower glucose in experimental diabetic models and human subjects with type 2 diabetes.
`
`CONCLUSIONS — GLP-1R agonists and DPP-IV inhibitors have shown promising results
`in clinical trials for the treatment of type 2 diabetes. The need for daily injections of potentially
`immunogenic GLP-1– derived peptides and the potential for unanticipated side effects with
`chronic use of DPP-IV inhibitors will require ongoing scrutiny of the risk-benefit ratio for these
`new therapies as they are evaluated in the clinic.
`
`Diabetes Care 26:2929 –2940, 2003
`
`A fter meal ingestion, nutrient entry
`
`into the stomach and transit
`through the proximal gastrointesti-
`nal (GI) tract stimulates activation of neu-
`ral and hormonal signals that control
`gastric emptying and gut motility, nutri-
`ent absorption, and hormonal regulation
`of energy disposal and storage. The mu-
`
`cosal epithelium of the GI tract is one of
`the earliest integrators of information rel-
`evant to digestion and assimilation of
`nutrient loads. Highly specialized en-
`teroendocrine cells dispersed along the
`length of the GI tract play an important
`role in controlling the rate of gastric emp-
`tying and small bowel motility, pancreatic
`
`● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ●
`
`From the Banting and Best Diabetes Centre, Department of Medicine, Toronto General Hospital, University
`of Toronto, Ontario, Canada.
`Address correspondence and reprint requests to Daniel J. Drucker, MD, Banting and Best Diabetes Centre,
`University of Toronto, Toronto General Hospital, 200 Elizabeth St., MBRW 4R-402, Toronto, Ontario,
`Canada M5G 2C4. E-mail: d.drucker@utoronto.ca.
`Received for publication 5 May 2003 and accepted in revised form 30 June 2003.
`D.J.D. is on the scientific advisory board for Amylin Pharmaceuticals, Conjuchem, and Transition Ther-
`apeutics and a consultant for Merck, Forest Labs, Bristol Myers Squibb, Triad Pharmaceuticals, Aventis,
`Novartis, Amylin Pharmaceuticals, and Conjuchem.
`Abbreviations: DPP, dipeptidyl peptidase IV; GI, gastrointestinal; GIP, glucose-dependent insulinotropic
`peptide; GIPR, GIP receptor; GLP, glucagon-like peptide; GLP-1R, GLP-1 receptor.
`A table elsewhere in this issue shows conventional and Syste`me International (SI) units and conversion
`factors for many substances.
`© 2003 by the American Diabetes Association.
`
`enzyme secretion, and the growth and
`differentiated absorptive function of the
`small and large bowel epithelium. The
`aim of this review is to examine our cur-
`rent understanding of the physiological
`actions of two gut hormones, glucagon-
`like peptide (GLP)-1 and glucose-
`dependent insulinotropic polypeptide
`(GIP), with an emphasis on the biological
`importance and pharmaceutical potential
`of these peptides for the treatment of type
`2 diabetes.
`
`INTRODUCTION TO THE
`INCRETIN CONCEPT — The devel-
`opment and application of the insulin ra-
`dioimmunoassay to clinical investigation
`has permitted the assessment of ␤-cell se-
`cretory function after meal ingestion in
`normal and diabetic subjects. The obser-
`vation that food ingestion or enteral glu-
`cose administration provoked a greater
`stimulation of insulin release compared
`with similar amounts of energy (glucose)
`infused intravenously (1,2) led to the de-
`velopment of the incretin concept. Hence,
`it was postulated that gut-derived signals
`stimulated by oral nutrient ingestion rep-
`resent potent insulin secretagogues re-
`sponsible for the augmentation of insulin
`release when energy is administered via
`the gut versus the parenteral route (3).
`Although several neurotransmitters and
`gut hormones possess incretin-like activ-
`ity, the considerable evidence from im-
`munoneutralization, antagonist, and
`knockout studies suggests that GIP and
`GLP-1 represent the dominant peptides
`responsible for the majority of nutrient-
`stimulated insulin secretion. The observa-
`tion that patients with type 2 diabetes
`exhibit a significant reduction in the mag-
`nitude of meal-stimulated insulin release
`underlies the interest in determining
`whether defective incretin release or resis-
`tance to incretin action contributes to the
`pathophysiology of ␤-cell dysfunction in
`diabetic subjects.
`
`INCRETIN SYNTHESIS,
`SECRETION, AND
`DEGRADATION — GIP and GLP-1
`are members of the glucagon peptide su-
`perfamily and share considerable amino
`
`DIABETES CARE, VOLUME 26, NUMBER 10, OCTOBER 2003
`
`2929
`
`MPI EXHIBIT 1023 PAGE 1
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1023, p. 1 of 12
`
`

`

`Incretin action in type 2 diabetes
`
`subjects using antisera capable of discrim-
`inating the full-length from the NH2-
`terminally cleaved peptides. The t1/2 of
`infused GIP is ⬃7 and 5 min in normal
`and diabetic human subjects, respectively
`(14). In contrast, the t1/2 of exogenously
`infused intact GLP-1 is considerably
`shorter (13), with intravenously adminis-
`tered GLP-1 eliminated with a half-life of
`⬃2 min in both normal and obese dia-
`betic human subjects (17). Although the
`NH2-terminally truncated peptides
`GIP(3-42) and GLP-1(9-36)amide func-
`tion as weak antagonists of their respec-
`tive receptors (18,19), there is little
`evidence that these truncated peptides ex-
`ert physiologically important actions in
`human subjects in vivo. Despite observa-
`tions that GLP-1(9-36)amide may func-
`tion as an activator of insulin-independent
`glucose clearance in pigs (20), this pep-
`tide does not exert significant glucose-
`lowering properties in human subjects
`(21).
`Circulating levels of GIP(1-42) are
`normal or slightly increased in type 2 di-
`abetic subjects in the basal or postpran-
`dial states (22). In contrast, subjects with
`diabetes or impaired glucose tolerance ex-
`hibit modest but significant reductions in
`levels of meal-stimulated circulating
`GLP-1 (22,23). Furthermore, meal-
`induced increases in GIP and GLP-1 se-
`cretion are inversely correlated with the
`extent of insulin resistance detected in
`human subjects (24). The lower levels of
`circulating GLP-1 detected in diabetic
`subjects are not attributable to altered
`GLP-1 clearance (17). Whether levels of
`meal-stimulated GLP-1 may be restored
`toward normal with improved control of
`diabetes remains unknown.
`
`GIP ACTION: INSIGHTS
`FROM PRECLINICAL AND
`HUMAN STUDIES — GIP was orig-
`inally observed to inhibit gastric acid se-
`cretion (gastric inhibitory polypeptide),
`predominantly at supraphysiological dos-
`ages. Subsequent studies have demon-
`strated potent glucose-dependent insulin
`stimulatory effects from GIP administra-
`tion in dogs and rodents. GIP also regu-
`lates fat metabolism in adipocytes,
`including stimulation of lipoprotein
`lipase activity, fatty acid incorporation,
`and fatty acid synthesis (25). Unlike
`GLP-1, GIP does not inhibit glucagon se-
`cretion or gastric emptying. GIP does pro-
`mote ␤-cell proliferation and cell survival
`
`Figure 1—Structure of preproglucagon and preproGIP encoding GLP-1 and GIP, respectively, is
`shown. The arrow designates the position of the DPP-IV–mediated cleavage after the position 2
`alanine residue. GRPP, glicentin-related pancreatic peptide; IP, intervening peptide.
`
`acid identity. GIP is a single 42⫺amino
`acid peptide encoded within a larger
`153⫺amino acid precursor (Fig. 1) (4).
`GIP-secreting enteroendocrine K-cells are
`concentrated in the duodenum and prox-
`imal jejunum; hence, these cells are ana-
`tomically situated in an ideal location for
`sensing and responding to nutrient inges-
`tion. GLP-1 is derived from a larger pro-
`glucagon precursor that encodes not only
`GLP-1 but also the related proglucagon-
`derived peptides glucagon, GLP-2, oxyn-
`tomodulin, and glicentin (Fig. 1) (5). The
`two forms of GLP-1 secreted after meal
`ingestion, GLP-1(7-37) and GLP-1(7-
`36)amide differ by a single amino acid.
`Both peptides are equipotent and exhibit
`identical plasma half-lives and biological
`activities acting through the same recep-
`tor (6,7); however, the majority (⬃80%)
`of circulating active GLP-1 appears to be
`GLP-1(7-36)amide (8). In contrast to the
`more proximal location of GIP-producing
`K-cells, the majority of GLP-1 is synthe-
`sized within L-cells located predomi-
`nantly in the ileum and colon, although
`GLP-1–producing L-cells have also been
`identified more proximally in the duode-
`num and jejunum. Despite the more dis-
`tal location of most L-cells, circulating
`levels of GLP-1 also increase rapidly
`within minutes of food ingestion. Hence,
`GLP-1 secretion from the distal gut is con-
`trolled by both neural and endocrine sig-
`nals initiated by nutrient entry in the
`proximal GI tract, as well as by subse-
`quent direct contact of open-type L-cells
`with digested nutrients. Ingestion of a
`mixed meal or a meal enriched with spe-
`cific fats and complex carbohydrates is
`
`particularly effective in stimulating GIP
`and GLP-1 release in human subjects
`(9,10). Although the vagal nerve, via M1
`muscarinic receptors, and several neu-
`roendocrine peptides contribute to the
`regulation of GLP-1 release in rodents
`(11,12), the factors responsible for rapid
`nutrient-stimulated GLP-1 release in hu-
`man subjects are largely unknown.
`The levels of total circulating GIP and
`GLP-1 immunoreactivity reflect a combi-
`nation of intact, full-length active and
`NH2-terminally truncated inactive pep-
`tides, with GIP(3-42) and GLP-1(9-
`36)amide contributing to ⬎50% of total
`immunoreactive GIP and GLP-1 in both
`the fasting and the postprandial states
`(13,14). Plasma levels of both GIP and
`GLP-1 immunoreactivity are low in the
`fasting state and rise rapidly within min-
`utes of food ingestion. Initial studies of
`circulating levels of GIP and GLP-1 relied
`principally on radioimmunoassays inca-
`pable of distinguishing the biologically
`active full-length peptides from inactive
`COOH-terminal peptide fragments gen-
`erated as a result of proteolytic cleavage.
`Studies have demonstrated that both GIP
`and GLP-1 were cleaved at the position 2
`alanine by the widely expressed amino-
`peptidase dipeptidyl peptidase IV (DPP-
`IV) (15,16). These findings have
`prompted a reanalysis of the circulating
`molecular forms of GIP and GLP-1 using
`newer radioimmunoassays more specific
`for the full-length bioactive peptides in
`normal and diabetic subjects.
`The disappearance of exogenously
`administered GIP and GLP-1 has been
`studied in normal and diabetic human
`
`2930
`
`DIABETES CARE, VOLUME 26, NUMBER 10, OCTOBER 2003
`
`MPI EXHIBIT 1023 PAGE 2
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1023, p. 2 of 12
`
`

`

`Drucker
`
`in islet cell line studies (26,27); whether
`GIP also induces ␤-cell growth or survival
`in diabetic rodents remains unclear.
`The physiological actions of GIP have
`been deduced using GIP peptide antago-
`nists, GIP receptor antisera, and GIP re-
`ceptor knockout mice. NH2-terminally
`truncated or modified GIP peptides such
`as GIP(6-30)amide, GIP(7-30)amide, or
`(Pro3)GIP block GIP binding to the GIP
`receptor with varying effectiveness, and
`attenuate the insulinotropic effects of ex-
`ogenous GIP in vitro and endogenous GIP
`in vivo (28 –30). Similarly, immunopuri-
`fied antisera against the extracellular do-
`main of the GIP receptor block GIP
`binding and attenuate glucose-dependent
`insulin secretion after oral glucose load-
`ing in rats and mice (31,32). Complemen-
`tary evidence for the incretin-like actions
`of GIP is derived from analysis of GIP
`receptor⫺null mice, which exhibit mild
`glucose intolerance after oral glucose
`loading (33). Surprisingly, GIPR⫺/⫺ mice
`exhibit resistance to diet-induced obesity
`after months of high-fat feeding. More-
`over, the GIPR⫺/⫺ genotype attenuates
`obesity in the ob/ob mouse, possibly be-
`cause of reduced fat storage and altered
`lipid metabolism as a direct result of ab-
`sent GIP receptor (GIPR) action in adipo-
`cytes (34). Whether GIPR action
`significantly modulates adipocyte biol-
`ogy, lipoprotein synthesis, and weight ac-
`cretion in humans is not known.
`In contrast to the potent glucose-
`lowering actions of GIP in normal ro-
`dents, exogenous GIP administration is
`comparatively less insulinotropic in obese
`diabetic rodents. GIP levels are increased
`in some models of experimental rodent
`diabetes, and continuous GIP infusion for
`4 h produces GIPR desensitization in nor-
`mal rats (35). ZDF rats exhibit normal
`levels of GIP, absent insulinotropic re-
`sponses to exogenous GIP and reduced
`expression of the GIPR in isolated islets
`(36). Recent studies with more potent GIP
`analogs engineered for resistance to
`DPP-IV have demonstrated improved in-
`sulinotropic and glucose-lowering prop-
`erties after peptide administration to both
`normal and diabetic rodents (37–39).
`Infusion of porcine or human GIP
`into patients with type 2 diabetes has pro-
`duced variable insulinotropic responses,
`ranging from preserved (40) to attenuated
`or near absent insulin secretion (41– 45).
`The potential for ␤-cell GIP responsivity
`to improve with treatment in type 2 dia-
`
`Figure 2—The major biological actions of GLP-1.
`
`betic subjects is intriguing, but has not
`been extensively examined (46). The GIP
`defect in insulin secretion seems most
`pronounced in the late phase of insulin
`secretion (47). Moreover, ⬃50% of nor-
`moglycemic first-degree relatives of type
`2 diabetic subjects exhibit reduced insu-
`lin secretion after exogenous GIP infusion
`(48). Hence the reduced insulinotropic
`action of GIP in diabetes likely reflects a
`combination of genetic and acquired de-
`fects. Whether the pancreatic effects of
`GIP on ␤-cell proliferation and survival
`are also diminished in experimental or
`clinical diabetes is not known.
`
`GLP-1 PRECLINICAL
`STUDIES AND
`PHYSIOLOGICAL ACTIONS —
`Original observations elucidating a role
`for GLP-1 in the potentiation of glucose-
`dependent insulin secretion (49 –51) and
`insulin gene expression (52) were fol-
`lowed by experiments demonstrating that
`GLP-1 also inhibits glucagon secretion
`(53,54) and gastric emptying (55) (Fig.
`2). Acute intracerebroventricular (56) in-
`jection of GLP-1 or GLP-1 receptor (GLP-
`1R) agonists produces transient reduction
`in food intake, whereas more prolonged
`intracerebroventricular or peripheral
`GLP-1R agonist administration is associ-
`ated with weight loss in some (57– 60),
`but not all (61) studies. GLP-1 actions on
`food intake appear related in part to over-
`lapping actions on central nervous system
`aversive signaling pathways, which re-
`mains a topic of intense interest (62– 66).
`In contrast to GIP, the spectrum of actions
`
`delineated for GLP-1 that promote glu-
`cose lowering (regulation of insulin and
`glucagon secretion, inhibition of gastric
`emptying, and reduction of food intake)
`appear comparable in diabetic versus
`nondiabetic animals of various ages.
`GLP-1 exerts actions on ␤-cells inde-
`pendent of acute stimulation of insulin se-
`cretion. Incubation of isolated rat islet
`cells with GLP-1 recruited nonresponsive
`glucose-resistant ␤-cells to a functional
`state of glucose-responsive insulin secre-
`tion, designated glucose competence
`(67,68). GLP-1R agonists also promote
`insulin biosynthesis, ␤-cell proliferation,
`and survival (69 –71), and stimulate dif-
`ferentiation of exocrine cells or islet pre-
`cursors toward a more differentiated
`␤-cell phenotype (72–74). The GLP-1R–
`dependent augmentation of ␤-cell mass
`has been demonstrated in diverse experi-
`mental models, including neonatal rats
`administered streptozotocin and ex-
`endin-4 (75) and normal Wistar rats ages
`6 and 22 months infused with native
`GLP-1 for 5 days (76). Similarly, GLP-1R
`agonists promote ␤-cell proliferation and
`expansion of functional islet mass after
`partial pancreatectomy in rats aged 4 –5
`weeks (69) or in neonatal rat pups sub-
`jected to experimental
`intrauterine
`growth retardation (77). The expansion
`of ␤-cell mass after GLP-1R agonist ad-
`ministration prevents or delays the occur-
`rence of diabetes in db/db mice (78) and
`GK diabetes-prone rats (79). Further-
`more, the induction of islet proliferation
`after GLP-1R activation has been seen
`with a broad range of GLP-1R agonists,
`
`DIABETES CARE, VOLUME 26, NUMBER 10, OCTOBER 2003
`
`2931
`
`MPI EXHIBIT 1023 PAGE 3
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1023, p. 3 of 12
`
`

`

`Incretin action in type 2 diabetes
`
`including native GLP-1 (76,79,80), ex-
`endin-4, NN2211 (81), and CJC-1131
`(82).
`GLP-1R agonists also activate anti-
`apoptotic pathways coupled to a reduc-
`tion in ␤-cell death. db/db mice treated
`with exendin-4 for 2 weeks exhibited de-
`creased numbers of apoptotic ␤-cells, re-
`duced pancreatic caspase-3 activation,
`and increased Akt1 expression (78). Re-
`duced islet apoptosis has been observed
`in GLP-1–treated Zucker diabetic rats
`(83) and in exendin-4 –treated mice after
`streptozotocin-induced ␤-cell injury
`(70). The anti-apoptotic actions of
`GLP-1R agonists are likely direct, as
`GLP-1 reduced peroxide-induced apo-
`ptosis in Min6 insulinoma cells (84) and
`exendin-4 significantly attenuated cyto-
`kine-induced apoptosis in cultures of pu-
`rified rat ␤-cells (70). Hence, the GLP-
`1R– dependent activation of both
`proliferative and anti-apoptotic pathways
`in the pancreas provides complementary
`mechanisms for preserving and enhanc-
`ing functional ␤-cell mass.
`The physiological
`importance of
`GLP-1 action has been studied using
`GLP-1R antagonists. Infusion of the pep-
`tide exendin(9-39) into rats, mice, ba-
`boons, and humans produces an increase
`in fasting glucose and glycemic excursion
`after oral glucose loading in association
`with reduced levels of circulating insulin
`(32,85– 87). Exendin(9-39) also pro-
`duces abnormal glycemic excursion after
`nonenteral glucose loading in mice (32).
`These findings illustrate that transient
`disruption of GLP-1 action consistently
`perturbs the incretin and nonincretin ac-
`tions of GLP-1 on glucoregulation. Acute
`intracerebroventricular injection of ex-
`endin(9-39) increases food intake in sati-
`ated rats (56), whereas repeated daily
`intracerebroventricular administration of
`exendin(9-39) increases food intake and
`weight gain (57). Similarly, acute ex-
`endin(9-39) administration increases gas-
`tric emptying after glucose ingestion in
`fistulized rats (88). Comparable studies
`with exendin(9-39) in humans have dem-
`onstrated the essential role of GLP-1 ac-
`tion for glucose control via regulation of
`glucagon and insulin secretion (89,90).
`Hence, the majority of actions observed
`after exogenous administration of GLP-
`1R agonists are also physiologically essen-
`tial, as revealed by acute interruption of
`GLP-1 action.
`Genetic disruption of GLP-1R expres-
`
`sion in mice has produced comparable in-
`sights into the physiological importance
`of GLP-1 action. GLP-1R⫺/⫺ mice exhibit
`abnormal glucose tolerance after both
`oral and intraperitoneal glucose challenge
`in association with diminished glucose-
`stimulated insulin secretion. In contrast,
`insulin sensitivity and the glucagon re-
`sponse to glucose loading or hypoglyce-
`mia are normal in the absence of GLP-1R
`signaling (91). Consistent with the car-
`diovascular effects of GLP-1 in rodents,
`GLP-1R⫺/⫺ mice exhibit defective cardio-
`vascular responses to stress (92). Despite
`the potential importance of GLP-1R cir-
`cuits for transducing the anorectic action
`of leptin (93), GLP-1R⫺/⫺ mice retain
`normal to enhanced leptin sensitivity
`(94,95). Similarly, food intake and body
`weight are not significantly perturbed in
`GLP-1R⫺/⫺ mice in the CD1 genetic
`background (96,97). In contrast, GLP-
`1R⫺/⫺ mice manifest subtle but detect-
`able abnormalities in islet number and
`size (98) and exhibit a defective ␤-cell re-
`generative response to partial pancreatec-
`tomy (99). Hence, GLP-1R actions are
`physiologically important for the growth
`and adaptive regeneration of murine
`␤-cells.
`
`GLP-1R agonists and experimental
`models of diabetes
`The glucose-lowering action of GLP-1 de-
`lineated in nondiabetic animals has been
`demonstrated in multiple models of ex-
`perimental diabetes. A 48-h infusion of
`native GLP-1 lowered blood glucose in
`association with increased levels of circu-
`lating insulin, islet insulin content, and
`insulin mRNA in Wistar rats aged 22
`months (100), and perfused pancreas
`studies have demonstrated GLP-1–
`dependent augmentation of insulin secre-
`tion in ZDF rats of diverse genetic
`backgrounds (101–103). Similarly, ex-
`endin-4 lowered glucose in db/db and
`ob/ob mice, ZDF rats, and diabetic rhesus
`monkeys in acute and chronic experi-
`ments (78,104,105) and the GLP-1 ana-
`log NN211 improved glycemic control in
`pigs, rats, and mice (60,81,106). Remark-
`ably, glucose tolerance remained signifi-
`cantly improved in ZDF rats for weeks
`after a 48-h infusion of native GLP-1
`(107). This “memory effect” for sustained
`improvement of glycemic control was
`also observed in db/db mice after discon-
`tinuation of therapy with CJC-1131, an
`albumin-bound GLP-1R agonist (82).
`
`GLP-1 action in human subjects
`The majority of GLP-1 actions delineated
`in preclinical experiments have also been
`demonstrated in human studies. Infusion
`of GLP-1(7-36)amide into normal human
`subjects stimulated insulin secretion, re-
`duced glucagon secretion, and signifi-
`cantly reduced blood glucose in the
`fasting state after glucose loading or meal
`ingestion (6,50,108). In contrast to GIP,
`the insulinotropic and glucose-lowering
`actions of GLP-1 are preserved in human
`subjects with type 2 diabetes (45,109) in
`both the fasting and the postprandial
`states (7). Similarly, GLP-1 inhibits gas-
`tric acid secretion (110,111) and gastric
`emptying in humans (55) and the GLP-1–
`dependent attenuation of gastric empty-
`ing contributes to decreased glycemic
`excursion and, consequently, reduced
`glucose-stimulated insulin secretion
`(112,113). Consistent with the impor-
`tance of gastric emptying and glucagon
`secretion for glycemic control, GLP-1 also
`lowers blood glucose in type 1 diabetic
`subjects (114 –116). Analogous to studies
`demonstrating the induction of glucose
`competence in rodent ␤-cells, GLP-1 in-
`fusion enhances ␤-cell function and insu-
`lin secretory dynamics in human subjects
`with impaired glucose tolerance or type 2
`diabetes (117–119). GLP-1 may also en-
`hance glucose clearance in humans
`(120,121); however, the majority of these
`actions are likely mediated indirectly
`through effects on insulin and glucagon
`(122–124). Although several reports have
`described the effects of GLP-1 on muscle,
`liver, and fat cells, experimental evidence
`demonstrating expression of the GLP-1
`receptor in these tissues in vivo is lacking.
`Hence, the indirect actions of GLP-1,
`leading to improvement in glycemic con-
`trol and reduction in free fatty acids, may
`explain observations of improved insulin
`sensitivity in GLP-1–treated diabetic sub-
`jects (125).
`The effect of GLP-1 in restoring glu-
`cose competence in rodent islets has
`prompted studies of GLP-1 action and
`␤-cell function in type 2 diabetic patients.
`Insulin–treated diabetic subjects previ-
`ously classified as “sulfonylurea non-
`responders” exhibited ␤-cell GLP-1
`responsivity, with lowering of fasting and
`postprandial glucose in association with
`enhanced insulin secretion (126). Pa-
`tients treated with both GLP-1 and glib-
`enclamide exhibited a greater degree of
`glucose reduction compared with the ef-
`
`2932
`
`DIABETES CARE, VOLUME 26, NUMBER 10, OCTOBER 2003
`
`MPI EXHIBIT 1023 PAGE 4
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1023, p. 4 of 12
`
`

`

`fect of either agent alone (127). Similarly,
`the combination of GLP-1 and metformin
`was shown in a 48-h crossover study to be
`more effective for lowering blood glucose
`than monotherapy with either agent alone
`(128).
`The GLP-1– dependent suppression
`of glucagon secretion raises the possibility
`that GLP-1 therapy will be associated with
`an increased risk of hypoglycemia and po-
`tentially defective counterregulation if
`glucagon secretion remains suppressed in
`the face of GLP-1–linked hypoglycemia.
`Rapid gastric emptying may be associated
`with enhanced GLP-1 release and an in-
`creased risk of hypoglycemia in postgas-
`trectomy patients (129). Similarly, acute
`administration of GLP-1 (80 nmol) to
`nondiabetic subjects in the fasted state
`produced mild relative hypoglycemia in
`some subjects (mean glucose ⬃3.5
`mmol/l) (130). Nevertheless, appropriate
`glucagon responses to hypoglycemia do
`not appear to be blunted in GLP-1–
`treated subjects (130), and GLP-1 in-
`fusion does not impair normal counter-
`regulatory responses to hypoglycemia in
`healthy human subjects (131). Hence, the
`risk of hypoglycemia seems modest in
`type 2 diabetic subjects treated with
`GLP-1R agonists alone.
`The demonstration that both intrace-
`rebroventricular and peripheral adminis-
`tration of GLP-1R agonists induces
`weight loss in preclinical experiments has
`fostered interest in the potential actions of
`these agents to diminish appetite and re-
`duce weight gain in overweight human
`subjects. The majority of human studies
`have examined appetite and food inges-
`tion over short (24-h) time periods after
`single-dose injection or continuous infu-
`sion of GLP-1. Small but statistically sig-
`nificant reductions in appetite and meal
`ingestion have been recorded in studies of
`normal, obese, and diabetic GLP-1–
`treated subjects (132–136). A meta-
`analysis of available data from 115
`subjects demonstrated significant GLP-1–
`dependent reductions in energy con-
`sumption in lean and overweight subjects
`(137). The acute reduction in food con-
`sumption and inhibition of gastric emp-
`tying has been detected even with
`physiological increases in levels of circu-
`lating GLP-1 (136). Administration of
`GLP-1 via continuous subcutaneous infu-
`sion for 6 weeks to obese diabetic subjects
`was associated with reduced appetite and
`a small but significant mean 1.9-kg
`
`weight loss (125). Hence, GLP-1 therapy
`in human subjects appears associated
`with prevention of weight gain or modest
`weight loss; however, long-term data are
`not yet available.
`Although single or repeated subcuta-
`neous injections of native GLP-1 decrease
`blood glucose in human subjects
`(138,139), the glucose-lowering effects
`are transient and no longer evident 1–2 h
`after peptide injection (140,141). Fur-
`thermore, continuous enhancement of
`GLP-1 action for 24 h/day appears supe-
`rior for glucose control compared with
`peptide infusion for 16 h (142). Contin-
`uous intravenous or subcutaneous infu-
`sion of GLP-1 in short- and long-term
`studies has been shown to be highly effec-
`tive in lowering blood glucose in diabetic
`subjects (125,143,144), but this intensive
`and expensive approach has major limita-
`tions for the treatment of large numbers of
`diabetic patients. The rapid degradation
`and clearance of native endogenous and
`exogenously administered GLP-1 (145)
`have spurred the clinical development of
`degradation-resistant GLP-1 analogs with
`longer durations of action in vivo.
`Exendin-4 is a naturally occurring
`39⫺amino acid GLP-1R agonist isolated
`from the salivary gland venom of the liz-
`ard Heloderma suspectum (146). Ex-
`endin-4 exhibits 53% amino acid identity
`to mammalian GLP-1 (146,147), yet
`binds to and activates the GLP-1 receptor.
`Furthermore, exendin-4 is highly resis-
`tant to the proteolytic activity of DPP-IV
`and exhibits a longer duration of action in
`vivo. Intravenous infusion of exendin-4
`lowered fasting and postprandial blood
`glucose in normal healthy volunteers and
`was associated with a 19% reduction in
`calorie consumption assessed during a
`single test meal (148). Exendin-4 exerted
`similar effects on insulin secretion after
`acute intravenous infusion in diabetic
`subjects (149), and subcutaneous daily
`administration of exendin-4 to subjects
`with type 2 diabetes significantly reduced
`blood glucose and HbA1c (a decline from
`9.1 to 8.3%) over a 1-month treatment
`period (150). Exendin-4 has been evalu-
`ated in eight phase 2 trials in 323 individ-
`uals with type 2 diabetes who received
`dosages of 0.05–2.0 ␮g/kg subcutane-
`ously. Nausea and vomiting were the
`principal side effects observed (151). A
`4-week treatment period produced a sig-
`nificant reduction in HbA1c levels, with
`sustained reduction in postprandial gly-
`
`Drucker
`
`cemia maintained over the 28-day treat-
`ment period.
`Exendin-4 treatment (0.08 ␮g/kg
`s.c., b.i.d. or t.i.d.) over 1 month was
`evaluated in 109 patients treated with sul-
`fonylureas or metformin, alone or in com-
`bination. The treatment was generally
`well tolerated, with three subjects with-
`drawing in the first 12 days because of
`nausea. At the end of the study period, a
`significant reduction was observed in lev-
`els of serum fructosamine, HbA1c, and
`mean postprandial glucose, but no signif-
`icant change was noted in body weight or
`serum lipids (152). Antibodies against ex-
`endin-4 were detected in 19% of treated
`subjects; however, the antibodies did not
`affect treatment responses. In all, 15% of
`patients experienced hypoglycemia; all of
`these subjects received sulfonylureas plus
`exendin-4 (152). Exendin-4, recently re-
`named exenatide, is currently being eval-
`uated for the treatment of type 2 diabetes
`in phase 3 trials in combination with met-
`formin, sulfonylurea agents, or both.
`NN2211 (liraglutide) is a fatty acid–
`linked DPP-IV–resistant derivative of
`GLP-1 designed for subcutaneous admin-
`istration that exhibits a pharmacokinetic
`profile compatible with once-daily injec-
`tion (153). NN2211 reduced fasting and
`postprandial glycemia in diabetic subjects
`after a single 10 ␮g/kg subcutaneous in-
`jection at 11:00 P.M., in association with
`inhibition of gastric emptying and re-
`duced levels of circulating glucagon (154).
`NN2211 has been tested in phase 2 clin-
`ical trials. Additional approaches for pro-
`longing the duration of action of GLP-1
`derivatives include the use of albumin-
`bound GLP-1 molecules (82) and sus-
`tained release exendin-4 preparations;
`however, human data with these pharma-
`ceutical approaches is currently limited.
`
`Inhibition of DPP-IV for the
`treatment of type 2 diabetes
`The observation that GLP-1 and GIP are
`rapidly cleaved at the position 2 alanine
`leading to inactivation of their biological
`activity (15,16) has fostered interest in the
`development of inhibitors of DPP-IV, the
`principal enzyme responsible for incretin
`inactivation (155,156). DPP-IV is but one
`member of a large family of related en-
`zymes with overlapping enzyme specific-
`ity; however, adenosine deaminase
`affinity chromatography that specifically
`binds DPP-IV removes 95% of DPP-IV–
`
`DIABETES CARE, VOLUME 26, NUMBER 10, OCTOBER 2003
`
`2933
`
`MPI EXHIBIT 1023 PAGE 5
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1023, p. 5 of 12
`
`

`

`Incretin action in type 2 diabetes
`
`Table 1—Properties and biological actions of GIP and GLP-1
`
`GIP
`
`GLP-1
`
`42–Amino acid peptide
`Released from duodenum
`NH2-terminal inactivation by DPP-IV
`Stimulates insulin secretion
`Minimal effect on gastric emptying
`No effect on glucagon secretion
`No regulation of satiety and body weight
`Promotes expansion of ␤-cell mass
`Normal GIP secretion in diabetic subjects
`Defective GIP response in type 2 diabetes
`
`30/31–Amino acid peptide
`Released from distal small bowel and colon
`NH2-terminal inactivation by DPP-IV
`Stimulates insulin secretion
`Inhibits gastric emptying
`Inhibits glucagon secretion
`Inhibits food intake and weight gain
`Promotes expansion of ␤-cell mass
`Reduced GLP-1 secretion in dia

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