throbber
Regulatory Peptides 117 (2004) 77 – 88
`
`Review
`
`www.elsevier.com/locate/regpep
`
`Pharmacology of exenatide (synthetic exendin-4): a potential therapeutic
`for improved glycemic control of type 2 diabetes
`
`Loretta L. Nielsen 1, Andrew A. Young, David G. Parkes*
`
`Amylin Pharmaceuticals Inc., 9360 Town Centre Dr. Ste 110, San Diego, CA 92121, USA
`
`Received 12 June 2003; received in revised form 7 October 2003; accepted 14 October 2003
`
`Abstract
`
`to
`Exenatide (synthetic exendin-4), glucagon-like peptide-1 (GLP-1), and GLP-1 analogues have actions with the potential
`significantly improve glycemic control in patients with diabetes. Evidence suggests that these agents use a combination of mechanisms
`which may include glucose-dependent stimulation of insulin secretion, suppression of glucagon secretion, enhancement of h-cell mass,
`slowing of gastric emptying, inhibition of food intake, and modulation of glucose trafficking in peripheral tissues. The short in vivo
`half-life of GLP-1 has proven a significant barrier to continued clinical development, and the focus of current clinical studies has shifted
`to agents with longer and more potent in vivo activity. This review examines recent exendin-4 pharmacology in the context of several
`known mechanisms of action, and contrasts exendin-4 actions with those of GLP-1 and a GLP-1 analogue. One of the most provocative
`areas of recent research is the finding that exendin-4 enhances h-cell mass, thereby impeding or even reversing disease progression.
`Therefore, a major focus of this is article an examination of the data supporting the concept that exendin-4 and GLP-1 may increase h-
`cell mass via stimulation of h-cell neogenesis, stimulation of h-cell proliferation, and suppression of h-cell apoptosis.
`D 2003 Elsevier B.V. All rights reserved.
`
`Keywords: Pharmacology; Exenatide; Type 2 diabetes
`
`1. Introduction
`
`Exendin-4, the naturally occurring form of exenatide
`(synthetic exendin-4; AC2993), was originally isolated from
`the salivary secretions of the lizard Heloderma suspectum
`(Gila monster; Fig. 1) [1]. In the Gila monster, exendin-4
`circulates after the lizard bites down on its prey (ingestion of a
`meal) and thus represents the first example of an endocrine
`hormone secreted from salivary glands. [2]. It is unknown
`whether exendin-4 has a role in fuel homeostasis in the Gila
`monster [2]. Exendin-4 has a 53% amino acid sequence
`overlap with mammalian glucagon-like peptide-1 (GLP-1).
`In mammals, GLP-1 is processed from the proglucagon gene
`in L-cells in the small intestine [3]. Exendin-4 is transcribed
`from a distinct gene, not the Gila monster homologue of the
`mammalian proglucagon gene from which GLP-1 is
`expressed [4]. In mammals, exendin-4 is resistant to degra-
`dation by dipeptidyl peptidase-IV (DPP-IV) and has a much
`
`* Corresponding author. Tel.: +1-858-642-7290; fax: +1-858-334-1290.
`E-mail addresses: lnielsen@amylin.com (L.L. Nielsen),
`dparkes@amylin.com (D.G. Parkes).
`1 Alternate contact: Tel.: +1-858-642-7203; fax: +1-858-334-1203.
`
`0167-0115/$ - see front matter D 2003 Elsevier B.V. All rights reserved.
`doi:10.1016/j.regpep.2003.10.028
`
`longer plasma half-life than GLP-1, which is degraded by
`DPP-IV with a half-life of less than 2 min [5,6].
`Exendin-4 is not an analogue of GLP-1. In other words,
`the structure of the synthetic exendin-4 peptide (exenatide)
`was not created by sequential modification of the structure
`of GLP-1. However, exendin-4 and GLP-1 do share many
`glucoregulatory actions which may be mediated by the
`known pancreatic GLP-1 receptor [7]. Glucoregulatory
`actions of exendin-4 include glucose-dependent enhance-
`ment of insulin secretion [8 – 11], glucose-dependent sup-
`pression of inappropriately high glucagon secretion [10,12],
`slowing of gastric emptying [10,13] which may be para-
`doxically accelerated in people with diabetes [14], and
`reduction of food intake ([15,16]; Fig. 2). In addition,
`exendin-4 has been shown to promote h-cell proliferation
`and islet neogenesis from precursor cells in both in vitro and
`in vivo models [17 – 19]. These glucoregulatory actions of
`exendin-4, combined with enhanced pharmacokinetics, re-
`sult in very high in vivo potency relative to native GLP-1
`[11,20,21]. The putative mechanisms of action of exendin-4
`are compared and contrasted with the actions of GLP-1 and
`a long-acting GLP-1 analogue in the following sections.
`One of the most provocative areas of recent research is
`
`MPI EXHIBIT 1037 PAGE 1
`
`MPI EXHIBIT 1037 PAGE 1
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 1 of 12
`
`

`

`78
`
`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`Fig. 1. Comparison of amino acid sequences for exendin-4, mammalian GLP-1, and Gila monster GLP-1.
`
`based on observations that exendin-4 may improve h-cell
`mass, thereby impeding or even reversing disease progres-
`sion. Therefore, a major focus of this article will be to
`examine in detail
`the published reports supporting the
`concept that exendin-4 and GLP-1 may increase h-cell mass
`via stimulation of h-cell neogenesis, stimulation of h-cell
`proliferation, and suppression of h-cell apoptosis.
`The onset of type 2 diabetes is characterized by the
`emergence of postprandial (post-meal) hyperglycemia and
`subsequently, fasting hyperglycemia [22]. In most individ-
`uals, hyperglycemia results from a failure of pancreatic h-
`cells to secrete adequate insulin to compensate for insulin-
`resistance in peripheral
`tissues [23,24]. The fraction of
`glycosylated hemoglobin (A1C) in circulating red blood
`cells provides an accurate indicator of average glucose
`concentrations in the blood for the previous 3 months.
`A1C levels in healthy humans typically comprises 5 – 6%
`of total hemoglobin, while A1C values in people with
`poorly controlled diabetes generally exceed 9% [25].
`
`Results from the United Kingdom Prospective Diabetes
`Study (UKPDS) showed that a reduction in A1C was
`associated with a reduced risk of vascular complications,
`and also reaffirmed that type 2 diabetes is a progressive
`disease characterized by a continuous loss of h-cell function
`that current therapies cannot rectify. [26,27]. Exenatide is
`the USAN generic drug name for synthetic exendin-4, an
`investigational therapeutic being studied by Amylin Phar-
`maceuticals in partnership with Eli Lilly and Company, that
`may have a beneficial impact on the course of this disease.
`
`2. GLP-1 receptor
`
`The GLP-1 receptor (GLP-1R) is a seven-transmembrane
`domain, G-protein coupled receptor, initially described as
`the exendin receptor [28 – 30]. Distribution of the mamma-
`lian GLP-1R includes pancreatic periductal- and h-cells,
`kidney, heart, stomach, and brain [31]. The pancreatic
`
`Fig. 2. Theoretical overview of the primary, anti-diabetic mechanisms of action for exendin-4 (or exenatide). Data suggest that exendin-4 enhances glycemic
`control by enhancing glucose-dependent insulin secretion, suppressing glucose-dependent glucagon secretion, slowing gastric emptying, reducing food intake,
`stimulating h-cell health, and increasing the insulin sensitivity of peripheral tissues.
`
`MPI EXHIBIT 1037 PAGE 2
`
`MPI EXHIBIT 1037 PAGE 2
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 2 of 12
`
`

`

`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`79
`
`GLP-1 receptor binds exendin-4 and GLP-1 with equal
`affinity in in vitro assays, and both peptides stimulate the
`receptor equipotently as demonstrated by the production of
`cyclic adenosine monophosphate (cAMP) in human and rat-
`based receptor systems [32 – 34]. Therefore, differences in
`receptor affinity and activation cannot explain the difference
`in in vivo potency between the two peptides. Under phys-
`iological conditions, the GLP-1R recognizes GLP-1 specif-
`ically, with no significant binding to secretin, vasoactive
`intestinal peptide (VIP), or other closely related, mammalian
`peptide hormones [28,31].
`Based on studies using GLP-1R knockout (GLP-1R / )
`mice and in vitro receptor blockade in islets from these mice,
`a GLP-1 receptor appears to be involved in, and necessary
`for, the incretin/insulinotropic actions of exenatide and GLP-
`1, and the hepatic portal glucose sensor actions of GLP-1
`[3,35 – 38]. GLP-1R / mice exhibit fasting hyperglyce-
`mia and abnormal blood glucose excursions in response to
`glucose challenge [39]. However, care must be taken when
`ascribing specific physiological functions to the GLP-1
`receptor based on GLP-1R / mice, as these mice also
`exhibit abnormalities in the hypothalamic – pituitary – adrenal
`axis and alterations in the GIP (glucose-dependent insulino-
`tropic polypeptide) hormone path- ways which may com-
`pensate for the absence of the GLP-1 signal transduction
`pathway [40,41]. Both GLP-1 and GIP stimulate insulin
`secretion under conditions of hyperglycemia (incretin
`actions) in GLP-1R / mice, resulting in lowered post-
`prandial glucose excursions [3]. Truncated exendin-4(9 –
`39)-NH2 binds to and antagonizes mammalian pancreatic
`GLP-1 receptors [7] and is associated with an increased
`postprandial glucose excursion when administered to wild-
`type, male CD1 mice (5 Ag/mouse IP 20 min before oral
`glucose challenge; [35]). In contrast, administration of trun-
`cated exendin-4(9 – 39)-NH2 to GLP-1R / mice had no
`statistically significant effect on postprandial glucose excur-
`sions, although acute blood glucose levels (10 – 30-min post-
`challenge) trended lower after both oral and intraperitoneal
`glucose administration [35].
`While exendin-4 and GLP-1 appear to share certain
`glucose-lowering actions, it is apparent that not all actions
`of exendin-4 are predictable based on the known pharma-
`cology of GLP-1 [2]. For example,
`intraportal GLP-1
`infusion triggers firing of the hepatic vagal afferent nerves,
`while exendin-4 does not [42]. Exendin-4 sensitized differ-
`entiated 3T3-L1 adipocytes to insulin-dependent glucose
`uptake, while GLP-1 had no effect in the same assay [43].
`Exendin-4 may therefore exert at least some of its actions
`through a functionally different receptor, although this
`putative receptor has not yet been identified [38,42,43].
`
`3. Glycemic control
`
`Exendin-4, GLP-1, and GLP-1 analogues such as
`NN2211 have demonstrated abilities to control fasting and
`
`postprandial glucose excursions. The effects of GLP-1 on
`glycemic control in nonhuman models of diabetes have been
`reviewed elsewhere [44] and will not be extensively covered
`here, except for comparisons with exendin-4.
`Exendin-4 had potent activity in reducing plasma glucose
`when administered as a single intraperitoneal dose of 0.001
`to 10 Ag to hyperglycemic db/db mice, a model of type 2
`diabetes [11]. The maximal glucose-lowering effect of
`exendin-4 was sustained through the last time point mea-
`sured (4 h). Comparable administration of GLP-1 in the
`range of 1 – 1000 Ag also lowered glucose acutely; however,
`plasma glucose concentrations rapidly returned to their
`usual hyperglycemic levels. Exendin-4 lowered plasma
`glucose concentrations in a dose-dependent manner within
`1 h after injection, with a maximum reduction of 37%. The
`half-maximal effective dose (ED50) for exendin-4 averaged
`0.06 Ag/kg compared to 329 Ag/kg for GLP-1. In the ob/ob
`mouse model of diabetes, exendin-4 had an average ED50 of
`0.136 Ag/kg compared to 744 Ag/kg for GLP-1. Overall,
`exendin-4 was greater than 5000-fold more potent than
`GLP-1 in controlling hyperglycemia. In both strains of
`diabetic mice, the magnitude of the glucose lowering effect
`following a single dose of exendin-4 was related to the pre-
`existing plasma glucose concentration, i.e., exendin-4 activ-
`ity was glucose-dependent.
`The effects of prolonged 12- to 13-week treatment with
`once-daily intraperitoneal exendin-4 (24 nmol/kg; 100 Ag/
`kg) has been studied in both diabetic db/db and nondiabetic
`mice [20]. After 1 week, fasting blood glucose concentra-
`tions in diabetic mice had declined from 232 to 90 mg/dl in
`the exendin-4-treated group, but remained at 238 mg/dl in
`the vehicle-control group. Fasting blood glucose concen-
`trations were also lower in nondiabetic mice treated with
`exendin-4 (70 mg/dl) compared with vehicle-treated mice
`(135 mg/dl). At the completion of treatment, A1C values
`were 47% lower in exendin-4-treated diabetic mice than in
`vehicle-treated, averaging 4.7% and 8.8%, respectively.
`Nondiabetic mice also had a significant lowering of A1C
`values after exendin-4 treatment (3.1 F 0.07% vs. 3.5 F
`0.08%. in vehicle-control mice). These A1C data affirmed
`the beneficial effects of exendin-4 on long-term glycemic
`control.
`The effects of chronic intraperitoneal administration of
`exendin-4 on A1C and insulin sensitivity were further
`studied in male obese, diabetic Fatty Zucker (ZDF) rats
`[11]. These animals exhibit overt hyperglycemia starting at
`8 to 10 weeks of age, are insulin-resistant, obese, dyslipi-
`demic, and succumb to h-cell failure at 12 to 14 weeks of
`age. In a 5-week study using twice daily intraperitoneal
`injections of 100 Ag exendin-4, initial pretreatment A1C
`values were approximately 73% greater in obese ZDF rats
`(6.0 – 6.9%) than in lean non-diabetic control rats (3.6 –
`3.9%; Fig. 3). After 35 days, a significant decline in A1C of
`41.3% was seen in the ZDF rats receiving exendin-4. This
`was 2.4-fold greater than the reduction observed in exendin-
`4-treated control lean rats. Measurements of glucose uptake
`
`MPI EXHIBIT 1037 PAGE 3
`
`MPI EXHIBIT 1037 PAGE 3
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 3 of 12
`
`

`

`80
`
`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`doses of NN2211 (2 Ag/kg; 0.6 nmol/kg) or vehicle under
`conditions of hyperglycemic glucose clamp. h-Cell-reduced
`pigs treated with the GLP-1 analogue required an approx-
`imately 200% increase in the glucose infusion rate com-
`pared to the vehicle-control group, and exhibited a tem-
`porally coincident increase in plasma insulin concentrations
`(AUC 72 F 28% greater than vehicle) which was glucose-
`dependent. These data indicate an improvement in insulin-
`sensitivity after treatment with the GLP-1 analogue.
`NN2211 treatment also suppressed plasma glucagon con-
`centrations during hyperglycemic clamp, but not during
`euglycemia following termination of the clamp, demonstrat-
`ing the glucose-dependency of this anti-diabetic activity
`mediated through the GLP-1 receptor. Longer-term admin-
`istration of subcutaneous NN2211 also had effects on
`glycemia in h-cell-reduced pigs. Once-daily NN2211 (3.3
`Ag/kg) was administered for 4 weeks. Treatment with the
`GLP-1 analogue reduced postprandial glucose excursions at
`both 2 and 4 weeks, and slowed gastric emptying. Overall,
`treatment with a GLP-1 analogue in a pig model of human
`diabetes confirmed several glucoregulatory mechanisms of
`action previously observed after administration of exendin-4
`or GLP-1.
`In fasted Rhesus monkeys with type 2 diabetes, a single
`subcutaneous injection of exendin-4 caused a reduction in
`plasma glucose that was dose-dependently accelerated, with
`a mean ED50 value of 0.25 Ag/kg and a maximal glucose
`nadir of approximately 37% at a dose of 100 Ag/kg [11].
`Subsequently, plasma glucose concentrations tended to rise
`again towards control
`levels 2 to 3 h after exenatide
`injection.
`The ability of GLP-1 to control glucose excursions in
`preclinical diabetes models led to a series of GLP-1 clinical
`trials in humans, which have been summarized [44]. How-
`ever, it has become apparent that the short plasma half-life
`of GLP-1 was a significant barrier to clinical development.
`In particular, Zander et al. [46] demonstrated that subcuta-
`neous infusion of GLP-1 efficiently lowers plasma glucose
`in patients with type 2 diabetes, but must be given contin-
`uously to be effective. The resistance of exendin-4 to
`degradation by dipeptidyl peptidase-IV and other putative
`mechanisms which together result in a better pharmacoki-
`netic profile for exendin-4 offered one possible solution to
`this dilemma. A series of phase II trials of exenatide
`(synthetic exendin-4) have now been completed in greater
`than 300 subjects with type 2 diabetes (reviewed by Nielsen
`and Baron [47]). In general, a consistent pattern of safety
`and pharmacodynamics was observed. Dose-ranging studies
`identified an optimal exenatide glucose-lowering dose range
`of 0.05 – 0.2 Ag/kg when injected subcutaneously. Fineman
`et al. [48] reported notable findings from a phase II study of
`exenatide in patients with type 2 diabetes not attaining A1C
`goals V 8% with oral sulfonylureas and/or metformin.
`Twenty-eight days of exenatide treatment reduced A1C by
`approximately 0.9% compared to baseline ( p V 0.006). In
`addition, the proportion of patients achieving A1C V 7%
`
`Fig. 3. Plasma A1C in diabetic and normoglycemic rats. Long-term
`administration of exendin-4 reduced plasma A1C in diabetic, obese ZDF
`rats and normoglycemic lean littermate control rats ( p < 0.01 for obese rats
`and p < 0.002 for lean rats, respectively, compared with vehicle-treated lean
`rats). By day 35, A1C values had decreased 41% in obese ZDF rats treated
`with exendin-4. The relative reduction in A1C was 2.4-fold greater in obese
`than in lean exendin-4-treated rats, consistent with exendin-4 having a
`greater effect under hyperglycemic than normoglycemic conditions.
`Exendin-4 (100 Ag) or vehicle was injected intraperitoneally, twice daily,
`for 5 weeks. Initial A1C values were 6.44 F 0.25% in obese ZDF rats and
`3.73 F 0.37% in lean rats. Lean/Vehicle group, open triangles. Lean/
`Exendin-4 group, closed triangles. Obese/Vehicle group, open circles.
`Obese/Exendin-4 group, closed circles. N = 6 rats per group. (Adapted from
`Ref. [11]. Copyright n1999 American Diabetes Association. Reprinted
`with permission from The American Diabetes Association).
`
`into tissues under euglycemic and hyperinsulinemic con-
`ditions at the end of this study demonstrated an increase in
`insulin sensitivity (defined as plasma insulin concentration
`divided by the rate of glucose infusion) in exendin-4-treated
`obese ZDF rats (76%, p < 0.02) and lean, non-diabetic rats
`(51%, p < 0.05), compared with the saline-treated controls.
`In a second study, ZDF rats injected intraperitoneally once
`or twice daily with exendin-4 for 6 weeks had dose-
`dependent arrest or reversal of the trend for A1C to increase
`with increasing age/weight. Exendin-4 doses as low as 0.1
`Ag/rat
`twice daily were effective. Insulin sensitivity in-
`creased dose-dependently by up to 49% in the exendin-4-
`treated animals at the end of this study, as measured by
`glucose uptake under euglycemic hyperinsulinemic condi-
`tions. Whether this improvement in insulin sensitivity is a
`consequence of reduced glucose toxicity, or via direct
`actions of exendin-4 to improve glucose uptake into insu-
`lin-sensitive tissues, remains to be elucidated. Increases in
`total cholesterol levels were also significantly reduced in the
`exendin-4 treatment group over the 6-week period.
`Analagous effects on glycemic control have also been
`reported in a larger mammalian model of human diabetes,
`using the GLP-1 analogue NN2211 to activate the GLP-1
`receptor signal transduction pathway [45]. Chemical de-
`struction of h-cells in male Go¨ttingen minipigs caused the
`pigs to have impaired glucose tolerance or to be outright
`diabetic. Fasted h-cell-reduced pigs received intravenous
`
`MPI EXHIBIT 1037 PAGE 4
`
`MPI EXHIBIT 1037 PAGE 4
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 4 of 12
`
`

`

`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`81
`
`was fourfold greater after exenatide treatment compared
`with placebo. Given that A1C only fully reflects a change
`in glycemia 3 months after a sustained change has occurred,
`this reduction in A1C and enhanced ability to achieve
`clinically relevant A1C target values after only one month
`of therapy is of great clinical interest. Glucose profiles
`during ingestion of a mixed meal demonstrated that the
`marked, acute ability of exenatide to reduce postprandial
`glycemia was sustained over the 28-day observation period.
`Another recent report [49] examined the impact of 1 month
`of SC exenatide dosing on blood glucose levels in poorly
`controlled, community dwelling, insulin-naive patients with
`type 2 diabetes. Although interpretation of these study
`results is limited by the lack of a placebo control, exenatide
`treatment resulted in reduced A1C compared with pretreat-
`ment values.
`
`4. Insulin secretion
`
`Glucose-dependent insulinotropism refers to the ability
`of agents such as exendin-4 and GLP-1 to stimulate insulin
`secretion during euglycemia or hyperglycemia, but not
`during hypoglycemia [2]. Glucose-dependent insulinotrop-
`ism has also been defined as the amplification of h-cell
`insulin secretion when circulating glucose concentrations
`are above, but not below, the normal range [8,9]. In animal
`models of diabetes, a predominant acute action of exendin-
`4 is glucose-dependent
`insulinotropism, resulting in an
`amplification of glucose/insulin secretion coupling. This
`action of exendin-4 contrasts with the action of non-
`glucose-dependent insulin secretagogues or hypoglycemic
`agents, such as sulfonylureas, which predominantly in-
`crease insulin secretion independent of prevailing glucose
`concentrations [50] and thus have a greater potential to
`induce hypoglycemia [27].
`Two in vitro models using pancreatic islets isolated from
`male Lewis rats have been used to examine the action of
`exendin-4 on insulin secretion [9]. In a static incubation
`model, islets were incubated in a medium containing either
`3 mmol/l (basal) or 10 mmol/l (elevated) glucose. Raising
`the glucose concentration from 3 to 10 mmol/l increased
`insulin secretion 9.8-fold ( p < 0.05). In the presence of
`elevated glucose (10 mmol/l) and a range of exendin-4
`doses (1 nmol/l – 1 Amol/l), insulin secretion was increased
`over basal levels by up to 19.6-fold ( p < 0.01). In a micro-
`physiometer model, increasing the glucose concentration
`from 3 mmol/l (basal) to 7.5 mmol/l for 15 min increased
`insulin secretion up to 6.4-fold. The addition of exendin-4
`(20 nmol/l) stimulated insulin secretion over and above the
`effect of 7.5 mmol/l glucose alone, to 13.5-fold over basal
`rate ( p < 0.01). Insulin secretion returned to previous levels
`within 5 min of cessation of exendin-4 perfusion.
`Thus, exendin-4 exerts direct effects on rat pancreatic
`islets in vitro to enhance glucose-stimulated insulin secre-
`tion. These effects do not appear to persist once exendin-4 is
`
`removed from the system, suggesting that the peptide may
`be binding and activating target receptors only while it is
`present
`in the perfusate. Furthermore,
`the insulinotropic
`action of exendin-4 in this system rapidly decreases when
`the ambient glucose is decreased back to 3 mmol/l, consis-
`tent with the glucose-dependence of insulinotropism being
`at least in part at the level of the pancreatic islet. Overall, the
`data support the interpretation that exendin-4 acts directly,
`but perhaps not exclusively, at the isolated pancreatic islet
`level to stimulate insulin secretion, and are consistent with
`published reports showing enhanced insulin secretion from
`islets exposed to GLP-1 [33,51 – 53].
`Intravenous administration of exendin-4 (0.4 nmol/kg)
`acutely increased plasma insulin concentrations in fasted
`Wistar rats, eliciting approximately double the insulin peak
`elicited by the same dose of GLP-1. The ED50 value for this
`insulinotropic activity was 0.014 nmol/kg for exendin-4
`compared with 0.19 nmol/kg for GLP-1 [20]. Similar results
`have been reported by Parkes et al. [9] examining the
`insulinotropic actions of exendin-4 during an intravenous
`glucose challenge (Fig. 4).
`Insulinotropic actions have also been reported for exen-
`din-4 in studies examining daily administration of the
`peptide (24 nmol/kg) to diabetic db/db mice for 12 – 13
`weeks [20]. Fasting plasma insulin concentrations in dia-
`betic mice treated with exendin-4 averaged 550% higher
`than vehicle-treated diabetic rats under conditions where the
`corresponding plasma glucose concentrations averaged 16
`and 29 mmol/l, respectively, compared to 7 – 8 mmol/l blood
`glucose in nondiabetic (young) db/db mice.
`In the human studies reported by Kolterman et al. [10],
`subcutaneous exenatide (synthetic exendin-4) rapidly low-
`ered both fasting and postprandial plasma glucose in
`patients with type 2 diabetes. The glucose-dependent insu-
`linotropism exhibited by exenatide was best illustrated by
`the data obtained in the fasting state, where there was a
`dose-dependent rise in serum insulin concentrations within
`the first 3 h after exenatide administration compared to
`placebo ( p < 0.001). In sharp contrast, placebo treatment
`resulted in relatively stable insulin concentrations through-
`out the 8-h period of observation. The rise and peak of
`serum insulin concentrations following exenatide adminis-
`tration coincided with the rapid decline of fasting glucose
`concentrations. After 3- to 4-h postdose, and coincident
`with reaching glucose nadir, mean serum insulin returned
`to baseline with little difference among groups. Insulin
`AUC(0 – 8 h) and Cmax values for all exenatide treatments
`increased in an apparently dose-dependent manner com-
`pared to placebo ( p < 0.05). Since exenatide concentrations
`remained elevated throughout the course of the assessment,
`the reduction in insulin beyond 3 h did not reflect a simple
`waning of exenatide effect due to lower circulating concen-
`trations of exenatide.
`Fineman et al. [48] used the homeostasis model assess-
`ment (HOMA) [54] methodology to assess h-cell function
`at baseline and at Days 14 and 28 in human subjects with
`
`MPI EXHIBIT 1037 PAGE 5
`
`MPI EXHIBIT 1037 PAGE 5
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 5 of 12
`
`

`

`82
`
`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`in fasting and postprandial glucagon levels in patients with
`type 2 diabetes [55] and the known activity of glucagon
`with respect to maintaining hepatic glucose output [56], it is
`reasonable to extrapolate that glucagon suppression by
`exenatide contributed to the overall effect of lowered plasma
`glucose in both the fasting and postprandial periods. Like
`insulin, glucagon concentrations also returned towards
`baseline beyond 3-h post-injection during the fasting state
`when the prevailing plasma glucose concentration reached
`its nadir. As the changes in concentrations of insulin and
`glucagon are coincident and reciprocal,
`the effects of
`exenatide on the enhancement of insulin secretion and the
`suppression of glucagon secretion may be considered glu-
`cose-dependent. It is noteworthy that currently available
`insulin secretagogues, as well as exogenous insulin, do not
`suppress the paradoxical postprandial glucagon rise ob-
`served in patients with diabetes [57]. This inappropriate
`elevation in postprandial plasma glucagon concentrations
`results in an inappropriately low insulin-to-glucagon ratio in
`the hepatic portal vein, contributing to sustained rates of
`excess hepatic glucose production [58]. Thus, by virtue of
`its ability to enhance endogenous insulin and lower gluca-
`gon secretion, exenatide would tend to re-establish a more
`physiologic and favorable portal vein ratio of insulin-to-
`glucagon compared to currently available agents.
`
`6. Enhanced B-cell mass
`
`One of the most provocative areas of recent research is
`based on observations that exendin-4 may improve h-cell
`health, thereby impeding or even reversing disease progres-
`sion. A number of published reports support the concept that
`exendin-4 and GLP-1 increase h-cell mass via stimulation
`of h-cell neogenesis, stimulation of h-cell proliferation, and
`suppression of h-cell apoptosis (Table 1).
`The development of the endocrine pancreas is under
`multifactorial control, and many of the key proteins or
`transcription factors involved in each developmental step
`have now been identified [59]. Expression of the transcrip-
`tion factor PDX-1/IDX-1 is essential for development and
`regeneration of the endocrine pancreas, and stimulation of
`PDX-1 may be one of the intracellular mechanisms by
`which exendin-4 and GLP-1 can enhance h-cell mass
`leading to long-term improvement
`in glucose control.
`Exendin-4 and GLP-1 have been proposed to produce
`differentiation of pancreatic non-h-cells including pancreat-
`ic ductal epithelial cells, acinar cells, and nestin-positive
`peri-ductal cells (potentially ‘‘functional’’ islet stem cells),
`into immunohistologically identifiable insulin-producing h-
`cell buds and subsequently, fully functional islets. In vitro
`studies in human and animal cell preparations support this
`hypothesis, and give insight into the molecular pathways
`mediating these changes.
`Multipotential progenitor cells in the pancreatic islets and
`ducts, so-called nestin-positive islet-derived progenitor cells
`
`Fig. 4. Insulinotropic actions of exendin-4 and GLP-1: Concentration
`response. Male normoglycemic Harlan – Sprague – Dawley rats were
`anesthetized, then given a 2-h intravenous infusion of exendin-4 (3, 30,
`300, or 3000 pmol/kg/min; n = 4 – 8 per dose), or GLP-1 (3, 30, 300, or
`3000 pmol/kg/min; n = 4 – 8 per dose), or vehicle (1 ml/h, n = 8) at t = 30
`min. At t = 0 min, D-glucose (5.7 mmol/kg) was infused at a rate of 0.5 ml/
`min over 1 – 2 min. This infusion regimen resulted in identical plasma
`glucose pharmacokinetic profiles in all treatment groups (see Ref. [9]).
`Plasma insulin was measured at 30, 15, 0, 5, 10, 15, 20, 30, 40, 50, 60,
`and 90 min. Peak insulinotropic effects of exendin-4 and GLP-1 occurred at
`a similar steady-state plasma concentration of approximately 1 to 2 nmol/l;
`thus, insulinotropic potencies were similar. However, the magnitude of the
`maximal effect was 63% greater with exendin-4 than with GLP-1
`( p < 0.03). Exendin-4, black circles. GLP-1, white rectangles. (Reprinted
`from Ref. [9]).
`
`type 2 diabetes receiving two or three daily subcutaneous
`injections of exenatide (0.08 Ag/kg). HOMA analysis
`revealed improved h-cell secretory function following exe-
`natide therapy. It is noteworthy that fasting values of plasma
`glucose and insulin, which were used to calculate HOMA,
`were obtained prior to the morning dose of exenatide when
`plasma concentrations of exenatide were negligible, sug-
`gesting a fundamental alteration in h-cell function following
`sustained exenatide exposure. Whether or not exenatide
`treatment increases pancreatic islet mass in humans awaits
`future studies, since h-cell function can improve indepen-
`dently of increased islet mass.
`
`5. Glucagon secretion
`
`Circulating glucagon was reduced after exenatide treat-
`ment in both the fasting and postprandial states in humans
`with type 2 diabetes [10]. The observation under fasting
`conditions supports the hypothesis that suppression of
`glucagon secretion is not merely a consequence of the
`slowing of nutrient presentation to the small
`intestine
`(gastric emptying). Given the well-documented elevations
`
`MPI EXHIBIT 1037 PAGE 6
`
`MPI EXHIBIT 1037 PAGE 6
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1037, p. 6 of 12
`
`

`

`L.L. Nielsen et al. / Regulatory Peptides 117 (2004) 77–88
`
`83
`
`Effects on pancreatic h-cells
`
`Agent
`
`Key findings
`
`Reference
`
`Nestin-positive islet-derived
`
`Exendin-4, GLP-1
`
`Exendin-4, GLP-1
`
`Exendin-4
`
`Exendin-4, GLP-1
`
`Exendin-4, GLP-1
`
`Intrauterine growth-retarded
`
`Exendin-4
`
`Exendin-4, GLP-1
`
`Exendin-4
`
`Exendin-4, tNN2211
`Exendin-4
`
`Stimulated differentiation of progenitor cells into
`insulin-producing cells.
`Stimulated differentiation into cells expressing insulin,
`pancreatic polypeptide, and glucagon.
`Attenuated the development of diabetes; increased h-cell
`mass via differentiation (neogenesis) and replication.
`Increased pancreatic insulin content, h-cell mass, and
`h-cell proliferation; decreased plasma glucose.
`Increased pancreatic insulin content and h-cell mass;
`decreased plasma glucose.
`Increased h-cell mass and proliferation; increased
`Pdx-1 expression; decreased plasma glucose.
`
`Increased h-cell mass, h-cell neogenesis, and Pdx-1
`expression; increased insulin secretion; decreased plasma
`glucose; lower A1C; improved oral glucose tolerance.
`Increased h-cell mass; decreased h-cell apoptosis;
`increased plasma insulin; decreased plasma glucose;
`improved oral glucose tolerance; decreased cell apoptosis in vitro.
`Increased h-cell mass and proliferation; decreased plasma glucose.
`Increased pancreatic insulin content and h-cell mass; decreased
`h-cell apoptosis; improved hyperinsulinemia; enhanced glucose-
`stimulated insulin secretion; improved oral glucose tolerance;
`decreased plasma glucose.
`
`[60]
`
`[61]
`
`[17]
`
`[18]
`
`[19]
`
`[66]
`
`[62]
`
`[63]
`
`[64]
`[65]
`
`(NIPs), have been previously identified by Habener et al. In
`a recent study, they now report the expression of functional
`GLP-1 receptors o

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