throbber
Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`Breakthroughs and Views
`
`BBRC
`
`www.elsevier.com/locate/ybbrc
`
`Glucose-dependent insulinotropic polypeptide analogues and
`their therapeutic potential for the treatment of obesity-diabetes
`
`Victor A. Gault,* Peter R. Flatt, and Finbarr P.M. O’Harte
`
`School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
`
`Received 17 June 2003
`
`Abstract
`
`Glucose-dependent insulinotropic polypeptide (GIP) is a key incretin hormone, released postprandially into the circulation in
`response to feeding, producing a glucose-dependent stimulation of insulin secretion. It is this glucose-dependency that has attracted
`attention towards GIP as a potential therapeutic agent for the treatment of type 2 diabetes. A major drawback to achieving this goal
`has been the rapid degradation of circulating GIP by the ubiquitous enzyme, dipeptidylpeptidase IV (DPP IV). However, recent
`studies have described a number of novel structurally modified analogues of GIP with enhanced plasma stability, insulinotropic and
`antihyperglycaemic activity. The purpose of this article was to provide an overview of the biological effects of several GIP modi-
`fications and to highlight the potential of such analogues in the treatment of type 2 diabetes and obesity.
`Ó 2003 Elsevier Inc. All rights reserved.
`
`Keywords: Dipeptidylpeptidase IV; Glucagon-like peptide-1; Glucose-dependent insulinotropic polypeptide; GIP analogues; Insulin secretion;
`Obesity; Type 2 diabetes
`
`Glucose-dependent insulinotropic polypeptide (GIP)
`is a 42 amino acid gastrointestinal hormone secreted
`from enteroendocrine K-cells in response to food and
`nutrient absorption [1]. Although initially characterized
`for its ability to inhibit histamine-induced gastric acid
`secretion, the primary role of GIP as an incretin hor-
`mone moderating glucose-induced insulin secretion is
`widely recognized [2]. In addition to its actions on the
`pancreatic beta cell, GIP is also known to exert various
`extrapancreatic effects, which further enhance its glucose
`lowering ability. In particular, GIP has been shown to
`augment insulin-dependent inhibition of glycogenolysis
`in the liver [3] and to exert stimulatory effects on glucose
`uptake and metabolism in muscle [4]. Furthermore,
`functional GIP receptors have been identified on
`adipocytes [5] and have been shown to stimulate glucose
`transport
`[6],
`increase fatty acid synthesis [7], and
`stimulate lipoprotein lipase activity [8]. More recent
`studies have shown GIP to stimulate beta cell mito-
`genesis and inhibit apoptosis [9,10]. Consequently, this
`
`* Corresponding author. Fax: +44-28-7032-4965.
`E-mail address: va.gault@ulster.ac.uk (V.A. Gault).
`
`0006-291X/$ - see front matter Ó 2003 Elsevier Inc. All rights reserved.
`doi:10.1016/S0006-291X(03)01361-5
`
`wide spectrum of biological activities has sparked the
`recent interest in GIP as a novel therapeutic candidate
`for the treatment of type 2 diabetes [11].
`
`Solutions to difficulties posed by GIP as a therapeutic
`agent
`
`Currently there are two main concerns in attempting
`to utilize GIP as a potential therapeutic agent. First, the
`native peptide has a short biological half-life in the cir-
`culation (approximately 3–5 min), due primarily to
`degradation by the ubiquitous enzyme, dipeptidylpep-
`tidase IV (DPP IV; EC 3.4.14.5). After release into the
`circulation, the native peptide (GIP 1-42) is rapidly hy-
`drolysed at the amino terminus removing Tyr1–Ala2 to
`produce the truncated metabolite GIP(3-42) [12]. This
`major degradation product was initially thought to be
`inactive [13], however, recent observations have shown it
`to function as a GIP receptor antagonist in vivo [14]. To
`circumvent degradation, DPP IV inhibitors are cur-
`rently being developed for in vivo administration. Sev-
`eral positive effects on glycaemic control have been
`
`MPI EXHIBIT 1084 PAGE 1
`
`MPI EXHIBIT 1084 PAGE 1
`
`

`

`208
`
`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`reported [15–17], but the effects of long-term interfer-
`ence on the metabolism of a plethora of other peptide
`substrates for DPP IV are as yet unknown [18]. Con-
`ceivably, a more attractive approach than widespread
`non-specific DPP IV inhibition involves the synthesis of
`specific GIP analogues modified at the enzyme cleavage
`site [11].
`Another concern regarding the use of GIP in diabetes
`therapy stems from a study in which GIP was shown to
`exhibit marked reduction of insulinotropic activity in
`type 2 diabetic subjects [19]. Indeed, several studies have
`shown blunted insulin responses to GIP infusion in type
`2 diabetes, albeit with differing degrees of beta cell re-
`sistance [19–22]. However, type 2 diabetes is associated
`with a global defect in insulin secretion not merely re-
`stricted to GIP, and encompassing other factors, in-
`cluding glucose and GLP-1 [23,24]. It has also been
`shown that beta cell sensitivity to GIP in type 2 diabetic
`patients improves with glyburide treatment [25]. Muta-
`tions of GIP receptors are rare in diabetes [21,26] and it
`is evident that any speculated abnormalities in GIP re-
`ceptor binding can be overcome by enzyme-resistant
`analogues of GIP [11,27–33]. Furthermore, a recent
`study has shown that an N-terminally modified ana-
`logue of GIP, Tyr1-glucitol-GIP, evokes a substantially
`larger and more protracted insulin response to oral
`glucose than the native GIP in type 2 diabetic patients
`[34].
`
`The attractiveness and potential of stable GIP analogues
`
`A number of significant features indicate that en-
`zyme-resistant analogues of GIP have unique potential
`for diabetes therapy. First, GIP is the major physio-
`logical
`incretin as indicated by greater hormone re-
`sponses to feeding [35] and comparative studies using
`either receptor antagonists [36,37] or receptor knockout
`mice [38,39]. Second, structural modification of the
`sister incretin, GLP-1, invariably compromises biologi-
`cal activity at the GLP-1 receptor [40–44], whereas the
`opposite is true for many analogues of GIP [11,27–33].
`Third, any effect of GIP on increasing glucagon con-
`centrations is only observed at normal glucose concen-
`trations [45,46] and thus is irrelevant following feeding
`or in treating type 2 diabetes [47]. Fourth, in contrast to
`GLP-1, GIP lacks significant effects on gastric emptying
`and is therefore well tolerated by human subjects. This
`has become an increasing and possibly damning diffi-
`culty with the use of GLP-1 and its analogues in pa-
`tients, as evidenced by increased gastrointestinal side
`effects and nausea [48]. This, therefore, focuses increas-
`ing attention on the antidiabetic potential of GIP. To
`date, several studies have been published examining the
`in vitro activities of a range of GIP fragments and an-
`alogues [27,49–54]. Moreover, as reviewed in the fol-
`
`lowing sections, the in vivo antidiabetic potential of a
`family of selective designer human GIP 1-42 analogues
`(Fig. 1) modified at positions Tyr1 [11,28–30], Ala2 [31–
`33], and Glu3 [37,55] have been tested in animal models
`of type 2 diabetes and obesity.
`
`Effects of modifications at position Tyr1
`
`Several novel Tyr1-modified analogues of GIP have
`been developed, modelled on previous studies with the
`glucagon-secretin family of gastrointestinal peptides and
`knowledge of the substrate-binding specificity of DPP IV
`(Fig. 1). These analogues include N-acetyl-, N-Fmoc-, N-
`glucitol-, N-palmitate-, and N-pyroglutamyl-GIP [11,27–
`30]. All analogues modified at Tyr1 exhibited complete
`resistance to DPP IV with in vitro half-lives greater than
`24 h compared with 2.3 h for native GIP (Table 1), as il-
`lustrated for N-acetyl-GIP in Figs. 2A–B. This is in
`agreement with DPP IV substrate-binding specificity,
`which predicts the requirement for a bulky N-terminal
`amino acid (such as tyrosine in the case of GIP) possessing
`a free protonated a-amino group. By attaching amino
`acids or functional groups to the N-terminus of GIP, Tyr1
`becomes unprotonated, therefore, removing the explicit
`prerequisite required for DPP IV to act [18].
`In assessing the biological activity of each analogue in
`vitro, cAMP formation in human GIP-receptor trans-
`fected cells [56] and insulinotropic activity in clonal
`BRIN-BD11 cells [57] was measured. All of the Tyr1-
`modified analogues
`studied exhibited an increased
`potency (2- to 10-fold increase in EC50 values) in stim-
`ulating cAMP production compared to native GIP
`(Table 1; Fig. 2C). Furthermore, the Tyr1-modified an-
`alogues exhibited significantly increased insulin secre-
`tory responses (1.1- to 1.4-fold) compared to the native
`peptide in BRIN-BD11 cells,
`indicative of improved
`biological activity (Table 1; Fig. 2D). However, subtle
`differences could be observed within this group, as both
`N-Fmoc- and N-palmitate-GIP appeared to be moder-
`ately (14–20%) less potent in vitro than N-acetyl-, N-
`glucitol-, and N-pyroglutamyl-GIP.
`In determining the insulin releasing and antihyper-
`glycaemic potential of the GIP analogues in vivo, we
`used ob/ob mice, an extensively studied animal model of
`spontaneous obesity and diabetes. Characteristically
`these mice exhibit hyperphagia, marked obesity, mod-
`erate hyperglycaemia, and severe hyperinsulinaemia
`[28]. All of the Tyr1-modified analogues were noticeably
`superior at stimulating insulin release (2.0- to 2.5-fold)
`and lowering blood glucose (1.4- to 1.9-fold) compared
`with native GIP (Table 1). Furthermore, it could be
`observed that N-acetyl-, N-glucitol-, and N-pyroglut-
`amyl-GIP were slightly more potent than both N-Fmoc-
`and N-palmitate-GIP. Of the Tyr1-modified analogues
`tested, N-acetyl-GIP [29] appeared to be the most
`
`MPI EXHIBIT 1084 PAGE 2
`
`MPI EXHIBIT 1084 PAGE 2
`
`

`

`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`209
`
`Fig. 1. A diagrammatic representation of the 42 amino acid sequence of human glucose-dependent insulinotropic polypeptide (GIP) with the solid
`arrow indicating the site of enzymatic degradation by dipeptidyl peptidase IV (DPP IV). The coloured amino acid residues (1–3) indicate the
`analogues synthesized and tested in animal models of obesity-diabetes [11,27–33], based on the entire human GIP 1-42 sequence.
`
`impressive (Table 1). As shown in Fig. 3, this stable
`analogue significantly augmented the plasma insulin
`response and curtailed the glycaemic excursion follow-
`ing conjoint administration with glucose to obese dia-
`betic ob/ob mice. The ability of N-acetyl-GIP to
`overcome the severe insulin resistance and beta cell
`defect (including poor response to native GIP) in this
`animal model is notable and affords proof of concept
`
`that such analogues offer potential as future therapeutic
`agents for type 2 diabetes.
`
`Effects of modifications at position Ala2
`
`A series of Ala2-substituted analogues of GIP have
`also been synthesized and tested for their DPP IV
`
`Table 1
`Summary of the biological properties of designer human GIP analogues modified at the N-terminal Tyr1, Ala2 or Glu3 amino acid residues
`
`DPP IV
`half-life
`(h)
`
`cAMP
`production EC50
`(nmol/liter)
`
`Plasma glucose
`AUC (% GIP max
`response)
`
`Plasma insulin
`AUC (% GIP max
`response)
`
`Modification
`
`Peptide
`
`Native hormone
`
`GIP
`
`Tyr1-modification
`
`Ala2-substitution
`
`N-Acetyl-GIP
`N-Glucitol-GIP
`N-Pyroglutamyl-GIP
`N-Palmitate-GIP
`N-Fmoc-GIP
`
`(Gly2)GIP
`(Ser2)GIP
`(Abu2)GIP
`(Sar2)GIP
`
`Maximal insulin
`response
`(% GIP max)
`100  3.1
`127  4.0
`141  8.1
`118  1.6
`122  3.3
`112  2.5
`107  6.4
`140  4.7
`62  0.6
`42  3.8
`52  2.2
`(Pro3)GIP
`Glu3-substitution
`>24
`Nil
`160
`43
`DPP IV half-lives were calculated by plotting the percentage of intact peptide remaining after incubation with DPP IV (n ¼ 3) versus incubation
`time. EC50 values were calculated from cAMP dose–response curves (n ¼ 3) in human GIP-receptor transfected CHL cells using Graph pad Prism.
`Maximal insulin response values in BRIN-BD11 cells (n ¼ 8) were calculated relative to the maximal percentage GIP response. Plasma glucose and
`insulin AUC values from ob/ob mice (n ¼ 7–8) were calculated and recorded as a percentage of the maximal GIP response. Values represent
`means SEM. Data taken from [11,27–33].
`
`2.3
`
`>24
`>24
`>24
`>24
`>24
`
`>8
`4.8
`1.9
`1.6
`
`18.2
`
`1.86
`2.03
`2.67
`10.0
`9.4
`
`15.0
`14.9
`38.5
`54.6
`
`100
`
`62
`57
`61
`69
`69
`
`81
`75
`94
`106
`
`100
`
`251
`236
`224
`206
`208
`
`144
`126
`79
`96
`
`MPI EXHIBIT 1084 PAGE 3
`
`MPI EXHIBIT 1084 PAGE 3
`
`

`

`210
`
`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`Fig. 2. (A,B) Representative HPLC profiles obtained after incubation of native GIP (A) and N-acetyl-GIP (B) with DPP IV for 2 h. Reaction
`products were separated on a Vydac C-18 column and peaks corresponding to intact GIP, N-acetyl-GIP, and GIP(3-42) are indicated. (C) Dose-
`dependent production of cAMP by native GIP and N-acetyl-GIP upon binding to CHL cells stably transfected with the human GIP receptor. (D)
`Dose-dependent effects of GIP and N-acetyl-GIP on insulin secretion from BRIN-BD11 cells. Values are means  SEM (n ¼ 3–8). **P < 0:01,
`***P < 0:001 compared to control. DP < 0:05 and DDDP < 0:01 compared to native GIP. Data taken from [29].
`
`Fig. 3. Plasma glucose and insulin responses of 18 h fasted obese diabetic (ob/ob) mice after intraperitoneal administration of glucose alone (2 g/kg
`body weight; open circles), or in combination with either 25 nmol/kg GIP (solid squares) or N-acetyl-GIP (open triangles). Values are means SEM
`(n ¼ 7–8). *P < 0:05, **P < 0:01, and ***P < 0:001 compared to glucose alone. DP < 0:05, DDP < 0:01, and DDDP < 0:001 compared to GIP. Data
`taken from [29].
`
`MPI EXHIBIT 1084 PAGE 4
`
`MPI EXHIBIT 1084 PAGE 4
`
`

`

`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`211
`
`stability and biological activity (Fig. 1). These include
`(Abu2)GIP,
`(Gly2)GIP,
`(Sar2)GIP,
`(Ser2)GIP, and
`(DD-Ala2)GIP [31–33]. Substitution of Ala2 with either
`2-aminobutyric acid (Abu) or sarcosine (Sar) did not
`appear to confer any increased resistance against DPP
`IV, as both analogues followed similar or slightly ac-
`celerated patterns of degradation to the native peptide
`(Table 1) [32]. Indeed, both of these substitutions im-
`paired biological activity compared with native GIP
`when tested using the in vitro insulin secretion and
`cAMP models. Surprisingly though, both analogues
`displayed antihyperglycaemic and insulinotropic activity
`comparable to native GIP when administered to obese
`diabetic ob/ob mice (Table 1) [32].
`In sharp contrast, substitution with either a glycine
`(Gly) or serine (Ser) residue for Ala2 [31] produced more
`stable analogues with significantly prolonged DPP IV
`half-lives compared with native GIP (Table 1). Unlike
`the Abu and Sar substitutions, both (Gly2)GIP and
`(Ser2)GIP displayed enhanced abilities to elevate cAMP
`(1.2-fold increase in EC50 values) and stimulate insulin
`secretion (1.2- to 1.4-fold) in vitro (Table 1). When
`tested in obese diabetic ob/ob mice, both analogues ex-
`hibited significantly improved insulinotropic activity
`(1.3- to 1.5-fold) and antihyperglycaemic activity (1.2- to
`1.3-fold) compared with native GIP (Table 1), further
`supporting the idea that DPP IV resistant analogues of
`GIP may prove useful in the treatment of type 2 diabetes
`[31].
`Using a similar approach, the research group of
`Pederson and McIntosh [33] recently investigated sub-
`stitution of the LL-alanine in position 2 of GIP with
`DD-alanine. This enzyme resistant analogue exhibited
`moderately reduced biological activity in vitro, although
`it significantly decreased the glycaemic excursion (1.6-
`fold) in fa/fa VDF Zucker rats with an efficacy similar to
`that seen with either (Gly2)GIP or (Ser2)GIP [31].
`However, while several of these Ala2-substituted ana-
`logues demonstrated significantly improved biological
`activity compared with native GIP, their efficacy was
`not as impressive as that of the Tyr1-modified analogues
`(Table 1).
`
`Effects of modification at position Glu3
`
`Substitution of Glu3 in GIP with a proline (Pro)
`residue [55] produced a novel GIP analogue, (Pro3)GIP
`(Fig. 1), completely resistant to DPP IV (Table 1).
`Surprisingly though, (Pro3)GIP inhibited GIP-stimu-
`lated cAMP production and insulin secretion with high
`sensitivity and specificity in vitro (Table 1). Further-
`more, studies using ob/ob mice showed that (Pro3)GIP
`effectively and specifically countered the insulin releasing
`and antihyperglycaemic actions of the native peptide
`in vivo [37,55], reminiscent of the effects of the major
`
`DPP IV degradation product, GIP(3-42) [14]. (Pro3)GIP
`has also recently been utilized to demonstrate that GIP
`is the major physiological
`incretin, accounting for
`approximately 80% of nutrient-induced enteroinsular
`pancreatic beta cell stimulation [37].
`Interestingly, the therapeutic potential of such a se-
`lective GIP receptor antagonist has recently been dem-
`onstrated in a study by Miyawaki and colleagues [58],
`where they showed that GIP plays a central role in lipid
`metabolism and in the development of both genetically
`inherited and diet-induced obesity. Thus, growing evi-
`dence supports the long-held view that GIP is an im-
`portant
`factor directly linking over-nutrition to fat
`deposition, obesity, and glucose intolerance [59,60].
`
`Conclusion
`
`Structural modification of GIP at Tyr1, Ala2 or Glu3
`resulted in analogues with greatly increased, moderately
`increased or antagonistic biological properties, respec-
`tively, both in vitro and in vivo. The Tyr1-modified
`analogues, especially N-acetyl-GIP, exhibit a substan-
`tially enhanced potency and duration of action com-
`pared to native GIP. Accordingly, these novel agents
`provide the basis for exploration to realize the potential
`of GIP in diabetes therapy. However, despite the tre-
`mendous potential for GIP analogues in the treatment
`of diabetes-obesity, their peptidic nature effectively rules
`out the option of straightforward oral administration.
`Therefore,
`in order to further develop rational drug
`design for GIP,
`information on appropriate delivery
`systems,
`three-dimensional
`structure and molecular
`interactions of the peptide with its receptor are essential.
`
`Acknowledgments
`
`We dedicate this paper to Professor Vincent Marks who has been a
`great inspiration and long-term enthusiast of GIP. The authors’ work
`was supported financially by University of Ulster Research Strategy
`Funding and the Research and Development Office of Health and
`Personal Social Services for Northern Ireland.
`
`References
`
`[1] J.C. Brown, Enteroinsular axis, in: G.J. Dockray, J.H. Walsh
`(Eds.), Gut Peptides: Biochemistry and Physiology, Raven Press,
`New York, 1994, pp. 765–784.
`[2] W. Creutzfeldt, The entero-insular axis in type 2 diabetes—
`incretins as therapeutic agents, Exp. Clin. Endocrinol. Diabetes
`109 (2001) 288–303.
`[3] D. Elahi, G.S. Meneilly, K.L. Minaker, J.W. Rowe, D.K.
`Andersen, Regulation of hepatic glucose production by gastric
`inhibitory polypeptide in man,
`in: Proceedings of the Sixth
`International Conference on Gastrointestinal Hormones, Ottawa,
`1986, p. 18.
`
`MPI EXHIBIT 1084 PAGE 5
`
`MPI EXHIBIT 1084 PAGE 5
`
`

`

`212
`
`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`[4] F.P.M. O’Harte, A.M. Gray, P.R. Flatt, Gastric inhibitory
`polypeptide and effects of glycation on glucose transport and
`metabolism in isolated mouse abdominal muscle, J. Endocrinol.
`156 (1998) 237–243.
`[5] R.G.-C. Yip, M.O. Boylan, T.J. Kieffer, M.M. Wolfe, Functional
`GIP receptors are present on adipocytes, Endocrinology 139
`(1998) 4004–4007.
`[6] R.H. Eckel, W.J. Fujimoto, J.D. Brunzell, Gastric inhibitory
`polypeptide enhances
`lipoprotein lipase activity in cultured
`preadipocytes, Diabetes 28 (1979) 1141–1142.
`[7] J. Oben, L. Morgan, J. Fletcher, V. Marks, Effects of the entero-
`pancreatic hormones, gastric inhibitory polypeptide and gluca-
`gon-like polypeptide-1 (7-36) amide on fatty acid synthesis in
`explants of rat adipose tissue, J. Endocrinol. 130 (1991) 267–272.
`[8] J.M. Knapper, S.M. Puddicombe, L.M. Morgan, J.M. Fletcher,
`Investigation into the actions of glucose-dependent insulinotropic
`polypeptide and glucagon-like peptide-1(7-36)amide on lipopro-
`tein lipase activity in explants of rat tissue, J. Nutr. 125 (1995)
`183–188.
`[9] A. Tr€uumper, K. Tr€uumper, H. Trusheim, R. Arnold, B. G€ooke, D.
`Horsch, Glucose-dependent insulinotropic polypeptide is a growth
`factor for beta (INS-1) cells by pleiotropic signaling, Mol.
`Endocrinol. 15 (2001) 1559–1570.
`[10] J. Ehses, V. Casilla, T. Doty, J.A. Pospisilik, H.-U. Demuth, R.A.
`Pederson, C.H.S. McIntosh, Glucose-dependent insulinotropic
`polypeptide (GIP) stimulates cell growth and promotes cell
`survival in INS-1 cells [abstract], Diabetes 51 (Suppl. 2) (2002)
`A339.
`[11] F.P.M. O’Harte, M.H. Mooney, P.R. Flatt, N-terminally mod-
`ified gastric inhibitory polypeptide exhibits amino-peptidase
`resistance and enhanced antihyperglycaemic activity, Diabetes
`48 (1999) 758–765.
`[12] T.J. Kieffer, C.H.S. McIntosh, R.A. Pederson, Degradation of
`glucose-dependent insulinotropic polypeptide and truncated glu-
`cagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase
`IV, Endocrinology 136 (1995) 3585–3596.
`[13] J.C. Brown, M. Dahl, S. Kwauk, C.H.S. McIntosh, S.C. Otte,
`R.A. Pederson, Actions of GIP, Peptides 2 (1981) 241–245.
`[14] V.A. Gault, J.C. Parker, P. Harriott, P.R. Flatt, F.P.M. O’Harte,
`Evidence that the major degradation product of glucose-depen-
`dent
`insulinotropic polypeptide (GIP), GIP(3-42),
`is a GIP
`receptor antagonist in vivo, J. Endocrinol. 175 (2002) 525–533.
`[15] C.F. Deacon, P. Danielsen, L. Klarskov, M. Olesen, J.J. Holst,
`Dipeptidyl peptidase IV inhibition reduces the degradation and
`clearance of GIP and potentiates its insulinotropic and antihy-
`perglycemic effects in anesthetized pigs, Diabetes 50 (2001) 1588–
`1597.
`[16] J.A. Pospisilik, S.G. Stafford, H.-U. Demuth, R. Brownsey, W.
`Parkhouse, D.T. Finegood, C.H.S. McIntosh, R.A. Pederson,
`Long-term treatment with the dipeptidyl peptidase IV inhibitor
`P32/98 causes sustained improvements in glucose tolerance,
`insulin sensitivity, hyperinsulinemia, and b-cell glucose respon-
`siveness in VDF (fa/fa) Zucker rats, Diabetes 51 (2002) 943–
`950.
`[17] B. Ahren, E. Simonsson, H. Larsson, M. Landin-Olsson, H.
`Torgeirsson, P.A. Jansson, M. Sandqvist, P. Bavenholm, S.
`Efendic, J.W. Eriksson, S. Dickinson, D. Holmes, Inhibition of
`dipeptidyl peptidase IV improves metabolic control over a 4-week
`study period in type 2 diabetes, Diabetes Care 25 (2002)
`869–875.
`[18] R. Mentlein, Dipeptidyl-peptidase IV (CD26)—role in the inacti-
`vation of regulatory peptides, Regul. Pept. 85 (1999) 9–24.
`[19] M.A. Nauck, M.M. Heimesaat, C. Ørskov, J.J. Holst, R. Ebert,
`W. Creutzfeldt, Preserved incretin activity of glucagon-like pep-
`tide-1 (7-36amide), but not of synthetic human gastric inhibitory
`polypeptide in patients with type 2 diabetes mellitus, J. Clin.
`Invest. 91 (1993) 301–307.
`
`[20] M. Nauck, R. Stockman, R. Ebert, W. Creutzfeldt, Reduced
`incretin effect in type 2 (non-insulin-dependent) diabetes, Diabe-
`tologia 29 (1986) 46–52.
`[21] D. Elahi, M. McAloon-Dyke, N. Fukagawa, G. Meneilly, A.
`Sclater, K. Minaker, J. Habener, D. Andersen, The insulinotropic
`actions of glucose-dependent insulinotropic polypeptide (GIP)
`and glucagon-like peptide-1 (7-37)
`in normal and diabetic
`subjects, Regul. Pept. 51 (1994) 63–74.
`[22] I.R. Jones, D.R. Owens, A.J. Moody, S.D. Luzio, T. Morris, T.M.
`Hayes, The effects of glucose-dependent insulinotropic polypep-
`tide infused at physiological concentrations in normal subjects and
`type 2 (non-insulin-dependent) diabetic patients on glucose
`tolerance and B-cell secretion, Diabetologia 30 (1987) 707–712.
`[23] L.L. Kjems, J.J. Holst, A. Volund, S. Madsbad, The influence of
`GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell
`sensitivity in type 2 and nondiabetic subjects, Diabetes 52 (2003)
`380–386.
`[24] W.K. Ward, D.C. Bolgiano, B. McKnight, J.B. Halter, D. Porte
`Jr., Diminished B cell capacity in patients with noninsulin-
`dependent diabetes mellitus, J. Clin. Invest. 74 (1984) 1318–1328.
`[25] G.S. Meneilly, M. Bryer-Ash, D. Elahi, The effect of glyburide on
`beta-cell sensitivity to glucose-dependent insulinotropic polypep-
`tide, Diabetes Care 16 (1993) 110–114.
`[26] K. Almind, L. Ambye, S.A. Urhammer, T. Hansen, S.M.
`Echwald, J.J. Holst, J. Gromada, B. Thorens, O. Pedersen, Dis-
`covery of amino acid variants in the human glucose-dependent
`insulinotropic polypeptide (GIP) receptor: the impact on the
`pancreatic beta cell responses and functional expression studies in
`Chinese hamster fibroblast cells, Diabetologia 41 (1998) 1194–
`1198.
`[27] F.P.M. O’Harte, Y.H.A. Abdel-Wahab, J.M. Conlon, P.R. Flatt,
`Amino-terminal glycation of gastric inhibitory polypeptide en-
`hances its insulinotropic action on clonal pancreatic B-cells,
`Biochim. Biophys. Acta 1425 (1998) 319–327.
`[28] F.P.M. O’Harte, M.H. Mooney, C.M.N. Kelly, P.R. Flatt,
`Improved glycaemic control in obese diabetic ob/ob mice using
`N-terminally modified gastric inhibitory polypeptide, J. Endocri-
`nol. 165 (2000) 639–648.
`[29] F.P.M. O’Harte, V.A. Gault, J.C. Parker, P. Harriott, M.H.
`Mooney, C.J. Bailey, P.R. Flatt, Improved stability,
`insulin-
`releasing activity and antidiabetic potential of
`two novel
`N-terminal analogues of glucose-dependent insulinotropic poly-
`peptide: N-acetyl-GIP and pGlu-GIP, Diabetologia 45 (2002)
`1281–1291.
`[30] V.A. Gault, P.R. Flatt, C.J. Bailey, P. Harriott, B. Greer, M.H.
`Mooney, F.P.M. O’Harte, Enhanced cyclic AMP generation and
`insulin-releasing potency of two novel N-terminal Tyr1-modified
`enzyme resistant forms of GIP,
`is associated with significant
`antihyperglycaemic activity in spontaneous obesity-diabetes, Bio-
`chem. J. 367 (2002) 913–920.
`[31] V.A. Gault, P.R. Flatt, P. Harriott, M.H. Mooney, C.J. Bailey,
`F.P.M. O’Harte, Improved biological activity of Gly2- and Ser2-
`substituted analogues of glucose-dependent insulinotropic poly-
`peptide, J. Endocrinol. 176 (2003) 133–141.
`[32] V.A. Gault, F.P.M. O’Harte, P. Harriott, P.R. Flatt, Degrada-
`tion, cyclic AMP production, insulin secretion and glycemic effects
`of two novel N-terminal Ala2-substituted analogs of GIP with
`preserved biological activity in vivo, Metabolism 52 (2003) 679–
`687.
`[33] S.A. Hinke, R.W. Gelling, R.A. Pederson, S. Manhart, C. Nian,
`H.-U. Demuth, C.H.S. McIntosh, Dipeptidyl peptidase IV-resis-
`tant [DD-Ala2]glucose-dependent insulinotropic polypeptide (GIP)
`improves glucose tolerance in normal and obese diabetic rats,
`Diabetes 51 (2002) 656–661.
`[34] J.R. Lindsay, S.T.B. Au, C.M.N. Kelly, F.P.M. O’Harte, P.R.
`Flatt, P.M. Bell, A novel amino-terminally glycated analogue
`of glucose-dependent
`insulinotropic polypeptide (GIP), with
`
`MPI EXHIBIT 1084 PAGE 6
`
`MPI EXHIBIT 1084 PAGE 6
`
`

`

`V.A. Gault et al. / Biochemical and Biophysical Research Communications 308 (2003) 207–213
`
`213
`
`prolonged insulinotropic activity in type 2 diabetes mellitus
`[abstract], Diabetes 51 (Suppl. 2) (2002) A85.
`[35] J.J. Meier, M.A. Nauck, W.E. Schmidt, B. Gallwitz, Gastric
`inhibitory polypeptide: the neglected incretin revisited, Regul.
`Pept. 107 (2002) 1–7.
`[36] C.-C. Tseng, X.-Y. Zhang, M.M. Wolfe, Effect of GIP and GLP-1
`antagonists on insulin release in the rat, Am. J. Physiol. 276 (1999)
`1049–1054.
`[37] V.A. Gault, P.R. Flatt, P. Harriott, B.D. Green, F.P.M. O’Harte,
`Effects of the novel (Pro3)GIP antagonist and exendin (9-39)
`amide on GIP and GLP-1 induced cyclic AMP generation, insulin
`secretion and postprandial insulin release in obese diabetic (ob/ob)
`mice: evidence that GIP is the major physiological
`incretin,
`Diabetologia 46 (2003) 222–230.
`[38] R.A. Pederson, M. Satkunarajah, C.H.S. McIntosh, L.A. Scroc-
`chi, D. Flamez, F. Schuit, D.J. Drucker, M.B. Wheeler, Enhanced
`glucose-dependent insulinotropic polypeptide secretion and insu-
`linotropic action in glucagon-like peptide 1 receptor = mice,
`Diabetes 47 (1998) 1046–1052.
`[39] K. Miyawaki, Y. Yamada, H. Yano, H. Niwa, N. Ban, Y. Ihara,
`A. Kubota, S. Fujimoto, M. Kajikawa, A. Kuroe, K. Tsuda, H.
`Hashimoto, T. Yamashita, T. Jomori, F. Tashiro, J.I. Miyazaki,
`Y. Seino, Glucose tolerance caused by a defect in the enteroinsular
`axis: a study in gastric inhibitory polypeptide receptor knockout
`mice, Proc. Natl. Acad. Sci. USA 96 (1999) 14843–14847.
`[40] R. Burcelin, W. Dolci, B. Thorens, Long-lasting antidiabetic
`effects of a dipeptidyl peptidase IV-resistant analog of glucagon-
`like peptide-1, Metabolism 48 (1999) 252–258.
`[41] E.G. Siegel, B. Gallwitz, G. Scharf, R. Mentlein, C. Morys-
`Wortmann, U.R. Folsch, J. Schrezenmeir, K. Drescher, W.E.
`Schmidt, Biological activity of GLP-1-analogues with N-terminal
`modifications, Regul. Pept. 79 (1999) 93–102.
`[42] Q. Xiao, J. Giguere, M. Parisien, W. Jeng, S.A. St-Pierre, P.L.
`Brubaker, M.B. Wheeler, Biological activities of glucagon-like
`peptide-1 analogues in vitro and in vivo, Biochemistry 40 (2001)
`2860–2869.
`[43] B. Gallwitz, T. Ropeter, C. Morys-Wortmann, R. Mentlein, E.G.
`Siegel, W.E. Schmidt, GLP-1 analogues resistant to degradation by
`dipeptidyl-peptidase IV in vitro, Regul. Pept. 86 (2000) 103–111.
`[44] C.F. Deacon, L.B. Knudsen, K. Madsen, F.C. Wiberg, O.
`Jacobsen, J.J. Holst, Dipeptidyl peptidase IV resistant analogues
`of glucagon-like peptide-1 which have extended metabolic stability
`and improved biological activity, Diabetologia 41 (1998) 271–278.
`[45] R.A. Pederson, J.C. Brown, Interaction of gastric inhibitory
`polypeptide, glucose, and arginine on insulin and glucagon
`secretion from the perfused rat pancreas, Endocrinology 103
`(1978) 610–615.
`[46] J.J. Meier, B. Gallwitz, N. Siepmann, J.J. Holst, C.F. Deacon,
`W.E. Schmidt, M.A. Nauck, Gastric inhibitory polypeptide (GIP)
`dose-dependently stimulates glucagon secretion in healthy human
`subjects as euglycaemia, Diabetologia 46 (2003) 798–801, DOI
`10.1007/s00125-003-1103-y.
`[47] B. Kreymann, G. Williams, M.A. Ghatei, S.R. Bloom, Glucagon-
`like peptide-1 7-36: a physiological incretin in man, Lancet 2
`(1987) 1300–1304.
`[48] C.B. Juhl, M. Hollingdal, J. Sturis, G. Jakobsen, H. Agersø, J.
`Veldhuis, N. Pørksen, O. Schmitz, Bedtime administration of
`NN2211, a long-acting GLP-1 derivative, substantially reduces
`
`fasting and postprandial glycemia in type 2 diabetes, Diabetes 51
`(2002) 424–429.
`[49] V.A. Gault, P. Harriott, P.R. Flatt, F.P.M. O’Harte, Cyclic AMP
`production and insulin releasing activity of synthetic fragment
`peptides of glucose-dependent insulinotropic polypeptide, Biosci.
`Rep. 22 (2002) 523–528.
`[50] V.A. Gault, N. Irwin, P. Harriott, P.R. Flatt, F.P.M. O’Harte,
`DPP IV resistance and insulin releasing activity of a novel di-
`substituted analogue of glucose-dependent insulinotropic poly-
`peptide, (Ser2-Asp13)GIP, Cell Biol. Int. 27 (2003) 41–46.
`[51] K. K€uuhn-Wache, S. Manhart, T. Hoffmann, S.A. Hinke, R.
`Gelling, R.A. Pederson, C.H.S. McIntosh, H.-U. Demuth, Syn-
`thesis of analogs of glucose-dependent insulinotropic polypeptide
`with increased dipeptidyl peptidase IV resistance, Adv. Exp. Med.
`Biol. 477 (2000) 187–195.
`[52] S.A. Hinke, R. Gelling, S. Manhart, F. Lynn, R.A. Pederson, K.
`K€uuhn-Wache, F. Rosche, H.-U. Demuth, D. Coy, C.H.S.
`McIntosh, Structure–activity relationships of glucose-dependent
`insulinotropic polypeptide (GIP), Biol. Chem. 384 (2003) 403–
`407.
`[53] S. Manhart, S.A. Hinke, C.H. McIntosh, R.A. Pederson, H.U.
`Demuth, Structure–function analysis of a series of novel GIP
`analogues containing different helical length linkers, Biochemistry
`42 (2003) 3081–3088.
`[54] S.A. Hinke, S. Manhart, N. Pamir, H. Demuth, R.W. Gelling,
`R.A. Pederson, C.H. McIntosh, Identification of a bioactive
`domain in the amino-terminus of glucose-dependent insulino-
`tropic polypeptide (GIP), Biochim. Biophys. Acta 1547 (2001)
`143–155.
`[55] V.A. Gault, F.P.M. O’Harte, P. Harriott, P.R. Flatt, Character-
`ization of
`the cellular and metabolic effects of a novel
`enzyme-resistant antagonist of glucose-dependent insulinotropic
`polypeptide, Biochem. Biophys. Res. Commun. 290 (2002) 1420–
`1426.
`[56] S. Gremlich, A. Porret, E.H. Hani, D. Cherif, N. Vionnet, P.
`Froguel, B. Thorens, Cloning, functional expression, and chro-
`mosomal
`localization of the human pancreatic islet glucose-
`dependent insulinotropic polypeptide receptor, Diabetes 44 (1995)
`1202–1208.
`[57] N.H. McClenaghan, C.R. Barnett, E. Ah-Sing, Y.H.A. Abdel-
`Wahab, F.P.M. O’Harte, T.-W. Yoon, S.K. Swanston-Flatt, P.R.
`Flatt, Characterization of a novel glucose-responsive insulin-
`secreting cell
`line, BRIN-BD11, produced by electrofusion,
`Diabetes 45 (1996) 1132–1140.
`[58] K. Miyawaki, Y. Yamada, N. Ban, Y. Ihara, K. Tsukiyama, K.
`Zhou, S. Fujimoto, A. Oku, K. Tsuda, S. Toyokuni, H. Hiai, W.
`Mizunoya, T. Fushiki, J.J. Holst, M. Makino, A. Tashita, Y.
`Kobara, Y.

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