US20080009603A1 - Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity - Google Patents

Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity Download PDF

Info

Publication number
US20080009603A1
US20080009603A1 US11/825,293 US82529307A US2008009603A1 US 20080009603 A1 US20080009603 A1 US 20080009603A1 US 82529307 A US82529307 A US 82529307A US 2008009603 A1 US2008009603 A1 US 2008009603A1
Authority
US
United States
Prior art keywords
gip
insulin
glucose
mice
pro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/825,293
Inventor
Victor Gault
Finbarr O'Harte
Nigel Irwin
Peter Flatt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UUTech Ltd
Original Assignee
UUTech Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB9907216A external-priority patent/GB9907216D0/en
Priority claimed from GB9917565A external-priority patent/GB9917565D0/en
Priority claimed from PCT/GB2000/001089 external-priority patent/WO2000058360A2/en
Priority claimed from GBGB0404124.0A external-priority patent/GB0404124D0/en
Application filed by UUTech Ltd filed Critical UUTech Ltd
Priority to US11/825,293 priority Critical patent/US20080009603A1/en
Publication of US20080009603A1 publication Critical patent/US20080009603A1/en
Assigned to UUTECH LIMITED reassignment UUTECH LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HARRIOTT, PATRICK, O'HARTE, FINBARR PAUL MARY, FLATT, PETER RAYMOND, GAULT, VICTOR A, IRWIN, NIGEL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T29/00Metal working
    • Y10T29/49Method of mechanical manufacture
    • Y10T29/4935Heat exchanger or boiler making

Definitions

  • the present invention relates to the release of insulin and the control of blood glucose concentration. More particularly the invention relates to antagonists of gastric inhibitory peptide (GIP) as pharmaceutical preparations for treatment of type 2 diabetes.
  • GIP gastric inhibitory peptide
  • Obesity and diabetes are predicted to reach epidemic proportions throughout the world in the next 20 years and current treatments do not restore normal insulin sensitivity or glucose homeostasis, therein resulting in debilitating diabetic complications and premature death.
  • GIP Gastric inhibitory polypeptide
  • tGLP-1 glucagon-like peptide-1(7-36)amide
  • GIP is released from intestinal endocrine K-cells into the bloodstream following ingestion of carbohydrate, protein and particularly fat (Meier, J. J. et al., 2002 , Regul. Pept. 107:1-13). GIP was initially discovered through its ability to inhibit gastric acid secretion (Brown, J. C. et al. 1969 , Can. J. Physiol. Pharmacol. 47:113-114) but its major physiological role is now generally believed to be that of an incretin hormone that targets pancreatic islets to enhance insulin secretion and help reduce postprandial hyperglycemia (Creutzfeldt, W., 2001 , Exp. Clin. Endocrinol. Diabetes 109:S288-S303).
  • GIP acts through binding to specific G-protein coupled GIP receptors located on pancreatic beta-cells (Wheeler, M. B. et al., 1995 , Endocrinology 136:4629-4639).
  • GLP-1 glucagon-like peptide-1
  • this ability to stimulate insulin secretion plus other potentially beneficial actions on pancreatic beta-cell growth and differentiation have led to much interest in using GIP or GLP-1 receptor agonists in the treatment of type 2 diabetes (Creutzfeldt, W., 2001 , Exp. Clin. Endocrinol. Diabetes 109:S288-S303; Holz, G. G. et al., 2003 , Curr. Med. Chem. 10:2471-2483).
  • GIP functions as a potent and natural stimulator of insulin secretion released from the intestine by feeding, it is widely expected that antagonists opposing GIP action will block the insulin-releasing actions of GIP and impair both oral glucose tolerance and the glycemic response to nutrient ingestion.
  • GIP is a key physiological component of the enteroinsular axis and that functional ablation of GIP leads to impaired glucose homeostasis moving the metabolic characteristic towards a type 2 diabetes phenotype (Gault, V. A. et al., 2002 , Biochem. Biophys. Res. Commun. 290:1420-1426).
  • Dipeptidyl peptidase IV (DPP IV; EC 3.4.14.5) has been identified as a key enzyme responsible for inactivation of GIP and tGLP-1 in serum. This occurs through the rapid removal of the N-terminal dipeptides Tyr 1 -Ala 2 and His 7 -Ala 8 giving rise to the main metabolites GIP(3-42) and GLP-1(9-36)amide, respectively. These truncated peptides are reported to lack biological activity or to even serve as antagonists at GIP or tGLP-1 receptors. The resulting biological half-lives of these incretin hormones in vivo are therefore very short, estimated to be no longer than 5 minutes. DPP IV is completely inhibited in serum by the addition of diprotin A (DPA, 0.1 mmol/1).
  • DPA diprotin A
  • tGLP-1 infusion restores pancreatic B-cell sensitivity, insulin secretory oscillations and improved glycemic control in various groups of patients with impaired glucose tolerance (IGT) or NIDDM.
  • ITT impaired glucose tolerance
  • NIDDM impaired glucose tolerance
  • Longer term studies also show significant benefits of tGLP-1 injections in NIDDM and possibly IDDM therapy, providing a major incentive to develop an orally effective or long-acting tGLP-1 analogue.
  • Several attempts have been made to produce structurally modified analogues of tGLP-1 which are resistant to DPP IV degradation.
  • GIP shares not only the same degradation pathway as tGLP-1 but many similar physiological actions, including stimulation of insulin and somatostatin secretion, and the enhancement of glucose disposal. These actions are viewed as key aspects in the antihyperglycemic properties of tGLP-1, and there is therefore good expectation that GIP may have similar potential as NIDDM therapy. Indeed, compensation by GIP is held to explain the modest disturbances of glucose homeostasis observed in tGLP-1 knockout mice. Apart from early studies, the anti-diabetic potential of GIP has not been explored and tGLP-1 may seem more attractive since it is viewed by some as a more potent insulin secretagogue when infused at so called physiological concentrations estimated by radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • GIP antagonist peptides which are resistant to rapid degradation by DPP IV.
  • the invention includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), which includes at least 12 amino acid residues from the N-terminal end of GIP(3-42).
  • the invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), which includes at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu 3 .
  • the amino acid substituted at Glu 3 can be selected from the group consisting of: proline, hydroxyproline, lysine, tyrosine, phenylalanine and tryptophan. Specifically, a proline can be substituted for Glu 3 .
  • the peptide analogue can further include modification by fatty acid addition at an epsilon amino group of at least one lysine residue.
  • the lysine residue can be Lys 16 , or Lys 37
  • the peptide analogue of GIP(1-42) can include at least 12 amino acid residues from the N-terminal end of GIP(1-42), and an amino acid modification at amino acid residues 1, 2 or 3.
  • the N-terminal amino acid residue can be acetylated. It can further comprising modification by fatty acid addition at an epsilon amino group of at least one lysine residue.
  • the modification can be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-16, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue.
  • the lysine residue can be Lys 16 , or Lys 37 .
  • the invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), wherein the analogue comprises a base peptide consisting of one of the following: GIP(1-12), GIP(1-13), GIP(1-14), GIP(1-15), GIP(1-16), GIP(1-17), GIP(1-18), GIP(1-19), GIP(1-20), GIP(1-21), GIP(1-22), GIP(1-23), GIP(1-24), GIP(1-25), GIP(1-26), GIP(1-27), GIP(1-28), GIP(1-29), GIP(1-30), GIP(1-31), GIP(1-32), GIP(1-33), GIP(1-34), GIP(1-35), GIP(1-36), GIP(1-37), GIP(1-38), GIP(1-39), GIP(1-40), GIP(1-41) and GIP(1-42), where the base peptide possesses one or more of the following modifications: (1) an amino acid substitution
  • Such a peptide analogue can have a proline substituted for Glu 3 . It can also have a modification in the form of a C-16 palmitate group linked to the epsilon amino group of a lysine residue.
  • the modification can be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-16, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue.
  • the lysine residue can be Lys 16 , or Lys 37 .
  • the invention further includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42), wherein the peptide analogue is resistant to degradation by enzyme DPP IV when compared to naturally-occurring GIP.
  • SEQ ID NO:1 a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42), wherein the peptide analogue is resistant to degradation by enzyme DPP IV when compared to naturally-occurring GIP.
  • peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu 3 , wherein the peptide analogue is resistant to degradation by enzyme DPP IV when compared to naturally-occurring GIP.
  • the invention includes a peptide analogue of GIP(142) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42), wherein the peptide analogue modulates insulin secretion.
  • the invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu 3 , wherein the peptide analogue modulates insulin secretion.
  • SEQ ID NO:1 a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu 3 , wherein the peptide analogue modulates insulin secretion.
  • the invention also includes use of any of the analogues in the preparation of a medicament for the treatment of obesity, insulin resistance, insulin resistant metabolic syndrome (Syndrome X) or type 2 diabetes.
  • the invention also includes a pharmaceutical composition including the peptide analogues.
  • the pharmaceutical composition can further comprise a pharmaceutically acceptable carrier.
  • the peptide analogue can be in the form of a pharmaceutically acceptable salt, or a pharmaceutically acceptable acid addition salt.
  • the invention includes a method of treating insulin resistance, obesity, or type 2 diabetes, where the method comprises administering to a mammal in need of such treatment a therapeutically effective amount of the pharmaceutical composition.
  • an effective peptide analogue of the biologically active GIP(1-42) which has improved characteristics for treatment of Type 2 diabetes wherein the analogue comprises at least 15 amino acid residues from the N terminus of GIP(1-42) and has at least one amino acid substitution or modification at position 1-3 and not including Tyr 1 glucitol GIP(1-42).
  • the structures of human and porcine GIP(1-42) are shown below.
  • the porcine peptide differs by just two amino acid substitutions at positions 18 and 34.
  • the analogue may include modification by fatty acid addition at an epsilon amino group of at least one lysine residue.
  • the invention includes Tyr 1 glucitol GIP(1-42) having fatty acid addition at an epsilon amino group of at least one lysine residue.
  • Analogues of GIP(1-42) may have an enhanced capacity to stimulate insulin secretion, enhance glucose disposal, delay glucose absorption or may exhibit enhanced stability in plasma as compared to native GIP. They also may have enhanced resistance to degradation.
  • Analogues having D-amino acid substitutions in the 1, 2 and 3 positions and/or N-glycated, N-alkylated, N-acetylated or N-acylated amino acids in the 1 position are resistant to degradation in vivo.
  • amino acid substitutions at second and third amino terminal residues are included, such as GIP(1-42)Gly 2 , GIP(142)Ser 2 , GIP(1-42)Abu 2 , GIP(1-42)Aib 2 , GIP(1-42)D-Ala 2 , GIP(1-42)Sar 2 , and GIP(1-42)Pro 3 .
  • Amino-terminally modified GIP analogues include N-glycated GIP(1-42), N-alkylated GIP(1-42), N-acetylated GIP(1-42), N-acetyl-GIP(1-42) and N-isopropyl GIP(1-42).
  • DPP IV dipeptidyl-peptidase IV
  • the invention provides a peptide which is more potent than human or porcine GIP in moderating blood glucose excursions, said peptide consisting of GIP(1-42) or N-terminal fragments of GIP(1-42) consisting of up to between 15 to 30 amino acid residues from the N-terminus (i.e., GIP(1-15) GIP(1-3)) with one or more modifications selected from the group consisting of:
  • the invention also provides the use of Tyr 1 -glucitol GIP in the preparation of a medicament for the treatment of diabetes.
  • the invention further provides improved pharmaceutical compositions including analogues of GIP with improved pharmacological properties.
  • analogues include certain commonly encountered amino acids, which are not encoded by the genetic code, for example, beta-alanine (beta-ala), or other omega-amino acids, such as 3-amino propionic, 4-amino butyric and so forth, ornithine (Orn), citrulline (Cit), homoarginine (Har), t-butylalanine (t-BuA), t-butylglycine (t-BuG), N-methylisoleucine (N-MeIle), phenylglycine (Phg), and cyclohexylalanine (Cha), norleucine (Nle), cysteic acid (Cya) and methionine sulfoxide (MSO), substitution of the D form of a neutral or acidic amino acid or the D form of tyrosine for tyrosine.
  • beta-alanine beta-ala
  • omega-amino acids such as 3-amino propionic, 4-amin
  • composition useful in the treatment of diabetes type II which comprises an effective amount of the peptide as described herein, in admixture with a pharmaceutically acceptable excipient.
  • the invention also provides a method of N-terminally modifying GIP or analogues thereof the method comprising the steps of synthesizing the peptide from the C terminal to the penultimate N terminal amino acid, adding tyrosine to a bubbler system as a F-moc protected Tyr(tBu)-Wang resin, deprotecting the N-terminus of the tyrosine and reacting with the modifying agent, allowing the reaction to proceed to completion, cleaving the modified tyrosine from the Wang resin and adding the modified tyrosine to the peptide synthesis reaction.
  • the agent is glucose, acetic anhydride or pyroglutamic acid.
  • FIG. 1 a illustrates degradation of GIP by DPP IV.
  • FIG. 1 b illustrates degradation of GIP and Tyr 1 glucitol GIP by DPP IV.
  • FIG. 2 a illustrates degradation of GIP human plasma.
  • FIG. 2 b illustrates degradation of GIP and Tyr 1 glucitol GIP by human plasma.
  • FIG. 3 illustrates electrospray ionization mass spectrometry of GIP, Tyr 1 -glucitol GIP and the major degradation fragment GIP(3-42).
  • FIG. 4 shows the effects of GIP and glycated GIP on plasma glucose homeostasis.
  • FIG. 5 shows effects of GIP on plasma insulin responses.
  • FIG. 6 illustrates DPP-IV degradation of GIP (1-42).
  • FIG. 7 illustrates DPP-IV degradation of GIP (Abu 2 ).
  • FIG. 8 illustrates DPP-IV degradation of GIP (Sar 2 ).
  • FIG. 9 illustrates DPP-IV degradation of GIP (Ser 2 ).
  • FIG. 10 illustrates DPP-IV degradation of N-Acetyl-GIP.
  • FIG. 11 illustrates DPP-IV degradation of glycated GIP.
  • FIG. 12 illustrates human plasma degradation of GIP.
  • FIG. 13 illustrates human plasma degradation of GIP (Abu 2 ).
  • FIG. 14 illustrates human plasma degradation of GIP (Sar 2 ).
  • FIG. 15 illustrates human plasma degradation of GIP (Ser 2 ).
  • FIG. 16 illustrates human plasma degradation of glycated GIP.
  • FIG. 17 shows the effects of various concentrations of GIP 1-42 and GIP (Abu 2 ) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 18 shows the effects of various concentrations of GIP 1-42 and GIP (Abu 2 ) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 19 shows the effects of various concentrations of GIP 1-42 and GIP (Sar 2 ) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 20 shows the effects of various concentrations of GIP 1-42 and GIP (Sar 2 ) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 21 shows the effects of various concentrations of GIP 1-42 and GIP (Ser 2 ) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 22 shows the effects of various concentrations of GIP 1-42 and GIP (Ser 2 ) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 23 shows the effects of various concentrations of GIP 1-42 and N-Acetyl-GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 24 shows the effects of various concentrations of GIP 1-42 and N-Acetyl-GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 25 shows the effects of various concentrations of GIP 1-42 and glycated GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 26 shows the effects of various concentrations of GIP 1-42 and glycated GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 27 shows the effects of various concentrations of GIP 1-42 and GIP (Gly 2 ) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 28 shows the effects of various concentrations of GIP 1-42 and GIP (Gly 2 ) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 29 shows the effects of various concentrations of GIP 1-42 and GIP (Pro 3 ) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 30 shows the effects of various concentrations of CIP 1-42 and GIP (Pro 3 ) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 31 a shows the primary structure of human gastric inhibitory polypeptide (GIP) (SEQ ID NO:1)
  • FIG. 31 b shows the primary structure of porcine gastric inhibitory polypeptide (GIP) (SEQ ID NO:2).
  • FIGS. 32A and 32B are a line graph and a bar graph, respectively, showing the effects of (Pro 3 )GIP on GIP-stimulated cyclic AMP generation and insulin secretion in vitro.
  • FIGS. 33A-33F are a set of six bar graphs showing the effects of Glu 3 -substituted forms of GIP and GIP(3-42) on GIP-stimulated insulin secretion in vitro.
  • FIGS. 34A through 34D are a set of two line graphs ( FIGS. 34A, 34C ) and two bar graphs ( FIGS. 34B, 34D ) showing that acute administration of (Pro 3 )GIP completely antagonises the actions of GIP on glucose tolerance ( FIGS. 34A, 34B ) and plasma insulin ( FIGS. 34C, 34D ) responses in obese diabetic ob/ob mice.
  • FIGS. 35A through 35D are a set of two line graphs ( FIGS. 35A, 35C ) and two bar graphs ( FIGS. 35B, 35D ) showing that acute administration of (Pro 3 )GIP impairs physiological meal-stimulated insulin release and worsens glycemic excursion in obese diabetic ob/ob mice.
  • FIGS. 36A and 36B are a set of two bar graphs showing that chronic administration of (Pro 3 )GIP for 11 days decreases plasma glucose and insulin concentrations of obese diabetic ob/ob mice.
  • FIGS. 37A through 37C are a set of three bar graphs showing that chronic administration of (Pro 3 )GIP for 11 days decreases glycated HbA 1c , pancreatic insulin content and associated islet hypertrophy of obese diabetic ob/ob mice.
  • FIGS. 38A through 38D are a set of two line graphs ( FIGS. 38A, 38C ) and two bar graphs ( FIGS. 38B, 38D ) showing that chronic administration of (Pro 3 )GIP for 11 days improves glucose tolerance of obese diabetic ob/ob mice without change of circulating insulin.
  • FIG. 39 is a line graph showing that chronic administration of (Pro 3 )GIP for 11 days improves insulin sensitivity in obese diabetic ob/ob mice.
  • FIG. 40 is a line graph showing that the beneficial effects of chronic administration of (Pro 3 )GIP for 11 days in obese diabetic ob/ob mice are reversed 9 days after cessation of treatment.
  • FIGS. 41A and 41B are a set of two line graphs showing that chronic administration of (Pro 3 )GIP for 11 days causes glucose intolerance in normal mice with reversal by 9 days after cessation of treatment.
  • FIGS. 42A through 42D are a set of two line graphs ( FIGS. 42A, 42C ) and two bar graphs ( FIGS. 42B, 42D ) showing the effects of (Pro 3 )GIP on plasma glucose and insulin response to native GIP 4 hours after administration.
  • FIGS. 43A through 43D are a set of two line graphs and two bar graphs showing the effects of daily (Pro 3 )GIP administration on food intake ( FIG. 43A ), body weight ( FIG. 43B ), plasma glucose ( FIG. 43C ) and insulin ( FIG. 43D ) concentrations in ob/ob mice.
  • FIGS. 44A through 44D are a set of four line graphs with inset bar graphs showing the effects of daily (Pro 3 )GIP administration on glucose tolerance and plasma insulin response to glucose in ob/ob mice.
  • FIGS. 45A through 45D are a set of two line graphs ( FIGS. 45A, 45C ) and two bar graphs ( FIGS. 45B, 45D ) showing the effects of daily (Pro 3 )GIP administration ( ⁇ ; black bars) or saline ( ⁇ ; white bars) on glucose ( FIGS. 45A, 45B ) and insulin ( FIGS. 45C, 45D ) responses to feeding in ob/ob mice fasted for 18 hours.
  • FIGS. 46A through 46D are a set of two line graphs ( FIGS. 46A, 46C ) and two bar graphs ( FIGS. 46B, 46D ) showing the effects of daily (Pro 3 )GIP administration on insulin sensitivity in ob/ob mice.
  • FIGS. 47A through 47D are a set of four bar graphs showing the effects of daily (Pro 3 )GIP administration on pancreatic weight ( FIG. 47A ), insulin content ( FIG. 47B ), islet number ( FIG. 47C ) and islet diameter ( FIG. 47D ) in ob/ob mice.
  • FIGS. 48A through 48F are a set of two bar graphs ( FIGS. 48A, 48D ) and four photomicrographs ( FIGS. 48B, 48C , 48 E, 48 F), showing the effects of daily (Pro 3 )GIP administration on islet size and morphology in ob/ob)mice.
  • FIG. 49 is an illustration of how the GIP receptor (“GIP-R”) antagonist, (Pro 3 )GIP, counters beta cell hyperplasia, hyperinsulinemia and insulin resistance lead to improved glucose intolerance and diabetes control.
  • GIP-R GIP receptor
  • Pro 3 GIP receptor
  • FIGS. 50A and 50B are a line graph and a bar graph, respectively, showing intracellular cyclic AMP production ( FIG. 50A ) by GIP ( ⁇ ) and fatty acid derivatised GIP analogues N-AcGIP(LysPAL 16 ) ( ⁇ ) and N-AcGIP(LysPAL 37 ) ( ⁇ ), and insulin-releasing activity of glucose (5.6 mmo/l glucose; white bars), GIP (lined bars) and fatty acid derivatised GIP analogues ( FIG. 50B ) N-AcGIP(LysPAL 16 ) (grey bars) and N-AcGIP(LysPAL 37 ) (black bars) in the clonal pancreatic beta cell line, BRIN-BD11.
  • FIGS. 51A through 51D are a set of two line graphs ( FIGS. 51A, 51C ) and two bar graphs ( FIGS. 51B, 51D ) showing glucose lowering effects ( FIGS. 51A, 51B ) and insulin-releasing activity ( FIGS. 51C, 51D ) of GIP and fatty acid derivatised GIP analogues in 18 hour-fasted ob/ob mice.
  • FIGS. 52A and 52B are a pair of bar graphs showing dose-dependent effects of GIP and N-AcGIP(LysPAL 37 ) in ob/ob mice fasted for 18 hours.
  • FIGS. 53A through 53E are a set of graphs showing the effects of daily N-AcGIP(LysPAL 37 ) ( ⁇ ; black bars) administration on food intake ( FIG. 53A ), body weight ( FIG. 53B ), plasma glucose ( FIG. 53C ), insulin ( FIG. 53D ) and glycated hemoglobin N-AcGIP(LysPAL 37 ) (12.5 nmoles/kg/day) ( FIG. 53E ).
  • FIGS. 54A through 54D are a set of two line graphs ( FIGS. 54A, 54C ) and two bar graphs ( FIGS. 54B, 54D ) showing the effects of daily N-AcGIP(LysPAL 37 ) administration on glucose tolerance ( FIGS. 54A, 54B ) and plasma insulin response ( FIGS. 54C, 54D ) to glucose.
  • FIGS. 55A through 55D are a line graph and three bar graphs showing the effects of daily N-AcGIP(LysPAL 37 ) administration on insulin sensitivity ( FIGS. 55A, 55B ) and pancreatic weight ( FIG. 55C ) and insulin content ( FIG. 55D ).
  • FIGS. 56A through 56D are a set of two line graphs ( FIGS. 56A, 56C ) and two bar graphs ( FIGS. 56B, 56D ) showing the retention of glucose lowering ( FIGS. 56A, 56B ) and insulin releasing ( FIGS. 56C, 56D ) activity of N-AcGIP(LysPAL 37 ) and native GIP after daily injection for 14 days.
  • FIGS. 57A through 57D are a set of two line graphs ( FIGS. 57A, 57C ) and two bar graphs ( FIGS. 57B, 57D ) showing the acute glucose lowering ( FIGS. 57A, 57B ) and insulin releasing ( FIGS. 57C, 57D ) effects of N-AcGIP(LysPAL 37 ) after 14 daily injections of either N-AcGIP(LysPAL 37 ) (12.5 nmoles/kg/day; ⁇ ; black bars), native GIP (12.5 nmoles/kg/day; ⁇ ; lined bars) or saline vehicle (control; ⁇ ; white bars).
  • the peptide analogues disclosed herein display resistance to degradation by the enzyme DPP IV. These analogues include those with alterations at residues 1, 2 and/or 3 of the native GIP(1-42) peptide, where the alterations interfere with or delay cleavage by DPP IV.
  • the alterations can include chemical modification of one or more of the first three amino acids, such as by acylation, acetylation, alkylation, glycation, conversion of a bond between two amino acids, such as to a psi-[CH 2 NH] bond, or to a stable isotere bond, or addition of an isopropyl group.
  • the alterations can also include amino acid substitutions at the 1, 2, and/or 3 position, to either a different naturally-occurring amino acid, or an amino acid not encoded by the genetic code.
  • Other alterations are also possible, and the object is to prevent cleavage of the peptide by DPP IV, yet still allow for insulin secretion. This may be accomplished by alterations at other regions of the peptide, such as by alterations that alter the three-dimensional structure to prevent DPP IV cleavage, yet still allow insulin secretion.
  • Preferred alterations include chemical modifications of residues 1, 2, and 3, amino acid substitutions at residues 1, 2, and 3, and chemical modifications of lysine residues throughout the protein.
  • Particularly preferred alterations include acetylation of Tyr 1 and linkage of a C-16 palmitate group to the epsilon amino group of a lysine residue (especially Lys 16 or Lys 37 ), or substitution of Glu 3 , especially by proline.
  • the modification can also be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue.
  • GIP receptor antagonism using Glu 3 -substituted forms of GIP, such as (Pro 3 )GIP, improve obesity-related insulin resistance and associated glucose intolerance. This has uncovered an unexpected approach to the therapy of obesity, insulin resistance and type 2 diabetes.
  • Example 2 As described in Example 1 below, an N-terminally modified version of the GIP protein was prepared, as were analogues of the modified protein. The protein and its analogues were then evaluated in Example 2 for their antihyperglycemic and insulin-releasing properties in vivo, and were found to exhibit a substantial resistance to amino peptidase degradation and increased glucose lowering activity relative to native GIP.
  • the 42 amino acid GIP is an important incretin hormone released into the circulation from endocrine K-cells of the duodenum and jejunum following ingestion of food.
  • the high degree of structural conservation of GIP among species supports the view that this peptide plays an important role in metabolism. Secretion of GIP is stimulated directly by actively transported nutrients in the gut lumen without a notable input from autonomic nerves.
  • the most important stimulants of GIP release are simple sugars and unsaturated long chain fatty acids, with amino acids exerting weaker effects.
  • the insulin-releasing effect of GIP is strictly glucose-dependent. This affords protection against hypoglycemia and thereby fulfills one of the most desirable features of any current or potentially new antidiabetic drug.
  • Tyr 1 -glucitol GIP significantly enhanced the antihyperglycemic activity and insulin-releasing action of the peptide when administered with glucose to rats.
  • Native GIP enhanced insulin release and reduced the glycemic excursion as observed in many previous studies.
  • amino-terminal glycation of GIP increased the insulin-releasing and antihyperglycemic actions of the peptide by 62% and 38% respectively, as estimated from insulin area under the curve (AUC) measurements.
  • AUC insulin area under the curve
  • GIP glycated GIP is enhancing insulin secretion in vivo both by enhanced potency at the receptor as well as improving DPP IV resistance.
  • GIP (3-42) and other N-terminally modified fragments, including GIP(4-42), and GIP(17-42) are either weakly effective or inactive in stimulating insulin release.
  • N-terminal deletions of GIP result in receptor antagonist properties in GIP receptor transfected Chinese hamster kidney cells [9], suggesting that inhibition of GIP catabolism would also reduce the possible feedback antagonism at the receptor level by the truncated GIP(3-42).
  • GIP In addition to its insulinotopic actions, a number of other potentially important extrapancreatic actions of GIP may contribute to the enhanced antihyperglycemic activity and other beneficial metabolic effects of Tyr 1 -glucitol GIP. These include the stimulation of glucose uptake in adipocytes, increased synthesis of fatty acids and activation of lipoprotein lipase in adipose tissue. GIP also promotes plasma triglyceride clearance in response to oral fat loading. In liver, GIP has been shown to enhance insulin-dependent inhibition of glycogenolysis. GIP also reduces both glucagon-stimulated lipolysis in adipose tissue as well as hepatic glucose production.
  • GIP has a potent effect on glucose uptake and metabolism in mouse isolated diaphragm muscle. This latter action may be shared with tGLP-1 and both peptides have additional benefits of stimulating somatostatin secretion and slowing down gastric emptying and nutrient absorption.
  • glycated GIP, acetylated GIP, GIP(Ser 2 ) are GIP(Abu 2 ) more resistant than native GIP to in vitro degradation with DPP IV. From these data GIP(Sar 2 ) appears to be less resistant. As shown in Table 2, all analogues tested exhibited resistance to plasma degradation, including GIP(Sar 2 ) which from DPP IV data appeared least resistant of the peptides tested. DPA substantially inhibited degradation of GIP and all analogues tested with complete abolition of degradation in the cases of GIP(Abu 2 ), GIP(Ser 2 ) and glycated GIP. This indicates that DPP IV is a key factor in the in vivo degradation of GIP.
  • the glycated GIP analogue exhibited a considerably greater insulinotropic response relative to native GIP.
  • N-terminal acetylated GIP exhibited a similar pattern and the GIP(Ser 2 ) analogue also evoked a strong response.
  • GIP(Gly 2 ) and GIP(Pro 3 ) appeared to the least potent analogues in terms of insulin release.
  • Other stable analogues tested namely GIP(Abu 2 ) and GIP(Sar 2 ), exhibited a complex pattern of responsiveness dependent on glucose concentration and dose employed.
  • very low concentrations were extremely potent under hyperglycemic conditions (16.7 mM glucose). This suggests that even these analogues may prove therapeutically useful in the treatment of type 2 diabetes where insulinotropic capacity combined with in vivo degradation dictates peptide potency.
  • DPP-IV dipeptidylpeptidase-IV
  • (Pro 3 )GIP other Glu 3 -substituted forms of GIP and GIP(3-42) are potent GIP receptor antagonists both in vitro and in vivo.
  • GLP-1 insulin secretory response to all secretagogues, including GLP-1 is compromised in type 2 diabetes (Kjems, L. L. et al., 2003 , Diabetes 52:380-386; Flatt, P. R. et al., “Defective insulin secretion in diabetes and insulinoma,” in Nutrient regulation of insulin secretion, Flatt P. R., ed. London, Portland Press, 1992, p. 341-386).
  • GLP-1 and GIP for diabetes therapy is reliant on peptide engineering to provide analogs of incretin hormones with improved potency due to DPP IV resistance, decreased renal clearance and/or enhanced GIP receptor and post-receptor activity (Gault, V. A. et al., 2003, Biochem Biophys Res Commun 308:207-213).
  • GIP-R GIP receptor
  • Example 5 daily injections of the stable and specific GIP-R antagonist, (Pro 3 )GIP can be used to chemically ablate the GIP-R and evaluate the role of endogenous circulating GIP in obesity-diabetes as manifested in ob/ob mice.
  • the results reveal a cardinal role for GIP in insulin resistance and associated metabolic disturbances, and provide the first experimental evidence that GIP-R antagonists might provide a novel and effective means of treating obesity-driven forms of type 2 diabetes.
  • the analogue (Pro 3 )GIP can be used as a specific and potent antagonist of the GIP-R that is highly stable and resistant to DPP IV-mediated degradation (Gault, V. A. et al., 2002 , Biochem. Biophys. Res. Commun. 290:1420-1426).
  • (Pro 3 )GIP acutely, the results disclosd herein highlight the involvement of GIP in the plasma insulin response to feeding and the enteroinsular axis of ob/ob mice (Gault, V. A. et al., 2003 , Diabetologia 46:222-230).
  • FIG. 49 is an illustration of how the GIP-R antagonist, (Pro 3 )GIP, counters beta cell hyperplasia, hyperinsulinemia and insulin resistance lead to improved glucose intolerance and diabetes control.
  • GIP-R antagonist Pro 3
  • GIP-R knockout ob/ob mice Possible longer-term direct actions of GIP on adipocyte function and fat stores, suggested by studies in GIP-R knockout ob/ob mice have been omitted.
  • Example 5 The results shown in Example 5 have demonstrated for the first time that daily administration of the GIP-R antagonist, (Pro 3 )GIP, improves glucose tolerance and ameliorates insulin resistance and abnormalities of islet structure and function in ob/ob mice. Notably, these effects were reversed by discontinuation of (Pro 3 )GIP for 9 days. Freedom from any obvious side effects also accords with earlier observations in normal mice (Irwin, N., 2004 , Biol. Chem. 385:845-852) and mice genetically engineered with life-long GIP-R deficiency (Miyawaki, K. et al., 2002 , Nat. Med. 8:738-742).
  • GIP-R antagonists such as (Pro 3 )GIP, provide a novel, physiological and effective means to treat obese type 2 diabetes through the alleviation of insulin resistance.
  • Example 6 fatty acid derivatisation was used to develop two novel long-acting, N-terminally modified GIP analogues (N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 )).
  • DPP IV dipeptidylpeptidase IV
  • Cyclic AMP production was assessed using GIP receptor transfected CHL fibroblasts.
  • In vitro insulin release was assessed in BRIN-BD11 cells.
  • Insulinotropic and glycaemic responses to acute and long-term peptide administration were evaluated in obese diabetic (ob/ob) mice.
  • Fatty acid derivatisation has previously been shown to prolong the half-life of insulin (Kurtzhals, P. et al., 1995 , Biochem. J. 312: 725-731) and the sister incretin glucagon-like peptide-1 (GLP-1) (Knudsen, L. B. et al., 2000 , J. Med. Chem. 43: 1664-1669; Green, B. D. et al., 2004 , Biol. Chem. 385: 169-177; Kim, J. G. et al., 2003 , Diabetes 52: 751-759).
  • N-terminally modified GIP analogues have been developed which exhibit profound resistance to DPP IV (Hinke, S. A. et al., 2002 , Diabetes 51: 656-661; Gault, V. A. et al., 2002 , Biochem. J. 367: 913-920; Gault, V. A. et al., 2003 , J. Endocrinol. 176: 133-141; O'Harte, F. P. M. et al., 1999 , Diabetes 48: 758-765).
  • these analogues display enhanced and protracted antihyperglycaemic and insulin-releasing activity when administered acutely to animals with obesity-diabetes (Hinke, S. A. et al., 2002 , Diabetes 51: 656-661; Gault, V. A. et al., 2002 , Biochem. J. 367: 913-920; Gault, V. A. et al., 2003 , J. Endocrinol. 176: 133-141; O'Harte, F. P. M. et al., 1999 , Diabetes 48: 758-765; O'Harte, F. P. M.
  • N-AcGIP has emerged as the most effective DPP IV-resistant analogue, substantially augmenting the plasma insulin response and curtailing the glycaemic excursion following conjoint administration with glucose to ob/ob mice (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291).
  • Example 6 was designed to evaluate the metabolic stability, biological activity and antidiabetic potential of novel second generation fatty acid derivatised, N-terminally modified N-AcGIP analogues, N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ).
  • Both GIP analogues contain a C-16 palmitate group linked to the epsilon-amino group of Lys at positions 16 or 37, in combination with an N-terminal (Tyr 1 ) acetyl group (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291).
  • N-AcGIP(LysPAL 37 ) The most effective analogue, N-AcGIP(LysPAL 37 ) was administered to ob/ob mice by once daily intraperitoneal injection for 14 days prior to evaluation of glucose homeostasis, pancreatic beta cell function and insulin sensitivity. Possible desensitization of GIP receptor action by prolonged exposure to elevated concentrations of N-AcGIP(LysPAL 37 ) was also examined. The results indicate the particular promise of the novel second generation N-terminally acetylated GIP analogue, N-AcGIP(LysPAL 37 ), as a potential therapeutic agent for the treatment of type 2 diabetes.
  • DPP IV-resistant analogues of GIP and GLP-1 are still subject to renal filtration.
  • fatty acid derivatisation has been used to improve the duration of action of GLP-1 (Knudsen, L. B. et al., 2000 , J. Med. Chem. 43: 1664-1669; Green, B. D. et al., 2004 , Biol. Chem. 385: 169-177; Kim, J. G. et al., 2003 , Diabetes 52: 751-759).
  • Example 6 describes the results of introducing two specific modifications to the native GIP hormone, namely N-terminal acetylation and C-terminal fatty acid derivatisation.
  • N-terminal acetylation was employed, as previously described (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291), to significantly enhance stability to DPP IV.
  • conjugation of a C-16 palmitate residue at the epsilon-amino group of Lys 16 or Lys 37 was introduced to extend the biological half-life through binding to circulating proteins (Kurtzhals, P. et al., 1995 , Biochem. J. 312: 725-731).
  • both GIP analogues appeared to be completely resistant to enzymatic breakdown by DPP IV, which corroborates previous observations with N-AcGIP(O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291). Furthermore, both analogues displayed similar or slightly better insulin-releasing and cyclic AMP generating properties to native GIP and N-AcGIP when tested in the in vitro cellular systems (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291).
  • ob/ob mice were employed.
  • the ob/ob syndrome is an extensively studied model of spontaneous obesity and diabetes, exhibiting hyperphagia, marked obesity, moderate hyperglycaemia and severe hyperinsulinemia (Bailey, C. J. et al., 1982 , Int. J. Obesity 6: 11-21).
  • ob/ob syndrome is an extensively studied model of spontaneous obesity and diabetes, exhibiting hyperphagia, marked obesity, moderate hyperglycaemia and severe hyperinsulinemia (Bailey, C. J. et al., 1982 , Int. J. Obesity 6: 11-21).
  • Gault, V. A. et al., 2002 Biochem. J. 367: 913-920
  • Gault, V. A. et al., 2003 J. Endocrinol.
  • N-AcGIP(LysPAL 37 ) appeared to be the best fatty acid derivatised analogue displaying a more protracted, significantly enhanced insulin-releasing potency over N-AcGIP(LysPAL 16 ) in vivo.
  • Reasons for the increased potency of N-AcGIP(LysPAL 37 ) remain unclear, but one explanation is an extended half-life.
  • Another possibility may be that a fatty acid chain linked to the Lys closer to the C-terminus of the peptide may have less of a detrimental effect upon the bioactive region of the molecule known to be located within the N-terminus (Gault, V. A. et al., 2002 , Biosci. Rep. 22: 523-528; Hinke, S. A.
  • N-AcGIP(LysPAL 37 ) was the more potent of the two analogues in vivo, it was further utilised in dose-response studies.
  • native GIP itself has only very modest effects in ob/ob mice, as sometimes observed with type 2 diabetic subjects (Nauck, M. A. et al., 1993 , J. Clin. Invest. 91: 301-307; Meier, J. J. et al., 2004 , Diabetes 53: 220-224; Vilsb ⁇ ll, T.
  • N-AcGIP(LysPAL 37 ) even at the lowest dose of 6.25 nmoles/kg, exhibited significant glucose-lowering and insulinotropic activity when administered with glucose.
  • N-AcGIP(LysPAL 37 ) is subject to albumin binding, the fact that it is still highly biologically active even at lower concentrations indicates striking potency.
  • N-AcGIP(LysPAL 37 ) Daily administration of N-AcGIP(LysPAL 37 ) to young adult ob/ob mice by intraperitoneal injection (12.5 nmoles/kg) resulted in a progressive lowering of plasma glucose concentrations and a significant decrease of glycated haemoglobin by 14 days. This was associated with a substantial improvement of glucose tolerance. Importantly food intake and body weight were unchanged ruling out the possibility that improvement of glucose homeostasis was merely the consequence of body weight loss. These observations also indicate that N-AcGIP(LysPAL 37 ) did not exert any untoward toxic actions affecting feeding over the study period. This is in harmony with recent studies showing that GIP does not inhibit gastric emptying (Meier, J. J.
  • N-AcGIP(LysPAL 37 ) a key component of the beneficial action of N-AcGIP(LysPAL 37 ) concerned effects on beta-cells.
  • native GIP is a weak stimulus to insulin secretion in ob/ob mice at the age studied, plasma and pancreatic insulin concentrations were raised in ob/ob mice receiving the novel fatty acid derivatised analogue.
  • This is consistent with the action of GIP as a promoter of proinsulin gene expression (Wang, Y. et al., 1996 , Mol. Cell. Endocrinol. 116:81-87) and exemplifies the increased potency reported for N-terminally modified GIP analogues in this animal model of diabetes (Hinke, S. A.
  • N-terminally acetylated GIP carrying a palmitate group linked to Lys at position 37 displays resistance to DPP IV and an impressive profile of bioactivity manifested by potent and long-acting glucose-lowering activity in a commonly employed animal model of obesity-diabetes.
  • This activity profile provides strong encouragement for the development of long-acting fatty acid derivatised N-terminally modified analogues of GIP for the once-daily treatment of type 2 diabetes.
  • the peptide analogues of the present invention have use in treating diseases and conditions caused by improper modulation of insulin levels, including diabetes, type 2 diabetes, insulin resistance, insulin resistant metabolic syndrome (Syndrome X), and obesity.
  • a peptide analogue produced by the methods of the present invention can be used in a pharmaceutical composition, wherein the analogue is combined with a pharmaceutically acceptable carrier.
  • a pharmaceutical composition may also contain (in addition to the analogue and a carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration.
  • Administration of the peptide analogue of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as by oral ingestion, inhalation, topical application or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Administration can be internal or external; or local, topical or systemic.
  • compositions containing a peptide analogue of this invention can be administered intravenously, as by injection of a unit dose, for example.
  • unit dose when used in reference to a therapeutic composition of the present invention refers to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent, i.e., carrier or vehicle.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • the composition of the present invention When a therapeutically effective amount of the composition of the present invention is administered orally, the composition of the present invention will be in the form of a tablet, capsule, powder, solution or elixir.
  • the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant.
  • the tablet, capsule, and powder contain from about 5 to 95% protein of the present invention, and preferably from about 25 to 90% protein of the present invention.
  • a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added.
  • the liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol.
  • the pharmaceutical composition When administered in liquid form, contains from about 0.5 to 90% by weight of the composition of the present invention, and preferably from about 1 to 50% of the composition of the present invention.
  • composition of the present invention When a therapeutically effective amount of the composition of the present invention is administered by intravenous, cutaneous or subcutaneous injection, the composition of the present invention will be in the form of a pyrogen-free, parenterally acceptable aqueous solution.
  • parenterally acceptable protein solutions having due regard to pH, isotonicity, stability, and the like, is within the skill in the art.
  • a preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the composition of the present invention, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art.
  • the pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
  • a sustained-release matrix is a matrix made of materials, usually polymers, which are degradable by enzymatic or acid/base hydrolysis or by dissolution. Once inserted into the body, the matrix is acted upon by enzymes and body fluids.
  • the sustained-release matrix desirably is chosen from biocompatible materials such as liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), polylactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, poly(ortho)esters, polyproteins, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • a preferred biodegradable matrix is a matrix of one of either polylactide, polyglycolide, or polylactide co-glycolide (co-polymers of lactic acid and glycolic acid).
  • compositions can include pharmaceutically acceptable salts of the components therein, e.g., which may be derived from inorganic or organic acids.
  • pharmaceutically acceptable salt is meant those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well-known in the art. For example, S. M. Berge, et al., describe pharmaceutically acceptable salts in detail in J Pharmaceutical Sciences (1977) 66:1 et seq., which is incorporated herein by reference in its entirety.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like. The salts may be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting a free base function with a suitable organic acid.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example
  • Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerophosphate, hemisulfate, heptonoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxymethanesulfonate (isethionate), lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate and undecanoate.
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such as dec
  • compositions, carriers, diluents and reagents are used interchangeably and represent that the materials are capable of administration to or upon a mammal with a minimum of undesirable physiological effects such as nausea, dizziness, gastric upset and the like.
  • physiological effects such as nausea, dizziness, gastric upset and the like.
  • the preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art and need not be limited based on formulation.
  • Such compositions are prepared as injectables either as liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared.
  • the preparation can also be emulsified.
  • the active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein.
  • Suitable excipients include, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient.
  • the amount of peptide analogue of the present invention in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, on the nature of prior treatments which the patient has undergone, and on a variety of other factors, including the type of injury, the age, weight, sex, medical condition of the individual. Ultimately, the attending physician will decide the amount of the analogue with which to treat each individual patient. Initially, the attending physician will administer low doses of peptide analogue and observe the patient's response. Larger doses of peptide analogue may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further.
  • the N-terminal modification of GIP is essentially a three step process. Firstly, GIP is synthesized from its C-terminal (starting from a Fmoc-Gln (Trt)-Wang resin (Calbiochem Novabiochem, Beeston, Nottingham, UK) up to the penultimate N-terminal amino-acid (Ala 2 ) on an automated peptide synthesizer (Applied Biosystems, California, USA). The synthesis follows standard Fmoc peptide chemistry protocols. Secondly, the N-terminal amino acid of native GIP (Tyr) is added to a manual bubbler system as a Fmoc-protected Tyr(tBu)-Wang resin.
  • Tyr native GIP
  • This amino acid is deprotected at its N-terminus (piperidine in DMF (20% v/v)) and allowed to react with a high concentration of glucose (glycation, under reducing conditions with sodium cyanoborohydride), acetic anhydride (acetylation), pyroglutamic acid (pyroglutamyl) etc. for up to 24 hours as necessary to allow the reaction to go to completion.
  • the completeness of reaction is monitored using the ninhydrin test which determines the presence of available free a-amino groups.
  • the now structurally modified Tyr is cleaved from the Wang resin (95% TFA, and 5% of the appropriate scavengers. N.B.
  • Tyr is considered to be a problematic amino acid and may need special consideration) and the required amount of N-terminally modified-Tyr consequently added directly to the automated peptide synthesiser, which will carry on the synthesis, thereby stitching the N-terminally modified-Tyr to the a-amino of GIP (Ala 2 ), completing the synthesis of the GIP analogue.
  • This peptide is cleaved off the Wang resin (as above) and then worked up using the standard Buchner filtering, precipation, rotary evaporation and drying techniques.
  • Reversed-phase Sep-Pak cartridges (C-18) were purchased from Millipore-Waters (Milford, Mass., USA). All water used in these experiments was purified using a Milli-Q, Water Purification System (Millipore Corporation, Milford, Mass., USA).
  • Electrospray ionization mass spectrometry (ESI-MS). Samples for ESI-MS analysis containing intact and degradation fragments of GIP (from DPP IV and plasma incubations) as well as Tyr 1 -glucitol GIP, were further purified by HPLC. Peptides were dissolved (approximately 400 pmol) in 100 ⁇ l of water and applied to the LCQ benchtop mass spectrometer (Finnigan MAT, Hemel Hempstead, UK) equipped with a microbore C-18 HPLC column (150 ⁇ 2.0 mm, Phenomenex, Ltd., Macclesfield, UK).
  • M r molecular mass
  • M i m/z ratio
  • i number of charges
  • M h mass of a proton.
  • Plasma glucose was assayed by an automated glucose oxidase procedure using a Beckman Glucose Analyzer II [33].
  • Plasma insulin was determined by dextran charcoal radioimmunoassay as described previously [34].
  • Incremental areas under plasma glucose and insulin area under the curve (AUC) were calculated using a computer program (CAREA) employing the trapezoidal rule [35] with baseline subtraction. Results are expressed as mean ⁇ SEM and values were compared using the Student's unpaired t-test. Groups of data were considered to be significantly different if P ⁇ 0.05.
  • FIG. 1 illustrates the typical peak profiles obtained from the HPLC separation of the products obtained from the incubation of GIP ( FIG. 1 a ) or Tyr 1 -glucitol GIP ( FIG. 1 b ) with DPP IV for 0, 2, 4 and 12 hours.
  • Degradation of GIP was evident after 4 hours incubation (54% intact), and by 12 hours the majority (60%) of intact GIP was converted to the single product with a retention time of 21.61 minutes.
  • Tyr 1 -glucitol GIP remained almost completely intact throughout 2-12 hours incubation. Separation was on a Vydac C-18 column using linear gradients of 0% to 31.5% acetonitrile over 15 minutes, to 38.5% over 30 minutes and from 38.5 to 70% acetonitrile over 5 minutes.
  • FIG. 2 shows a set of typical HPLC profiles of the products obtained from the incubation of GIP or Tyr 1 -glucitol GIP with human plasma for 0 and 4 hours.
  • GIP FIG. 2 a
  • the incubation of Tyr 1 -glucitol GIP under similar conditions did not result in the formation of any detectable degradation fragments during this time with only a single peak being observed with a retention time of 21.77 minutes.
  • FIG. 3 shows the monoisotopic molecular masses obtained for GIP ( FIG. 3A ), Tyr 1 -glucitol GIP ( FIG. 3B ) and the major plasma degradation fragment of GIP ( FIG. 3C ) using ESI-MS.
  • the peptides analyzed were purified from plasma incubations as shown in FIG. 2 .
  • Peptides were dissolved (approximately 400 pmol) in 100 ⁇ l of water and applied to the LC/MS equipped with a microbore C-18 HPLC column. Samples (30 ⁇ l direct loop injection) were applied at a flow rate of 0.2 ml/min, under isocratic conditions 35% acetonitrile/water.
  • Mass spectra were recorded using a quadripole ion trap mass analyzer. Spectra were collected using full ion scan mode over the mass-to-charge (m/z) range 150-2000.
  • FIG. 3C shows the prominent multiply charged species (M+3H) 3+ and (M+4H) 4+ detected from the major fragment of GIP at m/z 1583.8 and 1188.1, corresponding to intact M r 4748.4 and 4748 Da, respectively ( FIG. 3C ). This corresponds with the theoretical mass of the N-terminally truncated form of the peptide GIP(3-42). This fragment was also the major degradation product of DPP IV incubations (data not shown).
  • FIGS. 4 and 5 show the effects of intraperitoneal (ip) glucose alone (118 mmol/kg) (control group), and glucose in combination with GIP or Tyr 1- glucitol GIP (10 nmol/kg) on plasma glucose and insulin concentrations.
  • FIG. 4A shows plasma glucose concentrations after i.p. glucose alone (18 mmol/kg) (control group), or glucose in combination with either GIP of Tyr 1 -glucitol GIP (10 nmol/kg). The time of injection is indicated by the arrow (0 minutes).
  • FIG. 4B shows plasma glucose AUC values for 0-60 minutes post injection. Values are mean ⁇ SEM for six rats. **P ⁇ 0.01, ***P ⁇ 0.001 compared with GIP and Tyr 1- glucitol GIP; ⁇ P ⁇ 0.05, ⁇ P ⁇ 0.01 compared with non-glucated GIP.
  • FIG. 5A shows plasma insulin concentrates after i.p.
  • FIG. 5B shows plasma insulin AUC values were calculated for each of the 3 groups up to 90 minutes post injection. The time of injection is indicated by the arrow (0 minutes). Plasma insulin AUC values for 0-60 minutes post injection. Values are mean ⁇ SEM for six rats. *P ⁇ 0.05, **P ⁇ 0.001 compared with GIP and Tyr 1- glucitol GIP; ⁇ P ⁇ 0.05, ⁇ P ⁇ 0.01 compared with non-glycated GIP.
  • FIGS. 4A , B Plasma glucose concentrations and area under the curve (AUC) were significantly lower following administration of either GIP or Tyr 1 -glucitol GIP ( FIGS. 4A , B). Furthermore, individual values at 15 and 30 minutes together with AUC were significantly lower following administration of Tyr 1 -glucitol GIP as compared to GIP. Consistent with the established insulin-releasing properties of GIP, plasma insulin concentrations of both peptide-treated groups were significantly raised at 15 and 30 minutes compared with the values after administration of glucose alone ( FIG. 5A ). The overall insulin responses, estimated as AUC were also significantly greater for the two peptide-treated groups ( FIG. 5B ).
  • This example further looked at the ability of additional N-terminal structural modifications of GIP in preventing inactivation by DPP and in plasma and their associated increase in both the insulin-releasing potency and potential therapeutic value.
  • Native human GIP, glycated GIP, acetylated GIP and a number of GIP analogues with N-terminal amino acid substitutions were tested.
  • High-performance liquid chromatography (HPLC) grade acetonitrile was obtained from Rathburn (Walkersbum, Scotland). Sequencing grade trifluoroacetic acid (TFA) was obtained from Aldrich (Poole, Dorset, UK). Dipeptidyl peptidase IV was purchased from Sigma (Poole, Dorset, UK), and Diprotin A was purchased from Calbiochem Novabiochem (Beeston, Nottingham, UK). RPMI 1640 tissue culture medium, foetal calf serum, penicillin and streptomycin were all purchased from Gibco (Paisley, Strathclyde, UK). All water used in these experiments was purified using a Milli-Q, Water Purification System (Millipore, Milford, Mass., USA). All other chemicals used were of the highest purity available.
  • GIP, GIP(Abu 2 ), GIP(Sar 2 ), GIP(Ser 2 ), GIP(Gly 2 ) and GIP(Pro 3 ) were sequentially synthesized on an Applied Biosystems automated peptide synthesizer (model 432A) using standard solid-phase Fmoc procedure, starting with an Fmoc-Gln-Wang resin. Following cleavage from the resin by trifluoroacetic acid:water, thioanisole, ethanedithiol (90/2.5/5/2.5, a total volume of 20 ml/g resin), the resin was removed by filtration and the filtrate volume was decreased under reduced pressure.
  • Electrospray ionization-mass spectrometry (ESI-MS) was carried out as described in Example 2.
  • Degradation of GIP and novel GIP analogues by DPP IV and human plasma was carried out as described in Example 2.
  • BRIN-BD11 cells [30] were cultured in sterile tissue culture flasks (Coming, Glass Works, UK) using RPMI-1640 tissue culture medium containing 10% (v/v) foetal calf serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin) and 11.1 mM glucose. The cells were maintained at 37° C. in an atmosphere of 5% CO 2 and 95% air using a LEEC incubator (Laboratory Technical Engineering, Nottingham, UK).
  • Results are expressed as mean ⁇ S.E.M. and values were compared using the Student's unpaired t-test. Groups of data were considered to be significantly different if P ⁇ 05.
  • FIGS. 6-11 illustrate the typical peak profiles obtained from the HPLC separation of the reaction products obtained from the incubation of GIP, GIP(Abu 2 ), GIP(Sar 2 ), GIP(Ser 2 ), glycated GIP and acetylated GIP with DPP IV, for 0, 2, 4, 8 and 24 hours.
  • the results summarized in Table 1 indicate that glycated GIP, acetylated GIP, GIP(Ser 2 ) are GIP(Abu 2 ) more resistant than native GIP to in vitro degradation with DPP IV. From these data GIP(Sar 2 ) appears to be less resistant.
  • Table represents the percentage of intact peptide (i.e., GIP 1-42) relative to the major degradation product GIP 3-42. Values were taken from HPLC traces performed in triplicate and the mean and S.E.M. values calculated. DPA is diprotin A, a specific inhibitor of DPPIV.
  • FIGS. 12-16 show a representative set of HPLC profiles obtained from the incubation of GIP and GIP analogues with human plasma for 0, 2, 4, 8 and 24 hours. Observations were also made after incubation for 24 hours in the presence of DPA. These results are summarized in Table 2 are broadly comparable with DPP IV incubations, but conditions which more closely mirror in vivo conditions are less enzymatically severe. GIP was rapidly degraded by plasma. In comparison, all analogues tested exhibited resistance to plasma degradation, including GIP(Sar 2 ) which from DPP IV data appeared least resistant of the peptides tested.
  • DPA substantially inhibited degradation of GIP and all analogues tested with complete abolition of degradation in the cases of GIP(Abu 2 ), GIP(Ser 2 ) and glycated GIP. This indicates that DPP IV is a key factor in the in vivo degradation of GIP. TABLE 2 Percent intact peptide remaining after incubation with human plasma.
  • Table represents the percentage of intact peptide (i.e., GIP 1-42) relative to the major degradation product GIP 3-42. Values were taken from HPLC traces performed in triplicate and the mean and S.E.M. values calculated. DPA is diprotin A, a specific inhibitor of DPPIV.
  • FIGS. 17-30 show the effects of a range of concentrations of GIP, GIP(Abu 2 ), GIP(Sar 2 ), GIP(Ser 2 ), acetylated GIP, glycated GIP, GIP(Gly 2 ) and GIP(Pro 3 ) on insulin secretion from BRIN-BD11 cells at 5.6 and 16.7 mM glucose.
  • Native GIP provoked a prominent and dose-related stimulation of insulin secretion.
  • the glycated GIP analogue exhibited a considerably greater insulinotropic response compared with native peptide.
  • GIP(Gly 2 ) and GIP(Pro 3 ) appeared to be the least potent analogues in terms of insulin release.
  • Other stable analogues tested namely GIP(Abu 2 ) and GIP(Sar 2 ), exhibited a complex pattern of responsiveness dependent on glucose concentration and dose employed. Thus very low concentrations were extremely potent under hyperglycemic conditions (16.7 mM glucose). This suggests that even these analogues may prove therapeutically useful in the treatment of type 2 diabetes where insulinotropic capacity combined with in vivo degradation dictates peptide potency.
  • Glu 3 Substituted GIP Improves Obesity-Related Insulin Resistance and Associated glucose intolerance
  • This example examines GIP receptor antagonism and obesity-related insulin resistance and associated glucose intolerance using a Glu 3 -substituted form of GIP, namely, (Pro 3 )GIP.
  • the molecular masses of the purified peptide analogues were determined using Matrix Assisted Laser Desorption Ionisation-Time of Flight (MALDI-TOF) mass spectrometry. Samples were dissolved in 10 ⁇ l H 2 O (approximately 40 pmol/l), placed on a stainless steel sample plate and allowed to dry at room temperature. Samples were then mixed with a matrix solution (10 mg/ml solution of ⁇ -cyano-4-hydroxycinnamic acid) in acetonitrile/ethanol (1/1) and allowed to dry at room temperature.
  • MALDI-TOF Matrix Assisted Laser Desorption Ionisation-Time of Flight
  • CHL Chinese hamster lung
  • fibroblast cells stably transfected with the human GIP receptor were cultured in DMEM tissue culture medium containing 10% (v/v) foetal bovine serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin).
  • BRIN-BD11 cells were cultured using RPMI-1640 tissue culture medium containing 10% (v/v) foetal bovine serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin).
  • Cells were maintained in sterile tissue culture flasks (Coming Glass Works, Sunderland, UK) at 37° C. in an atmosphere of 5% CO 2 and 95% air using an LEEC incubator (Laboratory Technical Engineering, Nottingham, UK).
  • Insulin release from BRIN-BD11 cells was determined using cell monolayers (McClenaghan, N. H. et al., 1996 , Diabetes 45:1132-1140). Cells were harvested with the aid of trypsin/EDTA (Gibco), seeded into 24-multiwell plates (Nunc, Roskilde, Denmark) at a density of 1.0 ⁇ 10 5 cells per well, and allowed to attach overnight at 37° C. Prior to acute test, cells were preincubated for 40 minutes at 37° C.
  • GIP receptor transfected CHL cells were seeded into 12-well plates (Nunc, Roskilde, Denmark) at a density of 1.0 ⁇ 10 5 cells per well. The cells were then allowed to grow for 48 hours before being loaded with tritiated adenine (2 ⁇ Ci; TRK311, Amersham, Buckinghamshire, UK) and incubated at 37° C. for 6 hours in 1 ml DMEM, supplemented with 0.5% (w/v) foetal bovine serum.
  • the cells were then washed twice with HBS buffer (130 mM NaCl, 20 mM HEPES, 0.9 mM NaHPO 4 , 0.8 mM MgSO 4 , 5.4 mM KCl, 1.8 mM CaCl 2 , 25 mM glucose, 25 ⁇ M phenol red, pH 7.4).
  • HBS buffer 130 mM NaCl, 20 mM HEPES, 0.9 mM NaHPO 4 , 0.8 mM MgSO 4 , 5.4 mM KCl, 1.8 mM CaCl 2 , 25 mM glucose, 25 ⁇ M phenol red, pH 7.4
  • the cells were then exposed for 10 minutes at 37° C. to forskolin (FSK, 10 ⁇ M) or varying concentrations of (Pro 3 )GIP in the absence (control) or presence of native GIP (10 ⁇ 7 M).
  • TCA trichloroacetic acid
  • Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 15, 30 and 60 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at ⁇ 20° prior to glucose and insulin determinations.
  • Plasma glucose and insulin responses were evaluated using 8- to 12-week old ob/ob mice where food was withdrawn for an 18-hour period prior to intraperitoneal injection of saline (0.9% (w/v) NaCl; control) or (Pro 3 )GIP (25 nmol/kg body weight). Following injection, the mice were allowed to re-feed for 15 minutes. Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 15, 30, 60 and 120 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at ⁇ 20° prior to glucose and insulin determinations.
  • pancreatic tissue was excised from non-fasted ob/ob mice after 11 days treatment with either saline (0.9% w/v NaCl) or (Pro 3 )GIP (25 nmol/kg body weight/day). Pancreatic samples were individually wrapped in aluminium foil and snap frozen in liquid nitrogen. Individual excised pancreatic samples were then either embedded, sectioned and immunohistochemically stained for insulin or permeabilised for determination of pancreatic insulin content.
  • HbA 1c Plasma glucose and insulin concentrations.
  • HbA 1c was measured in whole blood by ion-exchange high-performance liquid chromatography using the Menari HA-8140 kit (BIOMEN, Berkshire, UK).
  • Plasma glucose was assayed by an automated glucose oxidase procedure using a Beckman Glucose Analyzer II (Stevens, J. F., 1971 , Clinica Chemica Acta 32:199-201) and plasma insulin was determined by RIA (Flatt, P. R. et al., 1981 , Diabetologia 20:573-577).
  • Incremental areas under plasma glucose and insulin curves were calculated using a computer generated program (CAREA) employing the trapezoidal rule (Burington, R. S., 1973 , Handbook of Mathematical Tables and Formulae , New York, McGraw Hill) with baseline subtraction.
  • Results are expressed as means ⁇ SEM. Values were compared using Student's unpaired t-test and groups of data were considered to be significantly different if P ⁇ 0.05.
  • FIG. 32A is a line graph showing 3 H-cAMP production as a percent of maximal response (y-axis) with increasing peptide concentration (M) (x-axis).
  • FIG. 32A is a line graph showing 3 H-cAMP production as a percent of maximal response (y-axis) with increasing peptide concentration (M) (x-axis).
  • 32B is a bar graph showing insulin secretion (y-axis) with increasing peptide concentration (M) (x-axis) for 5.6 mM glucose (control) (white bar), GIP (gray bars), (Pro 3 )GIP (lined bars) and (Pro 3 )GIP+GIP(10 7 M) (black bars).
  • FIGS. 33A through 33F are bar charts.
  • FIG. 33A shows 3 H-cAMP production as a percent of 10 ⁇ 7 M GIP (y-axis) versus log 10 of GIP (110-M) (white bar, control) and GIP (10 ⁇ 7 M)+GIP(3-42) (black bars).
  • FIGS. 33A through 33F show 3 H-cAMP production as a percent of 10 ⁇ 7 M GIP (y-axis) versus log 10 of GIP (110-M) (white bar, control) and GIP (10 ⁇ 7 M)+GIP(3-42) (black bars).
  • FIGS. 33A shows 3 H-cAMP production as a percent of 10 ⁇ 7 M GIP (y-axis) versus log 10 of GIP (110-M) (white bar, control) and GIP (10 ⁇ 7 M)+GIP(3-42) (black bars).
  • FIGS. 33A shows 3 H-cAMP production as a percent of 10 ⁇ 7 M GIP (y-axi
  • FIG. 33B through 33F show insulin secretion (in ng/10 6 cells/20 minutes) (y-axis) as a function of peptide concentration (M) (x-axis) for GIP (10 ⁇ 7 M) (white bar, control) and a Glu 3 -substituted form of GIP (black bars), including (Hyp 3 )GIP ( FIG. 33B ), (Lys 3 )GIP ( FIG. 33C ), (Tyr 3 )GIP ( FIG. 33D ), (Trp 3 )GIP ( FIG. 33E ), and (Phe 3 )GIP ( FIG. 33F ).
  • FIGS. 34A through 34D are a set of two line graphs ( FIGS. 34A, 34C ) and two bar graphs ( FIGS. 34B, 34D ) showing that acute administration of (Pro 3 )GIP completely antagonises the actions of GIP on glucose tolerance ( FIGS. 34A, 34B ) and plasma insulin ( FIGS. 34C, 34D ) responses in obese diabetic ob/ob mice.
  • FIGS. 34A and 34C are line graphs show plasma glucose levels ( FIG. 34A , y-axis) and plasma insulin levels ( FIG.
  • FIGS. 34B and 34D are bar graphs showing plasma glucose AUC for glucose alone (white bars), GIP (grey bars) and glucose+(GIP+Pro 3 GIP)) (black bars).
  • FIGS. 35A through 35D show the effects of (Pro 3 )GIP on physiological meal-stimulated insulin release and glycemic excursion in obese diabetic ob/ob mice.
  • Plasma glucose and insulin concentrations were measured in mice allowed to re-feed for 15 minutes prior to i.p. administration of saline (0.9% (w/v) NaCl) as control or (Pro 3 )GIP (25 nmol/kg body weight). The time of injection is indicated by the arrow (15 minutes).
  • FIGS. 35A through 35D are a set of two line graphs ( FIGS. 35A, 35C ) and two bar graphs ( FIGS. 35B, 35D ).
  • the figures show plasma insulin ( FIGS. 35A ) and plasma glucose ( FIG. 35C ) over time for saline control ( ⁇ ) and (Pro 3 )GIP ( ⁇ ), and plasma insulin AUC ( FIG. 35B ) and plasma glucose AUC ( FIG. 35D ) for saline control (white bars) and (Pro 3 )GIP (black bars), respectively.
  • Values are means ⁇ SEM for 8 mice. *P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001 compared with saline alone.
  • FIG. 36 is a set of two bar graphs showing plasma glucose ( FIG. 36A ) and insulin ( FIG. 36B ) concentrations after daily subcutaneous administration of saline alone (0.9% (w/v) NaCl; as control; white bars) or (Pro 3 )GIP (25 nmol/kg body weight; black bars) for 11 days. Values are means ⁇ SEM for 6 mice and *P ⁇ 0.05 compared with saline alone. Chronic administration of (Pro 3 )GIP (black bars) for 11 days decreases plasma glucose and plasma insulin concentrations of obese diabetic ob/ob mice, relative to controls.
  • FIG. 39 shows the effects of chronic administration of (Pro 3 )GIP for 11 days on insulin sensitivity in obese diabetic ob/ob mice.
  • chronic administration of (Pro 3 )GIP caused a significant improvement of insulin sensitivity.
  • GIP Gastric inhibitory polypeptide
  • GIP-R GIP receptor
  • GIP-R ablation also significantly lowered overall plasma glucose (1.4-fold; P ⁇ 0.05) and insulin (1.5-fold; P ⁇ 0.05) responses to feeding. These changes were associated with significantly enhanced (1.6-fold; P ⁇ 0.05) insulin sensitivity in the (Pro 3 )GIP-treated group.
  • Daily injection of (Pro 3 )GIP reduced pancreatic insulin content (1.3-fold; P ⁇ 0.05) and partially corrected the obesity-related islet hypertrophy and beta cell hyperplasia of ob/ob mice.
  • These comprehensive beneficial effects of (Pro 3 )GIP were reversed following 9 days cessation of treatment and were independent of food intake and body weight, which were unchanged.
  • Obese diabetic mice derived from the colony maintained at Aston University, UK (Bailey, C. J., et al., 1982 , Int. J. Obes. 6:11-21) were used at 12-16 weeks of age. Animals were age-matched, divided into groups and housed individually in an air-conditioned room at 22 ⁇ 2° C. with a 12 hour light:12 hour dark cycle. Drinking water and a standard rodent maintenance diet (Trouw Nutrition, Cheshire, UK) were freely available. All animal experiments were carried out in accordance with the UK Animals (Scientific Procedures) Act 1986. No adverse effects were observed following administration of (Pro 3 )GIP.
  • (Pro 3 )GIP was sequentially synthesized on an Applied Biosystems automated peptide synthesizer (Model 432 A).
  • (Pro 3 )GIP was purified by reversed-phase HPLC on a Waters Millenium 2010 chromatography system (Software version 2.1.5) and subsequently characterized using electrospray ionization mass spectrometry (ESI-MS).
  • mice received once daily i.p. injections (17:00 hours) of either saline vehicle (0.9% (w/v), NaCl) or (Pro 3 )GIP (25 nmol/kg body wt).
  • saline vehicle (0.9% (w/v), NaCl
  • Pro 3 )GIP 25 nmol/kg body wt.
  • Food intake and body weight were recorded daily whilst plasma glucose and insulin concentrations were monitored at intervals of 2-6 days.
  • Whole blood for the measurement of glycated hemoglobin was taken on days 11 and 20.
  • Intraperitoneal glucose tolerance (18 mmol/kg body wt), metabolic response to native GIP (25 nmol/kg body wt) and insulin sensitivity (50 U/kg body wt) tests were performed on days 11 and 20. Mice fasted for 18 hours were used to examine the metabolic response to 15 minutes feeding. In a separate series, pancreatic tissues were excised at the end of the 11-day treatment period or 9 days following discontinuation of (Pro 3 )GIP and processed for immunohistochemistry or measurement of insulin following extraction with 5 ml/g of ice-cold acid ethanol (750 ml ethanol, 235 ml water, 15 ml concentrated HCl).
  • Plasma glucose was assayed by an automated glucose oxidase procedure (Stevens, J. F., 1971 , Clin. Chem. Acta 32:199-201) using a Beckman Glucose Analyzer II (Beckman Instruments, Galway, Ireland). Plasma and pancreatic insulin were assayed by a modified dextran-coated charcoal radioimmunoassay (Flatt, P. R. et al., 1981 , Diabetologia 20:573-577).
  • Glycated hemoglobin was determined using cation-exchange columns (Sigma, Poole, Dorset, UK) with measurement of absorbance (415 nm) in wash and eluting buffer using a VersaMax Microplate Spectrophotometer (Molecular Devices, Wokingham, Berkshire, UK).
  • Tissue fixed in 4% paraformaldehyde/PBS and embedded in paraffin was sectioned at 8 ⁇ m. After de-waxing, sections were incubated with blocking serum (Vector Laboratories, CA, USA) prior to exposure to insulin antibody. Tissue samples were then incubated consecutively with secondary biotinylated universal, pan-specific antibody (Vector Laboratories, CA, USA) and streptavidin/peroxidase preformed complex (Vector Laboratories, CA, USA).
  • pancreatic tissue was counterstained with hematoxylin (BDH Chemicals, Dorset, UK) and then plunged into acid methanol (500 ml methanol, 500 ml H 2 O and 2.5 ml concentrated HCl) prior to dehydration and mounting in Depex (BDH Chemicals, Dorset, UK).
  • acid methanol 500 ml methanol, 500 ml H 2 O and 2.5 ml concentrated HCl
  • Depex BDH Chemicals, Dorset, UK
  • the stained slides were viewed under a microscope (Nikon Eclipse E2000, Diagnostic Instruments Incorporated, Michigan, USA) attached to a JVC camera Model KY-F55B (JVC, London, UK) and analyzed using Kromoscan imaging software (Kinetic Imaging Limited, Faversham, Kent, UK).
  • the average number and diameter of every islet in each section was estimated in a blinded manner using an eyepiece graticule calibrated with a stage micrometer (Graticules Limited, Tonbridge, Kent, UK). The longest and shortest diameters of each islet were determined with the graticule. Half of the sum of these two values was then considered to be the average islet diameter. Approximately 60-70 random sections were examined from the pancreas of each mouse.
  • FIGS. 42A through 42D are a set of two line graphs ( FIGS. 42A, 42C ) and two bar graphs ( FIGS. 42B, 42D ) showing the effects of (Pro 3 )GIP on plasma glucose and insulin response to native GIP 4 hours after administration.
  • Tests were conducted 4 hours after administration of (Pro 3 )GIP (25 nmoles/kg body weight) or saline (0.9% NaCl) in 18 hour-fasted ob/ob mice. Plasma glucose and insulin concentrations were measured prior to and after i.p.
  • FIGS. 43A through 43D are a set of two line graphs and two bar graphs showing the effects of daily (Pro 3 )GIP administration on food intake ( FIG. 43A ), body weight ( FIG. 43B ), plasma glucose ( FIG. 43C ) and insulin ( FIG. 43D ) concentrations in ob/ob mice.
  • Parameters were measured for 5 days prior to, 11 days during (indicated by black bar) and 9 days after treatment with saline or (Pro 3 )GIP (25 nmol/kg bw/day). Values are mean ⁇ SEM for eight mice. *P ⁇ 0.05 compared with saline group.
  • FIGS. 44A through 44D are a set of four line graphs with inset bar graphs showing the effects of daily (Pro 3 )GIP administration on glucose tolerance and plasma insulin response to glucose in ob/ob mice.
  • Tests were conducted after daily treatment with (Pro 3 )GIP (25 nmoles/kg body weight/day; ⁇ ; black bars) or saline (control; ⁇ ; white bars) for 11 days ( FIG. 44A, 44C ) or 9 days after cessation of treatment ( FIG. 44B, 44B ).
  • Glucose 18 mmoles/kg body weight was administered at the time indicated by the arrow.
  • Plasma glucose FIG.
  • FIG. 44A, 44B and insulin ( FIG. 44C, 44D ) AUC values for 0-60 minutes post injection, with identical baseline subtractions in each case to demonstrate the complete effect of (Pro 3 )GIP treatment, are shown in insets. Values are mean ⁇ SEM for eight mice. *P ⁇ 0.05, **P ⁇ 0.01 and ***P ⁇ 0.001 compared with saline group.
  • FIGS. 45A through 45D are a set of two line graphs ( FIGS. 45A , 45 C) and two bar graphs ( FIGS. 45B, 45D ) showing the effects of daily (Pro 3 )GIP administration ( ⁇ ; black bars) or saline ( ⁇ ; white bars) on glucose ( FIGS. 45A, 45B ) and insulin ( FIGS. 45C, 45D ) responses to feeding in ob/ob mice fasted for 18 hours.
  • Tests were conducted after daily treatment with (Pro 3 )GIP (25 nmol/kg body weight/day) or saline for 11 days.
  • the arrow indicates the time of feeding (15 minutes).
  • AUC values for 0-105 minutes post-feeding are also shown. Values are mean ⁇ SEM for eight mice. *P ⁇ 0.05 compared with saline group.
  • FIGS. 46A through 46D are a set of two line graphs ( FIGS. 46A, 46C ) and two bar graphs ( FIGS. 46B, 46D ) showing the effects of daily (Pro 3 )GIP administration on insulin sensitivity in ob/ob mice.
  • Tests were conducted after daily treatment with (Pro 3 )GIP (25 nmol/kg body weight/day; ⁇ ; black bars) or saline (o; white bars) for 111 days ( FIG. 46A, 46B ) or 9 days after cessation of treatment ( FIG. 46C, 46D ).
  • Insulin 50 U/kg body weight
  • AUC values for 0-60 minutes post-injection are also shown. Values are mean ⁇ SEM for eight mice. *P ⁇ 0.05 compared with saline group.
  • Plasma glucose and insulin responses to 15 minutes feeding were significantly lowered (P ⁇ 0.05) at 30 and 60 minutes in ob/ob mice treated with (Pro 3 )GIP for 11 days ( FIG. 45 ).
  • AUC glucose and insulin were significantly (P ⁇ 0.05) decreased in (Pro 3 )GIP treated ob/ob mice, despite similar food intakes of 0.3-0.5 g/mouse/15 minutes.
  • FIGS. 46A and 45B the hypoglycemic action of insulin was significantly (P ⁇ 0.05) augmented in terms of AUC measures and post injection values in ob/ob mice treated with (Pro 3 )GIP for 11 days.
  • the responses following 9 days discontinuation of (Pro 3 )GIP treatment were similar to saline treated controls ( FIG. 45C, 45D ).
  • FIGS. 47A through 47D are a set of four bar graphs showing the effects of daily (Pro 3 )GIP administration on pancreatic weight ( FIG. 47A ), insulin content ( FIG. 47B ), islet number ( FIG. 47C ) and islet diameter ( FIG. 47D ) in ob/ob mice.
  • Parameters were measured after daily treatment with (Pro 3 )GIP (25 nmol/kg body weight/day; black bars) or saline (white bars) for 11 days and 9 days after cessation of treatment (day 20). Values are mean ⁇ SEM for eight mice.
  • FIGS. 48A through 48F are a set of two bar graphs ( FIGS. 48A, 48D ) and four photomicrographs ( FIGS. 48B, 48C , 48 E, 48 F), showing the effects of daily (Pro 3 )GIP administration on islet size and morphology in ob/ob)mice.
  • FIG. 47A pancreatic insulin content was significantly (P ⁇ 0.05) decreased in ob/ob mice receiving (Pro 3 )GIP for 11 days compared to controls ( FIG. 47B ). No significant differences were observed in islet number per pancreatic section ( FIG. 47C ), but average islet diameter was markedly and significantly reduced (P ⁇ 0.001) in (Pro 3 )GIP treated ob/ob mice ( FIG. 47D ). These effects were effectively reversed by discontinuation of (Pro 3 )GIP on day 20, however average islet diameter was still significantly reduced (P ⁇ 0.05). As shown in FIG.
  • FIG. 48A presents similar analysis following cessation of treatment, with a significant (P ⁇ 0.05) increase in the percentage of small islets still apparent.
  • Representative images ( ⁇ 40 magnification) of pancreata immunohistologically stained for insulin from 11-day (Pro 3 )GIP treated ob/ob mice ( FIG. 48B ) and saline treated controls ( FIG. 48C ) illustrate the dramatic changes in pancreatic islet morphology induced by (Pro 3 )GIP treatment. Pancreata immunohistologically stained for insulin on day 20 are also shown ( FIG. 48E, 48F ).
  • FIGS. 48B, 48C , 48 E and 48 F are representative images ( ⁇ 40 magnification) of pancreata stained for insulin following 11 days treatment with (Pro 3 )GIP ( FIG. 48B ) or saline ( FIG. 48C ).
  • Corresponding images 9 days after cessation of treatment with (Pro 3 )GIP ( FIG. 48E ) or saline ( FIG. 48F ) are also shown.
  • the arrows indicate islets.
  • Obese diabetic mice derived from the colony maintained at Aston University, UK were used at 12-17 weeks of age. The genetic background and characteristics of the colony used have been outlined in detail elsewhere (Bailey, C. J. et al., 1982 , Int. J. Obesity 6:11-21; Gault, V. A. et al., 2003 , J. Endocrinol. 176: 133-141). Animals were housed in an air-conditioned room at 22 ⁇ 2° C. with a 12 hours light:12 hours dark cycle. Drinking water and standard rodent maintenance diet (Trouw Nutrition, Cheshire, UK) were freely available. All test solutions were administered by i.p.
  • High performance liquid chromatography (HPLC) grade acetonitrile was obtained from Rathburn (Walkersbum, UK).
  • Trifluoroacetic acid (TFA) and trichloroacetic acid (TCA) were obtained from Aldrich (Poole, Dorset, UK).
  • DPP IV, isobutylmethylxanthine (IBMX), alpha-cyano-4-hydroxycinnamic acid, cyclic AMP and ATP were all purchased from Sigma (Poole, Dorset, UK).
  • Fmoc-protected amino acids were from Calbiochem Novabiochem (Nottingham, UK).
  • RPMI-1640 and DMEM tissue culture medium, foetal bovine serum, penicillin and streptomycin were all purchased from Gibco (Paisley, Strathclyde, UK).
  • the chromatography columns used for cyclic AMP assay, Dowex AG50 WX and neutral alumina AG7 were obtained from Bio-Rad (Life Science Research, Alpha Analytical, Larne, UK). All water used in these experiments was purified using a Milli-Q Water Purification System (Millipore, Milford, Mass., USA). All other chemicals used were of the highest purity available.
  • N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) were synthesised in the same way as native GIP but with the exception that the epsilon-amino groups of Lys at positions 16 or 37 were conjugated with a C-16 palmitate fatty acid.
  • GIP and fatty acid derivatised GIP analogues were incubated at 37° C. with purified porcine dipeptidylpeptidase IV (5 mU in 50 mmol/l triethanolamine-HCl; pH 7.8) for 0, 2, 4, 8 and 24 hours (final peptide concentration 2 mmol/l).
  • the reactions were subsequently terminated by addition of 10% (v/v) TFA/water and the reaction products separated using HPLC. Reaction products were applied to a Vydac C-4 column (4.6 ⁇ 250 mm; The Separations Group, Hesparia, Calif.) and the major degradation product GIP(3-42) separated from intact GIP.
  • the column was equilibrated with 0.12% (v/v) TFA/water at a flow rate of 1.0 ml/minute using 0.1% (v/v) TFA in 70% acetonitrile/water with the concentration of acetonitrile in the eluting solvent being raised from 0% to 40% over 10 minutes, and then from 40% to 75% over 35 minutes.
  • the absorbance was monitored at 206 nm using a SpectraSystem UV 2000 Detector (Thermoquest Limited, Manchester, UK) and the peaks collected manually prior to MALDI-ToF MS analysis. HPLC peak area data were used to calculate % intact peptide remaining throughout the incubation.
  • CHL Chinese hamster lung
  • fibroblasts stably transfected with the human GIP receptor (Gremlich, S. et al., 1995 , Diabetes 44: 1202-1208) were cultured in DMEM tissue culture medium containing 10% (v/v) FBS, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin).
  • DMEM tissue culture medium containing 10% (v/v) FBS, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin).
  • Clonal pancreatic BRIN-BD11 cells (McClenaghan, N. H.
  • Intracellular cyclic AMP production was measured using GIP-receptor transfected CHL fibroblasts (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291).
  • CHL cells were seeded into 12-well plates (Nünc, Roskilde, Denmark) at a density of 10 5 cells per well and allowed to grow for 48 hours before being loaded with tritiated adenine (2 ⁇ Ci; TRK311; Amersham, Buckinghamshire, UK). The cells were then incubated at 37° C.
  • BRIN-BD11 cells Insulin-release studies were carried out using clonal pancreatic BRIN-BD11 cells as described previously (O'Harte, F. P. M. et al., 2002 , Diabetologia 45: 1281-1291). Briefly, BRIN-BD11 cells were seeded into 24-well plates at a density of 10 5 cells per well, and allowed to attach overnight at 37° C. Acute tests for insulin release were preceded by 40 minutes pre-incubation at 37° C. in 1.0 ml Krebs Ringer bicarbonate buffer supplemented with 1.1 mmol/l glucose.
  • Test incubations were performed in the presence of 5.6 mmol/l glucose with a range of concentrations (10 ⁇ 13 to 10 ⁇ 6 mol/l) of GIP and GIP analogues. After 20 minutes incubation, the buffer was removed from each well and aliquots (200 ⁇ l) used for measurement of insulin.
  • N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) Effects of N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) in ob/ob mice.
  • Metabolic and dose-response effects of GIP and N-AcGIP(LysPAL) analogues (at 6.25-25 nmoles/kg bw) following glucose administration (18 mmoles/kg bw) were examined in mice fasted for 18 hours.
  • mice received once daily intraperitoneal injections (17:00 h) for 14 days of either saline vehicle (0.9%, w/v, NaCl), native GIP or N-AcGIP(LysPAL 37 ) (both at 12.5 nmoles/kg body weight/day). Food intake and body weight were recorded daily. Plasma glucose and insulin concentrations were monitored at 2-6 day intervals. At 14 days, groups of animals were used to evaluate intraperitoneal glucose tolerance (18 mmoles/kg) and insulin sensitivity (50 U/kg). In a separate series, two experimental protocols were employed to examine the possibility of GIP receptor desensitization after 14 days treatment.
  • Plasma glucose was assayed by an automated glucose oxidase procedure (Stevens, J. F., 1971, Clin. Chem. Acta 32:199-201) using a Beckman Glucose Analyser II (Beckman, Galway, Ireland). Plasma insulin was determined by dextran-charcoal RIA as described previously (Flatt, P. R. et al., 1981 , Diabetologia 20: 573-577).
  • Glycated hemoglobin was determined using cation-exchange columns (Sigma, Poole, Dorset, UK) with measurement of absorbance (415 nm) in wash and eluting buffers using a VersaMax microplate spectrophotometer (Molecular Devices, Wokingham, Berkshire, UK).
  • Results are expressed as mean ⁇ SEM. Data were compared using the unpaired Student's t-test. Where appropriate, data were compared using repeated measures ANOVA or one-way ANOVA, followed by the Student-Newman-Keuls post hoc test. Incremental areas under plasma glucose and insulin curves (AUC) were calculated using a computer-generated program employing the trapezoidal rule (Burington, R. S., 1973 , Handbook of Mathematical Tables and Formulae , McGraw-Hill, New York) with baseline subtraction. Groups of data were considered to be significantly different if p ⁇ 0.05.
  • Peptide samples were mixed with matrix (alpha-cyano-4-hydroxycinnamic acid) and m/z ratio vs. relative peak intensity recorded using a Voyager-DE BioSpectrometry Workstation.
  • FIG. 50B shows insulin-releasing activity of glucose (5.6 mmo/l glucose; white bars), GIP (lined bars) and fatty acid derivatised GIP analogues N-AcGIP(LysPAL 16 ) (grey bars) and N-AcGIP(LysPAL 37 ) (black bars) in the clonal pancreatic beta cell line, BRIN-BD11.
  • N-AcGIP(LysPAL 16 ) grey bars
  • N-AcGIP(LysPAL 37 ) black bars
  • both GIP analogues appear to be at least equipotent to the native peptide.
  • FIGS. 51A through 51D are a set of two line graphs ( FIGS. 51A, 51C ) and two bar graphs ( FIGS. 51B, 51D ) showing glucose lowering effects ( FIGS. 51A, 51B ) and insulin-releasing activity ( FIGS. 51C, 51D ) of GIP and fatty acid derivatised GIP analogues in 18 hour-fasted ob/ob mice. Plasma glucose and insulin concentrations were measured prior to and after i.p.
  • Basal blood glucose levels of the experimental groups were not significantly different at the start of the study (p>0.05). After injection of glucose alone, plasma glucose levels increased rapidly, attaining values of 40.3 ⁇ 1.5 mmol/l at 60 min. Native GIP reduced plasma glucose at each of the time points monitored, however, this failed to reach significance in terms of overall glucose excursion as revealed by the AUC values ( FIG. 52B ).
  • Administration of N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) produced a significant reduction in plasma glucose at each time point (p ⁇ 0.01 to p ⁇ 0.001) and significantly lowered glucose AUC (p ⁇ 0.001 to p ⁇ 0.001) when compared to glucose alone. Additionally, N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) decreased the overall glucose excursion (p ⁇ 0.05 to p ⁇ 0.001) when compared to native GIP.
  • FIGS. 51C and 51D The corresponding plasma insulin responses are illustrated in FIGS. 51C and 51D .
  • the maximal rise in plasma insulin was observed at 15 minutes, which then fell towards basal levels over the remaining 45 minutes.
  • Administration of native GIP significantly elevated the overall insulinotropic response (p ⁇ 0.05) compared with glucose alone.
  • N-AcGIP(LysPAL 16 ) or N-AcGIP(LysPAL 37 ) where administered together with glucose a maximum plasma insulin concentration was observed at 15 minutes. Protracted biological activity for both analogues was clearly evident from 30 to 60 minutes.
  • Glucose-mediated plasma insulin concentrations were significantly higher compared in both control (p ⁇ 0.01 to p ⁇ 0.001) and GIP-treated animals (p ⁇ 0.05 to p ⁇ 0.001).
  • N-AcGIP(LysPAL 16 ) and N-AcGIP(LysPAL 37 ) revealed significant enhancements in overall glucose-mediated insulin release compared to native GIP (1.5-fold and 2.3-fold, respectively; p ⁇ 0.01 to p ⁇ 0.001).
  • N-AcGIP(LysPAL 37 ) was significantly more potent (1.5-fold: p ⁇ 0.001) than N-AcGIP(LysPAL 16 ) at stimulating insulin secretion.
  • FIGS. 52A and 52B illustrate the dose-dependent antihyperglycaemic and insulinotropic effects of GIP and the more potent analogue N-AcGIP(LysPAL 37 ) when administered with glucose to ob/ob mice. They are a pair of bar graphs showing dose-dependent effects of GIP and N-AcGIP(LysPAL 37 ) in ob/ob mice fasted for 18 hours. The incremental area under the curve (AUC) for glucose ( FIG. 52A ) and insulin ( FIG. 52B ) between 0 and 60 minutes after i.p.
  • AUC area under the curve
  • N-AcGIP(LysPAL 37 ) was substantially more potent than native GIP (p ⁇ 0.01 to p ⁇ 0.001) and exhibited prominent dose-dependent antihyperglycaemic and insulinotropic actions at all doses administered ( FIGS. 52A, 52B ).
  • the lowest concentration of N-AcGIP(LysPAL 37 ) tested (6.25 nmoles/kg) had highly significant antihyperglycaemic properties compared to glucose alone (p ⁇ 0.001). Consistent with this observation, 6.25 nmoles/kg N-AcGIP(LysPAL 37 ) elicited a prominent insulin response (2.0-fold; p ⁇ 0.01) compared to glucose alone.
  • FIGS. 53A through 53E are a set of graphs showing the effects of daily N-AcGIP(LysPAL 37 ) ( ⁇ ; black bars) administration on food intake ( FIG. 53A ), body weight ( FIG. 53B ), plasma glucose ( FIG. 53C ), insulin ( FIG. 53D ) and glycated hemoglobin N-AcGIP(LysPAL 37 ) (12.5 nmoles/kg/day) ( FIG.
  • GIP or N-AcGIP(LysPAL 37 ) had no effect on body weight or food intake ( FIGS. 53A, 53B ).
  • daily injection of N-AcGIP(LysPAL 37 ) resulted in a progressive lowering of plasma glucose, resulting in significantly (p ⁇ 0.05) lowered concentrations at 14 days ( FIG. 53C ).
  • glycated hemoglobin was also significantly (p ⁇ 0.01) decreased in N-AcGIP(LysPAL 37 ) treated ob/ob mice ( FIG. 53E ). These changes were accompanied by a tendency towards elevated insulin concentrations, but these did not achieve statistical significance over the time frame studies ( FIG. 53D ).
  • FIGS. 54A through 54D are a set of two line graphs ( FIGS. 54A, 54C ) and two bar graphs ( FIGS. 54B, 54D ) showing the effects of daily N-AcGIP(LysPAL 37 ) administration on glucose tolerance ( FIGS. 54A, 54B ) and plasma insulin response ( FIGS. 54C, 54D ) to glucose.
  • Tests were conducted after 14 daily injections of either N-AcGIP(LysPAL 37 ) (12.5 nmoles/kg/day; ⁇ ; black bars), native GIP (12.5 nmoles/kg/day; ⁇ ; lined bars) or saline vehicle (control; ⁇ ; white bars).
  • Glucose (18 mmoles/kg) was administered by intraperitoneal injection at the time indicated by the arrow. Plasma glucose and insulin AUC values for 0-60 minutes post injection are shown in the right panels. Values are means SEM for 8 mice. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001 compared to control. ⁇ p ⁇ 0.05, ⁇ p ⁇ 0.01, ⁇ p ⁇ 0.001 compared to native GIP.
  • FIGS. 54A, 54B Plasma glucose concentrations throughout the test and the overall 0-60 minutes AUC values were decreased (p ⁇ 0.01 to p ⁇ 0.001). This was accompanied by increased insulin concentrations during the latter stages (p ⁇ 0.05) and a greater (p ⁇ 0.01) overall AUC insulin response ( FIGS. 54C, 54D ).
  • daily administration of native GIP had no effect on glucose tolerance or the plasma insulin response to glucose compared with control ob/ob mice receiving saline injections for 14 days ( FIG. 54 ).
  • FIGS. 55A through 55D are a line graph and three bar graphs showing the effects of daily N-AcGIP(LysPAL 37 ) administration on insulin sensitivity ( FIGS. 55A, 55B ) and pancreatic weight ( FIG. 55C ) and insulin content ( FIG. 55D ).
  • N-AcGIP(LysPAL 37 ) (12.5 nmoles/kg/day; ⁇ ; black bars), native GIP (12.5 nmoles/kg/day; ⁇ ; lined bars) or saline vehicle (control; ⁇ ; white bars).
  • insulin 50 U/kg was administered by intraperitoneal injection at the time indicated by the arrow.
  • Plasma glucose AUC values for 0-60 minutes post injection are shown in the right panels. Values are means ⁇ SEM for 8 mice. *p ⁇ 0.05, **p ⁇ 0.01 compared to control. ⁇ p ⁇ 0.05, ⁇ p ⁇ 0.01 compared to native GIP.
  • FIGS. 55A, 55B Insulin sensitivity of the 3 groups of mice after 14 days treatment is shown in FIGS. 55A, 55B .
  • N-AcGIP(LysPAL 37 ) prompted a significant improvement of insulin sensitivity.
  • Both the individual glucose concentrations and 0-60 minutes AUC values were significantly different (p ⁇ 0.01) from the other two groups.
  • daily treatment with native GIP did not affect the characteristic insulin resistance of ob/ob mice ( FIG. 55A, 55B ).
  • FIGS. 56A through 56D are a set of two line graphs ( FIGS. 56A, 56C ) and two bar graphs ( FIGS. 56B, 56D ) showing the retention of glucose lowering ( FIGS. 56A, 56B ) and insulin releasing ( FIGS. 56C, 56D ) activity of N-AcGIP(LysPAL 37 ) and native GIP after daily injection for 14 days.
  • FIGS. 57A through 57D are a set of two line graphs ( FIGS.
  • FIGS. 57A, 57C and two bar graphs ( FIGS. 57B, 57D ) showing the acute glucose lowering ( FIGS. 57A, 57B ) and insulin releasing ( FIGS. 57C, 57D ) effects of N-AcGIP(LysPAL 37 ) after 14 daily injections of either N-AcGIP(LysPAL37) (12.5 nmoles/kg/day; ⁇ ; black bars), native GIP (12.5 nmoles/kg/day; ⁇ ; lined bars) or saline vehicle (control; ⁇ ; white bars).
  • N-AcGIP(LysPAL 37 ) (25 nmoles/kg) was administered by intraperitoneal injection with glucose (18 mmoles/kg) at the time indicated by the arrow.
  • Plasma glucose and insulin AUC values for 0-60 minutes post injection are shown in the right panels. Values are means ⁇ SEM for 8 mice. *p ⁇ 0.05, **p ⁇ 0.01 compared to mice receiving control injections. ⁇ p ⁇ 0.05, ⁇ p ⁇ 0.01 compared to group receiving injections of native GIP.

Abstract

The present invention provides peptide analogues which are antagonists of gastric inhibitory peptide (GIP). The peptides, based on GIP 1-42 include substitutions and/or modifications which have enhanced resistance to degradation by the enzyme dipeptidyl peptidase IV (DPP IV). The invention also provides a process of N terminally modifying GIP and the use of the peptide analogues for treatment of diabetes.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 11/090,787, filed Mar. 25, 2005, which is a continuation-in-part application of International Application No. PCT/GB2005/000710, which was filed on Feb. 25, 2005, designated the United States and was published in English, which claims benefit of U.K. Application No. GB 0404124.0, filed on Feb. 25, 2004. application Ser. No. 11/090,787 is also a continuation-in-part of U.S. application Ser. No. 09/937,687, filed Jan. 8, 2002, which is the U.S. National Phase Application of International Application No. PCT/GB00/01089, which designated the United States and was filed on Mar. 29, 2000 and published in English, which in turn claims the benefit of GB9907216.7, filed on Mar. 29, 1999, and GB9917565.5, filed Jul. 27, 1999. The entire teachings of the above applications are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to the release of insulin and the control of blood glucose concentration. More particularly the invention relates to antagonists of gastric inhibitory peptide (GIP) as pharmaceutical preparations for treatment of type 2 diabetes.
  • BACKGROUND
  • Obesity and diabetes are predicted to reach epidemic proportions throughout the world in the next 20 years and current treatments do not restore normal insulin sensitivity or glucose homeostasis, therein resulting in debilitating diabetic complications and premature death.
  • Gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1(7-36)amide (truncated GLP-1; tGLP-1) are two important insulin-releasing hormones secreted from endocrine cells in the intestinal tract in response to feeding. Together with autonomic nerves they play a vital supporting role to the pancreatic islets in the control of blood glucose homeostasis and nutrient metabolism.
  • GIP is released from intestinal endocrine K-cells into the bloodstream following ingestion of carbohydrate, protein and particularly fat (Meier, J. J. et al., 2002, Regul. Pept. 107:1-13). GIP was initially discovered through its ability to inhibit gastric acid secretion (Brown, J. C. et al. 1969, Can. J. Physiol. Pharmacol. 47:113-114) but its major physiological role is now generally believed to be that of an incretin hormone that targets pancreatic islets to enhance insulin secretion and help reduce postprandial hyperglycemia (Creutzfeldt, W., 2001, Exp. Clin. Endocrinol. Diabetes 109:S288-S303). GIP acts through binding to specific G-protein coupled GIP receptors located on pancreatic beta-cells (Wheeler, M. B. et al., 1995, Endocrinology 136:4629-4639). Like its sister incretin hormone, glucagon-like peptide-1 (GLP-1), this ability to stimulate insulin secretion plus other potentially beneficial actions on pancreatic beta-cell growth and differentiation have led to much interest in using GIP or GLP-1 receptor agonists in the treatment of type 2 diabetes (Creutzfeldt, W., 2001, Exp. Clin. Endocrinol. Diabetes 109:S288-S303; Holz, G. G. et al., 2003, Curr. Med. Chem. 10:2471-2483).
  • Since GIP functions as a potent and natural stimulator of insulin secretion released from the intestine by feeding, it is widely expected that antagonists opposing GIP action will block the insulin-releasing actions of GIP and impair both oral glucose tolerance and the glycemic response to nutrient ingestion. In fact, all studies published to date indicate that GIP is a key physiological component of the enteroinsular axis and that functional ablation of GIP leads to impaired glucose homeostasis moving the metabolic characteristic towards a type 2 diabetes phenotype (Gault, V. A. et al., 2002, Biochem. Biophys. Res. Commun. 290:1420-1426).
  • Dipeptidyl peptidase IV (DPP IV; EC 3.4.14.5) has been identified as a key enzyme responsible for inactivation of GIP and tGLP-1 in serum. This occurs through the rapid removal of the N-terminal dipeptides Tyr1-Ala 2 and His7-Ala8 giving rise to the main metabolites GIP(3-42) and GLP-1(9-36)amide, respectively. These truncated peptides are reported to lack biological activity or to even serve as antagonists at GIP or tGLP-1 receptors. The resulting biological half-lives of these incretin hormones in vivo are therefore very short, estimated to be no longer than 5 minutes. DPP IV is completely inhibited in serum by the addition of diprotin A (DPA, 0.1 mmol/1).
  • In situations of normal glucose regulation and pancreatic B-cell sensitivity, this short duration of action is advantageous in facilitating momentary adjustments to homeostatic control. However, the current goal of a possible therapeutic role of incretin hormones, particularly tGLP-1 in non-insulin dependent diabetes (NIDDM) therapy is frustrated by a number of factors in addition to finding a convenient route of administration. Most notable of these are rapid peptide degradation and rapid absorption (peak concentrations are reached in 20 minutes) and the resulting need for both high dosage and precise timing with meals. Recent therapeutic strategies have focused on precipitated preparations to delay peptide absorption and inhibition of GLP-1 degradation using specific inhibitors of DPP IV. A possible therapeutic role is also suggested by the observation that a specific inhibitor of DPP IV, isoleucine thiazolidide, lowered blood glucose and enhanced insulin secretion in glucose-treated diabetic obese Zucker rats presumably by protecting against catabolism of the incretin hormones tGLP-1 and GIP.
  • Studies have indicated that tGLP-1 infusion restores pancreatic B-cell sensitivity, insulin secretory oscillations and improved glycemic control in various groups of patients with impaired glucose tolerance (IGT) or NIDDM. Longer term studies also show significant benefits of tGLP-1 injections in NIDDM and possibly IDDM therapy, providing a major incentive to develop an orally effective or long-acting tGLP-1 analogue. Several attempts have been made to produce structurally modified analogues of tGLP-1 which are resistant to DPP IV degradation. A significant extension of serum half-life is observed with His7-glucitol tGLP-1 and tGLP-1 analogues substituted at position 8 with Gly, Aib (amino isobutyric acid), Ser or Thr. However, these structural modifications seem to impair receptor binding and insulinotrophic activity thereby compromising part of the benefits of protection from proteolytic degradation. In recent studies using His7-glucitol tGLP-1, resistance to DPP IV and serum degradation was accompanied by severe loss of insulin releasing activity.
  • GIP shares not only the same degradation pathway as tGLP-1 but many similar physiological actions, including stimulation of insulin and somatostatin secretion, and the enhancement of glucose disposal. These actions are viewed as key aspects in the antihyperglycemic properties of tGLP-1, and there is therefore good expectation that GIP may have similar potential as NIDDM therapy. Indeed, compensation by GIP is held to explain the modest disturbances of glucose homeostasis observed in tGLP-1 knockout mice. Apart from early studies, the anti-diabetic potential of GIP has not been explored and tGLP-1 may seem more attractive since it is viewed by some as a more potent insulin secretagogue when infused at so called physiological concentrations estimated by radioimmunoassay (RIA).
  • There is therefore a need for a diabetes treatment that includes an analogue of GIP which can cause release of insulin, yet also be resistant to rapid degradation by DPP IV.
  • SUMMARY OF THE INVENTION
  • Disclosed herein are GIP antagonist peptides which are resistant to rapid degradation by DPP IV.
  • The invention includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), which includes at least 12 amino acid residues from the N-terminal end of GIP(3-42). The invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), which includes at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu3.
  • The amino acid substituted at Glu3 can be selected from the group consisting of: proline, hydroxyproline, lysine, tyrosine, phenylalanine and tryptophan. Specifically, a proline can be substituted for Glu3. The peptide analogue can further include modification by fatty acid addition at an epsilon amino group of at least one lysine residue. The lysine residue can be Lys16, or Lys37
  • The peptide analogue of GIP(1-42) (SEQ ID NO:1) can include at least 12 amino acid residues from the N-terminal end of GIP(1-42), and an amino acid modification at amino acid residues 1, 2 or 3. The N-terminal amino acid residue can be acetylated. It can further comprising modification by fatty acid addition at an epsilon amino group of at least one lysine residue. The modification can be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-16, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue. The lysine residue can be Lys16, or Lys37.
  • The invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), wherein the analogue comprises a base peptide consisting of one of the following: GIP(1-12), GIP(1-13), GIP(1-14), GIP(1-15), GIP(1-16), GIP(1-17), GIP(1-18), GIP(1-19), GIP(1-20), GIP(1-21), GIP(1-22), GIP(1-23), GIP(1-24), GIP(1-25), GIP(1-26), GIP(1-27), GIP(1-28), GIP(1-29), GIP(1-30), GIP(1-31), GIP(1-32), GIP(1-33), GIP(1-34), GIP(1-35), GIP(1-36), GIP(1-37), GIP(1-38), GIP(1-39), GIP(1-40), GIP(1-41) and GIP(1-42), where the base peptide possesses one or more of the following modifications: (1) an amino acid substitution at Glu3; (2) a modification by fatty acid addition at an epsilon amino group of at least one lysine residue; and (3) a modification by N-terminal acetylation. Such a peptide analogue can have a proline substituted for Glu3. It can also have a modification in the form of a C-16 palmitate group linked to the epsilon amino group of a lysine residue. The modification can be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-16, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue. The lysine residue can be Lys16, or Lys37.
  • The invention further includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42), wherein the peptide analogue is resistant to degradation by enzyme DPP IV when compared to naturally-occurring GIP.
  • Also included is a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu3, wherein the peptide analogue is resistant to degradation by enzyme DPP IV when compared to naturally-occurring GIP.
  • In addition, the invention includes a peptide analogue of GIP(142) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42), wherein the peptide analogue modulates insulin secretion.
  • The invention also includes a peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(1-42) and having an amino acid substitution at Glu3, wherein the peptide analogue modulates insulin secretion.
  • The invention also includes use of any of the analogues in the preparation of a medicament for the treatment of obesity, insulin resistance, insulin resistant metabolic syndrome (Syndrome X) or type 2 diabetes.
  • The invention also includes a pharmaceutical composition including the peptide analogues. The pharmaceutical composition can further comprise a pharmaceutically acceptable carrier. The peptide analogue can be in the form of a pharmaceutically acceptable salt, or a pharmaceutically acceptable acid addition salt.
  • In a further aspect, the invention includes a method of treating insulin resistance, obesity, or type 2 diabetes, where the method comprises administering to a mammal in need of such treatment a therapeutically effective amount of the pharmaceutical composition.
  • According to the present invention there is provided an effective peptide analogue of the biologically active GIP(1-42) which has improved characteristics for treatment of Type 2 diabetes wherein the analogue comprises at least 15 amino acid residues from the N terminus of GIP(1-42) and has at least one amino acid substitution or modification at position 1-3 and not including Tyr1 glucitol GIP(1-42).
  • The structures of human and porcine GIP(1-42) are shown below. The porcine peptide differs by just two amino acid substitutions at positions 18 and 34.
  • The analogue may include modification by fatty acid addition at an epsilon amino group of at least one lysine residue.
  • The invention includes Tyr1 glucitol GIP(1-42) having fatty acid addition at an epsilon amino group of at least one lysine residue.
  • Analogues of GIP(1-42) may have an enhanced capacity to stimulate insulin secretion, enhance glucose disposal, delay glucose absorption or may exhibit enhanced stability in plasma as compared to native GIP. They also may have enhanced resistance to degradation.
  • Any of these properties will enhance the potency of the analogue as a therapeutic agent.
  • Analogues having D-amino acid substitutions in the 1, 2 and 3 positions and/or N-glycated, N-alkylated, N-acetylated or N-acylated amino acids in the 1 position are resistant to degradation in vivo.
  • Various amino acid substitutions at second and third amino terminal residues are included, such as GIP(1-42)Gly2, GIP(142)Ser2, GIP(1-42)Abu2, GIP(1-42)Aib2, GIP(1-42)D-Ala2, GIP(1-42)Sar2, and GIP(1-42)Pro3.
  • Amino-terminally modified GIP analogues include N-glycated GIP(1-42), N-alkylated GIP(1-42), N-acetylated GIP(1-42), N-acetyl-GIP(1-42) and N-isopropyl GIP(1-42).
  • Other stabilized analogues include those with a peptide isostere bond between amino terminal residues at position 2 and 3. These analogues may be resistant to the plasma enzyme dipeptidyl-peptidase IV (DPP IV) which is largely responsible for inactivation of GIP by removal of the amino-terminal dipeptide Tyr1-Ala2.
  • In particular embodiments, the invention provides a peptide which is more potent than human or porcine GIP in moderating blood glucose excursions, said peptide consisting of GIP(1-42) or N-terminal fragments of GIP(1-42) consisting of up to between 15 to 30 amino acid residues from the N-terminus (i.e., GIP(1-15) GIP(1-3)) with one or more modifications selected from the group consisting of:
  • (a) substitution of Ala2 by Gly;
  • (b) substitution of Ala2 by Ser;
  • (c) substitution of Ala2 by Abu;
  • (d) substitution of Ala2 by Aib;
  • (e) substitution of Ala2 by D-Ala;
  • (f) substitution of Ala2 by Sar (sarcosine);
  • (g) substitution of Glu3 by Pro;
  • (h) modification of Tyr1 by acetylation;
  • (i) modification of Tyr1 by acylation;
  • (j) modification of Tyr1 by alkylation;
  • (k) modification of Tyr1 by glycation;
  • (l) conversion of Ala 2-Glu3 bond to a psi [CH2NH] bond;
  • (m) conversion of Ala2-Glu3 bond to a stable peptide isotere bond; and
  • (n) (n-isopropyl-H) 1GIP.
  • The invention also provides the use of Tyr1-glucitol GIP in the preparation of a medicament for the treatment of diabetes.
  • The invention further provides improved pharmaceutical compositions including analogues of GIP with improved pharmacological properties.
  • Other possible analogues include certain commonly encountered amino acids, which are not encoded by the genetic code, for example, beta-alanine (beta-ala), or other omega-amino acids, such as 3-amino propionic, 4-amino butyric and so forth, ornithine (Orn), citrulline (Cit), homoarginine (Har), t-butylalanine (t-BuA), t-butylglycine (t-BuG), N-methylisoleucine (N-MeIle), phenylglycine (Phg), and cyclohexylalanine (Cha), norleucine (Nle), cysteic acid (Cya) and methionine sulfoxide (MSO), substitution of the D form of a neutral or acidic amino acid or the D form of tyrosine for tyrosine.
  • According to the present invention there is also provided a pharmaceutical composition useful in the treatment of diabetes type II which comprises an effective amount of the peptide as described herein, in admixture with a pharmaceutically acceptable excipient.
  • The invention also provides a method of N-terminally modifying GIP or analogues thereof the method comprising the steps of synthesizing the peptide from the C terminal to the penultimate N terminal amino acid, adding tyrosine to a bubbler system as a F-moc protected Tyr(tBu)-Wang resin, deprotecting the N-terminus of the tyrosine and reacting with the modifying agent, allowing the reaction to proceed to completion, cleaving the modified tyrosine from the Wang resin and adding the modified tyrosine to the peptide synthesis reaction.
  • Preferably the agent is glucose, acetic anhydride or pyroglutamic acid.
  • The invention will now be demonstrated with reference to the following non-limiting examples and the accompanying figures wherein.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 a illustrates degradation of GIP by DPP IV.
  • FIG. 1 b illustrates degradation of GIP and Tyr1 glucitol GIP by DPP IV.
  • FIG. 2 a illustrates degradation of GIP human plasma.
  • FIG. 2 b illustrates degradation of GIP and Tyr1 glucitol GIP by human plasma.
  • FIG. 3 illustrates electrospray ionization mass spectrometry of GIP, Tyr1-glucitol GIP and the major degradation fragment GIP(3-42).
  • FIG. 4 shows the effects of GIP and glycated GIP on plasma glucose homeostasis.
  • FIG. 5 shows effects of GIP on plasma insulin responses.
  • FIG. 6 illustrates DPP-IV degradation of GIP (1-42).
  • FIG. 7 illustrates DPP-IV degradation of GIP (Abu2).
  • FIG. 8 illustrates DPP-IV degradation of GIP (Sar2).
  • FIG. 9 illustrates DPP-IV degradation of GIP (Ser2).
  • FIG. 10 illustrates DPP-IV degradation of N-Acetyl-GIP.
  • FIG. 11 illustrates DPP-IV degradation of glycated GIP.
  • FIG. 12 illustrates human plasma degradation of GIP.
  • FIG. 13 illustrates human plasma degradation of GIP (Abu2).
  • FIG. 14 illustrates human plasma degradation of GIP (Sar2).
  • FIG. 15 illustrates human plasma degradation of GIP (Ser2).
  • FIG. 16 illustrates human plasma degradation of glycated GIP.
  • FIG. 17 shows the effects of various concentrations of GIP 1-42 and GIP (Abu2) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 18 shows the effects of various concentrations of GIP 1-42 and GIP (Abu2) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 19 shows the effects of various concentrations of GIP 1-42 and GIP (Sar2) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 20 shows the effects of various concentrations of GIP 1-42 and GIP (Sar2) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 21 shows the effects of various concentrations of GIP 1-42 and GIP (Ser2) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 22 shows the effects of various concentrations of GIP 1-42 and GIP (Ser2) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 23 shows the effects of various concentrations of GIP 1-42 and N-Acetyl-GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 24 shows the effects of various concentrations of GIP 1-42 and N-Acetyl-GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 25 shows the effects of various concentrations of GIP 1-42 and glycated GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 26 shows the effects of various concentrations of GIP 1-42 and glycated GIP 1-42 on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 27 shows the effects of various concentrations of GIP 1-42 and GIP (Gly2) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 28 shows the effects of various concentrations of GIP 1-42 and GIP (Gly2) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 29 shows the effects of various concentrations of GIP 1-42 and GIP (Pro3) on insulin release from BRIN-BD11 cells incubated at 5.6 mM glucose.
  • FIG. 30 shows the effects of various concentrations of CIP 1-42 and GIP (Pro3) on insulin release from BRIN-BD11 cells incubated at 16.7 mM glucose.
  • FIG. 31 a shows the primary structure of human gastric inhibitory polypeptide (GIP) (SEQ ID NO:1), and FIG. 31 b shows the primary structure of porcine gastric inhibitory polypeptide (GIP) (SEQ ID NO:2).
  • FIGS. 32A and 32B are a line graph and a bar graph, respectively, showing the effects of (Pro3)GIP on GIP-stimulated cyclic AMP generation and insulin secretion in vitro.
  • FIGS. 33A-33F are a set of six bar graphs showing the effects of Glu3-substituted forms of GIP and GIP(3-42) on GIP-stimulated insulin secretion in vitro.
  • FIGS. 34A through 34D are a set of two line graphs (FIGS. 34A, 34C) and two bar graphs (FIGS. 34B, 34D) showing that acute administration of (Pro3)GIP completely antagonises the actions of GIP on glucose tolerance (FIGS. 34A, 34B) and plasma insulin (FIGS. 34C, 34D) responses in obese diabetic ob/ob mice.
  • FIGS. 35A through 35D are a set of two line graphs (FIGS. 35A, 35C) and two bar graphs (FIGS. 35B, 35D) showing that acute administration of (Pro3)GIP impairs physiological meal-stimulated insulin release and worsens glycemic excursion in obese diabetic ob/ob mice.
  • FIGS. 36A and 36B are a set of two bar graphs showing that chronic administration of (Pro3)GIP for 11 days decreases plasma glucose and insulin concentrations of obese diabetic ob/ob mice.
  • FIGS. 37A through 37C are a set of three bar graphs showing that chronic administration of (Pro3)GIP for 11 days decreases glycated HbA1c, pancreatic insulin content and associated islet hypertrophy of obese diabetic ob/ob mice.
  • FIGS. 38A through 38D are a set of two line graphs (FIGS. 38A, 38C) and two bar graphs (FIGS. 38B, 38D) showing that chronic administration of (Pro3)GIP for 11 days improves glucose tolerance of obese diabetic ob/ob mice without change of circulating insulin.
  • FIG. 39 is a line graph showing that chronic administration of (Pro3)GIP for 11 days improves insulin sensitivity in obese diabetic ob/ob mice.
  • FIG. 40 is a line graph showing that the beneficial effects of chronic administration of (Pro3)GIP for 11 days in obese diabetic ob/ob mice are reversed 9 days after cessation of treatment.
  • FIGS. 41A and 41B are a set of two line graphs showing that chronic administration of (Pro3)GIP for 11 days causes glucose intolerance in normal mice with reversal by 9 days after cessation of treatment.
  • FIGS. 42A through 42D are a set of two line graphs (FIGS. 42A, 42C) and two bar graphs (FIGS. 42B, 42D) showing the effects of (Pro3)GIP on plasma glucose and insulin response to native GIP 4 hours after administration.
  • FIGS. 43A through 43D are a set of two line graphs and two bar graphs showing the effects of daily (Pro3)GIP administration on food intake (FIG. 43A), body weight (FIG. 43B), plasma glucose (FIG. 43C) and insulin (FIG. 43D) concentrations in ob/ob mice.
  • FIGS. 44A through 44D are a set of four line graphs with inset bar graphs showing the effects of daily (Pro3)GIP administration on glucose tolerance and plasma insulin response to glucose in ob/ob mice.
  • FIGS. 45A through 45D are a set of two line graphs (FIGS. 45A, 45C) and two bar graphs (FIGS. 45B, 45D) showing the effects of daily (Pro3)GIP administration (▴; black bars) or saline (□; white bars) on glucose (FIGS. 45A, 45B) and insulin (FIGS. 45C, 45D) responses to feeding in ob/ob mice fasted for 18 hours.
  • FIGS. 46A through 46D are a set of two line graphs (FIGS. 46A, 46C) and two bar graphs (FIGS. 46B, 46D) showing the effects of daily (Pro3)GIP administration on insulin sensitivity in ob/ob mice.
  • FIGS. 47A through 47D are a set of four bar graphs showing the effects of daily (Pro3)GIP administration on pancreatic weight (FIG. 47A), insulin content (FIG. 47B), islet number (FIG. 47C) and islet diameter (FIG. 47D) in ob/ob mice.
  • FIGS. 48A through 48F are a set of two bar graphs (FIGS. 48A, 48D) and four photomicrographs (FIGS. 48B, 48C, 48E, 48F), showing the effects of daily (Pro3)GIP administration on islet size and morphology in ob/ob)mice.
  • FIG. 49 is an illustration of how the GIP receptor (“GIP-R”) antagonist, (Pro3)GIP, counters beta cell hyperplasia, hyperinsulinemia and insulin resistance lead to improved glucose intolerance and diabetes control.
  • FIGS. 50A and 50B are a line graph and a bar graph, respectively, showing intracellular cyclic AMP production (FIG. 50A) by GIP (▴) and fatty acid derivatised GIP analogues N-AcGIP(LysPAL16) (□) and N-AcGIP(LysPAL37) (●), and insulin-releasing activity of glucose (5.6 mmo/l glucose; white bars), GIP (lined bars) and fatty acid derivatised GIP analogues (FIG. 50B) N-AcGIP(LysPAL16) (grey bars) and N-AcGIP(LysPAL37) (black bars) in the clonal pancreatic beta cell line, BRIN-BD11.
  • FIGS. 51A through 51D are a set of two line graphs (FIGS. 51A, 51C) and two bar graphs (FIGS. 51B, 51D) showing glucose lowering effects (FIGS. 51A, 51B) and insulin-releasing activity (FIGS. 51C, 51D) of GIP and fatty acid derivatised GIP analogues in 18 hour-fasted ob/ob mice.
  • FIGS. 52A and 52B are a pair of bar graphs showing dose-dependent effects of GIP and N-AcGIP(LysPAL37) in ob/ob mice fasted for 18 hours.
  • FIGS. 53A through 53E are a set of graphs showing the effects of daily N-AcGIP(LysPAL37) (●; black bars) administration on food intake (FIG. 53A), body weight (FIG. 53B), plasma glucose (FIG. 53C), insulin (FIG. 53D) and glycated hemoglobin N-AcGIP(LysPAL37) (12.5 nmoles/kg/day) (FIG. 53E).
  • FIGS. 54A through 54D are a set of two line graphs (FIGS. 54A, 54C) and two bar graphs (FIGS. 54B, 54D) showing the effects of daily N-AcGIP(LysPAL37) administration on glucose tolerance (FIGS. 54A, 54B) and plasma insulin response (FIGS. 54C, 54D) to glucose.
  • FIGS. 55A through 55D are a line graph and three bar graphs showing the effects of daily N-AcGIP(LysPAL37) administration on insulin sensitivity (FIGS. 55A, 55B) and pancreatic weight (FIG. 55C) and insulin content (FIG. 55D).
  • FIGS. 56A through 56D are a set of two line graphs (FIGS. 56A, 56C) and two bar graphs (FIGS. 56B, 56D) showing the retention of glucose lowering (FIGS. 56A, 56B) and insulin releasing (FIGS. 56C, 56D) activity of N-AcGIP(LysPAL37) and native GIP after daily injection for 14 days.
  • FIGS. 57A through 57D are a set of two line graphs (FIGS. 57A, 57C) and two bar graphs (FIGS. 57B, 57D) showing the acute glucose lowering (FIGS. 57A, 57B) and insulin releasing (FIGS. 57C, 57D) effects of N-AcGIP(LysPAL37) after 14 daily injections of either N-AcGIP(LysPAL37) (12.5 nmoles/kg/day; ●; black bars), native GIP (12.5 nmoles/kg/day; ▴; lined bars) or saline vehicle (control; □; white bars).
  • DETAILED DESCRIPTION
  • The peptide analogues disclosed herein display resistance to degradation by the enzyme DPP IV. These analogues include those with alterations at residues 1, 2 and/or 3 of the native GIP(1-42) peptide, where the alterations interfere with or delay cleavage by DPP IV. The alterations can include chemical modification of one or more of the first three amino acids, such as by acylation, acetylation, alkylation, glycation, conversion of a bond between two amino acids, such as to a psi-[CH2NH] bond, or to a stable isotere bond, or addition of an isopropyl group. The alterations can also include amino acid substitutions at the 1, 2, and/or 3 position, to either a different naturally-occurring amino acid, or an amino acid not encoded by the genetic code. Other alterations are also possible, and the object is to prevent cleavage of the peptide by DPP IV, yet still allow for insulin secretion. This may be accomplished by alterations at other regions of the peptide, such as by alterations that alter the three-dimensional structure to prevent DPP IV cleavage, yet still allow insulin secretion.
  • Preferred alterations include chemical modifications of residues 1, 2, and 3, amino acid substitutions at residues 1, 2, and 3, and chemical modifications of lysine residues throughout the protein.
  • Particularly preferred alterations include acetylation of Tyr1 and linkage of a C-16 palmitate group to the epsilon amino group of a lysine residue (especially Lys16 or Lys37), or substitution of Glu3, especially by proline. The modification can also be the linking of, e.g., a C-8, a C-10, a C-12, a C-14, a C-18 or a C-20 palmitate group to the epsilon amino group of a lysine residue.
  • It has been found that longer-term, as opposed to acute, GIP receptor antagonism using Glu3-substituted forms of GIP, such as (Pro3)GIP, improve obesity-related insulin resistance and associated glucose intolerance. This has uncovered an unexpected approach to the therapy of obesity, insulin resistance and type 2 diabetes.
  • As described in Example 1 below, an N-terminally modified version of the GIP protein was prepared, as were analogues of the modified protein. The protein and its analogues were then evaluated in Example 2 for their antihyperglycemic and insulin-releasing properties in vivo, and were found to exhibit a substantial resistance to amino peptidase degradation and increased glucose lowering activity relative to native GIP.
  • The 42 amino acid GIP is an important incretin hormone released into the circulation from endocrine K-cells of the duodenum and jejunum following ingestion of food. The high degree of structural conservation of GIP among species supports the view that this peptide plays an important role in metabolism. Secretion of GIP is stimulated directly by actively transported nutrients in the gut lumen without a notable input from autonomic nerves. The most important stimulants of GIP release are simple sugars and unsaturated long chain fatty acids, with amino acids exerting weaker effects. As with tGLP-1, the insulin-releasing effect of GIP is strictly glucose-dependent. This affords protection against hypoglycemia and thereby fulfills one of the most desirable features of any current or potentially new antidiabetic drug.
  • The present results demonstrate for the first time that Tyr1-glucitol GIP displays profound resistance to serum and DPP IV degradation. Using ESI-MS the present study showed that native GIP was rapidly cleaved in vitro to a major 4748.4 Da degradation product corresponding to GIP(3-42), which confirmed previous findings using matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Serum degradation was completely inhibited by diprotin A (Ile-Pro-Ile), a specific competitive inhibitor of DPP IV, confirming this as the main enzyme for GIP inactivation in vivo. In contrast, Tyr1-glucitol GIP remained almost completely intact after incubation with serum or DPP IV for up to 12 hours. This indicates that glycation of GIP at the amino-terminal Tyr1 residue masks the potential cleavage site from DPP IV and prevents removal of the Tyr1-Ala2 dipeptide from the N-terminus preventing the formation of GIP(3-42).
  • Consistent with in vitro protection against DPP IV, administration of Tyr1-glucitol GIP significantly enhanced the antihyperglycemic activity and insulin-releasing action of the peptide when administered with glucose to rats. Native GIP enhanced insulin release and reduced the glycemic excursion as observed in many previous studies. However, amino-terminal glycation of GIP increased the insulin-releasing and antihyperglycemic actions of the peptide by 62% and 38% respectively, as estimated from insulin area under the curve (AUC) measurements. Detailed kinetic analysis is difficult due to necessary limitation of sampling times, but the greater insulin concentrations following Tyr1-glucitol GIP as opposed to GIP at 30 minutes post-injection is indicative of a longer half-life. The glycemic rise was modest in both peptide-treated groups and glucose concentrations following injection of Tyr1-glucitol GIP were consistently lower than after GIP. Since the insulinotropic actions of GIP are glucose-dependent, it is likely that the relative insulin-releasing potency of Tyr1-glucitol GIP is greatly underestimated in the present in vivo experiments.
  • In vitro studies in the laboratory of the present inventors using glucose-responsive clonal B-cells showed that the insulin-releasing potency of Tyr1glucitol GIP was several orders of magnitude greater than GIP and that its effectiveness was more sensitive to change of glucose concentrations within the physiological range. Together with the present in vivo observations, this suggests that N-terminal glycation of GIP confers resistance to DPP IV degradation whilst enhancing receptor binding and insulin secretory effects on the B-cell. These attributes of Tyr1-glucitol GIP are fully expressed in vivo where DPP IV resistance impedes degradation of the peptide to GIP(3-42), thereby prolonging the half-life and enhancing effective concentrations of the intact biologically active peptide. It is thus possible that glycated GIP is enhancing insulin secretion in vivo both by enhanced potency at the receptor as well as improving DPP IV resistance. Thus numerous studies have shown that GIP (3-42) and other N-terminally modified fragments, including GIP(4-42), and GIP(17-42) are either weakly effective or inactive in stimulating insulin release. Furthermore, evidence exists that N-terminal deletions of GIP result in receptor antagonist properties in GIP receptor transfected Chinese hamster kidney cells [9], suggesting that inhibition of GIP catabolism would also reduce the possible feedback antagonism at the receptor level by the truncated GIP(3-42).
  • In addition to its insulinotopic actions, a number of other potentially important extrapancreatic actions of GIP may contribute to the enhanced antihyperglycemic activity and other beneficial metabolic effects of Tyr1-glucitol GIP. These include the stimulation of glucose uptake in adipocytes, increased synthesis of fatty acids and activation of lipoprotein lipase in adipose tissue. GIP also promotes plasma triglyceride clearance in response to oral fat loading. In liver, GIP has been shown to enhance insulin-dependent inhibition of glycogenolysis. GIP also reduces both glucagon-stimulated lipolysis in adipose tissue as well as hepatic glucose production. Finally, recent findings indicate that GIP has a potent effect on glucose uptake and metabolism in mouse isolated diaphragm muscle. This latter action may be shared with tGLP-1 and both peptides have additional benefits of stimulating somatostatin secretion and slowing down gastric emptying and nutrient absorption.
  • This study demonstrates that the glycation of GIP at the aminoterminal Tyr1 residue limits GIP catabolism through impairment of the proteolytic actions of serum peptidases and thus prolongs its half-life in vivo. This effect is accompanied by enhanced antihyperglycemic activity and raised insulin concentrations in vivo, suggesting that such DPP IV resistant analogues are potentially useful therapeutic agents for NIDDM. Tyr1-glucitol GIP appears to be particularly interesting in this regard since such amino-terminal modification of GIP enhances rather than impairs glucose-dependent insulinotropic potency as was observed recently for tGLP-1.
  • As shown in Table 1 in Example 3, glycated GIP, acetylated GIP, GIP(Ser2) are GIP(Abu2) more resistant than native GIP to in vitro degradation with DPP IV. From these data GIP(Sar2) appears to be less resistant. As shown in Table 2, all analogues tested exhibited resistance to plasma degradation, including GIP(Sar2) which from DPP IV data appeared least resistant of the peptides tested. DPA substantially inhibited degradation of GIP and all analogues tested with complete abolition of degradation in the cases of GIP(Abu2), GIP(Ser2) and glycated GIP. This indicates that DPP IV is a key factor in the in vivo degradation of GIP.
  • As shown in FIGS. 17-30, the glycated GIP analogue exhibited a considerably greater insulinotropic response relative to native GIP. N-terminal acetylated GIP exhibited a similar pattern and the GIP(Ser2) analogue also evoked a strong response. From these tests, GIP(Gly2) and GIP(Pro3) appeared to the least potent analogues in terms of insulin release. Other stable analogues tested, namely GIP(Abu2) and GIP(Sar2), exhibited a complex pattern of responsiveness dependent on glucose concentration and dose employed. Thus, very low concentrations were extremely potent under hyperglycemic conditions (16.7 mM glucose). This suggests that even these analogues may prove therapeutically useful in the treatment of type 2 diabetes where insulinotropic capacity combined with in vivo degradation dictates peptide potency.
  • A major limitation to the possible therapeutic use of both GIP and GLP-1 as insulin-releasing agents for the treatment of diabetes is their rapid degradation in vivo by dipeptidylpeptidase-IV (DPP-IV; EC 3.4.14.5). This enzyme rapidly removes the amino-terminal dipeptide from the two peptides producing GIP(3-42) and GLP-1(9-36), which lack biological activity (Gault, V. A. et al., 2002, Biochem. Biophys. Res. Commun. 290:1420-1426). In searching for stable amino-terminally modified forms of GIP and GLP-1, it was discovered that a novel synthetic GIP analogue with a single proline substitution at position 3 close to the cleavage site, (Pro3)GIP, functioned as a potent GIP receptor antagonist.
  • As shown in Example 4, below, (Pro3)GIP, other Glu3-substituted forms of GIP and GIP(3-42) are potent GIP receptor antagonists both in vitro and in vivo. Experiments evaluating the effects of chronic GIP receptor antagonism in normal mice using (Pro3)GIP demonstrated a substantial but reversible deterioration of glucose tolerance. This is entirely consistent with the widely recognised physiological role of GIP as an important insulin-releasing intestinal hormone involved in the regulation of glucose disposal following feeding (Meier, J. J. et al., 2002, Regul. Pept. 107:1-13).
  • Most notably, and in complete contrast to normal mice, the experiments disclosed herein show that chronic (Pro3)GIP administration to obese diabetic ob/ob mice for 11 days does not worsen glucose intolerance and diabetes status at all. Surprisingly, GIP receptor antagonism in this obese insulin resistant model was associated with highly substantial improvements of glycated HbA1c, plasma glucose and insulin concentrations, glucose tolerance and insulin sensitivity. Pancreatic insulin content was also decreased and the characteristic islet hypertrophy of the obese mutant was partially reversed. These latter observations indicate a decreased secretory demand for endogenous insulin following (Pro3)GIP as a result of improved insulin resistance.
  • Indeed, insulin sensitivity tests conducted in ob/ob mice 11 days into (Pro3)GIP treatment revealed a substantial improvement in tissue insulin insensitivity, which more than compensated for the functional ablation of the insulin-releasing GIP component of the enteroinsular axis. The exact mechanism responsible for this effect on insulin sensitivity is unknown but ablation of direct action of circulating GIP on adipose tissue metabolism is a likely candidate. Also noteworthy was the fact that all these beneficial actions of (Pro3)GIP in obese diabetic ob/ob mice were reversed within 9 days cessation of treatment.
  • These results clearly indicate that (Pro3)GIP and other analogues based on Glu3-substituted or N-terminally truncated forms of the gastrointestinal hormone GIP can offer an important therapeutic means of alleviating insulin resistance for the treatment of obesity, the so-called insulin resistant (metabolic) syndrome and type 2 diabetes in humans.
  • Some studies have attempted to enhance incretin action using DPP IV inhibitors or stable analogs of GLP-1 and GIP for the treatment of type 2 diabetes (Green, B. D. et al., 2004, Curr. Pharm. Des. 10:In Press; Drucker, D. J. et al., Diabetes Care 10:2929-2940). Such an approach is reliant on the possibility that incretin action is defective in diabetes and that the underlying defects responsible for metabolic disarray might be over-ridden by exogenous GLP-1 or GIP administration. There is some evidence for a beneficial and possibly therapeutic role of both GLP-1 and GIP analogs in diabetes (Meier, J. J. et al., 2002, Regul. Pept. 107:1-13; Gault, V. A. et al., 2003, Biochem Biophys Res Commun 308:207-213; Holst, J. J. et al., 2004, Am. J. Physiol. Endocrinol. Metab. 287:E199-E206; Green, B. D. et al., 2004, Curr. Pharm. Des. 10:In Press; Drucker, D. J. et al., Diabetes Care 10:2929-2940). Nevertheless, understanding of the possible involvement of incretin hormones in the pathophysiology of diabetes is lacking, partly due to cross-reaction of classical GLP-1 and GIP radioimmunoassays with the predominant DPP IV-generated truncated peptide forms, GLP-1(9-36) and GIP(3-42), which circulate at particularly high concentrations (Meier, J. J. et al., 2002, Regul. Pept. 107:1-13). Some clinical studies seems to suggest existence of a defect in the secretion of GLP-1 and a defect in the action of GIP in type 2 diabetes (Hoist, J. J. et al., 2004, Am. J. Physiol. Endocrinol. Metab. 287:E199-E206). However, the basis for such a conclusion is not impressive given the many previous contradictory human studies (Morgan, L. M., “Insulin secretion and the enteroinsular axis,” In: Nutrient regulation of insulin secretion, Flatt, P. R., ed., London, Portland Press, 1992, p. 1-22), and the likelihood that the reported insensitivity of pancreatic beta cells to GIP (Vilsboll, T. et al., 2002, Diabetologia 45:1111-1119) may reflect a generalized secretory dysfunction rather than a specific cellular defect (Meier, J. J. et al., 2003, Metabolism 52:1579-1585). Indeed, the insulin secretory response to all secretagogues, including GLP-1 is compromised in type 2 diabetes (Kjems, L. L. et al., 2003, Diabetes 52:380-386; Flatt, P. R. et al., “Defective insulin secretion in diabetes and insulinoma,” in Nutrient regulation of insulin secretion, Flatt P. R., ed. London, Portland Press, 1992, p. 341-386). Thus the proposed use of GLP-1 and GIP for diabetes therapy is reliant on peptide engineering to provide analogs of incretin hormones with improved potency due to DPP IV resistance, decreased renal clearance and/or enhanced GIP receptor and post-receptor activity (Gault, V. A. et al., 2003, Biochem Biophys Res Commun 308:207-213).
  • Although no single animal model can match the complex etiology of type 2 diabetes in man, studies of the ob/ob syndrome in mice have highlighted notable abnormalities of GIP in relation to the interplay between hyperphagia, hyperinsulinemia and the metabolic demise associated with progressive obesity-diabetes (Flatt, P. R. et al., 1983, Diabetes 32:433-435; Flatt, P. R. et al., 1984, J. Endocrinol. 101:249-256; Bailey, C. J. et al., 1986, Acta Endocrinol. (Copenh) 112:224-229). These animals constitute a model of non-insulin dependent diabetes associated with gross obesity and severe insulin resistance, driven by leptin deficiency (Bailey, C. J. et al., “Animal syndromes resembling type 2 diabetes,” in Textbook of Diabetes, 3rd ed. Pickup J. C. & Williams G., eds. Oxford, Blackwell Science Ltd., 2003, p. 25.1-25.30). Furthermore, recent research suggests an interaction between leptin and the enteroinsular axis (Anini, Y. et al., 2003, Diabetes 52:252-259) and that over-stimulation of the GIP receptor (“GIP-R”) on adipocytes appears to be an important contributor to fat deposition in ob/ob mice (Miyawaki, K. et al., 2002, Nat. Med. 8:738-742).
  • As shown in Example 5, below, daily injections of the stable and specific GIP-R antagonist, (Pro3)GIP can be used to chemically ablate the GIP-R and evaluate the role of endogenous circulating GIP in obesity-diabetes as manifested in ob/ob mice. The results reveal a cardinal role for GIP in insulin resistance and associated metabolic disturbances, and provide the first experimental evidence that GIP-R antagonists might provide a novel and effective means of treating obesity-driven forms of type 2 diabetes.
  • Knockout of the GIP-R in normal mice has been shown to result in significant impairment of glucose tolerance and meal-induced insulin secretion without appreciable effects on food intake, body weight or basal glucose or insulin concentrations (Miyawaki, K. et al., 1999, Proc. Nat. Acad. Sci. USA 96:14843-14847). More recent studies with genetic GIP-R knockout mice have corroborated these findings and additionally shown that GIP has a significant involvement in the enteroinsular axis (Pederson, R. A. et al., 1998, Diabetes 47:1046-1052; Pamir, N. et al., 2003, Am. J. Physiol. Endocrinol. Metab. 284:E931-939). However, double knockout of receptors for GLP-1 and GIP results in a surprisingly modest deterioration of glucose homeostasis (Hansotia, T., et al., 2004, Diabetes 53:1326-1335; Preitner, F., et al., 2004, J. Clin. Invest. 113:635-645), indicating possible up-regulation of compensatory mechanisms during life-long deletion of GLP-1 and GIP receptors.
  • The analogue (Pro3)GIP can be used as a specific and potent antagonist of the GIP-R that is highly stable and resistant to DPP IV-mediated degradation (Gault, V. A. et al., 2002, Biochem. Biophys. Res. Commun. 290:1420-1426). Using (Pro3)GIP acutely, the results disclosd herein highlight the involvement of GIP in the plasma insulin response to feeding and the enteroinsular axis of ob/ob mice (Gault, V. A. et al., 2003, Diabetologia 46:222-230). Comparison with the effects of the GLP-1-R antagonist, exendin(9-39), indicates that GIP contributes substantially to the insulin releasing actions of the enteroinsular axis and represents the major physiological incretin (Gault, V. A. et al., 2003, Diabetologia 46:222-230). Once daily administration of (Pro3)GIP to normal mice for 11 days results in the reversible impairment of glucose tolerance associated with decreased insulin sensitivity (Irwin, N., 2004, Biol. Chem. 385:845-852). Basal and postprandial insulin secretion together with pancreatic insulin content and islet morphology were unchanged. Thus longer-term chemical ablation of GIP-R function with daily (Pro3)GIP can mimic the phenotype induced by genetic GIP-R knockout in mice with the exception of revealing a potentially important additional effect of endogenous GIP on insulin action, which appears to be independent of enhanced insulin secretion.
  • Far from reproducing this predicted scenario and the metabolic deterioration observed following genetic or chemical knockout of the GIP-R in normal mice (Miyawaki, K. et al., 1999, Proc. Nat. Acad. Sci. USA 96:14843-14847; Irwin, N., 2004, Biol. Chem. 385:845-852), ob/ob mice treated with daily (Pro3)GIP for 11 days exhibited a marked improvement in diabetic status. This included decreased fasting and basal hyperglycemia, lowered glycated hemoglobin, improved glucose tolerance and a significantly diminished glycemic excursion following feeding. Notably, basal and glucose-stimulated plasma insulin concentrations were decreased, suggesting that insulin sensitivity must have improved significantly following (Pro3)GIP in order to restrain the hyperglycemia. Indeed, insulin sensitivity tests conducted after 11 days of (Pro3)GIP administration revealed a 57% increase in the glucose-lowering action of exogenous insulin. Bearing in mind that the severity of the ob/ob syndrome represents a tough test for current antidiabetic drugs, including insulin, sulfonylureas, metformin and thiazolidenediones (Flatt, P. R. et al., “Defective insulin secretion in diabetes and insulinoma,” in Nutrient regulation of insulin secretion, Flatt P. R., ed. London, Portland Press, 1992, p. 341-386; Stevenson, R. W. et al., 1995, The Diabetes Annual 9:175-191; Wiernsperger, N. F., “Preclinical pharmacology of biguanides,” Handbook of Experimental Pharmacology 119:305-358, 1996), induction of such rapid and reversible changes by GIP-R blockade using (Pro3)GIP is unprecedented.
  • It is important to note that the above effects were observed independently of any change in food intake or body weight in (Pro3)GIP treated ob/ob mice. This accords with the view that endogenous GIP lacks effects on feeding activity (Meier, J. J. et al., 2002, Regul. Pept. 107:1-13). However, the observation on body weight contrasts with findings in ob/ob mice cross-bred to genetically knockout GIP-R function (Miyawaki, K. et al., 2002, Nat. Med. 8:738-742). Thus in these transgenic mice, life-long depletion of GIP-R function was associated with decreased body weight gain and significant amelioration of both adiposity and insulin resistance compared with control (Lepob/Lepob) mice (Miyawaki, K. et al., 2002, Nat. Med. 8:738-742). In this previous study, the improvement of insulin sensitivity may have been a simple consequence of reduced adipose tissue mass as this would significantly enhance peripheral glucose disposal (Bailey, C. J. et al., “Animal syndromes resembling type 2 diabetes,” in Textbook of Diabetes, 3rd ed. Pickup J. C. & Williams G., eds. Oxford, Blackwell Science Ltd., 2003, p. 25.1-25.30). However, the present results observed in rapid time and without effects on feeding or body weight clearly indicate the involvement of an alternative mechanism.
  • The most plausible explanation for the present data stem from appreciation of the key milestones in the age-dependent progression of the ob/ob syndrome on the Aston background as depicted in FIG. 49, which is an illustration of how the GIP-R antagonist, (Pro3)GIP, counters beta cell hyperplasia, hyperinsulinemia and insulin resistance lead to improved glucose intolerance and diabetes control. Possible longer-term direct actions of GIP on adipocyte function and fat stores, suggested by studies in GIP-R knockout ob/ob mice have been omitted.
  • Due to double recessive ob mutation and resulting leptin deficiency, young ob/ob mice develop a profound early hyperphagia (Bailey, C. J., et al., 1982, Int. J. Obes. 6:11-21). Substantial enteroendocrine stimulation results in K-cell hyperplasia and markedly elevated concentrations of intestinal and circulating GIP (Flatt, P. R. et al., 1983, Diabetes 32:433-435; Flatt, P. R. et al., 1984, J. Endocrinol. 101:249-256; Bailey, C. J. et al., 1986, Acta Endocrinol. (Copenh) 112:224-229). This in turn promotes islet hypertrophy and beta cell hyperplasia (Bailey, C. J., et al., 1982, Int. J. Obes. 6:11-21) together with marked hyperinsulinemia and mounting insulin resistance (Flatt, P. R., et al., 1981, Horm Metab Res 13:556-560). This process manifests itself in terms of rising basal hyperglycemia and glucose intolerance. A vicious spiral is thus established wherein beta cell compensation results in marked hyperinsulinemia which attempts to moderate increasing insulin resistance (Bailey, C. J., et al., 1982, Int. J. Obes. 6:11-21; Flatt, P. R., et al., 1981, Horm Metab Res 13:556-560). Viewed in this context, it is clear that chemical ablation of GIP-R function with daily (Pro3)GIP will decrease beta cell stimulation and hyperinsulinemia. However, instead of causing further impairment of glucose homeostasis, a preferentially marked improvement of insulin sensitivity results in a substantial improvement of the metabolic syndrome. Further support for this scenario, is the partial amelioration of islet hypertrophy and beta cell hyperplasia in (Pro3)GIP treated ob/ob mice (FIG. 48). Notably, average islet diameter was diminished with the largest islets (>15 mm) being replaced by a greater proportion with small or medium diameters (0.1-15 mm). These effects were largely reversed by 9 day cessation of treatment, supporting the idea of active islet and beta cell growth in adult ob/ob mice (Bailey, C. J., et al., 1982, Int. J. Obes. 6:11-21). Recent observations indicate that GIP acts as a mitotic stimulus and anti-apoptotic agent to the beta cell (Pospisilik, J. A. et al., 2003, Diabetes 52:741-750; Trumper, A. et al., 2001, Mol. Endocrinol. 15:1559-1570; Ehses, J. A. et al., 2003, Endocrinology 144:4433-4445, Trumper, A. et al., 2002, J. Endocrinol. 174:233-246). Thus, it is believed that negative effects of (Pro3)GIP on islet size reflects a combination of decreased proliferation and increased apoptosis of beta cells.
  • The results shown in Example 5 have demonstrated for the first time that daily administration of the GIP-R antagonist, (Pro3)GIP, improves glucose tolerance and ameliorates insulin resistance and abnormalities of islet structure and function in ob/ob mice. Notably, these effects were reversed by discontinuation of (Pro3)GIP for 9 days. Freedom from any obvious side effects also accords with earlier observations in normal mice (Irwin, N., 2004, Biol. Chem. 385:845-852) and mice genetically engineered with life-long GIP-R deficiency (Miyawaki, K. et al., 2002, Nat. Med. 8:738-742). The present observations point to a cardinal role of endogenous GIP in the pathogenesis of obese-insulin resistant-diabetes. More importantly, the data indicate that GIP-R antagonists, such as (Pro3)GIP, provide a novel, physiological and effective means to treat obese type 2 diabetes through the alleviation of insulin resistance.
  • In Example 6, fatty acid derivatisation was used to develop two novel long-acting, N-terminally modified GIP analogues (N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37)).
  • Degradation studies were carried out with dipeptidylpeptidase IV (DPP IV). Cyclic AMP production was assessed using GIP receptor transfected CHL fibroblasts. In vitro insulin release was assessed in BRIN-BD11 cells. Insulinotropic and glycaemic responses to acute and long-term peptide administration were evaluated in obese diabetic (ob/ob) mice.
  • In contrast to GIP both analogues displayed resistance to DPP IV degradation. The analogues also stimulated cyclic AMP production and exhibited significantly improved in vitro insulin secretion compared to control. Administration of N-AcGIP(LysPAL16) or N-AcGIP(LysPAL37) together with glucose in ob/ob mice significantly reduced the glycaemic excursion and improved the insulinotropic response compared to GIP. Dose-response studies with N-AcGIP(LysPAL37) revealed highly significant decreases in the overall glycaemic excursion and increases in circulating insulin even with 6.25 nmoles/kg. Once daily injection of ob/ob mice with N-AcGIP(LysPAL37) over 14 days significantly decreased plasma glucose, glycated haemoglobin and improved glucose tolerance compared with saline or native GIP. Plasma and pancreatic insulin were significantly increased, together with a significant enhancement in the insulin response to glucose and a notable improvement of insulin sensitivity. No evidence was found for GIP-receptor desensitization and the metabolic effects of N-AcGIP(LysPAL37) were independent of any change in feeding or body weight.
  • These results show that novel fatty acid derivatised, N-terminally modified analogues of GIP such as N-AcGIP(LysPAL37), may have significant potential for the treatment of type 2 diabetes.
  • One approach to counter both renal clearance and enzyme degradation of GIP concerns the utilisation of fatty acid derivatisation together with N-terminal modification. Fatty acid derivatisation has previously been shown to prolong the half-life of insulin (Kurtzhals, P. et al., 1995, Biochem. J. 312: 725-731) and the sister incretin glucagon-like peptide-1 (GLP-1) (Knudsen, L. B. et al., 2000, J. Med. Chem. 43: 1664-1669; Green, B. D. et al., 2004, Biol. Chem. 385: 169-177; Kim, J. G. et al., 2003, Diabetes 52: 751-759). A number of N-terminally modified GIP analogues have been developed which exhibit profound resistance to DPP IV (Hinke, S. A. et al., 2002, Diabetes 51: 656-661; Gault, V. A. et al., 2002, Biochem. J. 367: 913-920; Gault, V. A. et al., 2003, J. Endocrinol. 176: 133-141; O'Harte, F. P. M. et al., 1999, Diabetes 48: 758-765). Several of these, most notably those modified at Tyr1 of GIP with an addition of an acetyl, glucitol, pyroglutamyl or Fmoc adduct, exhibit enhanced activity at the GIP receptor in vitro (Gault, V. A. et al., 2002, Biochem. J. 367: 913-920; O'Harte, F. P. M. et al., 1999, Diabetes 48: 758-765; O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). As a result of degradation resistance and enhanced cellular activity, these analogues display enhanced and protracted antihyperglycaemic and insulin-releasing activity when administered acutely to animals with obesity-diabetes (Hinke, S. A. et al., 2002, Diabetes 51: 656-661; Gault, V. A. et al., 2002, Biochem. J. 367: 913-920; Gault, V. A. et al., 2003, J. Endocrinol. 176: 133-141; O'Harte, F. P. M. et al., 1999, Diabetes 48: 758-765; O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). Of these, N-AcGIP has emerged as the most effective DPP IV-resistant analogue, substantially augmenting the plasma insulin response and curtailing the glycaemic excursion following conjoint administration with glucose to ob/ob mice (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291).
  • Example 6 was designed to evaluate the metabolic stability, biological activity and antidiabetic potential of novel second generation fatty acid derivatised, N-terminally modified N-AcGIP analogues, N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37). Both GIP analogues contain a C-16 palmitate group linked to the epsilon-amino group of Lys at positions 16 or 37, in combination with an N-terminal (Tyr1) acetyl group (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). The relative stability to DPP IV degradation, insulin secretion and cyclic AMP properties were examined in vitro together with acute and dose-response studies in obese diabetic ob/ob mice. The most effective analogue, N-AcGIP(LysPAL37) was administered to ob/ob mice by once daily intraperitoneal injection for 14 days prior to evaluation of glucose homeostasis, pancreatic beta cell function and insulin sensitivity. Possible desensitization of GIP receptor action by prolonged exposure to elevated concentrations of N-AcGIP(LysPAL37) was also examined. The results indicate the particular promise of the novel second generation N-terminally acetylated GIP analogue, N-AcGIP(LysPAL37), as a potential therapeutic agent for the treatment of type 2 diabetes.
  • Despite their many attributes, DPP IV-resistant analogues of GIP and GLP-1, like their native counterparts, are still subject to renal filtration. To circumvent this problem, fatty acid derivatisation has been used to improve the duration of action of GLP-1 (Knudsen, L. B. et al., 2000, J. Med. Chem. 43: 1664-1669; Green, B. D. et al., 2004, Biol. Chem. 385: 169-177; Kim, J. G. et al., 2003, Diabetes 52: 751-759). The most promising analogue, NN2211 (Liraglutide), appears effective in improving blood glucose control in type 2 diabetic subjects despite a tendency towards promotion of nausea possibly due to slowing of gastric emptying (Agersø, H. et al., 2002, Diabetologia 45: 195-202).
  • Example 6 describes the results of introducing two specific modifications to the native GIP hormone, namely N-terminal acetylation and C-terminal fatty acid derivatisation. N-terminal acetylation was employed, as previously described (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291), to significantly enhance stability to DPP IV. In contrast, conjugation of a C-16 palmitate residue at the epsilon-amino group of Lys16 or Lys37 was introduced to extend the biological half-life through binding to circulating proteins (Kurtzhals, P. et al., 1995, Biochem. J. 312: 725-731). Unlike the native peptide, both GIP analogues appeared to be completely resistant to enzymatic breakdown by DPP IV, which corroborates previous observations with N-AcGIP(O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). Furthermore, both analogues displayed similar or slightly better insulin-releasing and cyclic AMP generating properties to native GIP and N-AcGIP when tested in the in vitro cellular systems (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291).
  • To assess the antihyperglycaemic and insulinotropic potential of the fatty acid derivatised GIP analogues in vivo, obese diabetic (ob/ob) mice were employed. The ob/ob syndrome is an extensively studied model of spontaneous obesity and diabetes, exhibiting hyperphagia, marked obesity, moderate hyperglycaemia and severe hyperinsulinemia (Bailey, C. J. et al., 1982, Int. J. Obesity 6: 11-21). As described in previous studies (Gault, V. A. et al., 2002, Biochem. J. 367: 913-920; Gault, V. A. et al., 2003, J. Endocrinol. 176: 133-141), native GIP only modestly reduced the glycaemic excursion in ob/ob mice reflecting the severe insulin resistance of this mutant animal model (Bailey, C. J. et al., 1982, Int. J. Obesity 6: 11-21). In sharp contrast, both N-acetylated GIP analogues additionally substituted with a palmitate molecule at Lys16 or Lys37 (N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37)) significantly lowered plasma glucose levels compared to the native peptide. This was accompanied by significantly enhanced insulin-releasing activity, especially in the case of N-AcGIP(LysPAL37). The significantly protracted insulinotropic response to both fatty acid derivatised GIP analogues at 60 minutes despite substantially lower plasma glucose is indicative of an extended plasma half-life. This may be due to binding of both palmitate derivatised GIP analogues to serum albumin, therefore significantly impairing their clearance via the kidneys (Meier, J. J. et al., 2004, Diabetes 53: 654-662). However, further studies including establishment of sensitive and specific immunoassays for the novel GIP analogues would be needed to confirm such actions.
  • N-AcGIP(LysPAL37) appeared to be the best fatty acid derivatised analogue displaying a more protracted, significantly enhanced insulin-releasing potency over N-AcGIP(LysPAL16) in vivo. Reasons for the increased potency of N-AcGIP(LysPAL37) remain unclear, but one explanation is an extended half-life. Another possibility may be that a fatty acid chain linked to the Lys closer to the C-terminus of the peptide may have less of a detrimental effect upon the bioactive region of the molecule known to be located within the N-terminus (Gault, V. A. et al., 2002, Biosci. Rep. 22: 523-528; Hinke, S. A. et al., 2001, Biochim. Biophys. Acta 1547: 143-55; Manhart, S. et al., 2003, Biochemistry 42: 3081-3088). However, similarities between the in vitro biological activities of the two palmitate substituted analogues make this less likely.
  • Given that N-AcGIP(LysPAL37) was the more potent of the two analogues in vivo, it was further utilised in dose-response studies. Considering that native GIP itself has only very modest effects in ob/ob mice, as sometimes observed with type 2 diabetic subjects (Nauck, M. A. et al., 1993, J. Clin. Invest. 91: 301-307; Meier, J. J. et al., 2004, Diabetes 53: 220-224; Vilsbøll, T. et al., 2002, Diabetologia 45: 1111-1119), it is remarkable that N-AcGIP(LysPAL37), even at the lowest dose of 6.25 nmoles/kg, exhibited significant glucose-lowering and insulinotropic activity when administered with glucose. Considering N-AcGIP(LysPAL37) is subject to albumin binding, the fact that it is still highly biologically active even at lower concentrations indicates striking potency.
  • Daily administration of N-AcGIP(LysPAL37) to young adult ob/ob mice by intraperitoneal injection (12.5 nmoles/kg) resulted in a progressive lowering of plasma glucose concentrations and a significant decrease of glycated haemoglobin by 14 days. This was associated with a substantial improvement of glucose tolerance. Importantly food intake and body weight were unchanged ruling out the possibility that improvement of glucose homeostasis was merely the consequence of body weight loss. These observations also indicate that N-AcGIP(LysPAL37) did not exert any untoward toxic actions affecting feeding over the study period. This is in harmony with recent studies showing that GIP does not inhibit gastric emptying (Meier, J. J. et al., 2003, Am. J. Physiol. Endocrinol. Metab. 286: 621-625). Daily administration of native GIP to ob/ob mice for 14 days had no effect on any of the parameters measure, consistent with the very short half-life of the native GIP in vivo.
  • As expected, a key component of the beneficial action of N-AcGIP(LysPAL37) concerned effects on beta-cells. Thus although native GIP is a weak stimulus to insulin secretion in ob/ob mice at the age studied, plasma and pancreatic insulin concentrations were raised in ob/ob mice receiving the novel fatty acid derivatised analogue. This is consistent with the action of GIP as a promoter of proinsulin gene expression (Wang, Y. et al., 1996, Mol. Cell. Endocrinol. 116:81-87) and exemplifies the increased potency reported for N-terminally modified GIP analogues in this animal model of diabetes (Hinke, S. A. et al., 2002, Diabetes 51: 656-661; Gault, V. A. et al., 2002, Biochem. J. 367: 913-920; Gault, V. A. et al., 2003, J. Endocrinol. 176: 133-141; O'Harte, F. P. M. et al., 1999, Diabetes 48: 758-765; O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). Furthermore, the insulin response to glucose was significantly enhanced in ob/ob mice receiving N-AcGIP(LysPAL37). This ability to augment or restore pancreatic beta cell glucose responsiveness has been similarly observed with GLP-1 (Holz, G. G. et al., 1993, Nature 28: 362-365; Flamez, D. et al., 1998, Diabetes 47: 646-652). As with observations on glycaemic control, none of these attributes were reproduced by daily injections of native GIP.
  • Results of insulin sensitivity tests conducted after 14 days treatment indicate that the improvement of diabetic status achieved in ob/ob mice with N-AcGIP(LysPAL37) was not solely due to the potentiation of insulin secretion. Thus, these animals also exhibited a significant improvement of insulin sensitivity compared to the GIP or saline treated groups. Given that hyperinsulinemia is generally believed to down-regulate insulin receptor function (Marshall, S. et al., 1981, Diabetes 30: 746-753), this suggests that N-AcGIP(LysPAL37) may exert other compensatory effects. Further study is necessary to evaluate this aspect but possibilities include inhibition of counter-regulatory hormones and effects on extrapancreatic sites such as muscle, adipose tissue and liver (Morgan, L. M. et al., 1996, Biochem. Soc. Trans. 24:585-591; O'Harte, F. P. M. et al., 1998, J. Endocrinol. 156: 237-243; Yip, R. G. et al., 1998, Endocrinology 139: 4004-4007).
  • Irrespective of knowledge of the full range of actions contributing to the antihyperglycaemic effect of N-AcGIP(LysPAL37), a currently envisaged problem of long-term treatment with stable analogues of GIP or GLP-1 concerns desensitization of hormone receptor action (Delmeire, D. et al., 2004, Biochem. Pharmacol. 68: 33-39). Although this has been observed during prolonged exposure of pancreatic beta cells to GIP in rats (Tseng, C. C. et al., 1996, Am. J. Physiol. 270: E661-E666), there was no evidence that treatment with N-AcGIP(LysPAL37) for 14 days compromised the glucose lowering or insulin releasing actions of N-AcGIP(LysPAL37). Thus the antidiabetic actions of N-AcGIP(LysPAL37) were clearly evident when the analogue was administered acutely together with glucose. Furthermore, the acute effects of N-AcGIP(LysPAL37) in such experiments were identical in groups of ob/ob mice receiving either N-AcGIP(LysPAL37), native GIP or saline injections for 14 days.
  • Such data clearly indicate that prolonged exposure to N-AcGIP(LysPAL37) does not induce and possibly overcomes inherent GIP receptor desensitization in ob/ob mice. Given the high circulating concentrations of GIP in these obese-diabetic rodents (Flatt, P. R. et al., 1983, Diabetes 32: 433-435; Flatt, P. R. et al., 1984, J. Endocrinol. 101: 249-256), it is tempting to link beta cell refractoriness to GIP evident in ob/ob mice to simple receptor desensitization at the hands of inappropriate secretion and metabolism of GIP.
  • The data shown herein demonstrate that N-terminally acetylated GIP carrying a palmitate group linked to Lys at position 37 displays resistance to DPP IV and an impressive profile of bioactivity manifested by potent and long-acting glucose-lowering activity in a commonly employed animal model of obesity-diabetes. This activity profile provides strong encouragement for the development of long-acting fatty acid derivatised N-terminally modified analogues of GIP for the once-daily treatment of type 2 diabetes.
  • The peptide analogues of the present invention have use in treating diseases and conditions caused by improper modulation of insulin levels, including diabetes, type 2 diabetes, insulin resistance, insulin resistant metabolic syndrome (Syndrome X), and obesity.
  • A peptide analogue produced by the methods of the present invention can be used in a pharmaceutical composition, wherein the analogue is combined with a pharmaceutically acceptable carrier. Such a composition may also contain (in addition to the analogue and a carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration.
  • Administration of the peptide analogue of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as by oral ingestion, inhalation, topical application or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Administration can be internal or external; or local, topical or systemic.
  • The compositions containing a peptide analogue of this invention can be administered intravenously, as by injection of a unit dose, for example. The term “unit dose” when used in reference to a therapeutic composition of the present invention refers to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent, i.e., carrier or vehicle.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • When a therapeutically effective amount of the composition of the present invention is administered orally, the composition of the present invention will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder contain from about 5 to 95% protein of the present invention, and preferably from about 25 to 90% protein of the present invention. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of the composition of the present invention, and preferably from about 1 to 50% of the composition of the present invention.
  • When a therapeutically effective amount of the composition of the present invention is administered by intravenous, cutaneous or subcutaneous injection, the composition of the present invention will be in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable protein solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the composition of the present invention, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
  • Use of timed release or sustained release delivery systems are also included. A sustained-release matrix, as used herein, is a matrix made of materials, usually polymers, which are degradable by enzymatic or acid/base hydrolysis or by dissolution. Once inserted into the body, the matrix is acted upon by enzymes and body fluids. The sustained-release matrix desirably is chosen from biocompatible materials such as liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), polylactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, poly(ortho)esters, polyproteins, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone. A preferred biodegradable matrix is a matrix of one of either polylactide, polyglycolide, or polylactide co-glycolide (co-polymers of lactic acid and glycolic acid).
  • The therapeutic compositions can include pharmaceutically acceptable salts of the components therein, e.g., which may be derived from inorganic or organic acids. By “pharmaceutically acceptable salt” is meant those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well-known in the art. For example, S. M. Berge, et al., describe pharmaceutically acceptable salts in detail in J Pharmaceutical Sciences (1977) 66:1 et seq., which is incorporated herein by reference in its entirety. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like. The salts may be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting a free base function with a suitable organic acid. Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerophosphate, hemisulfate, heptonoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxymethanesulfonate (isethionate), lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained. Examples of acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, hydrobromic acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid.
  • As used herein, the terms “pharmaceutically acceptable”, “physiologically tolerable” and grammatical variations thereof as they refer to compositions, carriers, diluents and reagents, are used interchangeably and represent that the materials are capable of administration to or upon a mammal with a minimum of undesirable physiological effects such as nausea, dizziness, gastric upset and the like. The preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art and need not be limited based on formulation. Typically such compositions are prepared as injectables either as liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared. The preparation can also be emulsified.
  • The active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein. Suitable excipients include, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof. In addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient.
  • The amount of peptide analogue of the present invention in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, on the nature of prior treatments which the patient has undergone, and on a variety of other factors, including the type of injury, the age, weight, sex, medical condition of the individual. Ultimately, the attending physician will decide the amount of the analogue with which to treat each individual patient. Initially, the attending physician will administer low doses of peptide analogue and observe the patient's response. Larger doses of peptide analogue may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further.
  • Additional guidance on methods of determining dosages can be found in standard references, for example, Spilker, Guide to Clinical Studies and Developing Protocols, Raven Press Books, Ltd., New York, 1984, pp. 7-13 and 54-60; Spilker, Guide to Clinical Trials, Raven Press, Ltd., New York, 1991, pp. 93-101; Craig et al., Modern Pharmacology, 2d ed., Little Brown and Co., Boston, 1986, pp. 127-133; Speight, Avery's Drug Treatment: Principles and Practices of Clinical Pharmacology and Therapeutics, 3d ed., Williams and Wilkins, Baltimore, 1987, pp. 50-56; Tallarida et al., Principles in General Pharmacology, Springer-Verlag, New York, 1998, pp. 18-20; and Olson, Clinical Pharmacology Made Ridiculously Simple, MedMaster, Inc., Miami, 1993, pp. 1-5.
  • EXAMPLES Example 1 Preparation of N-terminally Modified GIP and Analogues Thereof
  • The N-terminal modification of GIP is essentially a three step process. Firstly, GIP is synthesized from its C-terminal (starting from a Fmoc-Gln (Trt)-Wang resin (Calbiochem Novabiochem, Beeston, Nottingham, UK) up to the penultimate N-terminal amino-acid (Ala2) on an automated peptide synthesizer (Applied Biosystems, California, USA). The synthesis follows standard Fmoc peptide chemistry protocols. Secondly, the N-terminal amino acid of native GIP (Tyr) is added to a manual bubbler system as a Fmoc-protected Tyr(tBu)-Wang resin. This amino acid is deprotected at its N-terminus (piperidine in DMF (20% v/v)) and allowed to react with a high concentration of glucose (glycation, under reducing conditions with sodium cyanoborohydride), acetic anhydride (acetylation), pyroglutamic acid (pyroglutamyl) etc. for up to 24 hours as necessary to allow the reaction to go to completion. The completeness of reaction is monitored using the ninhydrin test which determines the presence of available free a-amino groups. Thirdly (once the reaction is complete), the now structurally modified Tyr is cleaved from the Wang resin (95% TFA, and 5% of the appropriate scavengers. N.B. Tyr is considered to be a problematic amino acid and may need special consideration) and the required amount of N-terminally modified-Tyr consequently added directly to the automated peptide synthesiser, which will carry on the synthesis, thereby stitching the N-terminally modified-Tyr to the a-amino of GIP (Ala2), completing the synthesis of the GIP analogue. This peptide is cleaved off the Wang resin (as above) and then worked up using the standard Buchner filtering, precipation, rotary evaporation and drying techniques.
  • Example 2 Preparation of Tyr1-Glucitol GIP and Its Properties In Vivo
  • The following example investigates preparation of Tyr1glucitol GIP together with evaluation of its antihyperglycemic and insulin-releasing properties in vivo. The results clearly demonstrate that this novel GIP analogue exhibits a substantial resistance to aminopeptidase degradation and increased glucose lowering activity compared with the native GIP.
  • Research Design and Methods
  • Materials. Human GIP was purchased from the American Peptide Company (Sunnyvale, Calif., USA). HPLC grade acetonitrile was obtained from Rathburn (Walkersburn, Scotland). Sequencing grade trifluoroacetic acid (TFA) was obtained from Aldrich (Poole, Dorset, UK). All other chemicals purchased including dextran T-70, activated charcoal, sodium cyanoborohydride and bovine serum albumin fraction V were from Sigma (Poole, Dorset, UK). Diprotin A (DPA) was purchased from Calbiochem-Novabiochem (UK) Ltd. (Beeston, Nottingham, UK) and rat insulin standard for RIA was obtained from Novo Industria (Copenhagen, Denmark). Reversed-phase Sep-Pak cartridges (C-18) were purchased from Millipore-Waters (Milford, Mass., USA). All water used in these experiments was purified using a Milli-Q, Water Purification System (Millipore Corporation, Milford, Mass., USA).
  • Preparation of Tyr1-Glucitol GIP. Human GIP was Incubated with Glucose Under reducing conditions in 10 mmol/l sodium phosphate buffer at pH 7.4 for 24 hours. The reaction was stopped by addition of 0.5 mol/l acetic acid (30 μl) and the mixture applied to a Vydac (C18) (4.6×250 mm) analytical HPLC column (The Separations Group, Hesperia, Calif., USA) and gradient elution conditions were established using aqueous/TFA and acetonitrile/TFA solvents. Fractions corresponding to the glycated peaks were pooled, taken to dryness under vacuum using an AES 1000 Speed-Vac concentrator (Life Sciences International, Runcorn, UK) and purified to homogeneity on a Supelcosil (C-8) (4.6×150 mm) column (Supelco Inc., Poole, Dorset, UK).
  • Degradation of GIP and Tyr1-glucitol GIP by DPP IV. HPLC-purified GIP or Tyr1-glucitol GIP were incubated at 37° C. with DPP-IV (5 mU) for various time periods in a reaction mixture made up to 500 μl with 50 mmol/l triethanolamine-HCl, pH 7.8 (final peptide concentration 1 umol/l). Enzymatic reactions were terminated after 0, 2, 4 and 12 hours by addition of 5 μl of 10% (v/v) TFA/water. Samples were made up to a final volume of 1.0 ml with 0.12% (v/v) TFA and stored at −20° C. prior to HPLC analysis.
  • Degradation of GIP and Tyr1-glucitol GIP by human plasma. Pooled human plasma (20 μl) taken from six healthy fasted human subjects was incubated at 37° C. with GIP or Tyr1-glucitol GIP (10 μg) for 0 and 4 hours in a reaction mixture made up to 500 μl, containing 50 mmol/l triethanolamine/HCL buffer pH 7.8. Incubations for 4 hours were also performed in the presence of diprotin A (5 mU). The reactions were terminated by addition of 5 μl of TFA and the final volume adjusted to 1.0 ml using 0.1% v/v TFA/water. Samples were centrifuged (13,000 g, 5 minutes) and the supernatant applied to a C-18 Sep-Pak cartridge (Millipore-Waters) which was previously primed and washed with 0.1% (v/v) TFA/water. After washing with 20 ml 0.12% TFA/water, bound material was released by elution with 2 ml of 80% (v/v) acetonitrile/water and concentrated using a Speed-Vac concentrator (Runcorn, UK). The volume was adjusted to 1.0 ml with 0.12% (v/v) TFA/water prior to HPLC purification.
  • HPLC analysis of degraded GIP and Tyr1-glucitol GIP. Samples were applied to a Vydac C-18 widepore column equilibriated with 0.12% (v/v) TFA/H2O at a flow rate of 1.0 ml/minute. Using 0.1% (v/v) TFA in 70% acetonitrile/H2O, the concentration of acetonitrile in the eluting solvent was raised from 0% to 31.5% over 15 min, to 38.5% over 30 minutes and from 38.5% to 70% over 5 minutes, using linear gradients. The absorbance was monitored at 206 nm and peak areas evaluated using a model 2221 LKB integrator. Samples recovered manually were concentrated using a Speed-Vac concentrator.
  • Electrospray ionization mass spectrometry (ESI-MS). Samples for ESI-MS analysis containing intact and degradation fragments of GIP (from DPP IV and plasma incubations) as well as Tyr1-glucitol GIP, were further purified by HPLC. Peptides were dissolved (approximately 400 pmol) in 100 μl of water and applied to the LCQ benchtop mass spectrometer (Finnigan MAT, Hemel Hempstead, UK) equipped with a microbore C-18 HPLC column (150×2.0 mm, Phenomenex, Ltd., Macclesfield, UK). Samples (30 μl direct loop injection) were injected at a flow rate of 0.2 ml/min, under isocratic conditions 35% (v/v) acetonitile/water. Mass spectra were obtained from the quadripole ion trap mass analyzer and recorded. Spectra were collected using full ion scan mode over the mass-to-charge (m/z) range 150-2000. The molecular masses of GIP and related structures were determined from ESI-MS profiles using prominent multiple charged ions and the following equation
    M r =iM i −iM h
  • where Mr=molecular mass; Mi=m/z ratio; i=number of charges; Mh=mass of a proton.
  • In vivo biological activity of GIP and Tyr1-glucitol GIP. Effects of GIP and Tyr1-glucitol GIP on plasma glucose and insulin concentrations were examined using 10-12 week old male Wistar rats. The animals were housed individually in an air conditioned room and 22±2° C. with a 12 hour light/12 hour dark cycle. Drinking water and a standard rodent maintenance diet (Trouw Nutrition, Belfast, Northern Ireland) were supplied ad libitum. Food was withdrawn for an 18 hour period prior to intraperitoneal injection of glucose alone (18 mmol/kq body weight) or in combination with either GIP or Tyr1-glucitol GIP (10 nmol/kg). Test solutions were administered in a final volume of 8 ml/kg body weight. Blood samples were collected at 0, 15, 30 and 60 minutes from the cut tip of the tail of conscious rats into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany). Samples were centrifuged using a Beckman microcentrifuge for about 30 seconds at 13,000 g. Plasma samples were aliquoted and stored at −20° C. prior to glucose and insulin determinations. All animal studies were done in accordance with the Animals (Scientific Procedures) Act 1986.
  • Analyses. Plasma glucose was assayed by an automated glucose oxidase procedure using a Beckman Glucose Analyzer II [33]. Plasma insulin was determined by dextran charcoal radioimmunoassay as described previously [34]. Incremental areas under plasma glucose and insulin area under the curve (AUC) were calculated using a computer program (CAREA) employing the trapezoidal rule [35] with baseline subtraction. Results are expressed as mean ±SEM and values were compared using the Student's unpaired t-test. Groups of data were considered to be significantly different if P<0.05.
  • Degradation of GIP and Tyr1-glucitol GIP by DPP IV FIG. 1 illustrates the typical peak profiles obtained from the HPLC separation of the products obtained from the incubation of GIP (FIG. 1 a) or Tyr1-glucitol GIP (FIG. 1 b) with DPP IV for 0, 2, 4 and 12 hours. The retention times of GIP and Tyr1-glucitol GIP at t=0 were 21.93 minutes and 21.75 minutes respectively. Degradation of GIP was evident after 4 hours incubation (54% intact), and by 12 hours the majority (60%) of intact GIP was converted to the single product with a retention time of 21.61 minutes. Tyr1-glucitol GIP remained almost completely intact throughout 2-12 hours incubation. Separation was on a Vydac C-18 column using linear gradients of 0% to 31.5% acetonitrile over 15 minutes, to 38.5% over 30 minutes and from 38.5 to 70% acetonitrile over 5 minutes.
  • Degradation of GIP and Tyr1-glucitol GIP by human plasma. FIG. 2 shows a set of typical HPLC profiles of the products obtained from the incubation of GIP or Tyr1-glucitol GIP with human plasma for 0 and 4 hours. GIP (FIG. 2 a) with a retention time of 22.06 minutes was readily metabolised by plasma within 4 hours incubation giving rise to the appearance of a major degradation peak with a retention time of 21.74 minutes. In contrast, the incubation of Tyr1-glucitol GIP under similar conditions (FIG. 2 b) did not result in the formation of any detectable degradation fragments during this time with only a single peak being observed with a retention time of 21.77 minutes. Addition of diprotin A, a specific inhibitor of DPP IV, to GIP during the 4 hours incubation completely inhibited degradation of the peptide by plasma. Peaks corresponding with intact GIP, GIP (3-42) and Tyr1-glucitol GIP are indicated. A major peak corresponding to the specific DPP IV inhibitor tripeptide DPA appears in the bottom peanels with retention time of 16-29 minutes.
  • Identification of GIP degradation fragments by ESI-MS. FIG. 3 shows the monoisotopic molecular masses obtained for GIP (FIG. 3A), Tyr1-glucitol GIP (FIG. 3B) and the major plasma degradation fragment of GIP (FIG. 3C) using ESI-MS. The peptides analyzed were purified from plasma incubations as shown in FIG. 2. Peptides were dissolved (approximately 400 pmol) in 100 μl of water and applied to the LC/MS equipped with a microbore C-18 HPLC column. Samples (30 μl direct loop injection) were applied at a flow rate of 0.2 ml/min, under isocratic conditions 35% acetonitrile/water. Mass spectra were recorded using a quadripole ion trap mass analyzer. Spectra were collected using full ion scan mode over the mass-to-charge (m/z) range 150-2000. The molecular masses (Mr) of GIP and related structures were determined from ESI-MS profiles using prominent multiple charged ions and the following equation Mr=iMi−iMh. The exact molecular mass (Mr) of the peptides were calculated using the equation Mr=iMi−iMh as defined above in Research Design and Methods. After spectral averaging was performed, prominent multiple charges species (M+3H)3+ and (M+4H)4+ were detected from GIP at m/z 1661.6 and 1246.8, corresponding to intact Mr 4981.8 and 4983.2 Da, respectively (FIG. 3A). Similarly, for Tyr1-glucitol GIP ((M+4H)4+ and (M+5H)5+) were detected at m/z 1287.7 and 1030.3, corresponding to intact molecular masses of Mr 5146.8 and 5146.5 Da, respectively (FIG. 3B). The difference between the observed molecular masses of the quadruply charged GIP and the N-terminally modified GIP species (163.6 Da) indicated that the latter peptide contained a single glucitol adduct corresponding to Tyr1-glucitol GIP. FIG. 3C shows the prominent multiply charged species (M+3H)3+ and (M+4H)4+ detected from the major fragment of GIP at m/z 1583.8 and 1188.1, corresponding to intact Mr 4748.4 and 4748 Da, respectively (FIG. 3C). This corresponds with the theoretical mass of the N-terminally truncated form of the peptide GIP(3-42). This fragment was also the major degradation product of DPP IV incubations (data not shown).
  • Effects of GIP and Tyr1-glucitol GIP on plasma glucose homeostasis. FIGS. 4 and 5 show the effects of intraperitoneal (ip) glucose alone (118 mmol/kg) (control group), and glucose in combination with GIP or Tyr1-glucitol GIP (10 nmol/kg) on plasma glucose and insulin concentrations.
  • FIG. 4A shows plasma glucose concentrations after i.p. glucose alone (18 mmol/kg) (control group), or glucose in combination with either GIP of Tyr1-glucitol GIP (10 nmol/kg). The time of injection is indicated by the arrow (0 minutes). FIG. 4B shows plasma glucose AUC values for 0-60 minutes post injection. Values are mean ±SEM for six rats. **P<0.01, ***P<0.001 compared with GIP and Tyr1-glucitol GIP; †P<0.05, ††P<0.01 compared with non-glucated GIP. FIG. 5A shows plasma insulin concentrates after i.p. glucose along (18 mmol/kg) (control group), or glucose in combination with either with GIP or glycated GIP (10 nmol/kq). The time of injection is indicated by the arrow. FIG. 5B shows plasma insulin AUC values were calculated for each of the 3 groups up to 90 minutes post injection. The time of injection is indicated by the arrow (0 minutes). Plasma insulin AUC values for 0-60 minutes post injection. Values are mean ±SEM for six rats. *P<0.05, **P<0.001 compared with GIP and Tyr1-glucitol GIP; †P<0.05, ††P<0.01 compared with non-glycated GIP.
  • Compared with the control group, plasma glucose concentrations and area under the curve (AUC) were significantly lower following administration of either GIP or Tyr1-glucitol GIP (FIGS. 4A, B). Furthermore, individual values at 15 and 30 minutes together with AUC were significantly lower following administration of Tyr1-glucitol GIP as compared to GIP. Consistent with the established insulin-releasing properties of GIP, plasma insulin concentrations of both peptide-treated groups were significantly raised at 15 and 30 minutes compared with the values after administration of glucose alone (FIG. 5A). The overall insulin responses, estimated as AUC were also significantly greater for the two peptide-treated groups (FIG. 5B). Despite lower prevailing glucose concentrations than the GIP-treated group, plasma insulin response, calculated as AUC, following Tyr1-glucitol GIP was significantly greater than after GIP (FIG. 5B). The significant elevation of plasma insulin at 30 minutes is of particular note, suggesting that the insulin-releasing action of Tyr1-glucitol GIP is more protracted than GIP even in the face of a diminished glycemic stimulus (FIGS. 4A, 5A).
  • Example 3 Additional N-Terminal Structural Modifications of GIP
  • This example further looked at the ability of additional N-terminal structural modifications of GIP in preventing inactivation by DPP and in plasma and their associated increase in both the insulin-releasing potency and potential therapeutic value. Native human GIP, glycated GIP, acetylated GIP and a number of GIP analogues with N-terminal amino acid substitutions were tested.
  • Materials and Methods. High-performance liquid chromatography (HPLC) grade acetonitrile was obtained from Rathburn (Walkersbum, Scotland). Sequencing grade trifluoroacetic acid (TFA) was obtained from Aldrich (Poole, Dorset, UK). Dipeptidyl peptidase IV was purchased from Sigma (Poole, Dorset, UK), and Diprotin A was purchased from Calbiochem Novabiochem (Beeston, Nottingham, UK). RPMI 1640 tissue culture medium, foetal calf serum, penicillin and streptomycin were all purchased from Gibco (Paisley, Strathclyde, UK). All water used in these experiments was purified using a Milli-Q, Water Purification System (Millipore, Milford, Mass., USA). All other chemicals used were of the highest purity available.
  • Synthesis of GIP and N-terminally modified GIP analogues. GIP, GIP(Abu2), GIP(Sar2), GIP(Ser2), GIP(Gly2) and GIP(Pro3) were sequentially synthesized on an Applied Biosystems automated peptide synthesizer (model 432A) using standard solid-phase Fmoc procedure, starting with an Fmoc-Gln-Wang resin. Following cleavage from the resin by trifluoroacetic acid:water, thioanisole, ethanedithiol (90/2.5/5/2.5, a total volume of 20 ml/g resin), the resin was removed by filtration and the filtrate volume was decreased under reduced pressure. Dry diethyl ether was slowly added until a precipitate was observed. The precipitate was collected by low-speed centrifugation, resuspended in diethyl ether and centrifuged again, this procedure being carried out at least five times. The pellets were then dried in vacuo and judged pure by reversed-phase HPLC on a Waters Millennium 2010 chromatography system (Software version 2.1.5.). N-terminal glycated and acetylated GIP were prepared by minor modification of a published method.
  • Electrospray ionization-mass spectrometry (ESI-MS) was carried out as described in Example 2. Degradation of GIP and novel GIP analogues by DPP IV and human plasma was carried out as described in Example 2.
  • Culture of insulin secreting cells. BRIN-BD11 cells [30] were cultured in sterile tissue culture flasks (Coming, Glass Works, UK) using RPMI-1640 tissue culture medium containing 10% (v/v) foetal calf serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin) and 11.1 mM glucose. The cells were maintained at 37° C. in an atmosphere of 5% CO2 and 95% air using a LEEC incubator (Laboratory Technical Engineering, Nottingham, UK).
  • Acute tests for insulin secretion. Before experimentation, the cells were harvested from the surface of the tissue culture flasks with the aid of trypsin/EDTA (Gibco), seeded into 24-multiwell plates (Nunc, Roskilde, Denmark) at a density of 1.5×105 cells per well, and allowed to attach overnight at 37° C. Acute tests for insulin release were preceded by 40 minutes pre-incubation at 37° C. in 1.0 ml Krebs Ringer bicarbonate buffer (115 mM NaCl, 4.7 mM KCl, 1.28 mM CaCl2, 1.2 mM KH2PO4, 1.2 mM MgSO4, 10 mM NaHCO3, 5 g/l bovine serum albumin, pH 7.4) supplemented with 1.1 mM glucose. Test incubations were performed (n=12) at two glucose concentrations (5.6 mM and 16.7 mM) with a range of concentrations (10-13 to 10−8 M) of GIP or GIP analogues. After 20 minutes incubation, the buffer was removed from each well and aliquots (200 μl) were used for measurement of insulin by radioimmunoassay [31]).
  • Statistical analysis. Results are expressed as mean ±S.E.M. and values were compared using the Student's unpaired t-test. Groups of data were considered to be significantly different if P<05.
  • Structural identification of GIP and GIP analogues by ESI-MS. The monoisotopic molecular masses of the peptides were determined using ESI-MS. After spectral averaging was performed, prominent multiple charged species (M+3H)3+ and (M+4H)4+ were detected for each peptide. Calculated molecular masses confirmed the structural identity of synthetic GIP and each of the N-terminal analogues.
  • Degradation of GIP and novel GIP analogues by DPP-IV. FIGS. 6-11 illustrate the typical peak profiles obtained from the HPLC separation of the reaction products obtained from the incubation of GIP, GIP(Abu2), GIP(Sar2), GIP(Ser2), glycated GIP and acetylated GIP with DPP IV, for 0, 2, 4, 8 and 24 hours. The results summarized in Table 1 indicate that glycated GIP, acetylated GIP, GIP(Ser2) are GIP(Abu2) more resistant than native GIP to in vitro degradation with DPP IV. From these data GIP(Sar2) appears to be less resistant.
    TABLE 1
    Percent intact peptide remaining after incubation with DPPIV.
    % Intact peptide remaining after time (h)
    Peptide 0 2 4 8 24
    GIP 1-42 100 52 ± 1 23 ± 1 0 0
    Glycated GIP 100 100 100 100  100
    GIP (Abu2) 100 38 ± 1 28 ± 2 0 0
    GIP (Ser2) 100 77 ± 2 60 ± 1 32 ± 4 0
    GIP (Sar2) 100 28 ± 2  8 0 0
    N-Acetyl-GIP 100 100 100 100 
  • Table represents the percentage of intact peptide (i.e., GIP 1-42) relative to the major degradation product GIP 3-42. Values were taken from HPLC traces performed in triplicate and the mean and S.E.M. values calculated. DPA is diprotin A, a specific inhibitor of DPPIV.
  • Degradation of GIP and GIP analogues by human plasma. FIGS. 12-16 show a representative set of HPLC profiles obtained from the incubation of GIP and GIP analogues with human plasma for 0, 2, 4, 8 and 24 hours. Observations were also made after incubation for 24 hours in the presence of DPA. These results are summarized in Table 2 are broadly comparable with DPP IV incubations, but conditions which more closely mirror in vivo conditions are less enzymatically severe. GIP was rapidly degraded by plasma. In comparison, all analogues tested exhibited resistance to plasma degradation, including GIP(Sar2) which from DPP IV data appeared least resistant of the peptides tested. DPA substantially inhibited degradation of GIP and all analogues tested with complete abolition of degradation in the cases of GIP(Abu2), GIP(Ser2) and glycated GIP. This indicates that DPP IV is a key factor in the in vivo degradation of GIP.
    TABLE 2
    Percent intact peptide remaining after
    incubation with human plasma.
    % Intact peptide remaining after incubations
    with human plasma
    Peptide
    0 2 4 8 24 DPA
    GIP 1-42 100 52 ± 1 23 ± 1 0 0 68 ± 2
    Glycated GIP 100 100 100 100  100 100
    GIP (Abu2) 100 38 ± 1 28 ± 2 0 0 100
    GIP (Ser2) 100 77 ± 2 60 ± 1 32 ± 4 0 63 ± 3
    GIP (Sar2) 100 28 ± 2  8 0 0 100
  • Table represents the percentage of intact peptide (i.e., GIP 1-42) relative to the major degradation product GIP 3-42. Values were taken from HPLC traces performed in triplicate and the mean and S.E.M. values calculated. DPA is diprotin A, a specific inhibitor of DPPIV.
  • Dose-dependent effects of GIP and novel GIP analogues on insulin secretion. FIGS. 17-30 show the effects of a range of concentrations of GIP, GIP(Abu2), GIP(Sar2), GIP(Ser2), acetylated GIP, glycated GIP, GIP(Gly2) and GIP(Pro3) on insulin secretion from BRIN-BD11 cells at 5.6 and 16.7 mM glucose. Native GIP provoked a prominent and dose-related stimulation of insulin secretion. Consistent with previous studies [28], the glycated GIP analogue exhibited a considerably greater insulinotropic response compared with native peptide. N-terminal acetylated GIP exhibited a similar pattern and the GIP(Ser2) analogue also evoked a strong response. From these tests, GIP(Gly2) and GIP(Pro3) appeared to be the least potent analogues in terms of insulin release. Other stable analogues tested, namely GIP(Abu2) and GIP(Sar2), exhibited a complex pattern of responsiveness dependent on glucose concentration and dose employed. Thus very low concentrations were extremely potent under hyperglycemic conditions (16.7 mM glucose). This suggests that even these analogues may prove therapeutically useful in the treatment of type 2 diabetes where insulinotropic capacity combined with in vivo degradation dictates peptide potency.
  • Example 4 Glu3 Substituted GIP Improves Obesity-Related Insulin Resistance and Associated glucose intolerance
  • This example examines GIP receptor antagonism and obesity-related insulin resistance and associated glucose intolerance using a Glu3-substituted form of GIP, namely, (Pro3)GIP.
  • Cell lines and animals. In vitro insulin secretion was evaluated using the clonal pancreatic beta-cell line, BRIN-BD11 (McClenaghan, N. H. et al., 1996, Diabetes 45:1132-1140). In vitro cyclic AMP generation was measured using Chinese hamster lung (CHL) fibroblast cells stably transfected with the human GIP receptor (Gremlich, S. et al., 1995, Diabetes 44:1202-1208). In vivo studies were conducted in 8-12 week-old obese diabetic ob/ob mice (Bailey C. J. et al., 1982, Int. J. Obesity 6:11-21) and normal control mice.
  • Peptide synthesis and characterisation. Glu3-substituted analogues were sequentially synthesised on an Applied Biosystems automated peptide synthesiser (Model 432A) using standard solid-phase Fmoc peptide chemistry (Fields, G. B. et al., 1990, Int. J. Pept. Protein Res. 35:161-214), from a pre-loaded Fmoc-Gln-Wang resin. The synthetic peptides were judged pure by reversed-phase HPLC on a Waters Millenium 2010 chromatography system (Software version 2.1.5). The molecular masses of the purified peptide analogues were determined using Matrix Assisted Laser Desorption Ionisation-Time of Flight (MALDI-TOF) mass spectrometry. Samples were dissolved in 10 μl H2O (approximately 40 pmol/l), placed on a stainless steel sample plate and allowed to dry at room temperature. Samples were then mixed with a matrix solution (10 mg/ml solution of α-cyano-4-hydroxycinnamic acid) in acetonitrile/ethanol (1/1) and allowed to dry at room temperature. The molecular masses were then recorded as mass-to-charge (m/z) ratio versus relative peak intensity and compared using theoretical values on a Voyager-DE BioSpectrometry Workstation (PerSeptive Biosystems, Framingham, Mass., USA).
  • Tissue culture. Chinese hamster lung (CHL) fibroblast cells stably transfected with the human GIP receptor were cultured in DMEM tissue culture medium containing 10% (v/v) foetal bovine serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin). BRIN-BD11 cells were cultured using RPMI-1640 tissue culture medium containing 10% (v/v) foetal bovine serum, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin). Cells were maintained in sterile tissue culture flasks (Coming Glass Works, Sunderland, UK) at 37° C. in an atmosphere of 5% CO2 and 95% air using an LEEC incubator (Laboratory Technical Engineering, Nottingham, UK).
  • Acute studies of insulin release. Insulin release from BRIN-BD11 cells was determined using cell monolayers (McClenaghan, N. H. et al., 1996, Diabetes 45:1132-1140). Cells were harvested with the aid of trypsin/EDTA (Gibco), seeded into 24-multiwell plates (Nunc, Roskilde, Denmark) at a density of 1.0×105 cells per well, and allowed to attach overnight at 37° C. Prior to acute test, cells were preincubated for 40 minutes at 37° C. in 1.0 ml Krebs Ringer bicarbonate buffer (115 mM NaCl, 4.7 mM KCl, 1.28 mM CaCl2, 1.2 mM KH2PO4, 10 mM MgSO4, 10 mM NaHCO3, 0.5% (w/v) bovine serum albumin, pH 7.4) supplemented with 1.1 mM glucose. Acute tests for insulin release were performed for 20 minutes at 37° C. at 5.6 mM glucose using various concentrations of Glu3-substituted analogues and GIP(3-42) in the presence of native GIP (10−7 M) as indicated in the Figures. After incubation, aliquots of buffer were removed and stored at −20° C. for insulin radioimmunoassay (Flatt, P. R. et al., 1981, Diabetologia 20:573-577).
  • Acute studies of cyclic AMP generation. GIP receptor transfected CHL cells were seeded into 12-well plates (Nunc, Roskilde, Denmark) at a density of 1.0×105 cells per well. The cells were then allowed to grow for 48 hours before being loaded with tritiated adenine (2 μCi; TRK311, Amersham, Buckinghamshire, UK) and incubated at 37° C. for 6 hours in 1 ml DMEM, supplemented with 0.5% (w/v) foetal bovine serum. The cells were then washed twice with HBS buffer (130 mM NaCl, 20 mM HEPES, 0.9 mM NaHPO4, 0.8 mM MgSO4, 5.4 mM KCl, 1.8 mM CaCl2, 25 mM glucose, 25 μM phenol red, pH 7.4). The cells were then exposed for 10 minutes at 37° C. to forskolin (FSK, 10 μM) or varying concentrations of (Pro3)GIP in the absence (control) or presence of native GIP (10−7 M). After removal of the medium, cells were lysed with 1 ml of 5% trichloroacetic acid (TCA) containing 0.1 mM unlabelled cAMP and 0.1 mM unlabelled ATP. The intracellular tritiated cAMP was then separated on Dowex and alumina exchange resins as previously described (Widmann, C. et al., 1993, Mol. Pharmacol. 45:1029-1035).
  • Acute in vivo effects of (Pro3)GIP administration in obese diabetic ob/ob mice. Plasma glucose and insulin responses were evaluated using 8- to 12-week old obese diabetic ob/ob mice following intraperitoneal (i.p.) injection of native GIP, (Pro3)GIP (25 nmol/kg body weight) or saline (0.9% (w/v) NaCl; control) immediately following the combined injection of GIP (25 nmol/kg body weight) with glucose (18 mmol/kg body weight). All test solutions were administered in a final volume of 8 ml/kg body weight. Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 15, 30 and 60 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at −20° prior to glucose and insulin determinations.
  • Acute in vivo effects of (Pro3)GIP on plasma glucose and insulin responses to feeding in obese diabetic ob/ob mice. Plasma glucose and insulin responses were evaluated using 8- to 12-week old ob/ob mice where food was withdrawn for an 18-hour period prior to intraperitoneal injection of saline (0.9% (w/v) NaCl; control) or (Pro3)GIP (25 nmol/kg body weight). Following injection, the mice were allowed to re-feed for 15 minutes. Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 15, 30, 60 and 120 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at −20° prior to glucose and insulin determinations.
  • Effects of chronic (Pro3)GIP administration on plasma glucose, insulin and glycated HbA1c in obese diabetic ob/ob mice and normal mice. Obese diabetic ob/ob mice and normal control mice aged 8-12 weeks were randomly divided into groups which received once daily subcutaneous injections (17:00 h) of either saline (0.9% w/v NaCl) or (Pro3)GIP (25 nmol/kg body weight in saline). After 11 days, treatment was ceased. Food intake and body weight were recorded daily. Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin coated glucose microcentrifuge tubes (Sarstedt, Numbrecht, Germany). Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g prior to glucose, insulin and HbA1c determinations.
  • Effects of chronic treatment with (Pro3)GIP on glucose tolerance in ob/ob mice and normal mice. Plasma glucose and insulin concentrations were measured following intraperitoneal administration of glucose (18 mmol/kg body weight) in ob/ob and normal mice treated with either saline (0.9% w/v NaCl) or (Pro3)GIP (25 nmol/kg body weight/day) for 11 days. This test was repeated 9 days after cessation of chronic (Pro3)GIP treatment. Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 15, 30 and 60 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at −20° prior to glucose and insulin determinations.
  • Effects of chronic treatment with (Pro3)GIP on the glucose lowering effects of exogenous insulin in ob/ob mice. The glucose lowering effects of insulin were evaluated by measuring plasma glucose response in 11-day saline (0.9% w/v NaCl) and (Pro3)GIP (25 nmol/kg body weight/day) treated ob/ob mice following acute intraperitoneal administration of insulin (50 U/kg bodyweight). Blood samples were collected from the cut tip of the tail of conscious mice into chilled fluoride/heparin microcentrifuge tubes (Sarstedt, Numbrecht, Germany) immediately prior to injection and at 30 and 60 minutes post injection. Blood samples were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13000 g and stored at −20° prior to glucose determination.
  • Effects of chronic treatment with (Pro3)GIP on pancreatic insulin content and associated islet hypertrophy in ob/ob mice. Pancreatic tissue was excised from non-fasted ob/ob mice after 11 days treatment with either saline (0.9% w/v NaCl) or (Pro3)GIP (25 nmol/kg body weight/day). Pancreatic samples were individually wrapped in aluminium foil and snap frozen in liquid nitrogen. Individual excised pancreatic samples were then either embedded, sectioned and immunohistochemically stained for insulin or permeabilised for determination of pancreatic insulin content.
  • Determination of HbA1c, plasma glucose and insulin concentrations. HbA1c was measured in whole blood by ion-exchange high-performance liquid chromatography using the Menari HA-8140 kit (BIOMEN, Berkshire, UK). Plasma glucose was assayed by an automated glucose oxidase procedure using a Beckman Glucose Analyzer II (Stevens, J. F., 1971, Clinica Chemica Acta 32:199-201) and plasma insulin was determined by RIA (Flatt, P. R. et al., 1981, Diabetologia 20:573-577). Incremental areas under plasma glucose and insulin curves (AUC) were calculated using a computer generated program (CAREA) employing the trapezoidal rule (Burington, R. S., 1973, Handbook of Mathematical Tables and Formulae, New York, McGraw Hill) with baseline subtraction.
  • Statistical analysis. Results are expressed as means ±SEM. Values were compared using Student's unpaired t-test and groups of data were considered to be significantly different if P<0.05.
  • Results
  • GIP-stimulated cyclic AMP production and insulin secretion were inhibited in dose-dependent fashion by (Pro3)GIP, showing that (Pro3)GIP is a potent functional GIP receptor antagonist.
  • GIP receptor transfected Chinese hamster lung (CHL) fibroblasts were incubated with 10−12 to 10−6 M (Pro3)GIP in the presence of native GIP (10−7 M). The results are shown in FIGS. 32A and 32B. FIG. 32A is a line graph showing 3H-cAMP production as a percent of maximal response (y-axis) with increasing peptide concentration (M) (x-axis). FIG. 32B is a bar graph showing insulin secretion (y-axis) with increasing peptide concentration (M) (x-axis) for 5.6 mM glucose (control) (white bar), GIP (gray bars), (Pro3)GIP (lined bars) and (Pro3)GIP+GIP(107M) (black bars). *P<0.05, **P<0.01, ***P<0.001 compared to glucose control. ΔΔP<0.01, ΔΔΔP<0.001 compared with native GIP at the same concentration. Values are means ±SEM for 3-8 observations.
  • (Pro3)GIP inhibited GIP-induced cAMP formation with an IC50 value of 2.6 μM. Insulin-releasing activity of BRIN-BD11 cells exposed to native GIP and (Pro3)GIP (in the absence and presence of 10−7 M GIP).
  • GIP-stimulated insulin secretion was inhibited in a dose-dependent fashion by GIP(3-42), (Hyp3)GIP, (Lys3)GIP, (Tyr3)GIP, (Trp3)GIP, and (Phe3)GIP, as shown in FIGS. 33A through 33F, which are bar charts. FIG. 33A shows 3H-cAMP production as a percent of 10−7M GIP (y-axis) versus log10 of GIP (110-M) (white bar, control) and GIP (10−7M)+GIP(3-42) (black bars). FIGS. 33B through 33F show insulin secretion (in ng/106 cells/20 minutes) (y-axis) as a function of peptide concentration (M) (x-axis) for GIP (10−7M) (white bar, control) and a Glu3-substituted form of GIP (black bars), including (Hyp3)GIP (FIG. 33B), (Lys3)GIP (FIG. 33C), (Tyr3)GIP (FIG. 33D), (Trp3)GIP (FIG. 33E), and (Phe3)GIP (FIG. 33F). *P<0.05, **P<0.01 compared to GIP (10−7 M) control. Values are means ±SEM for 3-8 observations.
  • FIGS. 34A through 34D are a set of two line graphs (FIGS. 34A, 34C) and two bar graphs (FIGS. 34B, 34D) showing that acute administration of (Pro3)GIP completely antagonises the actions of GIP on glucose tolerance (FIGS. 34A, 34B) and plasma insulin (FIGS. 34C, 34D) responses in obese diabetic ob/ob mice. FIGS. 34A and 34C are line graphs show plasma glucose levels (FIG. 34A, y-axis) and plasma insulin levels (FIG. 34C, y-axis) over time (x-axis) for glucose (control; ▾), glucose+GIP (♦) and glucose+(GIP+Pro3GIP)) (Δ). FIGS. 34B and 34D are bar graphs showing plasma glucose AUC for glucose alone (white bars), GIP (grey bars) and glucose+(GIP+Pro3GIP)) (black bars).
  • Plasma glucose and insulin concentrations after i.p. administration of glucose alone (18 mmol/kg body weight) or in combination with either native GIP or native GIP plus (Pro3)GIP (25 nmol/kg body weight). The time of injection is indicated by the arrow (0 minutes). Plasma glucose and insulin AUC values are given for 0-60 minutes post-injection. Values are means ±SEM for 8 mice. *P<0.05, **P<0.01, ***P<0.001 compared with glucose alone. ΔΔP<0.01, ΔΔΔP<0.001 compared with native GIP.
  • Acute administration of (Pro3)GIP completely antagonised the insulin-releasing action of GIP and the associated improvement of glucose tolerance in ob/ob mice. Indeed, the glycemic excursion following (Pro3)GIP (Δ) was worse than when glucose was administered alone (▾).
  • FIGS. 35A through 35D show the effects of (Pro3)GIP on physiological meal-stimulated insulin release and glycemic excursion in obese diabetic ob/ob mice. Plasma glucose and insulin concentrations were measured in mice allowed to re-feed for 15 minutes prior to i.p. administration of saline (0.9% (w/v) NaCl) as control or (Pro3)GIP (25 nmol/kg body weight). The time of injection is indicated by the arrow (15 minutes).
  • The results are shown in FIGS. 35A through 35D, which are a set of two line graphs (FIGS. 35A, 35C) and two bar graphs (FIGS. 35B, 35D). The figures show plasma insulin (FIGS. 35A) and plasma glucose (FIG. 35C) over time for saline control (▾) and (Pro3)GIP (⋄), and plasma insulin AUC (FIG. 35B) and plasma glucose AUC (FIG. 35D) for saline control (white bars) and (Pro3)GIP (black bars), respectively. Values are means ±SEM for 8 mice. *P<0.05, **P<0.01, ***P<0.001 compared with saline alone.
  • Acute administration of (Pro3)GIP decreased the insulin response to feeding and worsened the associated glycemic excursion in ob/ob mice. These effects of functional ablation of endogenous GIP by the (Pro3)GIP antagonist are fully consistent with the accepted role of GIP in the regulation of insulin secretion and glycemic excursion following feeding.
  • The effects of chronic administration of (Pro3)GIP for 11 days on plasma glucose and insulin concentrations of obese diabetic ob/ob mice were also studied. According to classical thinking and the experiments described above and the results shown in FIGS. 32-35, functional ablation of endogenous GIP by daily administration of (Pro3)GIP over 11 days would be expected to inhibit insulin secretion and cause a marked deterioration in glucose tolerance.
  • However, the exact opposite occurred during chronic treatment with (Pro3)GIP in ob/ob mice. This is shown in FIG. 36, which is a set of two bar graphs showing plasma glucose (FIG. 36A) and insulin (FIG. 36B) concentrations after daily subcutaneous administration of saline alone (0.9% (w/v) NaCl; as control; white bars) or (Pro3)GIP (25 nmol/kg body weight; black bars) for 11 days. Values are means ±SEM for 6 mice and *P<0.05 compared with saline alone. Chronic administration of (Pro3)GIP (black bars) for 11 days decreases plasma glucose and plasma insulin concentrations of obese diabetic ob/ob mice, relative to controls.
  • The effects of chronic administration of (Pro3)GIP for 11 days on HbA1C, (FIG. 37A), pancreatic insulin content (FIG. 37B) and associated islet hypertrophy (FIG. 37C) were examined in obese diabetic ob/ob mice treated with saline (control, white bars) and (Pro3)GIP were examined. HbA1c, pancreatic insulin content and average islet diameter were measured after 11 daily subcutaneous injections of either saline alone (white bars) or (Pro3)GIP (25 nmol/kg body weight; black bars) to obese diabetic ob/ob mice. Values are means ±SEM for 6 mice and *P<0.05, ***P<0.001 compared with saline-treated group.
  • Beneficial effects of chronic (Pro3)GIP administration in ob/ob mice were associated with significant decreases in HbA1c and pancreatic insulin stores, with partial correction of the marked islet hypertrophy of the ob/ob mutant. There was also an approximate 7% decrease in body weight in (Pro3)GIP-treated ob/ob mice without any change in food intake. This effect did not achieve significance over the short study period, but this observation clearly suggests that GIP antagonism may also have a longer-term anti-obesity action.
  • The effects of chronic administration of (Pro3)GIP for 11 days on glucose tolerance and plasma insulin in obese diabetic ob/ob mice is shown in FIGS. 38A-38D, which are a set of line graphs (FIGS. 38A, 38C) and bar graphs (FIGS. 38B, 38C) showing plasma glucose levels (FIGS. 38A, 38B) and plasma insulin levels (FIGS. 38C, 38D) in obese diabetic ob/ob mice treated with saline (control, white) or (Pro3)GIP (black). Plasma glucose and insulin concentrations were measured prior to and at intervals after intraperitoneal administration of glucose (18 mmol/kg body weight). Arrow indicates time of injection (t=0). Values are means ±SEM for 6 mice and *P<0.05, **P<0.01, ***P<0.001 compared with saline-treated group.
  • After 11 days treatment with (Pro3)GIP, glucose tolerance of ob/ob mice was substantially improved without change of circulating insulin (FIG. 38).
  • FIG. 39 shows the effects of chronic administration of (Pro3)GIP for 11 days on insulin sensitivity in obese diabetic ob/ob mice. Plasma glucose concentrations of saline and (Pro3)GIP treated ob/ob mice were measured prior to and at intervals after intraperitoneal administration of exogenous insulin (50 U/kg body weight; t=0). Values are means ±SEM for 6 mice and *P<0.05 compared with saline-treated group. As shown in FIG. 39, chronic administration of (Pro3)GIP caused a significant improvement of insulin sensitivity.
  • Interestingly, the beneficial effects of chronic administration of (Pro3)GIP for 11 days in obese diabetic ob/ob mice was reversed 9 days after cessation of treatment. This is consistent with a physiological effect, and is shown in FIG. 40. Plasma glucose concentrations were measured prior to and after intraperitoneal administration of glucose (18 mmol/kg body weight) for mice that had been treated with saline (control, □) or (Pro3)GIP (▴). Arrow indicates time of injection (t=0). Values are means ±SEM for 6.
  • FIGS. 41A and 41B are a pair of line graphs showing the effects of chronic administration of (Pro3)GIP for 11 days on glucose tolerance in normal mice. Plasma glucose concentrations were measured prior to and after intraperitoneal administration of glucose (18 mmol/kg body weight). Arrow indicates time of injection (t=0). Values are means ±SEM for 6 and *P<0.05, **P<0.01 compared to saline-treated group.
  • In total contrast to beneficial actions in ob/ob mice, chronic daily treatment of normal mice with (Pro3)GIP (Δ) for 11 days resulted in a marked deterioration of glucose tolerance (FIG. 41A) relative to controls (▪), which was reversed 9 days after cessation of treatment (FIG. 41B).
  • Example 5 Chemical Ablation of Gastric Inhibitory Polypeptide Receptor Action by Daily (Pro3)GIP Administration Improves Glucose Tolerance and Ameliorates Insulin Resistance and Abnormalities of Islet Structure in Obesity-Diabetes
  • Gastric inhibitory polypeptide (GIP) is an important incretin hormone secreted by endocrine K-cells in response to nutrient ingestion. This study investigated the effects of chemical ablation of GIP receptor (GIP-R) action on aspects of obesity-diabetes using a stable and specific GIP-R antagonist, (Pro3)GIP. Young adult ob/ob mice received once daily i.p. injections of saline vehicle or (Pro3)GIP over an 11-day period. Non-fasting plasma glucose levels and the overall glycemic excursion (AUC) to a glucose load were significantly reduced (1.6-fold; P<0.05) in (Pro3)GIP-treated mice compared to controls. GIP-R ablation also significantly lowered overall plasma glucose (1.4-fold; P<0.05) and insulin (1.5-fold; P <0.05) responses to feeding. These changes were associated with significantly enhanced (1.6-fold; P<0.05) insulin sensitivity in the (Pro3)GIP-treated group. Daily injection of (Pro3)GIP reduced pancreatic insulin content (1.3-fold; P<0.05) and partially corrected the obesity-related islet hypertrophy and beta cell hyperplasia of ob/ob mice. These comprehensive beneficial effects of (Pro3)GIP were reversed following 9 days cessation of treatment and were independent of food intake and body weight, which were unchanged. These studies highlight a role for GIP in obesity-related glucose intolerance and emphasize the potential of specific GIP-R antagonists as a new class of drugs for the alleviation of insulin resistance and treatment of type 2 diabetes.
  • Research Design and Methods
  • Animals. Obese diabetic (ob/ob) mice derived from the colony maintained at Aston University, UK (Bailey, C. J., et al., 1982, Int. J. Obes. 6:11-21) were used at 12-16 weeks of age. Animals were age-matched, divided into groups and housed individually in an air-conditioned room at 22±2° C. with a 12 hour light:12 hour dark cycle. Drinking water and a standard rodent maintenance diet (Trouw Nutrition, Cheshire, UK) were freely available. All animal experiments were carried out in accordance with the UK Animals (Scientific Procedures) Act 1986. No adverse effects were observed following administration of (Pro3)GIP.
  • Synthesis, purification and characterization of (Pro3)GIP. (Pro3)GIP was sequentially synthesized on an Applied Biosystems automated peptide synthesizer (Model 432 A). (Pro3)GIP was purified by reversed-phase HPLC on a Waters Millenium 2010 chromatography system (Software version 2.1.5) and subsequently characterized using electrospray ionization mass spectrometry (ESI-MS).
  • Experimental protocols for ob/ob mouse studies. Initially, extended biological activity of (Pro3)GIP was examined in 18-hour fasted ob/ob mice 4 hours after administration. Thereafter, over an 11-day period, mice received once daily i.p. injections (17:00 hours) of either saline vehicle (0.9% (w/v), NaCl) or (Pro3)GIP (25 nmol/kg body wt). During a subsequent 9-day period, observations were continued following discontinuation of (Pro3)GIP administration. Food intake and body weight were recorded daily whilst plasma glucose and insulin concentrations were monitored at intervals of 2-6 days. Whole blood for the measurement of glycated hemoglobin was taken on days 11 and 20. Intraperitoneal glucose tolerance (18 mmol/kg body wt), metabolic response to native GIP (25 nmol/kg body wt) and insulin sensitivity (50 U/kg body wt) tests were performed on days 11 and 20. Mice fasted for 18 hours were used to examine the metabolic response to 15 minutes feeding. In a separate series, pancreatic tissues were excised at the end of the 11-day treatment period or 9 days following discontinuation of (Pro3)GIP and processed for immunohistochemistry or measurement of insulin following extraction with 5 ml/g of ice-cold acid ethanol (750 ml ethanol, 235 ml water, 15 ml concentrated HCl). Blood samples taken from the cut tip of the tail vein of conscious mice at the times indicated in the Figures were immediately centrifuged using a Beckman microcentrifuge (Beckman Instruments, UK) for 30 seconds at 13,000 g. The resulting plasma was then aliquoted into fresh Eppendorf tubes and stored at −20° C. prior to glucose and insulin determinations.
  • Biochemical analysis. Plasma glucose was assayed by an automated glucose oxidase procedure (Stevens, J. F., 1971, Clin. Chem. Acta 32:199-201) using a Beckman Glucose Analyzer II (Beckman Instruments, Galway, Ireland). Plasma and pancreatic insulin were assayed by a modified dextran-coated charcoal radioimmunoassay (Flatt, P. R. et al., 1981, Diabetologia 20:573-577). Glycated hemoglobin was determined using cation-exchange columns (Sigma, Poole, Dorset, UK) with measurement of absorbance (415 nm) in wash and eluting buffer using a VersaMax Microplate Spectrophotometer (Molecular Devices, Wokingham, Berkshire, UK).
  • Immunocytochemistry. Tissue fixed in 4% paraformaldehyde/PBS and embedded in paraffin was sectioned at 8 μm. After de-waxing, sections were incubated with blocking serum (Vector Laboratories, CA, USA) prior to exposure to insulin antibody. Tissue samples were then incubated consecutively with secondary biotinylated universal, pan-specific antibody (Vector Laboratories, CA, USA) and streptavidin/peroxidase preformed complex (Vector Laboratories, CA, USA). Following washing, the stained pancreatic tissue was counterstained with hematoxylin (BDH Chemicals, Dorset, UK) and then plunged into acid methanol (500 ml methanol, 500 ml H2O and 2.5 ml concentrated HCl) prior to dehydration and mounting in Depex (BDH Chemicals, Dorset, UK). The stained slides were viewed under a microscope (Nikon Eclipse E2000, Diagnostic Instruments Incorporated, Michigan, USA) attached to a JVC camera Model KY-F55B (JVC, London, UK) and analyzed using Kromoscan imaging software (Kinetic Imaging Limited, Faversham, Kent, UK). The average number and diameter of every islet in each section was estimated in a blinded manner using an eyepiece graticule calibrated with a stage micrometer (Graticules Limited, Tonbridge, Kent, UK). The longest and shortest diameters of each islet were determined with the graticule. Half of the sum of these two values was then considered to be the average islet diameter. Approximately 60-70 random sections were examined from the pancreas of each mouse.
  • Statistics. Results are expressed as mean ±SEM. Data were compared using ANOVA, followed by a Student-Newman-Keuls post hoc test. Area under the curve (AUC) analyzes were calculated using the trapezoidal rule with baseline subtraction (Burington, R. S., Handbook of Mathematical Tables and Formulae, New York, McGraw-Hill, 1973). P <0.05 was considered to be statistically significant.
  • Results
  • Effects of (Pro3)GIP on plasma glucose and insulin concentrations 4 hours after administration were examined. The results are shown in FIGS. 42A through 42D, which are a set of two line graphs (FIGS. 42A, 42C) and two bar graphs (FIGS. 42B, 42D) showing the effects of (Pro3)GIP on plasma glucose and insulin response to native GIP 4 hours after administration. Tests were conducted 4 hours after administration of (Pro3)GIP (25 nmoles/kg body weight) or saline (0.9% NaCl) in 18 hour-fasted ob/ob mice. Plasma glucose and insulin concentrations were measured prior to and after i.p. administration of glucose (18 mmoles/kg body weight) in combination with native GIP (25 nmoles/kg body weight). The incremental area under the glucose or insulin curves (AUC) between 0 and 60 min are shown in the right panels. Values represent means ±SEM for 8 mice. *P<0.05 and **P<0.01 compared with saline alone group.
  • As shown in FIGS. 42A through 42D, administration of (Pro3)GIP for 4 hours previously impaired the plasma glucose and insulin responses to native GIP, given together with glucose. AUC glucose and insulin values were increased by 151% (P<0.05) and decreased by 25% (P<0.05); respectively, compared with saline-treated controls. This supports a protracted biological half-life and forms the basis of the once-daily injection.
  • The effects of (Pro3)GIP on food intake, body weight and non-fasting plasma glucose and insulin concentrations were studied. The results are shown in FIGS. 43A through 43D, which are a set of two line graphs and two bar graphs showing the effects of daily (Pro3)GIP administration on food intake (FIG. 43A), body weight (FIG. 43B), plasma glucose (FIG. 43C) and insulin (FIG. 43D) concentrations in ob/ob mice. Parameters were measured for 5 days prior to, 11 days during (indicated by black bar) and 9 days after treatment with saline or (Pro3)GIP (25 nmol/kg bw/day). Values are mean ±SEM for eight mice. *P<0.05 compared with saline group.
  • Administration of (Pro3)GIP had no effect on food intake and body weight (FIGS. 43A and 43B). On day 11, plasma glucose had declined to significantly reduced (P<0.05) concentrations in ob/ob mice receiving (Pro3)GIP (FIG. 43C). Cessation of treatment returned plasma glucose concentrations towards control levels. Consistent with this pattern, glycated hemoglobin was significantly lower (P<0.05) after 11 days treatment with (Pro3)GIP than either before or 9 days following cessation of daily injection (8.0±0.3%, 6.9±0.2%, 7.7±0.4%, respectively). No significant changes in plasma insulin levels were noted during or after the treatment period. However, there was a general trend for plasma insulin concentrations to decrease progressively during (Pro3)GIP treatment (FIG. 43D).
  • The effects of (Pro3)GIP on glucose tolerance are shown in FIGS. 44A through 44D, which are a set of four line graphs with inset bar graphs showing the effects of daily (Pro3)GIP administration on glucose tolerance and plasma insulin response to glucose in ob/ob mice. Tests were conducted after daily treatment with (Pro3)GIP (25 nmoles/kg body weight/day; ▴; black bars) or saline (control; □; white bars) for 11 days (FIG. 44A, 44C) or 9 days after cessation of treatment (FIG. 44B, 44B). Glucose (18 mmoles/kg body weight) was administered at the time indicated by the arrow. Plasma glucose (FIG. 44A, 44B) and insulin (FIG. 44C, 44D) AUC values for 0-60 minutes post injection, with identical baseline subtractions in each case to demonstrate the complete effect of (Pro3)GIP treatment, are shown in insets. Values are mean ±SEM for eight mice. *P<0.05, **P<0.01 and ***P<0.001 compared with saline group.
  • Daily administration of (Pro3)GIP for 11 days resulted in significantly reduced (P <0.001) plasma glucose concentrations at 15, 30 and 60 minutes following intraperitoneal glucose (FIG. 44A). This was corroborated by a significantly decreased 0-60 minutes AUC value (FIG. 44A) which was 2.1-fold reduced (P<0.01) compared to controls. Plasma insulin concentrations were also significantly (P<0.05) reduced 15, 30 and 60 minutes following intraperitoneal glucose injection in the (Pro3)GIP treated group (FIG. 44A). AUC, 0-60 minutes values were also significantly decreased (P<0.001). Interestingly, an almost identical pattern was observed when 11 day treated ob/ob mice were administered glucose together with native GIP (25 nmoles/kg body weight) (data not shown). This supports the view that GIP action was effectively antagonized in the (Pro3)GIP treated group. Discontinuation of (Pro3)GIP treatment for 9 days (day 20 of study) resulted in almost identical plasma glucose and insulin responses to intraperitoneal glucose (FIG. 44), with lower glucose-mediated plasma insulin concentrations noted at one time point (15 minutes; P<0.05).
  • The effects of (Pro3)GIP on metabolic response to feeding and insulin sensitivity are shown in FIGS. 45 and 46. FIGS. 45A through 45D are a set of two line graphs (FIGS. 45A, 45C) and two bar graphs (FIGS. 45B, 45D) showing the effects of daily (Pro3)GIP administration (▴; black bars) or saline (□; white bars) on glucose (FIGS. 45A, 45B) and insulin (FIGS. 45C, 45D) responses to feeding in ob/ob mice fasted for 18 hours. Tests were conducted after daily treatment with (Pro3)GIP (25 nmol/kg body weight/day) or saline for 11 days. The arrow indicates the time of feeding (15 minutes). AUC values for 0-105 minutes post-feeding are also shown. Values are mean ±SEM for eight mice. *P<0.05 compared with saline group.
  • FIGS. 46A through 46D are a set of two line graphs (FIGS. 46A, 46C) and two bar graphs (FIGS. 46B, 46D) showing the effects of daily (Pro3)GIP administration on insulin sensitivity in ob/ob mice. Tests were conducted after daily treatment with (Pro3)GIP (25 nmol/kg body weight/day; ▾; black bars) or saline (o; white bars) for 111 days (FIG. 46A, 46B) or 9 days after cessation of treatment (FIG. 46C, 46D). Insulin (50 U/kg body weight) was administered by intraperitoneal injection at the time indicated by the arrow. AUC values for 0-60 minutes post-injection are also shown. Values are mean ±SEM for eight mice. *P<0.05 compared with saline group.
  • Plasma glucose and insulin responses to 15 minutes feeding were significantly lowered (P<0.05) at 30 and 60 minutes in ob/ob mice treated with (Pro3)GIP for 11 days (FIG. 45). Similarly, AUC glucose and insulin were significantly (P<0.05) decreased in (Pro3)GIP treated ob/ob mice, despite similar food intakes of 0.3-0.5 g/mouse/15 minutes. As shown in FIGS. 46A and 45B, the hypoglycemic action of insulin was significantly (P<0.05) augmented in terms of AUC measures and post injection values in ob/ob mice treated with (Pro3)GIP for 11 days. The responses following 9 days discontinuation of (Pro3)GIP treatment were similar to saline treated controls (FIG. 45C, 45D).
  • The effects of (Pro3)GIP on pancreatic insulin and islet morphology are shown in FIGS. 47A through 47D, and 48A through 48F. FIGS. 47A through 47D are a set of four bar graphs showing the effects of daily (Pro3)GIP administration on pancreatic weight (FIG. 47A), insulin content (FIG. 47B), islet number (FIG. 47C) and islet diameter (FIG. 47D) in ob/ob mice. Parameters were measured after daily treatment with (Pro3)GIP (25 nmol/kg body weight/day; black bars) or saline (white bars) for 11 days and 9 days after cessation of treatment (day 20). Values are mean ±SEM for eight mice. *P<0.05 and ***P<0.001 compared with saline group. FIGS. 48A through 48F are a set of two bar graphs (FIGS. 48A, 48D) and four photomicrographs (FIGS. 48B, 48C, 48E, 48F), showing the effects of daily (Pro3)GIP administration on islet size and morphology in ob/ob)mice.
  • (Pro3)GIP treatment had no effect on pancreatic weight (FIG. 47A). However, pancreatic insulin content was significantly (P<0.05) decreased in ob/ob mice receiving (Pro3)GIP for 11 days compared to controls (FIG. 47B). No significant differences were observed in islet number per pancreatic section (FIG. 47C), but average islet diameter was markedly and significantly reduced (P<0.001) in (Pro3)GIP treated ob/ob mice (FIG. 47D). These effects were effectively reversed by discontinuation of (Pro3)GIP on day 20, however average islet diameter was still significantly reduced (P<0.05). As shown in FIG. 48A, more detailed analysis revealed that the reduction is islet diameter on day 11 was due to a significant decrease (P<0.001) in the percentage of larger diameter (>0.15 mm) islets with increases in the proportion of islets with small (<0.10 mm) and medium (0.1-0.15 mm) diameters. FIG. 48D presents similar analysis following cessation of treatment, with a significant (P<0.05) increase in the percentage of small islets still apparent. Representative images (×40 magnification) of pancreata immunohistologically stained for insulin from 11-day (Pro3)GIP treated ob/ob mice (FIG. 48B) and saline treated controls (FIG. 48C) illustrate the dramatic changes in pancreatic islet morphology induced by (Pro3)GIP treatment. Pancreata immunohistologically stained for insulin on day 20 are also shown (FIG. 48E, 48F).
  • Parameters were measured after daily treatment with (Pro3)GIP (25 nmol/kg body weight/day) or saline for 11 days (FIG. 48A) and 9 days after cessation of treatment (FIG. 48D). Proportion of islets classified as large (>0.15 mm) diameter, medium (0.1-0.15 mm) diameter and small (<0.1 mm) diameter are shown. Values are mean ±SEM for eight mice FIGS. 48B, 48C, 48E and 48F are representative images (×40 magnification) of pancreata stained for insulin following 11 days treatment with (Pro3)GIP (FIG. 48B) or saline (FIG. 48C). Corresponding images 9 days after cessation of treatment with (Pro3)GIP (FIG. 48E) or saline (FIG. 48F) are also shown. The arrows indicate islets.
  • Example 6 N-Terminally Acetylated and Lys16 and Lys37-Substituted GIP
  • This example examines the metabolic stability, biological activity and antidiabetic potential of fatty acid derivatized N-terminally modified GIP analogues. These are N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37), which have an N-terminal Tyr1 acetyl group, and a C-16 palmitate group linked to the epsilon-amino group of the lysine at either position 16 or position 37 of the GIP protein.
  • Materials and Methods
  • Animals. Obese diabetic (ob/ob) mice derived from the colony maintained at Aston University, UK were used at 12-17 weeks of age. The genetic background and characteristics of the colony used have been outlined in detail elsewhere (Bailey, C. J. et al., 1982, Int. J. Obesity 6:11-21; Gault, V. A. et al., 2003, J. Endocrinol. 176: 133-141). Animals were housed in an air-conditioned room at 22±2° C. with a 12 hours light:12 hours dark cycle. Drinking water and standard rodent maintenance diet (Trouw Nutrition, Cheshire, UK) were freely available. All test solutions were administered by i.p. injection in a final volume of 5 ml/kg bw. Blood was collected from the cut tip of the tail vein of conscious mice into chilled fluoride/heparin microcentrifuge tubes immediately prior to injection and at the times indicated in the Figures. Plasma was separated using a Beckman microcentrifuge (Beckman Instruments, UK) at 13,000 g for 30 second and stored at −20° C. prior to glucose and insulin determinations. All animal experiments were carried out in accordance with the UK Animals (Scientific Procedures) Act 1986. No adverse effects were observed following acute or long-term administration of any of the peptides.
  • Materials. High performance liquid chromatography (HPLC) grade acetonitrile was obtained from Rathburn (Walkersbum, UK). Trifluoroacetic acid (TFA) and trichloroacetic acid (TCA) were obtained from Aldrich (Poole, Dorset, UK). DPP IV, isobutylmethylxanthine (IBMX), alpha-cyano-4-hydroxycinnamic acid, cyclic AMP and ATP were all purchased from Sigma (Poole, Dorset, UK). Fmoc-protected amino acids were from Calbiochem Novabiochem (Nottingham, UK). RPMI-1640 and DMEM tissue culture medium, foetal bovine serum, penicillin and streptomycin were all purchased from Gibco (Paisley, Strathclyde, UK). The chromatography columns used for cyclic AMP assay, Dowex AG50 WX and neutral alumina AG7 were obtained from Bio-Rad (Life Science Research, Alpha Analytical, Larne, UK). All water used in these experiments was purified using a Milli-Q Water Purification System (Millipore, Milford, Mass., USA). All other chemicals used were of the highest purity available.
  • Synthesis, purification and characterisation of GIP and related analogues. Native GIP was sequentially synthesised using standard solid-phase Fmoc peptide chemistry (ABI 432A Peptide Synthesiser) as described previously (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) were synthesised in the same way as native GIP but with the exception that the epsilon-amino groups of Lys at positions 16 or 37 were conjugated with a C-16 palmitate fatty acid. In addition, an acetyl adduct was incorporated at the N-terminal Tyr1. The synthetic peptides were judged pure by reversed-phase HPLC on a Waters Millenium 2010 chromatography system (Software version 2.1.5) and subsequently characterised using matrix assisted laser desorption ionisation-time of flight mass spectrometry (MALDI-ToF MS) as described previously (Gault, V. A. et al., 2002, Cell. Biol. Int. 27: 41-46).
  • DPP IV degradation studies. GIP and fatty acid derivatised GIP analogues were incubated at 37° C. with purified porcine dipeptidylpeptidase IV (5 mU in 50 mmol/l triethanolamine-HCl; pH 7.8) for 0, 2, 4, 8 and 24 hours (final peptide concentration 2 mmol/l). The reactions were subsequently terminated by addition of 10% (v/v) TFA/water and the reaction products separated using HPLC. Reaction products were applied to a Vydac C-4 column (4.6×250 mm; The Separations Group, Hesparia, Calif.) and the major degradation product GIP(3-42) separated from intact GIP. The column was equilibrated with 0.12% (v/v) TFA/water at a flow rate of 1.0 ml/minute using 0.1% (v/v) TFA in 70% acetonitrile/water with the concentration of acetonitrile in the eluting solvent being raised from 0% to 40% over 10 minutes, and then from 40% to 75% over 35 minutes. The absorbance was monitored at 206 nm using a SpectraSystem UV 2000 Detector (Thermoquest Limited, Manchester, UK) and the peaks collected manually prior to MALDI-ToF MS analysis. HPLC peak area data were used to calculate % intact peptide remaining throughout the incubation.
  • Cells and cell culture. Chinese hamster lung (CHL) fibroblasts stably transfected with the human GIP receptor (Gremlich, S. et al., 1995, Diabetes 44: 1202-1208) were cultured in DMEM tissue culture medium containing 10% (v/v) FBS, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin). Clonal pancreatic BRIN-BD11 cells (McClenaghan, N. H. et al., 1996, Diabetes 45: 1132-1140) were cultured using RPMI-1640 culture medium containing 10% (v/v) FBS, 1% (v/v) antibiotics (100 U/ml penicillin, 0.1 mg/ml streptomycin) and 11.1 mmol/l glucose. Cells were maintained at 37° C. in an atmosphere of 5% CO2 and 95% air using an LEEC incubator (Laboratory Technical Engineering, Nottingham, UK).
  • In vitro biological activity. Intracellular cyclic AMP production was measured using GIP-receptor transfected CHL fibroblasts (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). In brief, CHL cells were seeded into 12-well plates (Nünc, Roskilde, Denmark) at a density of 105 cells per well and allowed to grow for 48 hours before being loaded with tritiated adenine (2 μCi; TRK311; Amersham, Buckinghamshire, UK). The cells were then incubated at 37° C. for 6 hours in 1 ml DMEM supplemented with 0.5% (w/v) BSA and subsequently washed twice with HBS buffer (pH 7.4). Cells were then exposed to GIP/GIP analogues (10−13 to 10−6 mol/l) in HBS buffer in the presence of 1 mmol/l IBMX for 15 minutes at 37° C. The medium was subsequently removed and the cells lysed with 1 ml of 5% TCA containing 0.1 mmol/l unlabelled cyclic AMP and 0.1 mmol/l unlabelled ATP. The intracellular cyclic AMP was then separated on Dowex and alumina exchange resins as described previously (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291).
  • Insulin-release studies were carried out using clonal pancreatic BRIN-BD11 cells as described previously (O'Harte, F. P. M. et al., 2002, Diabetologia 45: 1281-1291). Briefly, BRIN-BD11 cells were seeded into 24-well plates at a density of 105 cells per well, and allowed to attach overnight at 37° C. Acute tests for insulin release were preceded by 40 minutes pre-incubation at 37° C. in 1.0 ml Krebs Ringer bicarbonate buffer supplemented with 1.1 mmol/l glucose. Test incubations were performed in the presence of 5.6 mmol/l glucose with a range of concentrations (10−13 to 10−6 mol/l) of GIP and GIP analogues. After 20 minutes incubation, the buffer was removed from each well and aliquots (200 μl) used for measurement of insulin.
  • Effects of N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) in ob/ob mice. Metabolic and dose-response effects of GIP and N-AcGIP(LysPAL) analogues (at 6.25-25 nmoles/kg bw) following glucose administration (18 mmoles/kg bw) were examined in mice fasted for 18 hours. To evaluate long-term effects, groups of ob/ob mice received once daily intraperitoneal injections (17:00 h) for 14 days of either saline vehicle (0.9%, w/v, NaCl), native GIP or N-AcGIP(LysPAL37) (both at 12.5 nmoles/kg body weight/day). Food intake and body weight were recorded daily. Plasma glucose and insulin concentrations were monitored at 2-6 day intervals. At 14 days, groups of animals were used to evaluate intraperitoneal glucose tolerance (18 mmoles/kg) and insulin sensitivity (50 U/kg). In a separate series, two experimental protocols were employed to examine the possibility of GIP receptor desensitization after 14 days treatment. Acute metabolic effects of the usual injection of either saline, GIP or N-AcGIP(LysPAL37) were monitored when administered together with glucose (18 mmoles/kg). In the second, acute effects of N-AcGIP(LysPAL37) given together with glucose were examined in all 3 groups of mice. At the end of the 14-day treatment period, pancreatic tissues were excised for measurement of insulin following extraction with 5 ml/g ice-cold acid ethanol (75% ethanol, 2.35% H20, 1.5% HCl). Whole blood was taken for determination of glycated hemoglobin.
  • Biochemical analyses. Plasma glucose was assayed by an automated glucose oxidase procedure (Stevens, J. F., 1971, Clin. Chem. Acta 32:199-201) using a Beckman Glucose Analyser II (Beckman, Galway, Ireland). Plasma insulin was determined by dextran-charcoal RIA as described previously (Flatt, P. R. et al., 1981, Diabetologia 20: 573-577). Glycated hemoglobin was determined using cation-exchange columns (Sigma, Poole, Dorset, UK) with measurement of absorbance (415 nm) in wash and eluting buffers using a VersaMax microplate spectrophotometer (Molecular Devices, Wokingham, Berkshire, UK).
  • Statistics. Results are expressed as mean ±SEM. Data were compared using the unpaired Student's t-test. Where appropriate, data were compared using repeated measures ANOVA or one-way ANOVA, followed by the Student-Newman-Keuls post hoc test. Incremental areas under plasma glucose and insulin curves (AUC) were calculated using a computer-generated program employing the trapezoidal rule (Burington, R. S., 1973, Handbook of Mathematical Tables and Formulae, McGraw-Hill, New York) with baseline subtraction. Groups of data were considered to be significantly different if p<0.05.
  • Results
  • Structural characterisation by MALDI-ToF MS. Following synthesis and HPLC purification, the molecular masses were obtained for GIP, N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) using MALDI-ToF MS (Table 3, below). The mass-to-charge (m/z) ratio for native GIP was calculated to be 4983.7 Da, corresponding very closely to the theoretical mass of 4982.4 Da. Similarly, the m/z ratios for N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) were 5268.9 Da and 5267.7 Da, respectively. These values correlate very closely to the theoretical mass (5266.1 Da), therefore, confirming the correct structures for each of the synthetic peptides.
    TABLE 3
    Structural characterisation of GIP
    and GIP analogues by MALDI-ToF MS.
    Experimental Theoretical Difference
    Peptide Mr (Da) Mr (Da) (Da)
    GIP 4983.7 4982.4 1.3
    N-AcGIP(LysPAL16) 5268.9 5266.1 2.8
    N-AcGIP(LysPAL37) 5267.7 5266.1 1.6
  • Peptide samples were mixed with matrix (alpha-cyano-4-hydroxycinnamic acid) and m/z ratio vs. relative peak intensity recorded using a Voyager-DE BioSpectrometry Workstation.
  • Degradation by DPP IV. Table 4, below, illustrates the % intact peptide remaining after incubation with DPP IV. Degradation of native GIP was evident after just 2 hours with only 52±3% of the peptide remaining intact. After 8 hours incubation the native peptide was entirely degraded to GIP(3-42). In contrast, both N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) remained completely intact (no degradation fragment evident) even after 24 hours incubation with DPP IV.
    TABLE 4
    Percentage intact peptide remaining after incubation with DPP IV.
    % Intact peptide remaining after time (hours)
    Peptide 0 2 8 24
    Native GIP 100 52 ± 3 0 0
    N-AcGIP(LysPALl16) 100 100 100 100
    N-AcGIP(LysPAL37) 100 100 100 100
  • Values represent the % intact peptide remaining relative to the major degradation product GIP(3-42) following incubation with DPP IV as determined from HPLC peak area data. The reactions were performed in triplicate and the means ±SEM values calculated.
  • Changes in Cyclic AMP production. FIG. 50A shows intracellular cyclic AMP production by GIP (▴) and fatty acid derivatised GIP analogues N-AcGIP(LysPAL16) (□) and N-AcGIP(LysPAL37) (●), as determined by column chromatography, in CHL cells stably expressing the human GIP receptor. Each experiment was performed in triplicate (n=3) and the results expressed (means ±SEM) as a percentage of the maximum GIP response.
  • A concentration-dependent (10−13 to 10−6 mol/l) increase in cyclic AMP production was observed with native GIP (EC50 value 18.2 nmol/l) using CHL cells transfected with the human GIP receptor (FIG. 50A). Likewise, both N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) followed a similar pattern of stimulation to that of native GIP with calculated EC50 values of 12.1 and 13.0 nmol/l, respectively. The lower EC50 values for both analogues suggest an enhanced cyclic AMP-stimulating potency.
  • In vitro insulin-releasing activity. FIG. 50B shows insulin-releasing activity of glucose (5.6 mmo/l glucose; white bars), GIP (lined bars) and fatty acid derivatised GIP analogues N-AcGIP(LysPAL16) (grey bars) and N-AcGIP(LysPAL37) (black bars) in the clonal pancreatic beta cell line, BRIN-BD11. After a pre-incubation (40 minutes), the effects of various concentrations of peptide were tested on insulin-release during a 20 minutes incubation. Values are means ±SEM for 8 separate observations. *p<0.05, **p<0.01, ***p<0.001 compared to control (5.6 mmol/l glucose alone).
  • Consistent with its role as a potent insulinotropic hormone, native GIP dose-dependently stimulated insulin secretion (p<0.01 to p<0.001) compared to control (5.6 mmol/l glucose alone) (FIG. 50B). Likewise, both N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) significantly stimulated glucose-induced insulin secretion (p<0.05 to p<0.001). On the basis of cyclic AMP and insulin secretory data, both GIP analogues appear to be at least equipotent to the native peptide.
  • Metabolic effects in ob/ob mice. FIGS. 51A through 51D are a set of two line graphs (FIGS. 51A, 51C) and two bar graphs (FIGS. 51B, 51D) showing glucose lowering effects (FIGS. 51A, 51B) and insulin-releasing activity (FIGS. 51C, 51D) of GIP and fatty acid derivatised GIP analogues in 18 hour-fasted ob/ob mice. Plasma glucose and insulin concentrations were measured prior to and after i.p. administration of glucose alone (18 mmoles/kg bw; ◯; white bars) as a control, or in combination with GIP (▴; lined bars) or GIP analogues N-AcGIP(LysPAL16) (□; grey bars) and N-AcGIP(LysPAL37) (●; black bars) (25 nmoles/kg bw). The incremental area under the glucose or insulin curves (AUC) between 0 and 60 minutes are shown in the right panels. Values represent means ±SEM for 8 mice. *p<0.05, **p<0.01, ***p<0.001 compared to glucose alone, Δp<0.05, ΔΔp<0.01 and ΔΔΔp<0.001 compared to native GIP, γγγp<0.001 compared with N-AcGIP(LysPAL16).
  • Basal blood glucose levels of the experimental groups were not significantly different at the start of the study (p>0.05). After injection of glucose alone, plasma glucose levels increased rapidly, attaining values of 40.3±1.5 mmol/l at 60 min. Native GIP reduced plasma glucose at each of the time points monitored, however, this failed to reach significance in terms of overall glucose excursion as revealed by the AUC values (FIG. 52B). Administration of N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) produced a significant reduction in plasma glucose at each time point (p<0.01 to p<0.001) and significantly lowered glucose AUC (p<0.001 to p<0.001) when compared to glucose alone. Additionally, N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) decreased the overall glucose excursion (p<0.05 to p<0.001) when compared to native GIP.
  • The corresponding plasma insulin responses are illustrated in FIGS. 51C and 51D. After administration of glucose alone (control) the maximal rise in plasma insulin was observed at 15 minutes, which then fell towards basal levels over the remaining 45 minutes. Administration of native GIP significantly elevated the overall insulinotropic response (p<0.05) compared with glucose alone. When N-AcGIP(LysPAL16) or N-AcGIP(LysPAL37) where administered together with glucose, a maximum plasma insulin concentration was observed at 15 minutes. Protracted biological activity for both analogues was clearly evident from 30 to 60 minutes. Glucose-mediated plasma insulin concentrations were significantly higher compared in both control (p<0.01 to p<0.001) and GIP-treated animals (p<0.05 to p<0.001). The corresponding AUC values for N-AcGIP(LysPAL16) and N-AcGIP(LysPAL37) revealed significant enhancements in overall glucose-mediated insulin release compared to native GIP (1.5-fold and 2.3-fold, respectively; p<0.01 to p<0.001). N-AcGIP(LysPAL37) was significantly more potent (1.5-fold: p<0.001) than N-AcGIP(LysPAL16) at stimulating insulin secretion.
  • Dose-dependent metabolic effects in ob/ob mice. FIGS. 52A and 52B illustrate the dose-dependent antihyperglycaemic and insulinotropic effects of GIP and the more potent analogue N-AcGIP(LysPAL37) when administered with glucose to ob/ob mice. They are a pair of bar graphs showing dose-dependent effects of GIP and N-AcGIP(LysPAL37) in ob/ob mice fasted for 18 hours. The incremental area under the curve (AUC) for glucose (FIG. 52A) and insulin (FIG. 52B) between 0 and 60 minutes after i.p. administration of glucose alone (18 mmoles/kg bw; white bars) or in combination with GIP (lined bars) or N-AcGIP(LysPAL37) (each at 6.25, 12.5 and 25 nmoles/kg bw; black bars). Values represent means ±SEM for 8 mice. **p<0.01 and ***p<0.001 compared to glucose alone. ΔΔp<0.01 and ΔΔΔp<0.001 compared to native GIP at the same dose.
  • Data are presented as overall AUC responses for convenience. Expressed in this manner, native GIP did not significantly affect AUC glucose and insulin at any of the doses tested. N-AcGIP(LysPAL37) was substantially more potent than native GIP (p<0.01 to p<0.001) and exhibited prominent dose-dependent antihyperglycaemic and insulinotropic actions at all doses administered (FIGS. 52A, 52B). Remarkably, even the lowest concentration of N-AcGIP(LysPAL37) tested (6.25 nmoles/kg) had highly significant antihyperglycaemic properties compared to glucose alone (p<0.001). Consistent with this observation, 6.25 nmoles/kg N-AcGIP(LysPAL37) elicited a prominent insulin response (2.0-fold; p<0.01) compared to glucose alone.
  • Long-acting effects in ob/ob mice. The effects of daily injection of N-AcGIP(LysPAL37) for 14 days on food intake, body weight, glycated hemoglobin and non-fasting plasma glucose and insulin concentrations of ob/ob mice are shown in FIGS. 53A through 53E, which are a set of graphs showing the effects of daily N-AcGIP(LysPAL37) (●; black bars) administration on food intake (FIG. 53A), body weight (FIG. 53B), plasma glucose (FIG. 53C), insulin (FIG. 53D) and glycated hemoglobin N-AcGIP(LysPAL37) (12.5 nmoles/kg/day) (FIG. 53E). Native GIP (12.5 mmoles/kg/day; ▴; lined bars) or saline vehicle (control; □; white bars) were administered for the 14-day period indicated by the horizontal black bar. Values are means ±SEM for 8 mice. *p<0.05, **p<0.01 compared to control. ΔΔp<0.01 compared to native GIP.
  • GIP or N-AcGIP(LysPAL37) had no effect on body weight or food intake (FIGS. 53A, 53B). Plasma glucose and insulin concentrations were also unchanged by treatment with native GIP for 14 days (FIGS. 53C, 53D). In contrast, daily injection of N-AcGIP(LysPAL37) resulted in a progressive lowering of plasma glucose, resulting in significantly (p<0.05) lowered concentrations at 14 days (FIG. 53C). At this time, glycated hemoglobin was also significantly (p<0.01) decreased in N-AcGIP(LysPAL37) treated ob/ob mice (FIG. 53E). These changes were accompanied by a tendency towards elevated insulin concentrations, but these did not achieve statistical significance over the time frame studies (FIG. 53D).
  • Effects of long term treatment of ob/ob mice with N-AcGIP(LysPAL37) on glucose tolerance. FIGS. 54A through 54D are a set of two line graphs (FIGS. 54A, 54C) and two bar graphs (FIGS. 54B, 54D) showing the effects of daily N-AcGIP(LysPAL37) administration on glucose tolerance (FIGS. 54A, 54B) and plasma insulin response (FIGS. 54C, 54D) to glucose. Tests were conducted after 14 daily injections of either N-AcGIP(LysPAL37) (12.5 nmoles/kg/day; ●; black bars), native GIP (12.5 nmoles/kg/day; ▴; lined bars) or saline vehicle (control; □; white bars). Glucose (18 mmoles/kg) was administered by intraperitoneal injection at the time indicated by the arrow. Plasma glucose and insulin AUC values for 0-60 minutes post injection are shown in the right panels. Values are means SEM for 8 mice. *p<0.05, **p<0.01, ***p<0.001 compared to control. Δp<0.05, ΔΔp<0.01, ΔΔΔp<0.001 compared to native GIP.
  • Consistent with effects on glycated hemoglobin, treatment of ob/ob mice for 14 days with N-AcGIP(LysPAL37) resulted in a significant improvement in glucose tolerance (FIGS. 54A, 54B). Plasma glucose concentrations throughout the test and the overall 0-60 minutes AUC values were decreased (p<0.01 to p<0.001). This was accompanied by increased insulin concentrations during the latter stages (p<0.05) and a greater (p<0.01) overall AUC insulin response (FIGS. 54C, 54D). In contrast, daily administration of native GIP had no effect on glucose tolerance or the plasma insulin response to glucose compared with control ob/ob mice receiving saline injections for 14 days (FIG. 54).
  • Effects long term treatment of ob/ob mice with N-AcGIP(LysPAL37) on insulin sensitivity, and effects of long term treatment of ob/ob mice with N-AcGIP(LysPAL37) on pancreatic insulin content. FIGS. 55A through 55D are a line graph and three bar graphs showing the effects of daily N-AcGIP(LysPAL37) administration on insulin sensitivity (FIGS. 55A, 55B) and pancreatic weight (FIG. 55C) and insulin content (FIG. 55D). Observations were conducted after 14 daily injections of either N-AcGIP(LysPAL37) (12.5 nmoles/kg/day; ●; black bars), native GIP (12.5 nmoles/kg/day; ▴; lined bars) or saline vehicle (control; □; white bars). In FIG. 55A, insulin (50 U/kg) was administered by intraperitoneal injection at the time indicated by the arrow. Plasma glucose AUC values for 0-60 minutes post injection are shown in the right panels. Values are means ±SEM for 8 mice. *p<0.05, **p<0.01 compared to control. Δp<0.05, ΔΔp<0.01 compared to native GIP.
  • Insulin sensitivity of the 3 groups of mice after 14 days treatment is shown in FIGS. 55A, 55B. Compared with ob/ob mice receiving daily injections of saline or native GIP, N-AcGIP(LysPAL37) prompted a significant improvement of insulin sensitivity. Both the individual glucose concentrations and 0-60 minutes AUC values were significantly different (p<0.01) from the other two groups. In contrast, daily treatment with native GIP did not affect the characteristic insulin resistance of ob/ob mice (FIG. 55A, 55B).
  • Treatment of ob/ob mice for 14 days with native GIP or N-AcGIP(LysPAL37) did not affect pancreatic weight compared with saline-treated controls (FIGS. 55C, 55D). Similarly, pancreatic insulin content was similar in the GIP and saline treated groups. However, daily administration of N-AcGIP(LysPAL37) significantly increased (p<0.01) insulin content compared with each of the other groups (FIGS. 55C, 55D).
  • Evaluation of GIP receptor desensitization after long term treatment of ob/ob mice with N-AcGIP(LysPAL37). FIGS. 56A through 56D are a set of two line graphs (FIGS. 56A, 56C) and two bar graphs (FIGS. 56B, 56D) showing the retention of glucose lowering (FIGS. 56A, 56B) and insulin releasing (FIGS. 56C, 56D) activity of N-AcGIP(LysPAL37) and native GIP after daily injection for 14 days. Glucose (18 mmoles/kg) was administered by intraperitoneal injection alone (□; white bars) or in combination with either N-AcGIP(LysPAL37) (●; black bars) or native GIP (▴; lined bars) (both at 25 nmoles/kg) at the time indicated by the arrow. Plasma glucose and insulin AUC values for 0-60 minutes post injection are shown in the right panels. Values are means ±SEM for 8 mice. *p<0.05, **p<0.01 compared to glucose alone. Δp<0.05, ΔΔp<0.01 compared to native GIP. FIGS. 57A through 57D are a set of two line graphs (FIGS. 57A, 57C) and two bar graphs (FIGS. 57B, 57D) showing the acute glucose lowering (FIGS. 57A, 57B) and insulin releasing (FIGS. 57C, 57D) effects of N-AcGIP(LysPAL37) after 14 daily injections of either N-AcGIP(LysPAL37) (12.5 nmoles/kg/day; ●; black bars), native GIP (12.5 nmoles/kg/day; ▴; lined bars) or saline vehicle (control; □; white bars). N-AcGIP(LysPAL37) (25 nmoles/kg) was administered by intraperitoneal injection with glucose (18 mmoles/kg) at the time indicated by the arrow. Plasma glucose and insulin AUC values for 0-60 minutes post injection are shown in the right panels. Values are means ±SEM for 8 mice. *p<0.05, **p<0.01 compared to mice receiving control injections. Δp<0.05, ΔΔp<0.01 compared to group receiving injections of native GIP.
  • As shown in FIGS. 56A through 56D, treatment of ob/ob mice with N-AcGIP(LysPAL37) for 14 days did not prevent the ability of the peptide to significantly moderate the glycaemic excursion (p<0.01) and enhance plasma insulin concentrations (p<0.01) when administered acutely with intraperitoneal glucose. In contrast, the responses of ob/ob mice to acute administration of native GIP were almost identical in mice receiving treatment with GIP or saline for 14 days (FIGS. 56A-56D). To further substantiate the lack of GIP receptor desensitization following chronic treatment with N-AcGIP(LysPAL37), the acute effects of the analogue, administered with glucose, were examined in each of the 3 groups after 14 days treatment with N-AcGIP(LysPAL37), native GIP or saline (FIGS. 57A-57D). Apart from lower basal values in the former group, the glucose and insulin responses were identical with similar 0-60 minutes AUC measures for both plasma glucose and insulin concentrations.
  • While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (2)

1. A peptide analogue of GIP(1-42) (SEQ ID NO:1), comprising at least 12 amino acid residues from the N-terminal end of GIP(3-42).
2-38. (canceled)
US11/825,293 1999-03-29 2007-07-03 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity Abandoned US20080009603A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/825,293 US20080009603A1 (en) 1999-03-29 2007-07-03 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
GB9907216A GB9907216D0 (en) 1999-03-29 1999-03-29 Peptide
GBGB9907216.7 1999-03-29
GBGB9917565.5 1999-07-27
GB9917565A GB9917565D0 (en) 1999-07-27 1999-07-27 Peptide
PCT/GB2000/001089 WO2000058360A2 (en) 1999-03-29 2000-03-29 Analogs of gastric inhibitory peptide and their use for treatment of diabetes
US09/937,687 US6921748B1 (en) 1999-03-29 2000-03-29 Analogs of gastric inhibitory polypeptide and their use for treatment of diabetes
GBGB0404124.0 2004-02-25
GBGB0404124.0A GB0404124D0 (en) 2004-02-25 2004-02-25 Antagonists of GIP
PCT/GB2005/000710 WO2005082928A2 (en) 2004-02-25 2005-02-25 Peptide analogues of gip for treatment of diabetes, insulin resistance and obesity
US11/090,825 US7326688B2 (en) 1999-03-29 2005-03-25 Analogs of gastric inhibitory polypeptide and their use of treatment for diabetes
US11/090,787 US20050272652A1 (en) 1999-03-29 2005-03-25 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US11/713,892 US7875587B2 (en) 1999-03-29 2007-03-05 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US11/825,293 US20080009603A1 (en) 1999-03-29 2007-07-03 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US11/090,825 Continuation-In-Part US7326688B2 (en) 1999-03-29 2005-03-25 Analogs of gastric inhibitory polypeptide and their use of treatment for diabetes
US11/713,892 Continuation US7875587B2 (en) 1999-03-29 2007-03-05 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity

Publications (1)

Publication Number Publication Date
US20080009603A1 true US20080009603A1 (en) 2008-01-10

Family

ID=35449761

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/090,787 Abandoned US20050272652A1 (en) 1999-03-29 2005-03-25 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US11/713,892 Active US7875587B2 (en) 1999-03-29 2007-03-05 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US11/825,293 Abandoned US20080009603A1 (en) 1999-03-29 2007-07-03 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/090,787 Abandoned US20050272652A1 (en) 1999-03-29 2005-03-25 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US11/713,892 Active US7875587B2 (en) 1999-03-29 2007-03-05 Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity

Country Status (1)

Country Link
US (3) US20050272652A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070167370A1 (en) * 1999-03-29 2007-07-19 Uutech Limited Peptide analogues of GIP for treatment of diapetes, insulin resistance and obesity
US20090170762A1 (en) * 2005-09-08 2009-07-02 Uutech Limited Treatment of Diabetes Related Obesity
US20090286722A1 (en) * 2005-09-08 2009-11-19 Utech Limited Analogs of Gastric Inhibitory Polypeptide as a Treatment for Age Related Decreased Pancreatic Beta Cell Function
WO2010022213A2 (en) * 2008-08-22 2010-02-25 Elixir Pharmaceuticals, Inc. Compositions and methods for preserving pancreatic islet mass
WO2010016938A3 (en) * 2008-08-07 2010-04-15 Ipsen Pharma S.A.S. Glucose-dependent insulinotropic polypeptide analogues
WO2010016940A3 (en) * 2008-08-07 2010-04-15 Ipsen Pharma S.A.S. Analogues of glucose-dependent insulinotropic polypeptide
US20110136725A1 (en) * 2008-08-07 2011-06-09 Zheng Xin Dong Analogues of glucose-dependent insulinotropic polypeptide (gip) modified at n-terminal
KR101419332B1 (en) * 2008-08-07 2014-07-15 입센 파마 에스.에이.에스 Truncated analogues of glucose-dependent insulinotropic polypeptide
JP2020521784A (en) * 2017-05-31 2020-07-27 ユニバーシティ オブ コペンハーゲン Long-acting GIP peptide analogues
US10940678B2 (en) 2013-11-15 2021-03-09 Performance Polyamides, Sas Polyamide compositions for metal coating and metal components coated with the same
US10968266B2 (en) 2014-09-05 2021-04-06 University Of Copenhagen GIP peptide analogues

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7091183B1 (en) * 1996-12-03 2006-08-15 Boston Medical Center Corporation Specific antagonists for glucose-dependent insulinotropic polypeptide (GIP)
MXPA01009805A (en) * 1999-03-29 2004-07-30 Univ Ulster Peptide.
BRPI0516775A (en) * 2004-10-25 2008-09-23 Cytos Biotechnology Ag gastric inhibitor antigen (gip) polypeptide assays and their uses
CA2789262C (en) 2005-04-28 2016-10-04 Proteus Digital Health, Inc. Pharma-informatics system
EP2224945B1 (en) 2007-11-23 2012-05-16 Michael Rothkopf Methods of enhancing diabetes resolution
WO2011014680A2 (en) * 2009-07-31 2011-02-03 The Board Of Trustees Of The Leland Stanford Junior University Gastric inhibitory peptide variants and their uses
AR080592A1 (en) * 2010-03-26 2012-04-18 Lilly Co Eli PEPTIDE WITH ACTIVITY FOR GIP-R AND GLP-1-R, FAMILY FORMULATION THAT UNDERSTANDS IT, ITS USE TO PREPARE A USEFUL MEDICINAL PRODUCT FOR THE TREATMENT OF MELLITUS DIABETES AND TO INDICATE WEIGHT LOSS
EP3099313B1 (en) * 2014-01-27 2021-04-21 Centre National De La Recherche Scientifique Retro-inverso analogs of spadin display increased antidepressant effects
JOP20200119A1 (en) * 2015-01-09 2017-06-16 Lilly Co Eli Gip and glp-1 co-agonist compounds
KR20220070057A (en) 2015-03-09 2022-05-27 인테크린 테라퓨틱스, 아이엔씨. Methods for the treatment of nonalcoholic fatty liver disease and/or lipodystrophy
KR20180108602A (en) 2015-12-23 2018-10-04 암젠 인크 Methods for treating or ameliorating a metabolic disorder using a binding protein for a gastric inhibitory peptide receptor (GIPR) in combination with a GLP-1 agonist
JOP20190177A1 (en) 2017-01-17 2019-07-16 Amgen Inc Method of treating or ameliorating metabolic disorders using glp-1 receptor agonists conjugated to antagonists for gastric inhibitory peptide receptor (gipr)
MX2019011867A (en) 2017-04-03 2020-01-09 Coherus Biosciences Inc Pparî³ agonist for treatment of progressive supranuclear palsy.
CN113366014A (en) 2018-12-03 2021-09-07 安泰博医药 Modified GIP peptide analogs

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5545618A (en) * 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
US5846937A (en) * 1997-03-03 1998-12-08 1149336 Ontario Inc. Method of using exendin and GLP-1 to affect the central nervous system
US5998204A (en) * 1997-03-14 1999-12-07 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
US6077822A (en) * 1993-09-14 2000-06-20 Dumex-Alpharma A/S Drug salts
US6087476A (en) * 1988-03-24 2000-07-11 Igen International, Inc. Luminescent chimeric proteins
US6303661B1 (en) * 1996-04-25 2001-10-16 Probiodrug Use of dipeptidyl peptidase IV effectors for lowering the blood glucose level in mammals
US6410508B1 (en) * 1998-10-07 2002-06-25 Med College Georgia Res Inst Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
US20020151495A1 (en) * 1996-12-03 2002-10-17 Wolfe M. Michael Specific antagonists for glucose-dependent insulinotropic polypeptide (GIP)
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US20030157107A1 (en) * 2000-05-16 2003-08-21 Kazumasa Miyawaki Agents for preventing or ameliorating insulin resistance and/or obesity
US20030204063A1 (en) * 2000-08-02 2003-10-30 Denis Gravel Modified biological peptides with increased potency
US20030221201A1 (en) * 2001-08-30 2003-11-27 Biorexis Pharmaceutical Corporation Modified transferrin fusion proteins
US20030232761A1 (en) * 2002-03-28 2003-12-18 Hinke Simon A. Novel analogues of glucose-dependent insulinotropic polypeptide
US20040029805A1 (en) * 2002-06-15 2004-02-12 Wolfe M. Michael Prevention and treatment of nonalcoholic fatty liver disease (NAFLD) by antagonism of the receptor to glucose-dependent insulinotropic polypeptide (GIP)
US20040228846A1 (en) * 1999-02-10 2004-11-18 Curis, Inc. Methods and reagents for treating glucose metabolic disorders
US20040242853A1 (en) * 2001-07-31 2004-12-02 Nigel Greig Glp-1 exendin-4 peptide analogs and uses thereof
US20050009147A1 (en) * 1992-07-15 2005-01-13 Wilfried Bauer Peptides
US6849714B1 (en) * 1999-05-17 2005-02-01 Conjuchem, Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US6852690B1 (en) * 1995-08-22 2005-02-08 Amylin Pharmaceuticals, Inc. Method and composition for enhanced parenteral nutrition
US20050049177A1 (en) * 2003-05-15 2005-03-03 Trustees Of Tufts College Stable analogs of peptide and polypeptide therapeutics
US6921748B1 (en) * 1999-03-29 2005-07-26 Uutech Limited Analogs of gastric inhibitory polypeptide and their use for treatment of diabetes

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991011457A1 (en) 1990-01-24 1991-08-08 Buckley Douglas I Glp-1 analogs useful for diabetes treatment
JPH04145099A (en) 1990-10-05 1992-05-19 Sanwa Kagaku Kenkyusho Co Ltd Polypeptide derivative having gip-like activity and use thereof
WO1992010576A1 (en) 1990-12-13 1992-06-25 The Upjohn Company Fusion polypeptides
US5705483A (en) 1993-12-09 1998-01-06 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
EP0658568A1 (en) 1993-12-09 1995-06-21 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
JPH09511384A (en) 1994-01-24 1997-11-18 リサーチ トライアングル ファーマシューティカルズ リミテッド Treatment of juvenile diabetes
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US5512549A (en) 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5827734A (en) 1995-01-20 1998-10-27 University Of Washington Materials and methods for determining ob protein in a biological sample
US5739106A (en) 1995-06-07 1998-04-14 Rink; Timothy J. Appetite regulating compositions
KR19990028388A (en) 1995-06-30 1999-04-15 피터 지. 스트링거 How to Treat Diabetes
DE19530865A1 (en) 1995-08-22 1997-02-27 Michael Dr Med Nauck Active ingredient and agent for parenteral nutrition
AU7522496A (en) 1995-10-25 1997-05-15 Regents Of The University Of California, The Methods for restoring or enhancing reproductive function in reproductively impaired hosts
AU731685C (en) 1996-01-23 2002-08-22 Indevus Pharmaceuticals, Inc. Methods for using the obese gene and its gene product to stimulate hematopoietic development
CA2243718A1 (en) 1996-02-06 1997-08-14 Eli Lilly And Company Diabetes therapy
US5912007A (en) 1996-02-29 1999-06-15 Warner-Lambert Company Delivery system for the localized administration of medicaments to the upper respiratory tract and methods for preparing and using same
ATE417622T1 (en) 1996-08-08 2009-01-15 Amylin Pharmaceuticals Inc REGULATION OF GASTROINTESTINAL MOBILITY
CA2266585A1 (en) 1996-09-20 1998-03-26 Hoechst Aktiengesellschaft Use of leptin antagonists for treating insulin resistance in type ii diabetes
AU5083098A (en) 1996-10-25 1998-05-22 Icogen Corporation Use of leptin to stimulate hematopoiesis
DK0996459T3 (en) 1997-01-07 2006-01-16 Amylin Pharmaceuticals Inc Use of exendins and agonists thereof to reduce food intake
WO1998036763A1 (en) 1997-02-25 1998-08-27 Eli Lilly And Company Treatment of infertility with leptin receptor ligands
DE69840146D1 (en) 1997-03-14 2008-12-04 Univ California FLUORESCENCE PROTEIN SENSORS FOR THE DETECTION OF ANALYTES
CA2283834A1 (en) 1997-03-31 1998-10-08 James Arthur Hoffmann Glucagon-like peptide-1 analogs
IL120733A0 (en) 1997-04-29 1997-08-14 Yeda Res & Dev Leptin as an inhibitor of cell proliferation
WO1998055139A1 (en) 1997-06-06 1998-12-10 Smithkline Beecham Plc Use of leptin antagonists for the treatment of diabetes
JP2001513512A (en) 1997-08-08 2001-09-04 アミリン・ファーマシューティカルズ,インコーポレイテッド New exendin agonist compounds
ES2294822T3 (en) 1997-11-14 2008-04-01 Amylin Pharmaceuticals, Inc. NEW COMPOUNDS OF EXENDINE AGONISTS.
BR9814189A (en) 1997-11-14 2000-10-03 Amylin Pharmaceuticals Inc "exendin agonist compounds"
US20050272652A1 (en) * 1999-03-29 2005-12-08 Gault Victor A Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
DE19921537A1 (en) 1999-05-11 2000-11-23 Dieter Hoersch Treating carbohydrate metabolism disorders, especially diabetes, comprises activating insulin-secreting b-cells using glucagon-related peptide, glucose-dependent insulinotropic polypeptide, exendin-4 or related drugs
US6245320B1 (en) * 1999-09-01 2001-06-12 University Of Maryland Inhibition of mucin release from airway goblet cells by polycationic peptides
EP1356295A2 (en) 2000-04-27 2003-10-29 Bionebraska, Inc. Gastric inhibitory polypeptide diagnostic test for detecting susceptibility to type-2 diabetes, impaired glucose tolerance, or impaired fasting glucos
GB0121709D0 (en) 2001-09-07 2001-10-31 Imp College Innovations Ltd Food inhibition agent
US7459432B2 (en) 2001-09-24 2008-12-02 Imperial College Innovations Ltd. Modification of feeding behavior
EP1474163A2 (en) 2002-01-10 2004-11-10 Imperial College Innovations Limited Modification of feeding behavior
US20060252916A1 (en) 2002-06-04 2006-11-09 Eli Lilly And Company Modified glucagon-like peptide-1 analogs
JP2006502100A (en) 2002-06-11 2006-01-19 エーザイ株式会社 Use of a compound having GIP activity for the treatment of disorders associated with abnormal loss of cells and / or for the treatment of obesity
DK1569680T3 (en) 2002-10-22 2009-05-18 Waratah Pharmaceuticals Inc Treatment of diabetes
US20050059605A1 (en) 2003-01-31 2005-03-17 Krishna Peri Chemically modified metabolites of regulatory peptides and methods of producing and using same
EP1626981A4 (en) 2003-03-04 2006-11-22 Biorexis Pharmaceutical Corp Dipeptidyl-peptidase protected proteins
CN1961000B (en) 2004-02-11 2011-05-04 安米林药品公司 Hybrid polypeptides with selectable properties
AU2006279276A1 (en) 2005-07-29 2007-02-15 Amprotein Corporation Chimeric therapeutic agents
CA2623412A1 (en) * 2005-09-08 2007-03-15 Uutech Limited Analogs of gastric inhibitory polypeptide as a treatment for age related decreased pancreatic beta cell function
CA2622069A1 (en) * 2005-09-08 2007-03-15 Uutech Limited Treatment of diabetes related obesity

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6087476A (en) * 1988-03-24 2000-07-11 Igen International, Inc. Luminescent chimeric proteins
US5545618A (en) * 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
US20050009147A1 (en) * 1992-07-15 2005-01-13 Wilfried Bauer Peptides
US6077822A (en) * 1993-09-14 2000-06-20 Dumex-Alpharma A/S Drug salts
US6852690B1 (en) * 1995-08-22 2005-02-08 Amylin Pharmaceuticals, Inc. Method and composition for enhanced parenteral nutrition
US6303661B1 (en) * 1996-04-25 2001-10-16 Probiodrug Use of dipeptidyl peptidase IV effectors for lowering the blood glucose level in mammals
US20020151495A1 (en) * 1996-12-03 2002-10-17 Wolfe M. Michael Specific antagonists for glucose-dependent insulinotropic polypeptide (GIP)
US5846937A (en) * 1997-03-03 1998-12-08 1149336 Ontario Inc. Method of using exendin and GLP-1 to affect the central nervous system
US5998204A (en) * 1997-03-14 1999-12-07 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
US6410508B1 (en) * 1998-10-07 2002-06-25 Med College Georgia Res Inst Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
US20040228846A1 (en) * 1999-02-10 2004-11-18 Curis, Inc. Methods and reagents for treating glucose metabolic disorders
US6921748B1 (en) * 1999-03-29 2005-07-26 Uutech Limited Analogs of gastric inhibitory polypeptide and their use for treatment of diabetes
US7326688B2 (en) * 1999-03-29 2008-02-05 Uutech Limited Analogs of gastric inhibitory polypeptide and their use of treatment for diabetes
US6849714B1 (en) * 1999-05-17 2005-02-01 Conjuchem, Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US20030157107A1 (en) * 2000-05-16 2003-08-21 Kazumasa Miyawaki Agents for preventing or ameliorating insulin resistance and/or obesity
US20030204063A1 (en) * 2000-08-02 2003-10-30 Denis Gravel Modified biological peptides with increased potency
US20040242853A1 (en) * 2001-07-31 2004-12-02 Nigel Greig Glp-1 exendin-4 peptide analogs and uses thereof
US20030221201A1 (en) * 2001-08-30 2003-11-27 Biorexis Pharmaceutical Corporation Modified transferrin fusion proteins
US20030232761A1 (en) * 2002-03-28 2003-12-18 Hinke Simon A. Novel analogues of glucose-dependent insulinotropic polypeptide
US20040029805A1 (en) * 2002-06-15 2004-02-12 Wolfe M. Michael Prevention and treatment of nonalcoholic fatty liver disease (NAFLD) by antagonism of the receptor to glucose-dependent insulinotropic polypeptide (GIP)
US20050049177A1 (en) * 2003-05-15 2005-03-03 Trustees Of Tufts College Stable analogs of peptide and polypeptide therapeutics

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7875587B2 (en) 1999-03-29 2011-01-25 Uutech Limited Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
US20070167370A1 (en) * 1999-03-29 2007-07-19 Uutech Limited Peptide analogues of GIP for treatment of diapetes, insulin resistance and obesity
US20090170762A1 (en) * 2005-09-08 2009-07-02 Uutech Limited Treatment of Diabetes Related Obesity
US20090286722A1 (en) * 2005-09-08 2009-11-19 Utech Limited Analogs of Gastric Inhibitory Polypeptide as a Treatment for Age Related Decreased Pancreatic Beta Cell Function
EA020005B1 (en) * 2008-08-07 2014-07-30 Ипсен Фарма С.А.С. Glucose-dependent insulinotropic polypeptide analogues
US8999940B2 (en) 2008-08-07 2015-04-07 Ipsen Pharma S.A.S. Analogues of glucose-dependent insulinotropic polypeptide (GIP) modified at N-terminal
WO2010016940A3 (en) * 2008-08-07 2010-04-15 Ipsen Pharma S.A.S. Analogues of glucose-dependent insulinotropic polypeptide
WO2010016938A3 (en) * 2008-08-07 2010-04-15 Ipsen Pharma S.A.S. Glucose-dependent insulinotropic polypeptide analogues
US20110136733A1 (en) * 2008-08-07 2011-06-09 Zheng Xin Dong Analogues of glucose-dependent insulinotropic polypeptide
US20110136725A1 (en) * 2008-08-07 2011-06-09 Zheng Xin Dong Analogues of glucose-dependent insulinotropic polypeptide (gip) modified at n-terminal
US20110144007A1 (en) * 2008-08-07 2011-06-16 Zheng Xin Dong Glucose-dependent insulinotropic polypeptide analogues
CN102171244A (en) * 2008-08-07 2011-08-31 益普生制药股份有限公司 Analogues of glucose-dependent insulinotropic polypeptide
KR101417873B1 (en) * 2008-08-07 2014-07-09 입센 파마 에스.에이.에스 Analogues of glucose-dependent insulinotropic polypeptide
KR101419332B1 (en) * 2008-08-07 2014-07-15 입센 파마 에스.에이.에스 Truncated analogues of glucose-dependent insulinotropic polypeptide
EP2987805A3 (en) * 2008-08-07 2016-04-13 Ipsen Pharma S.A.S. Analogues of glucose-dependent insulinotropic polypeptide
EA020019B1 (en) * 2008-08-07 2014-08-29 Ипсен Фарма С.А.С. Analogues of glucose-dependent insulinotropic polypeptide
EA020018B1 (en) * 2008-08-07 2014-08-29 Ипсен Фарма С.А.С. Truncated analogues of glucose-dependent insulinotropic polypeptide
KR101593158B1 (en) 2008-08-07 2016-02-12 입센 파마 에스.에이.에스. Analogues of glucose-dependent insulinotropic polypeptide (gip) modified at n-terminal
KR20150056668A (en) * 2008-08-07 2015-05-26 입센 파마 에스.에이.에스. Analogues of glucose-dependent insulinotropic polypeptide (gip) modified at n-terminal
KR20150056667A (en) * 2008-08-07 2015-05-26 입센 파마 에스.에이.에스. Glucose-dependent insulinotropic polypeptide analogues
US9074014B2 (en) 2008-08-07 2015-07-07 Ipsen Pharma S.A.S. Analogues of glucose-dependent insulinotropic polypeptide
US9072703B2 (en) 2008-08-07 2015-07-07 Ipsen Pharma S.A.S. Glucose-dependent insulinotropic polypeptide analogues
KR101593155B1 (en) 2008-08-07 2016-02-12 입센 파마 에스.에이.에스. Glucose-dependent insulinotropic polypeptide analogues
WO2010022213A3 (en) * 2008-08-22 2010-04-15 Elixir Pharmaceuticals, Inc. Compositions and methods for preserving pancreatic islet mass
WO2010022213A2 (en) * 2008-08-22 2010-02-25 Elixir Pharmaceuticals, Inc. Compositions and methods for preserving pancreatic islet mass
US10940678B2 (en) 2013-11-15 2021-03-09 Performance Polyamides, Sas Polyamide compositions for metal coating and metal components coated with the same
US10968266B2 (en) 2014-09-05 2021-04-06 University Of Copenhagen GIP peptide analogues
JP2020521784A (en) * 2017-05-31 2020-07-27 ユニバーシティ オブ コペンハーゲン Long-acting GIP peptide analogues
US11572399B2 (en) 2017-05-31 2023-02-07 University Of Copenhagen Long-acting GIP peptide analogues

Also Published As

Publication number Publication date
US7875587B2 (en) 2011-01-25
US20050272652A1 (en) 2005-12-08
US20070167370A1 (en) 2007-07-19

Similar Documents

Publication Publication Date Title
US7875587B2 (en) Peptide analogues of GIP for treatment of diabetes, insulin resistance and obesity
CA2557151C (en) Peptide analogues of gip for treatment of diabetes, insulin resistance and obesity
US6921748B1 (en) Analogs of gastric inhibitory polypeptide and their use for treatment of diabetes
US5604203A (en) Analogs of peptide YY and uses thereof
KR101399178B1 (en) Hybrid polypeptides with selectable properties
EP0966297B1 (en) Regulation of gastrointestinal motility
US20090170762A1 (en) Treatment of Diabetes Related Obesity
JP5485876B2 (en) Unacylated ghrelin as a therapeutic agent in the treatment of metabolic diseases
CZ315594A3 (en) Peptide of glucagon type, insulinotropic derivatives, process of their preparation, pharmaceutical composition containing such compounds and use
SG181430A1 (en) Oxyntomodulin peptide analogue
AU2007221366A1 (en) Oxyntomodulin derivatives
KR20110043688A (en) Glucose-dependent insulinotropic polypeptide analogues
CA2271788A1 (en) Analogs of peptide yy and uses thereof
KR102230368B1 (en) Acylated oxyntomodulin analogues
JP2023544959A (en) hAM15-52 analog with improved amylin receptor (hAMY3R) potency
CN115461360A (en) Sustained GLP-1 and glucagon receptor dual agonists
HUT70176A (en) Amylin antagonists and agonists
WO2024003359A1 (en) AMYLIN RECEPTOR (hAMY3R) AGONISTS WITH IMPROVED CHEMICAL STABILITY
TWI428139B (en) A novel glucagon-like peptide analogue, a composition and use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: UUTECH LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAULT, VICTOR A;O'HARTE, FINBARR PAUL MARY;IRWIN, NIGEL;AND OTHERS;REEL/FRAME:022218/0019;SIGNING DATES FROM 20050729 TO 20050805

Owner name: UUTECH LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAULT, VICTOR A;O'HARTE, FINBARR PAUL MARY;IRWIN, NIGEL;AND OTHERS;SIGNING DATES FROM 20050729 TO 20050805;REEL/FRAME:022218/0019

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION