Title of Invention

"GIP ANALOG AND HYBRID POLYPEPTIDES WITH SELECTABLE PROPERTIES"

Abstract The present invention relates generally to novel GIP analogs and GIIP hybrid polypeptides with selectable properties, useful as agents for the treatment and prevention of metabolic diseases and disorders, for example those which can be alleviated by control plasma glucose levels, insulin levels, and/or insulin secretion, positive inotropic effects, reduction of catabolic effects, slowing of gastric emptying. Such conditions and disorders include, but are not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, critical care, insulin-resistance, obesity, and diabetes mellitus of any kind, including type 1, type 2, and gestational diabetes.
Full Text GIP ANALOG AND HYBRID P(XYPEPTIDES WITH SELECTABLE PROPERTIES
RELATED APPLICATIONS
[0001] The present application claims priority to commonly-owned and pending United States provisional applications: U.S. AppI. 60/652,662 filed Feb. 11, 2005; U.S. AppI. 60/653,433 filed Feb. 15, 2005; U.S. Provisional Application No. 60/651,647 filed February 11, 2005; U.S. Provisional Application Nos. 60/707,2~ and 60/707,369 filed August 11, 2005; U.S. Provisional Application No. 60/709,320 and 60/709,316 filed August 17, 2005, each of which are hereby incorporated by reference in their entirety. The present application hereby incorporates by reference in their entirety commonly-owned:
PCT/U505/04 178 filed February 11, 2005 and published as W02005/077072; PCTIUS2005/004351 filed February 11, 2005; PCTIUS2005/004631 filed February 11, 2005; PCT/U52005/045471 filed December 15, 2005; U.S. Patent Application No. 11/055,093; U.S. Patent Application No. 11/201664 filed August
10, 2005; and USSN 206,903 filed August 17, 2005. The applications teach compounds and uses related to and useful for the present invention. A paper copy of the Sequence Listing and a computer readable form of the Sequence Listing on two discs labeled "File Copy 1,' and "File Copy 2", each containing a file of 215 KB in size named "060 1-PCT.txt" created on Nov. 3, 2006, are hereby incorporated by reference.
FIELD OF THE INVENTION
[0002] The present invention relates to peptide chemistry and pharmaceuticals, and more particularly to novel gastric inhibitory peptide ("GIP") analog and GIP-containing hybrid polypeptides with selectable properties. It further relates to the use of these and other GIP compounds either alone or in adjunct with other compounds such as glucagon-like peptide 1 ("GLPl"), exendin-4, or amylin polypeptides, to treat metabolic disorders and conditions.
BACKGROUND OF THE INVENTION
[0003] Incretin peptides are hormones and peptide mimetics that cause an increase in the amount of insulin released when glucose levels are normal or particularly when they are elevated. These incretin peptides have other actions beyond the initial incretin action defined by insulin secretion. For instance, they may also have actions to reduce glucagon production and delay gastric emptying. In addition, they may have actions to improve insulin sensitivity, and they may increase islet cell neogenesis — the formation of new islets.
[0004] The concept of the incretin effect developed from the observation that insulin responses to oral glucose exceeded those measured after intravenous administration of equivalent amounts of glucose. It was concluded that gut-derived factors, or incretins, influenced postprandial insulin release. Nuttient entry into the stomach and proximal gastrointestinal tract causes release of incretin hormones, which theL stimulate insulin secretion. This insulinotropism, or ability to stimulate insulin secretion, can be
quantified by comparing insulin or C-peptide responses to oral vs. intravenous glucose loads. In this way, it has been shown that the incretin effect is responsible for about 50% to 70% of the insulin response to oral glucose in healthy individuals.

[0005] Although many postprandial hormones have incretin-like activity, predominant incretin peptides include glucose-dependent insulinotropic polypeptide, also known as gastric inhibitory polypeptide (GIP), glucagon-like peptide-1 (GLP-l), and exendin peptides (which are non-endogenous incretin mimetics). GIP and GLP-l both belong to the glucagon peptide superfamily and thus share amino acid sequence homology. GIP and GLP-l are secreted by specialized cells in the gastrointestinal tract and have receptors located on islet cells as well as other tissues. As incretins, both are secreted from the intestine in response to ingestion of nutrients, which results in enhanced insulin secretion. The insulinotropic effect of GIP and GLP-1 is dependent on elevations in ambient glucose. Both are rapidly inactivated by the ubiquitous enzyme dipeptidyl peptidase IV (DPP-IY).
[0006] Native human GIP is a single 42-amino acid peptide synthesized in and secreted by specialized enteroendocrine K-cells. These cells are concentrated primarily in the duodenum and proximal jejunum, although they also can be found throughout the intestine. The main stimulant for Gil' secretion is. ingestion of carbohydrate- and lipid-rich meals. Following ingestion, circulating plasma GIP levels increase 10- to 20-fold. The half-life of intact Gil' is estimated to be approximately 7.3 minutes in healthy subjects and 5.2 minutes in diabetic subjects.
[00071 The physiologic effects of GIP have been elucidated using GIP receptor antagonists, Gil' peptide antagonists, and Gil' receptor knockout mice, in addition to Gil' infusion protocols. Blocking GIP binding to its receptor results in attenuated glucose-dependent insulin secretion following oral glucose load in rats and mice. Similarly, administration of Gil' antagonists or GIP antisera markedly reduces the postprandial insulin release in rats. Gil' receptor knockout mice demonstrate normal fasting glucose levels but mild glucose intolerance following oral glucose loads. Interestingly, they also exhibit resistance to diet-induced obesity following months of high-fat feeding. Additionally, in the leptin-deficient ob/ob mouse, the Gil' receptor knockout genotype appears to decrease the extent of obesity that develops.
[0008] Gil' infusion has consistently demonstrated stimulation of insulin secretion in isolated rat islets, isolated perfused rat pancreas, dogs, and humans. During stepwise euglycemic, mild hyperglycemic (54 mg/dL above basal), and moderate hyperglycemic (143 mg/dL above basal) clamps, it has been demonstrated that Gil' infusion results in insulin secretion only in the presence of elevated glucose concentrations. Furthermore, it has been demonstrated that Gil' is not glucagonotropic in normal humans during either euglycemic or hyperglycemic conditions. Thus, the effect of endogenously released Gil' appears to be an important mechanism of postprandial insulin secretion and does not appear to play a role in the fasting state.
L0009] GIP has many non-incretin effects as well. Unlike other insulin secretagogues, GIP stimulates beta-cell proliferation and cell survival in INS-i islet cell-line studies. Furthermore, animal studies have suggested a role for GIP in lipid metabolism by stimulating lipoprotein lipase activity, inducing fatty acid incorporation into adipose tissue and stimulating fatty acid synthesis. However, in humans, there is no clear evidence for an effect of GIP on lipid metabolism. Gil' also appears to stimulate glucagon secretion from the isolated perfused rat pancreas, although human studies have not demonstrated any significant influence on glucagon secretion. Furthermore, unlike GLP- 1, Gm appears to act by accelerating emptying of the stomach rather than by inhibiting gastrointestinal motility.
[0010] The Gm-receptor, a member of the G-protein-coupled receptor family has a high specificity for Gil' and does not bind other peptides of the glucagon family. For this reason, GLP- 1/Gil' chimeric peptides show nearly no affinity for the Gm-receptor. From such studies it has been concluded that the GIP( 1-30) sequence of the GIP( 1-42) is sufficient for recognizing the receptor. Gm(6-30)-amide contains the high affinity binding region of GIP(I-42) but exhibits antagonist activity.
10011] Despite potent glucoregulatory actions through glucose-dependant stimulation of insulin secretion, the insulinotropic effect of Gil' is significantly reduced in diabetic subjects compared to normal individuals (16-18). Consequently, clinical use of Gil' has not been significantly advanced. Further, there remains a need to develop additional diabetic treatment modalities as well as treatments for metabolic diseases, conditions, and disorders. Accordingly, it is an object of the present invention to provide GIP analog and Gm-containing hybrid polypeptides and methods for their use to treat or prevent metabolic diseases, conditions, and disorders.
[0012] All documents referred to herein are incorporated by reference into the present application as though fully set forth herein.
SUMMARY OF THE INVENTION
[00131 Provided are methods for treating or preventing metabolic diseases and disorders including those which can be alleviated by control of plasma glucose levels, insulin levels, and/or insulin secretion, such as diabetes and diabetes-related conditions, and conditions and disorders including, but not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind, including type 1, type 2, and gestational diabetes. The methods comprise administering a therapeutically or prophylactically effective amount of a Gil' or GIP analog, fragment or derivative thereof, or a Gil' hybrid or derivatives thereof as described herein, alone (monotherapy) or in combination with another agent or therapy (adjunct therapy), for example a glucose lowering agent (e.g., antidiabetic) or agents or methods that inhibit or reduce gastric emptying (examples of such agents are presented herein), to a subject in need thereof. By providing a Gil' bioactivity as part of a Gm hybrid having one or more other hormonal bio-activities, e.g., pramlintide, exendin, BNP, compounds with one or more selectable functions will have the additional benefit of a property of a Gil' bio-activity such as
lowering plasma glucose, increasing insulin secretion, without the side effects associated with othe~
incretin molecules. For example, a GIP-sCT hybrid compound of the invention can have a selectable property of a salmon calcitonin, such as decreasing bone loss and bone resorption or reducing cartilage turnover (chondroprotection), and a property of a Gil', such as plasma glucose lowering (concomitant with an anti-catabolic aspect as described herein) and/or inhibiting bone resorption and maintaining or increasing bone density. A GIP hybrid with such selectable properties can enhance treatment of osteoporosis or conditions of high cartilage turnover, particularly in those who can also benefit from glycemic control, such as subjects with diabetes or under going critical care.
[0014] In one embodiment are provided novel Gil' analogs having one or more enhanced properties. The Gil' analogs have increased resistance to DPP-IV or to other proteases, such as those found in human plasma and/or on kidney brush border membranes, which increases in vivo half-life. In a further embodiment the Gil' analogs have at least one substitution, modification, insertion or deletion in amino acids 4-30. These changes can provide enhanced properties such as increased Gil' receptor binding, increased receptor selectivity, and/or increased resistance to degradation by chemical and/or proteolytic means. In another embodiment the Gil' analog further has least 50% sequence identity to native GIP( 1-30), native Gil'( 1-14), native GIP(19-30) or native GIP( 1-42) over the entire length of each molecule, and at least one biological property of a Gil'. In certain embodiments, novel Gil' analog polypeptides include unnatural amino acids, such as a D amino acid. In a further embodiment a GIP analog or hybriij is modified to have reduced renal clearance, such as by fatty acyl derivitization.
[0015] In another embodiment are provided novel GIP-containing hybrid polypeptides with selectable properties. The hybrid polypeptides exhibit at least one hormonal activity. In one embodiment the GIPhybrid polypeptides comprise at least two biologically active ("bio-active") peptide hormone modules covalently linked together, wherein at least one of the bio-active peptide hormone modules comprises a Gil' polypeptide. The other bio-active peptide hormone module can be: (a) a native component peptide hormone, (b) an analog or derivative of a native component peptide hormone that retains hormonal activity, (c) a fragment of a native component peptide hormone that retains hormonal activity, (d) a fragment of analogs or derivatives of a native component peptide hormone that retains hormonal activity, (e) a structural motif of a native component peptide hormone that imparts a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic to the hybrid polypeptide; or (0 a structural motif of analogs or derivatives of a native component peptide hormone that imparts a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic to the hybrid polypeptide. The structural motifs of (e) and (17) will collectively be referred to herein as "peptidic enhancers". An example of a peptidic enhancer is a Tr~ cage sequence, particularly one derived from exendin4.
[0016] In a further embodiment a GIP-hybrid polypeptide comprises at least two bio-active peptide hormone modules covalently linked together, wherein at least one of the bio-active peptide hormone modules is comprised of a GIP polypeptide, and the second exhibits at least one hormonal activity of a component peptide hormone. The bio-active peptide hormone modules are independently selected from:
component peptide hormones, fragments of component peptide hormones that exhibit at least one hormonal activity of the component peptide hormones, analogs and derivatives of component peptide hormones that exhibit at least one hormonal activity of the component peptide hormones, and fragments of analogs and derivatives of component peptide hormones that exhibit at least one hormonal activity of the component peptide hormones.
[0017] In one embodiment the GIP-hybrid polypeptides comprise a novel Gil' analog polypeptide of the invention covalently linked to at least one additional bio-active peptide hormone module. In a further embodiment the bio-active peptide hormone module is a peptidic enhancer. In one embodiment the GIP hybrid polypeptide of the invention will exhibit at least 50% sequence identity to a native Gil'( 1-30), native Gil'( 1-14), native Gil'( 19-30) or native GIP(1 -42) over the entire length of the GIP portion. In certain embodiments the Gil' portion can comprise a novel Gil' analog further comprising a peptidic enhancer, such as a trp-cage motif. Accordingly, a Gil' hybrid can comprise a Gil' portion that is a Gil' analog, fragment or derivative thereof with a peptidic enhancer, such as a trp-cage motif, and having enhanced properties, that is linked covalently to an additional bio-active peptide hormone module, such as another hormone or growth factor or fragment thereof.
[0018] Component peptide hormones for use in a GIP-hybrid polypeptide include: amylin, adrenomedullin (ADM), calcitonin (Cr), calcitonin gene related peptide (CGRP), intermedin, cholecystokinin ("CCK"), leptin, peptide YY (PYY), glucagon-like peptide-l (GLP-1), glucagon-like peptide 2 (GLP-2), oxyntomodulin (OXM), natriuretic peptides (e.g. AN?, BNP, CNP or urodilatin), urocortin family peptide, e.g., Ucn-2 and Ucn—3, neuromedin family peptide, e.g. neuromedin U25 or splice variants, exendin-3 and exendin-4.
[0019] In other Gil' hybrid embodiments the GIP portion is combined with a gastrin /CCK receptor ligand; an amylin receptor ligand; a c~lcitonin receptor ligand; a CGRP receptor ligand, a PYY receptor ligand, an EGF receptor ligand; a Glucagon-like peptide 1 receptor ligand; a Glucagon-like peptide 2 receptor ligand; a gastric inhibitory polypeptide (Gil') receptor ligand; a keratinocyte growth factor (KGF) receptor 1 ligand; a dipeptidyl peptidase IV inhibitor; a REG protein receptor ligand; a Growth Hormone receptor ligand; a Prolactin (PRL) receptor ligand; an Insulin-like Growth Factor (IGF) receptor ligan~ PTH-related protein (PTHrP) receptor ligand; hepatocyte growth factor (HGF) receptor ligand; a bone

morphogenetic protein (BMP) receptor ligand, a transforming growth factor (TGF receptor ligand; a laminin receptor ligand; a vasoactive intestinal peptide (VIP) receptor ligand; a fibroblast growth factor (FGF) receptor ligand; a nerve growth factor (NGF) receptor ligand; an islet neogenesis associated protein (INGAP) receptor ligand; an Activin-A receptor ligand; a vascular endothelial growth factor (VEGF) receptor ligand; an erythropoietin (EPO) receptor ligand; a pituitary adenylate cyclase activating polypeptide (PACAP) receptor ligand; a granulocyte colony stimulating factor (G-CSF) receptor ligand; a granulocyte- macrophage colony stimulating factor (GM-CSF); a platelet-derived growth factor (PDGF) receptor ligand, a cannabinoid CB 1 receptor antagonist, and a secretin receptor ligand.
[00201 In one embodiment desirable GIP hybrid polypeptides include an N-terminal GIP or novel Gil' analog fragment in combination with a C-terminal polypeptide or fragment thereof having a glucose lowering activity (e.g., antidiabetics, exendin) or the ability to inhibit or reduce gastric emptying. Such desirable Gil' hybrids include an N-terminal Gil' fragment or novel Gil' analog or derivative fragment in combination with a C-terminal exendin, GLP1, symiin (pramlintide), amylin, CCK, gastrmn, PYY, secretin, GRP, neuromedins, urocortin, calcitonin, or salmon calcitonin, a natriuretic peptide (e.g., ANP, BNP, CNP, urodilatin) or analog (e.g. amylin-sCT-amylin chimera), derivative or fragment thereof. In other embodiments desirable Gil' hybrids include a C-terminal Gil' or novel Gil' analog fragment in combination with an N-terminal polypeptide or fragment thereof having a glucose lowering activity (e.g., antidiabetics, exendin) or the ability to inhibit or reduce gastric emptying. In such embodiments, the chimeric polypeptides can include a C-terminal GIP, a novel Gil' analog, or fragment thereof, in combination with a N-terminal exendin, GLPI, symlin (pramlintide), amylin, CCK, gastrin, PYY, secretin, GRP, neuromedins, urocortin, calcitonin, or salmon calcitonin, a natriuretic peptide or analog, derivative or fragment thereof.
(0021] In other embodiments the peptidic enhancer is a tail or terminal extension derived from a second hormone, such as exendin, human GLP-1, or frog GLP-1, or is empirically determined. In one embodiment the peptidic enhancer is a heterologous C-terminal tail or terminal extension to the GIP portion. As with the other Gil' hybrids described herein, in one embodiment of the peptidic-enhancer containing hybrid, the Gil' portion can be native GIP, an active fragment thererof, or their analogs or derivatives. In another aspect the GIP portion of the hybrid comprises at least one modification, substitution, deletion or addition that provides one or more enhanced properties, e.g. increased resistance to proteolytic digestion (thus prolonging half-life), fatty acyl derivitization that reduces renal clearai~ce. In one embodiment the tail comprises a Trp-cage motif sequence. In another embodiment the GIP analog polypeptide portion includes unnatural amino acids, such as a D amino acid, e.g. that inhibits to reduces the rate of proteolysis by DPP-LV.
[0022] The present invention also provides for the treatment and prevention of metabolic diseases and disorders, particularly those which can be alleviated by control of plasma glucose levels, insulin levels, and/or insulin secretion, such as diabetes and diabetes-related conditions. Such conditions and disorders include, but are not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind, including type 1, type 2, and gestational diabetes, diabetes complications (e.g. neuropathy (treating with a Gil' hybrid containing an exendin family hormone module for example), neuropathic pain (treating with GIP hybrids comprising an amylin family hormone module for example), retinopathy, nephropathy, conditions of insufficient pancreatic beta cell mass (based on, e.g., islet neogenesis actions of exendin-4 and GLP-1). Accordingly, provided are methods for treating or preventing such conditions, wherein the method comprises administering a therapeutically or prophylactically effective amount of a GIP or an analog or derivative thereof, including a novel GIP analog of the invention, or a GIP-hybrid of the invention, including one having a peptidic enhancer, to a subject in need thereof. In one embodiment the polypeptides of the invention can be provided as monotherapy. In another embodiment for treating diabetes or conditions associated with elevated glucose levels, the Gil' compound can be administered in adjunct therapy with a glucose lowering agents (e.g., antidiabetics) or agents or methods that inhibit or reduce gastric emptying. Examples of such agents are presented herein. For example, in one embodiment is provided an adjunct therapy method for reducing blood glucose levels of a subject, e.g., one having type 1, type 2 or gestational diabetes mellitus, comprising administering to the subject a therapeutically effective amount of an exendin or an exendin agonist, such as wherein said exendin agonist is a peptide, in adjunct therapy with a Gil' or novel GIP analog of the invention, or an effective amount of a GIP-exendin hybrid.
[0023] Compounds of the invention, alone or in combination with a glucose lowering agent (e.g., antidiabetics) or with agents or methods that inhibit or reduce gastric emptying, can also be useful for potentiating, inducing, enhancing or restoring glucose responsivity in pancreatic islets or cells. These actions may also be used to treat or prevent conditions associated with metabolic disorders such as those described above and in U.S. Patent Application No. U520040228846, incorporated herein by reference in its entirety.
[0024] In another aspect methods for treating or preventing obesity are provided, wherein the method comprises administering a therapeutically or prophylactically effective amount of a GIl' or an analog or derivative thereof, including a novel Gil' analog of the invention, or a GIP-hybrid of the invention, including those having a peptidic enhancer, to a subject in need thereof. In one embodiment, the subject is an obese or overweight subject. While "obesity" is generally defined as a body mass index over 30, for purposes of this disclosure, any subject, including those with a body mass index of less than 30, who needs or wishes to reduce body weight is included in the scope of "obese." Subjects who are insuli,~ resistant, glucose intolerant, or have any form of diabetes mellitus (e.g., type 1, 2 or gestational diabetes)
can benefit from this method. Compounds of the invention can also be useful in treating or preventing other conditions associated with obesity including stroke, cancer (e.g.,. endometrial, breast, prostate, and colon cancer), gallbladder disease, sleep apnea, reduced fertility, and osteoarthritis, (see Lyznicki et al, Am. Fain. Phys. 63:2 185, 2001). Where conditions are associated with elevated glucose or hyperglycemia, the method comprises administering a therapeutically or prophylactically effective amount of a GIP compound, alone or in combination with a glucose lowering agent (e.g., antidiabetic) or agent or method that inhibits or reduces gastric emptying.
[0025] In yet another aspect, Gil' compounds, particularly Gil' hybrids of the invention, can be used in methods of reducing food intake, reducing appetite, inducing satiety, reducing nutrient availability, reducing caloric efficiency, causing weight loss, affecting body composition, altering body energy content or energy expenditure, and improving lipid profile (including reducing LDL cholesterol and triglyceride levels and/or changing HDL cholesterol levels) wherein the methods comprise administering to a subject an effective amount of a Gil' compound, particularly a Gil' hybrid compound, of the invention. In one embodiment, the methods of the invention are used to treat or prevent conditions or disorders which can be alleviated by reducing nutrient availability in a subject in need thereof, comprising administering to said subject a therapeutically or prophylactically effective amount of a Gil' compound of the invention. Conditions and disorders include, but are not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind, including type I, type 2, and gestational diabetes, diabetes complications (neuropathy (based on, e.g., neurotrophic actions of exendin-4), neuropathic pain (based on, e.g., amylin action), retinopathy, nephropathy, conditions of insufficient pancreatic beta cell mass (based on, e.g., islet neogenesis actions of exendin-4 and GLP-1). Where conditions are associated with elevated glucose or hyperglycemia, the method comprises administering a therapeutically or prophylactically effective amount of a Gil' compound, alone or in combination with a glucose lowering agent (e.g., antidiabetic) or agent or method that inhibits or reduces gastric emptying.
[0026] In addition to the amelioration of hypertension in subjects in need thereof as a result of reduced food intake, weight loss, and/or treating obesity, compounds of the invention may be used to treat or prevent hypotension and conditions associated therewith.
[00271 In another aspect Gil' analogs and hybrids are useful for decreasing or inhibiting bone resorption and maintaining or increasing bone density. When combined with an appropriate second hormonal module, Gil' hybrids are useful to treat these conditions as well as decreasing plasma calcium, and/or
inducing an analgesic effect, particularly to treat bone disorders such as osteopenia and osteoporosis, an~ treating painful neuropathy. In one embodiment such hybrids contain an exendin, GLPI, amylin and/or
sCT portion. For example, a GIP-sCT or GIP-amylinlsCT hybrid compound of the invention can have a selectable property of a salmon calcitonin or amylin/sCT/Amylin chimera, such as decreasing bone loss and bone resorption or reducing cartilage turnover (chondroprotection), and a property of a Gil', such as plasma glucose lowering (concomitant with an anabolic aspect as described herein) and/or inhibiting bone resorption and maintaining or increasing bone density. A Gil' hybrid with such selectable properties can enhance treatment of osteoporosis or conditions of high cartilage turnover, particularly in those who can also benefit from glycemic control, such as subjects with diabetes or under going critical care.
[0028] Gil' compounds, particularly GIP analogs, extended half-life GIl' hybrids (e.g. DPP-IV cleavage resistant (such as a D-Ala2, N-Acetyl or N-pyroglutamyl analogs) optionally further comprising a peptidic enhancer such as a heterologous C-terminal tail, and GIP hybrids comprising other hormone modules known to provide beneficial cardiovascular effects, are useful to treat cardiovascular disease and related conditions. As demonstrated herein Gil' compounds increase cardiac contractility (dp/dt), decrease blood pressure (for example by acute vasodilatation), decrease systolic pressure, decrease diastolic pressure, and can provide a direct beneficial action on cardiac cells. GIl' compounds also improve cardiac function via metabolic actions, e.g. glucose lowering, insulin secretion, beta cell proliferation. However, by also providing direct effects on cardiovascular system, the Gil' compounds are surprisingly even more beneficial.
[00291 Compounds of the invention can also be useful in the treatment or prevention of any number of gastrointestinal disorders that are associated with excess gastric secretion, excess intestinal electrolytes and water secretion as well as decreased absorption, e.g., infectious (e.g., viral or bacterial) diarrhea, inflammatory diarrhea, short bowel syndrome, or the diarrhea which typically occurs following surgical procedure, e.g., ileostomy (see e.g., Harrison's principles of Internal Medicine, McGraw Hill Inc., New York, 12th ed.). Examples of infectious diarrhea include, without limitation, acute viral diarrhea, acute bacterial diarrhea (e.g., salmonella, campylobacter, and clostridium) or diarrhea due to protozoal infections, or travellers diarrhea (e.g., Norwalk virus or rotavirus). Examples of inflammatory diarrhea include, without limitation, malabsorption syndrome, tropical spue, chronic pancreatitis, Crohn' s disease, diarrhea, and irritable bowel syndrome. Gil' and Gil' compounds of the invention can be used to treat or prevent an emergency or life-threatening situation involving a gastrointestinal disorder, e.g., after surgery or due to cholera. Furthermore, the compounds can be used to treat intestinal dysfunction in patients with Acquired Immune Deficiency Syndrome (AIDS), especially during cachexia. The compounds may also be useful for inhibiting small intestinal fluid and electrolyte secretion, and augmenting nutrient transport, as well as increasing cell proliferation in the gastrointestinal tract, regulating lipolysis in, e.g., adipase tissue and regulating blood flow in a mammal. Gil' compounds of the invention may also be useful for treating or preventing the above conditions by their gastrointestinal protective activity (e.g., inhibition of gastric secretion). Accordingly, a Gil' compound of the invention may be used to treat gastrointestinal or
muscosal damage. Exemplary types of damage include, but are not limited to, inflammatory bowel disease, bowel atrophy, conditions characterized by loss of bowel mucosa or bowel mucosal function, and other conditions of the gastrointestinal tract, including those which may be brought about by exposure to cytotoxic agents, radiation, toxicity, infection and/or injuly. Moreover, these compounds of the invention may be combined with analgesics, anti-inflammatory agents, growth hormone, heparin, or any other therapies that may be used to treat inflammatory bowel disease or other conditions listed above.
[0030] In another embodiment Gil' compounds can be useful for treating or preventing gastritis, pancreatitis, Barrett's esophagus, Gastroesophageal Reflux Disease (GERD) and conditions associated therewith. Such conditions can include, but are not limited to, heartburn, heartburn accompanied by regurgitation of gastric/intestinal contents into the mouth or the lungs, difficulty in swallowing, coughing, intermittent wheezing and vocal cord inflammation (conditions associated with GERD), esophageal erosion, esophageal ulcer, esophageal stricture, Barrett's metaplasia (replacement of normal esophageal epithelium with abnormal epithelium), and pulmonary aspiration. Gil' compounds can have antisecretory properties, such as inhibition of gastric acids, inhibition of bile acids, and inhibition of pancreatic enzymes. Moreover, GIP compounds can also have gastroprotective effects. Accordingly, GIP compounds of the invention may be particularly useful in the treatment or prevention of gastritis, pancreatitis, Barrett's esophagus, and/or GERD and related or associated conditions.
[0031] The present invention also relates to pharmaceutical compositions comprising a therapeutically or prophylactically effective amount of at least one novel Gil' analog or GJP hybrid polypeptide of the invention, or a pharmaceutically acceptable salt thereof, together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers useful in the delivery of the GIP compound.



[00321 These and other aspects of the invention will be more clearly understood with reference to the following embodiments and detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS

[0033] Figures lA and lB present views of the exendin-4 Trp-cage. Figure IA shows an NMR-derived ensemble structure of exendin-4 in 30% aqueous trifluoroethanol. The Trp-cage can be seen folding Wack towards the central region of exendin. Figure lB shows a CPK view of the 'Trp-cage" motif (residues 21-38) of a representative structure from the solution-state NMR structure ensemble of exendin-4.
[0034] Figure 2 presents sequences and receptor binding and glucose lowering (oral glucose tolerance test (OGiT) in non-diabetic NIH/Swiss mice) activities of reference sequences and novel GIP analog sequences of the invention (SEQ ID NOS 2, 413,3, 293, 414, 294, 415, 295-296, 416, 297, 417, 298, 291-292, 418-419 & 299 are disclosed respectively in order of appearance).
[0035] Figures 3A and 3B present suppression of glucose excursion following an 0011', and basal glucose lowering activity of GIP and Compound G in NIH/Swiss mice. In Figure 3A, bars represent mean ± sd, n=6- 10. Peptide was injected IP at t=-5 into overnight-fasted NIH/Swiss mice. Gavage (1 .Sglkg) was given at t=0. Sample was taken at t=30 mm. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA) * p [0036] Figure 4 presents a comparison of the glucose lowering action of Compound 0 with full length
GIP and exendin4 in diabetic db/db mice. Points represent mean ± sem. n= 6-10. Peptide was injected
II' at t=0 immediately following baseline sample into 2-hour fasted db/db mice. Samples were taken at
60, 120, and 180 mm. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson
& Johnson Company, Milpitas, CA) * p [0037] Figures 5A-SD present examples of Gil' phybrids comprising Gil' and amylin/calcitonin-like analog peptides. The compounds would have both GIP activity (e.g., glucose lowering) and reduction of gastric emptying (SEQ ID NOS 100-182 are disclosed respectively in order of appearance).

[0038] Figure 6 depicts the structures of various chemical groups mentioned herein.
[00391 Figure 7 depicts the structures of various chemical Fmoc derivatives mentioned herein.
[0040] Figures 8A and 8B depict glucose lowering effect of novel Gil' analogs. The figures demonstrate that a D-alanine substitution at position 2 in the analogs herein improves glucose lowering ability. Points represent mean ± sem. Peptide was injected lIP at t=0 immediately following baseline sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60, 120, 180 and 240 minutes. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA). pO.O5 vs. vehicle control; ANOVA, Dunnett' s test.
[0041] Figure 9 depicts glucose lowering effect of novel GIP analogs, particularly the effect of a Trpcage. Points represent mean ± sem. Peptide was injected Il' at t=0 immediately following baseline
sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60, 120, 180 and 240mm. Bloo,,

glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA). p005 vs. vehicle control; ANOVA, Dunnett's test.
[0042] Figures 1OA and lOB depict glucose lowering effect of various analogs. In this example, Ac modification and a Pro3 substitution did not significantly enhance glucose lowering ability. Points represent mean ± sem. Peptide was injected 1? at t=0 immediately following baseline sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60, 120, 180 and 240mm. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA). po.05 vs. vehicle control; ANOVA, Dunnett's test.
[0043] Figure 11 demonstrates superior protease resistance of an exemplary GIP hybrid, 0601 GIP3 794 versus a native Gil'.
[00] Figures 1 2A- 1 2AT depict further exemplary analogs and reference peptides of the invention. It is intended that the various modifications and variants shown are to be used in the present invention, and may be combined as discussed herein. For example, the terms exendin tail or exendin trp-cage motif includes any of the exendin tail variants depicted, which are useful as shield sequences (peptidic enhancers). Of further interest are the frog GLP1 C-terminal extensions as shown in the figures, which are yet another example of a shield sequence that can be used in place of an exendin tail (SEQ ID NOS 307-412 are disclosed respectively in order of appearance).
[0045] Figures 13A and 13B demonstrate food intake inhibtion and lowering of blood glucose by Gil' hybrids.
[0046] Figure 14. Effect of Gil' hybrids in food intake assay.
[0047] Figure 15. Effect of GIP hybrid in food intake assay.
[0048] Figures 16A and 16B demonstrate effect of Compound 10 (Fig. 16A) and sCT (Fig. 16B) in food intake assay.
[0049] Figure 17 demonstrates effect of Gil' hybrids on lowering of blood glucose.
[00501 Figures 1 8A and 1 8B depict cAMP production in whole cardiomyocytes from receptor activation in response to varying doses of test compound, human Gil'( 1-42) free acid form (Figure 1 8A) and Gil' hybrid compound G (Figure 18B).

[0051] Figures 19A-E depict the response of mean arterial pressure (Figure 19A), heart rate (Figure 19B), and rate of change in blood pressure (dP/dt, Figure 19C), systolic pressure (Figure 19D) and diastolic pressure (Figure 19E) as determined by telemetry in conscious rats to administration of GIl' compounds. Mean aiterial pressure is presented as % of predose values measured over the 30 minutes prior to drug administration. Figure 19C reflects the inotropic response to Gil' compounds. The rate of change of blood pressure (dP/dt) is indicative of cardiac contractility.
[0052] Figures 20A, 20B and 20C depict the lack an acute effect of GIP(I-42) and a GIl' DPP-lVresistant-analog/exend in-tail hybrid on food intake in contrast to exendin-4. The pancreatic hormone amylin produced a significant effect as expected.
[0053] Figure 21 depicts the lack of effect on weight loss in diet-induced obesity mice, in contrast to the effect of exendin-4.
[0054] Figure 22 provides an alignment of mamalian and non-mammalian Gil'. Positions Y1, E3, D9, SiI, D15, F22, V23, L26, L27 and K32 are conserved across all species (SEQ ID NOS 13, 420, 10, 12, 300-304, 2, 305, 11 & 306 are disclosed respectively in order of appearance).
[0055] Figure 23A and B depict beneficial activity of a GIP-amylin/sCT/amylin hybrid in slowing of gastric emptying and reducing intracellular calcium levels.
DETAILED DESCRIPTION OF THE INVENTION
[0056] Gastric inhibitory polypeptide (GIl') and glucagon-like peptide 1 (GLP-I) are gut peptide hormones that exert potent glucoregulatory action through their glucose-dependant stimulation of insulin secretion. Consequently, these incretin hormones have attracted great interest as potential anti-diabetic agents with reduced risk for hypoglycemia. Whereas GLP- 1, GLP- I analogs and mimetics have been shown to be efficacious in controlling glucose levels in type 2 diabetic patients, the insulinotropic effect of Gil' is reportedly significantly reduced in diabetic subjects, compared to normal individuals (16-18). The preservation of insulinotropic action of GLP- 1 but not of GIP in the same diabetic subjects suggests that GIP signal transduction is impaired in type 2 diabetes. Reduced Gil' receptor expression in pancreatic beta cells has been proposed to contribute to overall reduced incretin effects in diabetic subjects (19). Despite the reduced insuliiiotropic response to Gil' in subjects with type 2 diabetes, it is possible that administration of elevated pharmacological doses of Gil' or analogues could have therapeutic utility. Of note, Gil' lacks the gastrointestinal effects of GLP- 1 (20) that has limited the latter peptide' s therapeutic window, thus permitting the possibility of higher dosing regimens (21).
[0057] One of the major hurdles in the therapeutic development of these incretin hormones is their short duration of action due to enzymatic degradation in vivo. The enzyme dipeptidyl peptidase IV (DPP-IV) plays a key role in the N-terminal cleavage of the peptides in vivo, and more recently, neutral endopeptidase 24.11 (NEP) has also be implicated in their degradation (22-26). Several studres have reported greater in vivo efficacy of DPP- lV resistant Gil' analogues in rodent diabetic models (27-28).


[0058] Provided herein are novel Gil' analogs and GIP-containing hybrid polypeptides, or derivatives thereof, which have enhanced or novel properties, including enhanced DPP-IV resistance, dual hormonal activity, and improved plasma half-life. Also provided are methods for treating or preventing metabolic diseases and disorders including those which can be alleviated by control of plasma glucose levels, insulin
levels, and/or insulin secretion, such as diabetes and diabetes-related conditions, and conditions and~
disorders including, but not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind, including type 1, type 2, and gestational diabetes. The methods comprise administering a therapeutically or prophylactically effective amount of a Gil' or Gil' analog, fragment or derivative thereof or a novel GIP analog or Gil' hybrid or derivatives thereof as described herein, alone (monotherapy) or in combination with another agent or therapy (adjunct therapy), for example a glucose lowering agent (e.g., antidiabetic) or agents or methods that inhibit or reduce gastric emptying (examples of such agents are presented herein), to a subject in need thereof.
[0059] In addition to novel GIl' analogs and derivatives, the present invention relates to novel, Gil'-containing selectable hybrid polypeptides useful as agents for the treatment and prevention of metabolic diseases and disorders which can be alleviated by control of plasma glucose levels, insulin levels, and/or insulin secretion, such as diabetes and diabetes-related conditions. Such conditions and disorders include, but are not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind, including type 1, type 2, and gestational diabetes.
[0060] In one aspect, the invention involves the modular assembly of physiologically, metabolically, and/or pharmacokinetically active peptidic modules that may be selectable based on "bio-activities", e.g., therapeutic efficacy, scope of function, duration of action, physicochemical properties, and/or other pharmacokinetic properties.
[0061] Without intending to be limited by theory, the present invention relates at least in part to a "toolbox" approach, wherein bio-active peptide hormone modules are linked in binary, tertiary or higher order combinations to create novel, efficacious therapeutic agents with selectable properties. The "bioactive peptide hormone modules" may be peptide hormones, peptide fragments with hormonal activity, or structural motifs of peptide hormones that impart chemical, metabolic, and/or other pharmacokinetic stability. The peptide hormones can include native peptide hormones, as well as peptide hormone analogs and derivatives, as known in the art and described herein.
[0062] In one aspect of the invention, it has been found that the combination of certain physicochemical characteristics of two or more peptide hormones into a single modality can facilitate intervention at several points in a dysfunctional metabolic circuit. As such, in one aspect of the invention, rationallydesigned hybrid polypeptides are provided that integrate selectable bio-activities into a single polypeptide agent. In one embodiment, the selectable hybrid polypeptides of the invention may involve the use of chemically stable linkers to covalently attach the bio-active modules. In another embodiment, the selectable hybrid polypeptides of the invention may involve the use of cleavable linkers, which themselves may be or form part of a bio-active module.
[0063] Again, without intending to be limited by theory, design of the hybrid polypeptides of the present invention may generally involve: (1) the identification, selection and pairing of bio-active peptide hormone modules for desired efficacy and therapeutic use, and (2) the covalent linking of the bio-active modules (e.g. native peptide hormones, peptide hormone analogs or derivatives with hormonal activity, peptide hormone fragments with hormonal activity, stabilizing motifs, etc.) either directly or via a linker without loss of bio-activity of the component modules. In certain embodiments, module selection criteria may include, but not be limited to: (a) desired in vivo efficacy for desired therapeutic or prophylactic indication, such as an additive or a synergistic effect; (b) optional synergism or dual action of the linked modules for multiple therapeutic or prophylactic indications; and/or (c) a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic.
[0064] Gil'. Gil' Analogs and Novel GIP Analogs. (The section headings are used herein for organizational purposes only, and are not to be construed as in any way limiting the subject matter described.) Reference sequences include human Gil', truncated GIP, human GLP-1, exendin4, an exemplary Trp-cage, and exemplary "shield" sequences (e.g.a short and a long "exendin tail"):
(Table Removed)
[0065] Useful in the therapies disclosed herein and in the novel GIP hybrids disclosed herein, are native Gil' peptide hormones, and functional peptide analogs and derivatives4hereof. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known Gil' peptide that exhibits hormonal activity known in the art may be used either as a component of a novel Gil' analog or hybrid herein or in the novel adjunct therapies disclosed herein. In one embodiment, the Gil' peptide analogs and derivatives have at least one hormonal activity of a native Gil' peptide. In certain embodiments, the Gil' peptide analogs are agonists of a receptor that a native Gil' peptide is capable of specifically binding. Exemplary GIP peptide analogs and derivatives include those described in the references herein, which are hereby incorporated by reference. While the present application describes GIP polypeptide compounds as GIP analogs, novel Gil' analogs, and novel Gil' hybrids for use in the methods and therapies described herein, it is further intended that any suitable GIP agonist can be administered in place of a GIP compound, such agonists include agonist antibodies and
antibody fragments and derivatives, and small molecule Gil' receptor agonists. Accordingly, when a GIP compound or polypeptide is indicated for use in a particular therapeutic method, in another embodiment it is intended that a GIl' agonist be used, particularly an agonist antibody or fragment or derivative thereof.
[0066] In serum. Gill' is degraded by dipeptidyl peptidase IV (DPl'-IV). The resulting short biological half-life (about 2 minutes in vivo) limits the therapeutic use of Gil'.

[0067] The following references relate to various GIP analogs that are useful to provide as compone.nts for the novel Gil' analogs and Gill' hybrids of the present invention and find use in the novel therapies of the present invention based on their function on various target organs.
[0068] German Patent Application 19921537 discloses a method for extending the survival of insulin producing beta-cells by stimulation of their proliferation and prevention of their programmed cell death. The specific goal is to increase the endogenous insulin content and insulin response to elevated blood glucose levels. An important component of this invention is the activation of protein kinase B/Akt in insulin producing beta-cells in response to the administration of effectors such as GLP- I, Gil', Exendin-4 or GLl'- 1 receptor agonists or GIP-receptor agonists.
[0069] European Patent Application 0479210 discloses Gil' analogs of the formula GIP(1-13)-X-GIP(15-30)-Y, wherein X is an amino acid residue other than Met, and Y is selected from homoserine (inclusive homoserine-lactone) (referred to as "Hse"), homoserine amide (Hse-NH2), H-Gly-Lys-Lys-Asn-Asp-TrpLys-His-Asn-Ile-Thr-Gln-Hse (SEQ ID NO: 8) or H-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-Ile-ThrGln-Hse-NH2 (SEQ ID NO: 9).
[0070] United States Patent Application 20030232761 by Hinke et al, published December 18, 2003, reports C-terminal truncated fragments and N-terminal modified analogs of Gil' as well as various GIl' analogs with a reduced peptide bond or alterations of the amino acids close to the dipeptidyl peptidase IV (DPl'-IV)-specific cleavage site providing DPP-IV-resistance and prolonged half-life. Also reported are analogs with different linkers between potential receptor binding sites of Gil'.
[0071] W098/24464 discloses an antagonist of glucose-dependent insulinotropic polypeptide (Gil') consisting essentially of a 24 amino acid polypeptide corresponding to positions 7-30 of the sequence of GIP, a method of treating non-insulin dependent diabetes mellitus and a method of improving glucose tolerance in a non-insulin dependent diabetes mellitus patient.
[0072] WO 00/58360 and EPi 171465 disclose peptides. which stimulate the release of insulin. This disclosure provides a process of N tenninally-modifying GIl' and the use of the peptide analogues for
treatment of diabetes. The specific peptide analog, which is disclosed in this invention, comprises at leasi
15 amino acid residues from the N terminal end of Gil' (1-42). In another embodiment, Tyrl-glucitol GIl' (1-42) is disclosed.
[0073] WO 00/20592 discloses GIl' or anti-idiotypic antibodies of GIP or fragments thereof as GIPanalogs for maintaining or increasing bone density or bone formation.
[0074] Kuhn-Wache et al. (2000) discloses analogs of GIP with increased dipeptidyl peptidase IV resistance (Kuhn-Wache et al, in Langner & Ansorge, Cellular peptidases in Immune Functions and Diseases 2. Kluwer Academic/Plenum Publishers, 187-195.)
[0075] O'Harte et al. and Ghault et al. have reported GIl'(1-30) analogs--Tyrl -glucitol-GIP arid (l'ro3)GIP-displaying DPP-IV resistance and enhanced bioactivity. (O'Harte et al., NH2-terminally modified gastric inhibitory polypeptide exhibits amino-peptidase resistance and enhanced antihyperglycemic activity, Diabetes 48, 758-765 (1999)); and see Gault et al. "Characterization of the cellular and metabolic effects of a novel enzyme-resistant antagonist of Glucose-dependent insulinotropic polypeptide." Biochemical and Biophysical Research Communications 290, 1420-1426 (2002)).
[0076] Specific active Gil' and GIP analogs known in the art include:
(Table Removed)
[0077] Of particular interest are analogs modified at or near the dipeptidyl peptidase IV (DPP-IV) specific cleavage site, which improve DPP-IV-resistance and consequently prolong half-life. Amino acid alterations include modifications of the first 3 or 4 residues of GIl' and/or the bond between residues 2 and 3, which is cleaved by DPl'-lV. Modifications and substitutions include N-terminal modifications, Lamino acids, D-amino acids, proteinogenic and non-proteinogenic amino acids. Proteinogenic amino acids are defined as natural protein-derived alpha-amino acids. Non-proteinogenic amino acids are defined as all other amino acids, which are not building blocks of common natural proteins.
[0078] Of further interest are novel GIl' analogs having one or more modifications as described herei'~ and that exhibit at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least
85%, at least 90%, at least 95% or at least 98% sequence identity to a native GIP(1-30), native GIP(1-26), native GIP(1-14), native GIP(1-39), native Gil'(19-30), native GIP(19-26), native GIP(19-39), native GIP( 19-42) or native Gil'( 1-42) over the entire length of the Gil' portion.
[0079] Of particular interest are those Gil' compounds comprising at least 11, 12, 13, 14 or-i5 amino acid residues from the N-terminal end of a Gil', e.g., GIP(1-42); having a least one amino acid substitution or modification at position 1-3 and being biologically active. This includes modification by fatty acid addition at an epsilon amino group of at least one lysine residue, either present in or introduced into the molecule. Particular modifications include Tyr 1 -glucitol of a GIP, for example Tyr 1 -Glucitol GIP( 1-42) or Tyri-glucitol GLP(1-30). GIP analogs of interest include those comprising a substitution or modification selected from the group comprising D-amino acid substitutions in 1, 2 and/or 3 positions and/or N-terminal glycation, alkylation, acetylation or acylation, or other N-terminal modifications described herein. Of further interest are analogs wherein the amino acid in the 2 or 3 position is substituted by lysine, serine, 4-amino butyric amino acid, Aib, D-alanine, Sarcosine or proline. Further exemplary subsitutions in the 1, 2, or 3 position, and more particularly in the 2 position of Gil' are dAla, Val, dnorVal, dSer, Abu, dAbu, homo-Ser, d-homoSer, dPro, cyclopropyl Ala, d-cyclopropyl Ala, cycloHexyl Ala, d-cyclohexyl Ala, A(NMe), Aib, and cyclpropGly.
[0080] Further exemplary Gil' analog compounds have a modification at the N-terminus, retaining their GIP Receptor binding, in which the N-terminus modification includes H, isocap, isoBuOCO, octylglycine, Y(NMe) and succinoyl. Further exemplary GIP compounds include those with fatty acid modifications or combinations of modifications as described herein, while retaining their GIP Receptor binding and receptor activation activity, For example, an N-terminus modification can be combined with a substitution or mdofication at positions 1, 2 or 3 (which imparts resistance to DPP-IV as described herein) or with a fatty acyl deriviative modification (which can reduce renal clearance). In another example, a substitution or modification at at positions 1, 2 or 3 is combined with a fatty acyl derivative. For example an N-terminus octylglycine is combined with a d-amino acid at 1, 2, or, particularly a D-Ala at positon 2. In one embodiment a fatty acyl substitution is a octylglycine for lysine at position 16 or methionine at position 14. In other embodiments the methionine at positon 14 is deleted, and which, for example, can be further combined with a octylglycine for lysine at position 16 or 30. Another subsitution is acylation of the lysine at position 16 or 30, for example with an octyl or palmitoyl group. Other embodiments include a fatty acyl substitution where an octylglycine is substituted for lysine at position 16 or 30 or for methionine at position 14. To eliminate or reduce oxidation, the methionine at position 14 is deleted or substituted, for example with a lecince or other small hydrophobic amino acid, and/or the tryptophan at position 25 is deletred or substitutired, for example with a phenylalanine.
[0081] In one embodiment are analogs having at least one or two amino acid deletions in amino acids
30 of GIP, those having one or two deletions in amino acids 4 to 30, and those having one or more
deletions in amino acids 4-15, and those having one amino acid deletion in amino acids 1-30, 4-30 or 4-15 of a Gil'. Of course it is intended that such a modification can be combined with at least one other change as described herein, such as a change that imparts Dl'l'-IV resistance, reduces or eliminates oxidation, reduces renal clearance, improves receptor binding, or improves receptor activation.
[0082] Further exemplary substitutions are those derived from GIl' of other (non-human) species, for example the methionine 14 replaced by lecuine, the D at position 9 or 21 replaced by E, the histidine 18 replaced by alanine, arginine or lysine, the lysine at position 30 replaced by alanine, arginine or histidine, the alanine at position 13 replaced by leucine, and the alanine at position 28 replaced by serine. -
[0083] In one embodiment the Gil' analogs have one or more of the following modifications: dAla2 to Abu, Ala, Gly, or Ser; Metl4 to Leu; Hisl8 to Ala, Mg, or Lys; Asp2l to Glu; Lys3O to Arg or His; and/or an N-terminus as Gly(Oct).
[0084] Further exemplary Gil' modifications and combinations are shown in the following compounds:
(Table Removed)
[0085] Accordingly, it is intended that the modifications described herein can be combined with at least one other change as described herein, such as a change that imparts Dl'P-IV resistance, reduces or eliminates oxidation, reduces renal clearance, improves receptor binding, or improves receptor activatior, For example, intended are specific analogs that have one or more replacements or modificationsT as
decribed herein, such as a GIP D-Ala2 or L-Ala2 analog that also has a Phe for Trp replacement at position 25.
GIP Hybrid l'olv~eptides.
[0086] Bio-Active PeDtide Hormone Modules. As discussed herein the Gil' hybrid polypeptides of the present invention (also referred to as "phybrids"), generally comprise at least two bio-active peptide hormone modules co~'alently linked together, with a Gil' peptide as one of the modules. The bio-active peptide hormone modules may be: (a) native component peptide hormones, (b) analogs or derivatives of native component peptide hormones that retain hormonal activity, (c) fragments of native component peptide hormones that retain hormonal activity, (d) fragments of analogs or derivatives of native component peptide hormones that retain hormonal activity, (e) structural motifs of native component peptide hormones that impart a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic to the hybrid polypeptide; or (17) structural motifs of analogs or derivatives of native component peptide hormones that impart a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic to the hybrid polypeptide. The structural motifs of (e) and (1) will collectively be referred to herein as "peptidic enhancers". An example of a peptidic enhancer is a Trp cage sequence, particularly one derived from exendin-4, such as the Ex-4 short or long tails.
[0087] Xemplary bio-active peptide hormone modules include native peptide hormones selected from:
amylin, ADM, CT, CGRP, intermedin, CCK(1-33), CCK-8, leptin, l'YY(1-36), PYY(3-36), GLP-1(l-37), GLP-1(7-37), GLl'-l(7-36), GLl'-2, OXM, Gil', exendin-3, exendin-4, natriuretic peptide hormones, urocortin family peptides, e.g., Ucn-2 and Ucn—3, neuromedin family peptides, e.g. neuromedin U25 or splice variants, and ANP, BNP, CNP or urodilatin.
[0088] Other xemplary bio-active peptide hormone modules include analogs and derivatives of a component peptide hormone selected from: amylin, ADM, Cr, CGRP, intermedin, CCK, leptin, PYY(l36), PYY(3-36), GLl'-1(1-37), GLP-1(7-37), GLl'-1(7-36), GLP-2, OXMA a natriuretic peptide hormone, a urocortin family peptide, e.g., Ucn-2 and Ucn—3, a neuromedin family peptide, e.g. neuromedin U25 or splice variants, exendin-3, and exendin-4, wherein the analog or derivative exhibits at least one hormonal activity of the component peptide hormone. The analog may comprise one or more insertions, deletions, or substitutions of the amino acid sequence of the component peptide hormone, and the derivative may comprise one or more chemical modifications of an amino acid residue of an analog or component peptide hormone, as described more fully herein and known in the art.
known in the art. Particularly xemplary analogs and derivatives that exhibit at least one hormonal activity useful as bio-active peptide hormone modules of the invention include the following:
(Table Removed)
[0090] As known in the art, such peptide compounds may preferably be amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.

[00911 Still other xemplary bioactive peptide hormone modules include fragments of a component peptide hormone selected from: amylin, ADM, CT, CGRP, intermedin, CCK, leptin, l'YY(1-36), l'YY(3-36), GLl'-l(1-37), GLl'-1(7-37), GLl'-1(7-36), GLl'-2, OXM, a natriuretic peptide, a urocortin family peptide, e.g., Ucn-2 and Ucn—3, a neuromedin family peptide, e.g. neuromedin U25 or splice variant, exendin-3, and exendin-4, wherein the fragment exhibits at least one hormonal activity of the component peptide hormone.
[0092] Yet other xemplary bioactive peptide hormone modules include fragments of analogs or derivatives of a component peptide hormone selected from: amylin, ADM, CT, CGRP, intermedin, CCK, leptin, PYY(1-36), l'YY(3-36),GLP-1(l-37), GLl'-1(7-37), GLl'-l(7-36), GLP-2, OXM, AN?, BNP, CNP, urodilatin, Ucn-2 and Ucn—3, neuromedin U25 or splice variant, neuromedin 5, exendin-3 and exendin-4, wherein the fragment exhibits at least one hormonal activity of the component peptide hormone. Again, the analog may comprise one or more insertions, deletions, or substitutions of the amino acid sequence of the component peptide hormone, and the derivative may comprise one or more chemical modifications of an amino acid residue of an analog or component peptide hormone, as described more fully herein and known in the art.
[0093] Certain exemplary fragments that exhibit at least one hormonal activity include the following.' However, it should be understood that combinations of the above-described analogs and derivatives taken with fragments known in the art, including the xemplary fragments described herein, are contemplated.
(Table Removed)
[0094] Again, as known in the art, such peptide compounds may preferably be amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified. Further the above exemplary fragments may be combined with any of the analogs or derivatives discussed herein
or known in the art. For example, exemplary analog fragments may include 5Ala,'4Leu,~Phe-exendin-
4(1-28), Leu, Phe-exendin-4( 1-27), 5Ala,14Leu,25l'he-exendin-4( 1-28), Leu, l'he-exendin-4( 1-27), or any other combinations of the disclosed fragments, analogs, and derivatives. Furthe embodiments include

NN221 1 and Zl'-10.
[0095] Yet other exemplary bio-active peptide modules include "peptidic enhancer", i.e., structural motifs of component peptide hormones (including analogs and derivatives thereof) that impart a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, and/or other pharmacokinetic characteristic to the hybrid polypeptide. Exemplary peptidic enhancers include the following.
(Table Removed)
[0096] Again, it should be understood that combinations of the above-described Gil' analogs and derivatives taken together with the bio-active peptide modules described herein are contemplated. For example, the last six amino acid residues of amylin family peptide hormone analogs and derivatives known in the art and/or described above are also contemplated as exemplary bio-active peptide modules. For example, as further discussed herein, the peptidic enhancer Ex-4 short tail, which is an exemplary Trp-Cage sequence, or analog thereof, is added to the C-terminus of any Gil' analog, and in further embodiments the peptidic enhancer is attached using a linker.


[0097] In one aspect, the novel Gil' hybrid include a GIP portion exhibiting at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to a native GIP(1-30), native GIP(1-26), native GIP(1-14), native GLP(1 -39), native GIP(19-30), native GIP(19-26), native GIP(19-39), native GIP(19-42) or native GIl'(1-42) over the entire length of that Gil' portion.
[0098] Accordingly, in certain embodiments the Gil' portion of a Gil' hybrid can comprise a trp-cage motif. Such desirable GIP hybrids include an N-terminal Gil' or novel Gil' analog fragment in combination with a C-terminal polypeptide or fragment thereof having a glucose lowering activity (e.g., antidiabetics, exendin) or the ability to inhibit or reduce gastric emptying. Such desirable Gil' hybrids include an N-terminal Gil' fragment or novel Gil' analog fragment in combination with a C-terminal exendin, GLl'1, symlin (pramlintide), amylin, CCK, gastrmn, PYY, secretin, GRP, neuromedins, urocortin, calcitonin, or salmon calcitonin, or fragment thereof. In other embodiments desirable GIl' hybrids include a C-terminal GIl' or novel Gil' analog fragment in combination with an N-terminal polypeptide or fragment thereof having a glucose lowering activity (e.g., antidiabetics, exendin) or the ability to inhibit or reduce gastric emptying. In such embodiments, the chimeric polypeptides can include a C-terminal GIl', a novel Gil' analog (in which case a Trp-cage forming sequence is present), or fragment thereof, in combination with a N-terminal exendin, GLP1, symlin (pramlintide), amylin, CCK, gastrin, PYY, secretin, GRP, neuromedins, urocortin, calcitonin, or salmon calcitonin, or fragment thereof.
[0099] In other embodiments the Gil' or novel GD' analog is combined with a gastrin /CCK receptor ligand; an amylin receptor ligand; a calcitonin receptor ligand; an CGRP receptor ligand, a PYY receptor ligand, an EGF receptor ligand; a Glucagon-like peptide I receptor ligand; a Glucagon-like peptide 2 receptor ligand; a gastric inhibitory polypeptide (GIl') receptor ligand; a keratinocyte growth factor (KGF) receptor 1 ligand; a dipeptidyl peptidase IV inhibitor; a REG protein receptor ligand; a Growth Hormone receptor ligand; a Prolactin (PRL) receptor ligand; an Insulin-like Growth Factor (IGF) receptor ligand; PTH-related protein (l'THrP) receptor ligand; hepatocyte growth factor (HGF) receptor ligand; a bone morphogenetic protein (BMP) receptor ligand, a transforming growth factor (TGF receptor ligand; a laminin receptor ligand; a vasoactive intestinal peptide (VIP) receptor ligand; a fibroblast growth factor (FGF) receptor ligand; a nerve growth factor (NGF) receptor ligand; an islet neogenesis associated protein (INGAP) receptor ligand; an Activin-A receptor ligand; a vascular endothelial growth factor (VEGF) receptor ligand; an erythropoietin (EPO) receptor ligand; a pituitary adenylate cyclase activating polypeptide (PACAP) receptor ligand; a granulocyte colony stimulating factor (G-CSF) receptor ligand; a granulocyte- macrophage colony stimulating factor (GM-CSF); a platelet-derived growth factor (PDGF) receptor ligand, and a secretin receptor ligand.
[00100] The polypeptides of the present invention will preferably retain, at least in part, a biological activity of native human Gil', e.g., the polypeptides of the present invention will generally be Gil' agonists or antagonists. In one embodiment, the polypeptides of the present invention will exhibit biological activity in the treatment and prevention of metabolic conditions and disorders. Further, the novel Gil' analog polypeptides of the invention may include internal linker compounds, may include chemical modifications at internal amino acid residues, or may be chemically modified at the N-terminal or C-terminal residue. In yet another embodiment,thepolypeptidesoftheinventionincludeonlynatur

Nd
L amino acid residues and/or modified natural L amino acid residues. Alternatively, in another mbodiment, the polypeptides of the invention do not include unnatural amino acid residues.
[001011 In exemplary GD' hybrid embodiments, the GD' portion comprises a GD' N-terminal region modified or substituted to provide DPP-IV resistance superior to that of native GD'.
Exemplary Peotide Comnonent Families.
[00102] Native peptide hormones are known in the art, as are their analogs and derivatives. For reference, the sequences of several native peptide hormones are provided herein.
Exemplary Peptide Hormones
SEQ ID
No:
DescriptionSequence
33 Rat Amylin
34 h-Amylin:
35 h-ADM:
36 s-CT:
37 h-CT:
38 h-CGRP a:
39 h-CGRP J3:
40 h-AFP-6 (1-
47)
41 h-AFP-6 (8-
47):
42 Mouse AFP-6
(1-47):
43 Mouse AFP-6
(8-47):
44 CCK-8 -
sulfated:
45 h-Leptin:
46 hPYY:
47 hPYY(3-36)
KCNTATCATQRLANFLVRSSNNLGPVLPPTNVGSNTY KCNTATCATQRLANFLVHSSNNFGAILSSTNVGSNTY
YRQSMNNFQGLRSFGCRFGTCTVQKLAHQIYQFTDKDKDNVAPRSKISPQGY
CSNLSTCVLGKLSQELHKLQTYPRTNTGSGTP
CGNLSTCMLGTYTQDFNKFHTFPQTAIGVGAP
ACDTATCVTHRLAGLLSRSGGVVKNNFVPTNVGSKAF
ACNTATCVTHRLAGLLSRSGGMVKSNFVPTNVGSKAF
TQAQLLRVGCVLGTCQVQNLSHRLWQLMGPAGRQDSAPVDPSSPHSY
VGCVLGTCQVQNLSHRLWQLMGPAGRQDSAPVDPSSPHSY
PHAQLLRVGCVLGTCQVQNLSHRLWQLVRPAGRRDSAPVDPSSPHSY
VGCVLGTCQVQNLSHRLWQLVRPAGRRDSAPVDPSSPHSY
DY(503)MGWMDF
MHWGTLCGFLWLWPYLFYVQAVPIQKVQDDTKThIKTIVTRINDISHTQ
SVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQISND
LENLRDLLHVLAFSKSCRLPWASGLETLDSLGGVLEASGYSTEVVALSR
LQGSLQDMLWQLDLSPGC
YPIKPEAPGEDASPEELNRYYASLRHYLNLVTRQRY IKPEAPGEDASPEELNRYYASLRHYLNLVTRQRY

48 hGLl'-I (1- HDEFERHAEGTFTSDVSSTLEGQAALEFLAWLVKGRG
37):
Frog GLl'- 1: HAEGTYTNDVTEYLEEKAAKEFWLIKGKPKKIRYSOW
50 HAEGTFfSDVTQQLDEKAAKEFIDWLINGGl'SKEUS-OH
51 h-GLl'-1 (7- HAEGTFTSDVSSYLEGQAALEFIAWLVKGR
36):
52 h-GLP-2 HADGSFSDEMNTILDNLAARDFINWLIETK1TD
53 Frog GLl'-2: HABGTFTNDMTNYLEEKAAKEFVGWLIKGRPOH
54 OXM: HSQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNRNNIA
55 Exendin-3: HSDGTFTSDLSKQMEEEAVR LFIEWLKNGG PSSGAPPPS
S Exendin-4 HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS
[00103] These peptides are generally C-terminally amidated when expressed physiologically, but need not be for the purposes of the instant invention. In other words, the C-terminus of these peptides, as well as the GD' hybrid polypeptides of the present invention, may have a free —OH or NH2 group. These peptides may also have other post-translational modifications. One skilled in the art will appreciate that the hybrid polypeptides of the present invention may also be constructed with an N-terminal methionine residue.
[00104] It is also intended that in addition to comprising a GD' hybrid, in other embodiments the hormones described herein can be used in adjunct therapy, co-administered with, a GD' analog of the invention.
[OOlO5IIThe Amylin Family. As discussed herein component peptide hormones useful in the present invention with a GD' or a novel GD' analog include amylin family peptide hormones including amylin, adrenomedullin ("ADM"), calcitonin ("CT"), calcitonin gene related peptide ("CGRP"), intermedin (also known as "AFP-6") and related peptides. Native amylin family peptide hormones are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known amylin family peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention. Any amylin analog or derivative known in the art may be used in conjunction with the present invention.
[00106] Another family of peptide hormones implicated in metabolic diseases and disorders is the amylin family of peptide hormones, including amylin, calcitonin, calcitonin gene related peptide, adrenomedullin, and intermedin (also known as "AFl'-6"). Amylin is a 37-amino acid peptide hormone. It was isolated, purified and chemically characterized as the major component of amyloid deposits in the islets of pancreases of human Type 2 diabeties (Cooper et al., Proc. NatI. Acad. Sci., USA, 84:8628-8632 (1987)1~
The amylin molecule has two post-translational modifications: the C-terminus is amidated, and the cysteines in positions 2 and 7 are cross-linked to form an N-terminal loop. The sequence of the open reading frame of the human amylin gene shows the presence of the Lys-Arg dibasic amino acid proteolytic cleavage signal, prior to the N-terminal codon for Lys, and the Gly prior to the Lys-Arg proteolytic signal at the CLAIMS-terminal position, a typical sequence for amidation by protein amidating enzyme, PAM (Cooper et al., Biochem. Biophys. Acta, 1014:247-258 (1989)).
[00107] Amylin is believed to regulate gastric emptying, and suppress glucagon secretion and food intake, thus regulating the rate of glucose appearance in the circulation. It appears to complement the actions of insulin, which regulates the rate of glucose disappearance from the circulation and its uptake by peripheral tissues. These actions are supported by experimental findings in rodents and humans, which indicate that amylin complements the effects of insulin in postprandial glucose control by at least three independent mechanisms, all of which affect the rate of glucose appearance. First, amylin suppresses postprandial glucagon secretion. Compared to healthy adults, patients with type 1 diabetes have no circulating amylin and patients with type 2 diabetes have diminished postprandial amylin concentrations. Furthermore, infusion of an amylin specific monoclonal antibody, which bound circulating amylin, again resulted in greatly elevated glucagon concentrations relative to controls. Both of these results point to a physiological role of endogenous amylin in the regulation of postprandial glucagon secretion. Second, amylin slows gastrointestinal motility and gastric emptying. Finally, intrahypothalamic injections of rat amylin were shown to reduce feeding in rats and alter neurotransmitter metabolism in the hypothalamus. In certain studies, food intake was significantly reduced for up to eight hours following the intrahypothalamic injection of rat amylin and rat CGRP. In human trials, an amylin analog, pramlintide, has been shown to reduce weight or weight gain. Amylin may be beneficial in treating metabolic conditions such as diabetes and obesity. Amylin may also be used to treat pain, bone disorders, gastritis, to modulate lipids, in particular triglycerides, or to affect body composition such as the preferential loss of fat and sparing of lean tissue.
[00108] The hormone calcitonin (CT) was named for its secretion in response to induced hypercalcemia and its rapid hypocalcemic effect. It is produced in and secreted from neuroendocrine cells in the thyroid that have since been termed C cells. The best-studied action of CT(1-32) is its effect on the osteoclast. In vitro effects of CT include the rapid loss of ruffled borders and decreased release of lysosomal enzymes. Ultimately, the inhibition of osteoclast functions by CT results in a decrease in bone resorption. However, neither a chronic reduction of serum CT in the case of thyroidectomy nor the increased serum CT found in niedullary thyroid cancer appears to be associated with changes in serum calcium or bone mass. It is thus most likely that a major function of CT(1-32) is to combat acute hypercalcemia in emergency situations and/or protect the skeleton during periods of "calcium stress" such as growth, pregnancy, and lactation. (Reviewed in Becker, JCEM, 89(4): 1512-1525 (2004) and Sexton, Current Medicinal Chemistry 6: 1067-1093 (1999)). Consistent with this is recent data from the calcitonin gene knockout mouse, which removes both the calcitonin and the CGRP-I peptides, that revealed that the mouse had normal levels o'IS
basal calcium-related values, but an increased calcemic response (Kurihara H, et al., Hypertens Res. 2003 Feb; 26 Suppl:S 105-8).
[00109] CT has an effect on plasma calcium levels and inhibits osteoclast function and is widely used for the treatment of osteoporosis. Therapeutically, salmon CT (sCT) appears to increase bone density and decrease fracture rates with minimal adverse effects. CT has also been successfully used over the past 25 years as a therapy for Paget' s disease of bone, which is a chronic skeletal disorder that may result in enlarged or deformed bones in one or more regions of the skeleton. CT is also widely used for its analgesic effect on bone pain experienced during osteoporosis, although the mechanism for this effect is not clearly understood.
[00110] Calcitonin gene related peptide (CGRP) is a neuropeptide whose receptors are widely distributed in the body, including the nervous system and the cardiovascular system. This peptide seems to modulate sensory neurotransmission and is one of the most potent endogenous vasodilatory peptide discovered to date. Reported biological effects for CGRP include: modulation of substance P in inflammation, nicotinic receptor activity at the neuromuscular junction, stimulation of pancreatic enzyme secretion, a reduction of gastric acid secretion, peripheral vasodilation, cardiac acceleration, neuro-modulation, regulation of calcium metabolism, osteogenic stimulation, insulin secretion, an increase in body temperature and a decrease in food intake. (Wimalawansa, Amylin, calcitonin gene-related peptide, calcitonin and ADM: a peptide superfamily. Crit Rev Neurobiol. 1997; 11(2-3): 167-239). An important role of CGRP is to control blood flow to various organs by its potent vasodilatory actions, as evidenced by a decrease of mean arterial pressure following intravenous administration of cr-CGRl'. The vasodilatory actions are also supported by recent analysis of homozygous knockout CGRP mice, which demonstrated elevated peripheral vascular resistance and high blood pressure caused by increased peripheral sympathetic activity (Kurihara H, et al., Targeted disruption of ADM and cCCGRP genes reveals their distinct biological roles. Hypertens Res. 2003 Feb; 26 Suppl:5105-8). Thus, CGRP appears to elicit vasodilatory effects, hypotensive effects and an increase in heart rate among other actions.
[00111] Prolonged infusion of CGRP into patients with congestive cardiac failure has shown a sustained beneficial effect on hernodynamic functions without adverse effects, suggesting a use in heart failure. Other indications of CGRP use include renal failure, acute and chronic coronary artery ischemia, treatment of cardiac arrhythmia, other peripheral vascular disease such as Raynaud' s phenomenon, subarachnoid hemorrhage, hypertension, and pulmonary hypertension. Preeclamptic toxemia of pregnancy and preterm labor are also potentially treatable. (Wimalawansa, 1997). Recent therapeutic uses include the use of CGRP antagonists for the treatment of migraine headaches.
[001121 Adrenomedullin (ADM) is almost ubiquitously expressed with many more tissues containing the peptide than not. A published review of ADM, (Hinson, J.P. et al., Endocrine Reviews (2000) 21(2): 138-167) details its effects on the cardiovascular system, cellular growth, the central nervous system and the
endocrine system, with a range of biological actions including vasodilation., cell growth, regulation hormone secretion, and natriuresis. Studies in rat, cat, sheep, and man confirm that intravenous infusion
of ADM results in potent and sustained hypotension, and is comparable to that of CGRLP. However, the hypotensive effect of ADM on mean arterial pressure in the anesthetized rat is not inhibited by the CGRP antagonist CGRl8-37 suggesting that this effect is not mediated via CGRI' receptors. Acute or chronic administration of human ADM in rats, anesthetized, conscious or hypertensive, results in a significant decrease in total peripheral resistance accompanied by a fall in blood pressure, with a concomitant rise in heart rate, cardiac output and stroke volume.
[00113] ADM has also been proposed as an important factor in embryogenesis and differentiation and as an apoptosis survival factor for rat endothelial cells. This is supported by recent mouse ADM knockout studies, in which mice homozygous for loss of the ADM gene demonstrated defective vascular formation during embryogenesis and thus died mid-gestation. It was reported that ADM +/- heterozygous mice had high blood pressure along with susceptibility to tissue injury (Kurihara H, et al., Hypertens Res. 2003 F.eb; 26 Suppl:Sl05-8).
[00114] ADM affects such endocrine organs as the pituitary, the adrenal gland, reproductive organs and the pancreas. The peptide appears to have a role in inhibiting ACTH release from the pituitary. In the adrenal gland, it appears to affect the secretory activity of the adrenal cortex in both rat and human and it increases adrenal blood flow, acting as a vasodilator in the adrenal vascular bed in intact rats. ADM has been shown to be present throughout the female reproductive tract and plasma levels are elevated in normal pregnancy. Studies in a rat model of preeclampsia show that ADM can reverse hypertension and decrease pup mortality when given to rats during late gestation. Because it did not have a similar effect in animals in early gestation or non-pregnant rats in the preeclampsia model, this suggests that ADM may play an important regulatory role in the utero-placental cardiovascular system. In the pancreas, ADM most likely plays an inhibitory role since it attenuated and delayed insulin response to an oral glucose challenge, resulting in initial elevated glucose levels. ADM can also affect renal function. A bolus administered peripherally can significantly lower mean arterial pressure and raise renal blood flow, glomerular filtration rate and urine flow. In some cases, there is also an increase in Na+ excretion. -
100115] ADM also has other peripheral effects on bone and on the lung. For bone, studies have supported a role beyond the cardiovascular system and fluid homeostasis and have demonstrated that ADM acts on fetal and adult rodent osteoblasts to increase cell growth comparable to those of known osteoblast growth factors such as transforming growth factor-alpha. This is important clinically as one of the major challenges in osteoporosis research is to develop a therapy that increases bone mass via osteoblastic stimulation. In the lung, ADM not only causes pulmonary vasodilation. but also inhibits bronchoconstriction induced by histamine or acetylcholine. Recent studies using aerosolized ADM to treat pulmonary hypertension in a rat model indicate that inhalation treatment of this condition is effective, as evidenced by the fact that mean pulmonary arterial pressure and total pulmonary resistance were markedly lower in rats treated with ADM than in those given saline. This result was achieved without an alteration in systemic arterial pressure or heart rate (Nagaya N et al., Am J Physiol Heart CirNd Physiol. 2003;285:H2125-3 1).
[001161 In healthy volunteers, i.v. infusion of ADM has been shown to reduce arterial pressure and to stimulate heart rate, cardiac output, plasma levels of cAMP, prolactin, norepinephrmne and rennin. In these patients, there was little or no increase in urine volume or sodium excretion observed. In patients with heart failure or chronic renal failure, i.v. ADM had similar effects to those seen in normal subjects, and also induced diuresis and natriuresis, depending on the dose administered (Nicholls, MG et al. Peptides. 2001; 22:1745-1752) Experimental ADM treatment has also been shown to be beneficial in arterial and pulmonary hypertension, septic shock and ischemia/reperfusion injury (Beltowski 1., Pol J Pharmacol. 2004;56:5-27). Other indications for ADM treatment include: peripheral vascular disease, subarachnoid hemorrhage, hypertension, preeclamptic toxemia of pregnancy and preterm labor, and osteoporosrs.
[00117] Expression of AFP-6 (i.e., intermedin) is primarily in the pituitary and gastrointestinal tract. A specific receptor for AFl'-6 has not been reported; however, binding studies indicate that AFP-6 binds to all the known receptors of the Amylin Family. AFP-6 has been shown to increase cAIvIP production in SK-N-MC and L6 cells expressing endogenous CGRP receptors and competes with labeled CGRP for binding to its receptors in these cells. In published in vivo studies, AFP-6 administration led to blood pressure reduction in both nonnal and spontaneously hypertensive rats, most likely via interactions with the CRLR/RAMIP receptors. In vivo administration in mice led to a suppression of gastric emptying and food intake. (Roh et al. J Biol Chem. 2004 Feb 20;279(8):7264-74.)
[00118] It has been reported that the biological actions of amylin family peptide hormones are generally mediated via binding to two closely related type II G protein-coupled receptors (Gl'CRs), the calcitonin receptor (CTR) and the calcitonin receptor like receptor (CRLR). Cloning and functional studies have shown that CGRP, ADM, and amylin interact with different combinations of CTR or the CRLR and the receptor activity modifying protein (RAMP). Many cells express multiple RAMPs. It is believed that coexpression of RAMPs and either the CTR or CRLR is required to generate functional receptors for calcitonin, CGRP, ADM, and amylin. The RAMP family comprises three members (RAMPl, -2, and :3), which share less then 30% sequence identity, but have a common topological organization. Coexpression of CRLR and RAMP I leads to the formation of a receptor for CGRP. Co-expression of CRLR and RAMP2 leads to the formation of a receptor for ADM. Co-expression of CRLR and RAMP3 leads to the formation of a receptor for ADM and CGRP. Co-expression of hCTR2 and RAMP I leads to the formation of a receptor for amylin and CGRP. Co-expression of hCTR2 and RAMP3 leads to the formation of a receptor for amylin.
[00119] Thus a GD' hybrid comprising an amylin family hormone module can provide the functions and uses associated with the amylin family module, e.g. amylin, amylin/sCT/amylin, ADM, CGRP, as discussed, in addition to a GD' function.
[00120] In one embodiment, the amylin analogs and derivatives have at least one hormonal activity of
native amylin. In certain embodiments, the amylin analogs are agonists of a receptor which native amyli
is capable of specifically binding. Exemplary amylin analogs and derivatives include those described in US 2003/0026812 Al, which is hereby incorporated by reference.
[00121] Exemplary amylin analogs include:
[00122]As known in the art, such amylin analogs are preferably amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.
[00123] Any ADM analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the ADM analogs and derivatives have at least one hormonal activity of native ADM. In certain embodiments, the ADM analogs are agonists of a receptor which native ADM is
capable of specifically binding.
[00124] Any CT analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the CT analogs and derivatives have at least one hormonal activity of native CT. In certain embodiments, the Cr analogs are agonists of a receptor which native CT is capable of specifically binding. Exemplary CT analogs and derivatives include those described in U.S. Patent
Nos. 4,652,627; 4,606,856; 4,604,238; 4,597,900; 4,537,716; 4,497,731; 4,495,097; 4,444,981; 4,414,149; 4,401,593; and 4,397,780, which are hereby incorporated by reference.
[00125] Exemplary CT analogs include:
[00126] As known in the art, such CT analogs are preferably amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.
[00127]Any CGRP analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the CGRP analogs and derivatives have at least one hormonal activity of native CGRP. In certain embodiments, the CGRP analogs are agonists of a receptor which native CGRP is capable of specifically binding. Exemplary CGRP analogs and derivatives include those described in U.S. Patent Nos. 4,697,002; and 4,687,839, which are hereby incorporated by reference.
[00128] Exemplary CGRP analogs include:
[00129] Any AFl'-6 analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the AFP-6 analogs and derivatives have at least one hormonal activity of native AFl'-6. In certain embodiments, the AFP-6 analogs are agonists of a receptor which native AFP-6 is capable of specifically binding. Exemplary AFP-6 analogs and derivatives include those described
WO 2003/022304, which is hereby incorporated by reference.

[00130] Exemplary AFl'-6 analogs include:
[00131] As known in the art, such AFl'-6 analogs are preferably amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.
[OOl32lThe CCK Family. CCKs, including hCCK (cholecystokinin) and species variants, and various analogs thereof are known in the art. Generally, CCK has a 33-amino acid sequence first identified in humans, and includes a 8-amino acid in vivo C-terminal fragment ("CCK-8") that has been reportedly demonstrated in pig, rat, chicken, chinchilla, dog and humans. Other species variants include a 39-amino acid sequence found in pig, dog and guinea pig, and a 58-amino acid found in cat, dog and humans, and a 47-amino acid sequences homologous to both CCK and gastrmn. The C-terminal tyrosine-sulfated octapeptide sequence (CCK-8) is relatively conserved across species, and may be the minimum sequence for biological activity in the periphery of rodents. Thus, the term CCK-33 will generally refer to human CCK(1-33), while CCK-8 (CCK(26-33)) will refer to the C-terminal octapeptide generically in both the sulfated and unsulfated unless otherwise specified. Further, pentagastrin or CCK-5 will refer to the Cterminal peptide CCK(29-33), and the CCK-4 will refer to the C-terminal tetrapeptide CCK(30-33).
[00133] CCK was reportedly identified in 1928 from preparations of intestinal extracts by its ability to stimulate gallb~adder contraction. Other biological actions of CCK have since been reported, including stimulation of pancreatic secretion, delayed gastric emptying, stimulation of intestinal motility an"
stimulation of insulin secretion. See Lieverse et al., Ann. N.Y. Acad. Sci. 713: 268-272 (1994). The
actions of CCK, also reportedly include effects on cardiovascular function, respiratory function, neurotoxicity and seizures, cancer cell proliferation, analgesia, sleep, sexual and reproductive behaviors, memory, anxiety and dopamine-mediated behaviors. Crawley and Corwin, l'eptides 15: 731-755 (1994). Other reported effects of CCK include stimulation of pancreatic growth, stimulation of gallbladder contraction, inhibition of gastric acid secretion, pancreatic polypeptide release and a contractile component of peristalsis. Additional reported effects of CCK include vasodilation. Walsh, "Gastrointestinal Hormones," In Physiology of the Gastrointestinal Tract (3d ed. 1994; Raven Press, New York).
[00134] It has been reported that injections of combinations of glucagon, CCK and bombesin potentiated the inhibition of intake of condensed milk test meals in nondeprived rats over the inhibitions observed with individual compounds. Hinton et al., Brain Res. Bull. 17:615-619 (1986). It has also been reported that glucagon and CCK synergistically inhibit sham feeding in rats. LeSauter and Geary, Am. J. Physiol. 253:R217-225 (1987); Smith and Gibbs, Annals N.Y. Acad. Sci. 713:236-241 (1994). It has also been suggested that estradiol and CCK can have a synergistic effect on satiety. Dulawa et al., Peptides 15:9 13-918 (1994); Smith and Gibbs, supra. It has also been proposed that signals arising from the small intestine in response to nutrients therein may interact synergistically with CCK to reduce food intake. Cox, Behav. Brain Res. 38:35-44 (1990). Additionally, it has been reported that CCK induces satiety in several species. For example, it has been reported that feeding depression was caused by CCK injected intraperitoneally in rats, intraarterially in pigs, intravenously in cats and pigs, into the cerebral ventricles in monkeys, rats, dogs and sheep, and intravenously in obese and non-obese humans. See Lieverse et al., supra. Studies from several laboratories have reportedly confirmed the behavioral specificity of low doses of CCK on inhibition in feeding, by comparing responding for food to responding for nonfood reinforcers in both monkeys and rats and by showing that CCK elicits the sequence of behaviors normally observed after meal ingestion (i.e., the postprandial satiety sequence). Additionally, comparison of behavior after CCK to behavior after food ingestion, alone or in combination with CCK has reportedly revealed behavioral similarities between CCK and food ingestion. Crawley and Corwin, supra. It has also been reported that CCK in physiological plasma concentrations inhibits food intake and increases satiety in both lean and obese humans. See Lieverse et al., supra.
[00135] CCK was characterized in 1966 as a 33-amino acid peptide. Crawley and Corwin, supra. Species-specific molecular variants of the amino acid sequence of CCK have been identified. The 33-amino acid sequence and a truncated peptide, its 8-amino acid C-terminal sequence (CCK-8) have been reportedly identified in pig, rat, chicken, chinchilla, dog and humans. A 39-amino acid sequence was reportedly found in pig, dog and guinea pig. A 58-amino acid sequence was reported to have been found in cat, dog and humans. Frog and turtle reportedly show 47-amino acid sequences homologous to both CCK and gastrin. Very fresh human intestine has been reported to contain small amounts of an even larger molecule, termed CCK-83. In the rat, a principal intermediate form has been reportedly identified, and is
termed CCK-22. Walsh, "Gastrointestinal Hormones," In Physiology of the Gastrointestinal Tract (3d ect

1994; Raven l'ress, New York). A non-sulfated CCK-8 and a tetrapeptide (termed CCK-4 (CCK(30-33)) have been reported in rat brain. The C-terminal pentapeptide (termed CCK-4 (CCK(29-33)) conserves the structural homology of CCK, and also homology with the neuropeptide, gastrmn. The C-terminal sulfated octapeptide sequence, CCK-8, is reportedly relatively conserved across species. Cloning and sequence analysis of a cDNA encoding preprocholecystokinin from rat thyroid carcinoma, porcine brain, and porcine intestine reportedly revealed 345 nucleotides coding for a precursor to CCK, which is 115 amino acids and contains all of the CCK sequences previously reported to have been isolated. Crawley and Corwin, supra.
[00136] CCK is said to be distributed throughout the central nervous system and in endocrine cells and enteric nerves of the upper small intestine. CCK agonists include CCK itself (also referred to as CCK-33), CCK-8 (CCK(26-33)), non-sulfated CCK-8, pentagastrmn (CCK-5 or CCK(29-33)), and the tetrapeptide, CCK-4 (CCK(30-33)). At the pancreatic CCK receptor, CCK-8 reportedly displaced binding with a 1000-5000 greater potency than unsulfated CCK-8 or CCK-4, and CCK-8 has been reported to be approximately 1000-fold more potent than unsulfated CCK-8 or CCK-4 in stimulating pancreatic amylase secretion. Crawley and Corwin, supra. In homogenates from the cerebral cortex, CCK receptor binding was said to be displaced by unsulfated CCK-8 and by CCK-4 at concentrations that were equimolar, 10-fold or 100-fold greater than sulfated CCK-8. Id. Receptors for CCK have been reportedly identified in a variety of tissues, and two primary subtypes have been described: type A receptors and type B receptors. Type A receptors have been reported in peripheral tissues including pancreas, gallbladder, pyloric sphincter and afferent vagal fibers, and in discrete areas of the brain. The type A receptor subtype (CCKA) has been reported to be selective for the sulfated octapeptide. The Type B receptor subtype (CCKB) has been identified throughout the brain and in the stomach, and reportedly does not require sulfation or all eight amino acids. See Reidelberger, J. Nutr. 124 (8 Suppl.) 1327S-1333S (1994); Crawley and Corwin, supra.
[00137] Various in vivo and in vitro screening methods for CCK analogs are known in the art. Examples include in vivo assays involving the contraction of the dog or guinea pig gallbladder after rapid intravenous injection of the compound to be tested for CCK-like activity, and in vitro assays using strips of rabbit gallbladder. See Walsh, "Gastrointestinal Hormones", In Physiology of the Gastrointestinal Tract (3d ed. 1994; Raven Press, New York).
[00138] Certain exemplary CCKs and CCK analogs with CCK activity include:


(Table Removed)
[00139] As known in the art, such CCK peptides are preferably amidated, but within the context of the present invention, may optionally be in the acid form anless otherwise specified.
[00140]The Leptin Family. Component peptide hormones useful in the present invention also include leptin family peptide hormones. Native leptin family peptide hormones are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known leptin family peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.
[001411 Yet another peptide hormone family implicated in metabolic diseases and disorders is the leptin family. The mature form of circulating leptin is a 146-amino acid protein that is normally excluded from the CNS by the blood-brain barrier (BBB) and the blood-CSF barrier. See, e.g., Weigle et al., 1995. J Clin Invest 96 : 2065-2070. Leptin is the afferent signal in a negative feedback loop regulating food intake and body weight. The leptin receptor is a member of the cytokine receptor family. Leptin's anorexigenic effect is dependent on binding to homodimer of the Ob-Rb isoform of this receptor which encodes a long intra-cytoplasmic domain that includes several motifs for protein-protein interaction. ObRb is highly expressed in the hypothalamus suggesting that this brain region is an important site of leptin action. Mutation of the mouse ob gene has been demonstrated to result in a syndrome that exhibitspathophysiology that includes: obesity, increased body fat deposition, hyperglycemia, hyperinsulinemia, hypothermia, and impaired thyroid and reproductive functions in both male and female homozygous ob/ob obese mice (see e.g., Ingalis, et al., 1950. J Hered 41: 317-318. Therapeutic uses for leptin or leptin receptor include (i) diabetes (see, e.g., PCT Patent Applications WO 98/55139, WO 98/12224, and WO 97/02004); (ii) hematopoiesis (see, e.g., PCT Patent Applications WO 97/27286 and WO 98/18486); (iii) infertility (see, e.g., PCT Patent Applications WO 97/15322 and WO 98/36763); and (iv) tumor suppression (see, e.g., PCT Patent Applications WO 98/48831), each of which are incorporated herein by reference in their entirety.
[00142]The leptin receptor (OB-R) gene has been cloned (GenBank Accession No. AF098792) and mapped to the db locus (see, e.g., Tartaglia, et al., 1995. Cell 83: 1263-1271). Several transcripts of the OB-R, resulting from alternative splicing, have also been identified. Defects in OB-R produce a syndrome in the mutant diabetic ob/ob mouse that is phenotypically identical to the ob/ob mouse (see, e.g., Ghilardi, et al., 1996. Proc. NatI. Acad. Sci. USA 93: 623 1-6235). In contrast to ob/ob mice, however, administration of recombinant leptin to C57BLKS/J-m ob/ob mice does not result in reduced food intake and body weight (see, e.g., Roberts and Greengerg, 1996. Nutrition Rev. 54: 41-49).
[00143] Most leptin-related studies able to report weight loss activity from administration of recombinant
leptin, leptin fragments and/or leptin receptor variants have administered said constructs directly into thNd
ventricles of the brain. See e.g., Weigle, et al., 1995. J Clin Invest 96: 2065-2070; Barash, et al., 1996. Endocrinology 137: 3144-3147.
[0O1] Other studies have shown significant weight loss activity due to administration of leptin tides through intraperitoneally (i.p.) administration to test subjects. See, Grasso et al., 1997. Endocrinology 138:1413-1418. Further, leptin fragments, and most particularly an 18 amino acid fragment comprising residues taken from full length human leptin, have been reported to function in weight loss, but only upon direct administration through an implanted cannula to the lateral brain ventricle of rats. See, e.g., l'CT Patent Applications WO 97/46585, which is incorporated herein by reference in its entirety.
[001451 Thus a GD' hybrid comprising a leptin family hormone module can provide the functions and uses associated with the leptin family module, e.g. leptin, leptin fragment, as discussed, in addition to a GD' function.
[00146] Any leptin analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the leptin analogs and derivatives have at least one hormonal activity of native leptin. In certain embodiments, the leptin analogs are agonists of a receptor which native leptin is capable of specifically binding. Exemplary leptin analogs and derivatives include those described in, e.g., WO 2004/039832, WO 98/55139, WO 98/12224, and WO 97/02004, all of which are hereby incorporated by reference.
[00147] Exemplary leptin analogs include those where the amino acid at position 43 is substituted with Asp or Glu; position 48 is substituted Ala; position 49 is substituted with Glu, or absent; position 75 is substituted with Ala; position 89 is substituted with Leu; position 93 is substituted with Asp or Glu; position 98 is substituted with Ala; position 117 is substituted with Ser, position 139 is substituted with Leu, position 167 is substituted with Ser, and any combination thereof.
[00148] Certain exemplary leptin and leptin analogs with leptin activity include:
[00166] The Pl'F or PYY Family. Component peptide hormones useful in the present invention also include Pancreatic Polypeptide Family (PPF) peptide hormones, including PP,-NPY and PYY. Native PPF peptide hormones are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known PYY family peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.

[00167] Yet another family of peptide hormones implicated in metabolic diseases and disorders is the pancreatic polypeptide family ("l'PF"). Pancreatic polypeptide ("PP") was discovered as a contaminant of insulin extracts and was named by its organ of origin rather than functional importance (Kimmel et al., Endocrinology 83: 1323-30 (1968)). PP is a 36-amino acid peptide containing distinctive structural motifs. A related peptide was subsequently discovered in extracts of intestine and named Peptide YY ("PYY") because of the N- and C-terminal tyrosines (Tatemoto, Proc. NatI. Acad. Sci. USA 79: 2514-8 (1982)). A third related peptide was later found in extracts of brain and named Neuropeptide Y ("NPY") (Tatemoto, Proc. Natl. Acad. Sci. USA 79: 5485-9 (1982); Tatemoto et al., Nature 296: 659-60(1982)).

[00168] These three related peptides have been reported to exert various biological effects. Effects of PP include inhibition of pancreatic secretion and relaxation of the gallbladder. Centrally administered-PP produces modest increases in feeding that may be mediated by receptors localized to the hypothalamus and brainstem (reviewed in Gehlert, Proc. Soc. Exp. Biol. Med. 218: 7-22 (1998)).

[00169] Release of PYY occurs following a meal. An alternate molecular form of PYY is PYY(3-36) (Eberlein et al., Peptides 10: 797-803 (1989); Grandt et al., Regul. Pept. 51: 15 1-9 (1994)). This fragment constitutes approximately 40% of total PYY-like immunoreactivity in human and canine intestinal extracts and about 36% of total plasma PYY immunoreactivity in a fasting state to slightly over 50% following a meal. It is apparently a dipeptidyl peptidase-IY (DPP4) cleavage product of PYY. PYY(3-36) is reportedly a selective ligand at the Y2 and YS receptors, which appear pharmacologically unique in preferring N-terminally truncated (i.e., C-terminal fragments of) NPY analogs. Peripheral administration of PYY reportedly reduces gastric acid secretion, gastric motility, exocrine pancreatic secretion (Yoshinaga et al., Am. J. Physiol. 263: G695-701 (1992); Guan et al., Endocrinology 128: 911-6 (1991)'~

38


Pappas et al., Gastroenterology 91:1386-9 (1986)), gallbladder contraction and intestinal motility (Savage et al., Gut 28: 166-70 (1987)). The effects of central injection of PYY on gastric emptying, gastric motility and gastric acid secretion, as seen after direct injection in or around the hindbrain/brainstem (Chen and Rogers, Am. J. Physiol. 269: R787-92 (1995); Chen et al., Regul. Pept. 61: 95-98 (1996); Yang and Tache, Am. J. Physiol. 268: G943-8 (1995); Chen et al., Neurogastroenterol. Motil. 9:109-16 (1997)), may differ from those effects observed after peripheral injection. For example, centrally administered PYY had some effects opposite to those described herein for peripherally injected PYY(3-36) in that gastric acid secretion was stimulated, not inhibited. Gastric motility was suppressed only in conjunction with TRH stimulation, but not when administered alone, and was indeed stimulatory at higher doses through presumed interaction with PP receptors. PYY has been shown to stimulate food and water intake after central administration (Morley et al., Brain Res. 341: 200-3 (1985); Corp et al., Am. J. Physiol. 259:

R3 17-23 (1990)).

[00170]Any PPF analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the PPF analogs and derivatives have at least one hormonal activity of a native PPF polypeptide. In certain embodiments, the PPF analogs are agonists of a receptor which native PPF polypeptide is capable of specifically binding. Exemplary PPF analogs and derivatives include those described in WO 03/026591 and WO 03/057235, which are herein incorporated by reference in their entirety. In one embodiment, exemplary PPF analogs and derivatives that exhibit at least one PPF hormonal activity generally comprise at least two PYY motifs including a polyproline motif and C-terminal tail motif. Such analogs are generally described in U.S. Provisional Application No. 60/543,406 flIed February 11, 2004, which is herein incorporated by reference. Other exemplary PPF analogs are disclosed in PCT/U52005/004351, entitled "Pancreatic Polypeptide Family Motifs and Polypeptides Comprising the Same", Attorney Docket 18528.832, the contents of which is hereby incorporated by reference. By way of background, the receptors to which PYY family peptides bind are generally referred to as Y receptors, and research has suggested that the differences in Y receptor binding affinities are correlated with secondary and tertiary structural differences. See, e.g., Keire et al., Biochemistry 2Q00, 39, 9935-9942. Native porcine PYY has been characterized as including two C-terminal helical segments from residues 17 to 22 and 25 to 33 separated by a kink at residues 23, 24, and 25, a turn centered around residues 12-14, and the N-terminus folded near residues 30 and 31. Further, full-length porcine PYY has been characterized as including the PP fold, stabilized by hydrophobic interactions among residues in the N- and C- termini. See id.
[00171] A "PYY motif' is generally a structural component, primary, secondary, or tertiary, of a native PP family polypeptide that is critical to biological activity, i.e., biological activity is substantially decreased in the absence or disturbance of the motif. Exemplary PYY motifs include the N-terminal polyproline type II motif of a native PP family polypeptide, the type II beta-turn motif of native PP family polypeptide, the a-helical motif at the C-terminal end of native PP family polypeptide, and the C-terminal tail motif of native PP family polypeptide. More particularly, in the N-terminal polyproline region, amino
acids corresponding to residues 5 and 8 of a native PP family polypeptide are generally conserved as a proline. The type II beta-turn motif will generally include amino acids corresponding to residues 12-14 of a native PP family polypeptide. The a-helical motif can generally extend from amino acids corresponding to approximately residue 14 of a native PP family polypeptide to any point up to and including the C-terminal end, so long as the a-helical motif includes a sufficient number of amino acid residues such that an a-helical turn is formed in solution. The a-helical motif can also include amino acid substitutions, insertions and deletions to the native PP family sequence, so long as the a-helical turn is still formed in solution. The C-terminal tail motif generally includes amino acids corresponding to approximately the last 10 residues of a native PP family polypeptide, more preferably the last 7, 6, or 5 residues of a native PP family polypeptide, and more preferably amino acid residues 32-35. Exemplary PYY analogs include those with internal deletions, insertions, and substitutions in areas of the PYY molecule not corresponding to the polyproline motif and/or the C-terminal tail motif. For instance, internal deletions at positions 4., 6, 7, 9, or 10 are envisioned.
[00172] Additional Incretins and Incretin Mimetics. Component peptide hormones useful in the present invention also include GLP- I peptide hormones. Native GLP- 1 peptide hormones, including GLP- 1(1 -37), GLP-1(7-37), and GLP-1(7-36)amide, are known in art, as are functional peptide analogs and derivatives. As used herein, GLP-1 refers to all native forms of GLP-1 peptide hormones. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known GLP-l peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.
[00173] Central to many metabolic diseases and disorders is the regulation of insulin levels and blood glucose levels. Insulin secretion is modulated in part by secretagogue hormones, termed as incretins, which are produced by enteroendocrine cells. The incretin hormone, glucagon-like peptide-I ("GLP-1") is a peptide hormone secreted by intestinal cells that has been shown in multiple studies to produce an enhancing effect on insulin secretion. GLP-1 is processed from proglucagon in the gut and enhances nutrient-induced insulin release (Krcymann B., et al., Lancet, 2:1300-1303 (1987)). Various truncated forms of GLP-1, are known to stimulate insulin secretion (insulinotropic action) and cAMP formation (see, e.g., Mojsov, S., Int. J. Pep. Pro. Res., 40:333-343 (1992)). A relationship between various in vitro laboratory experiments and mammalian, especially human, insulinotropic responses to exogenous administration of GLP- 1, GLP- 1(7-36) amide, and GLP- 1(7-37) acid has been established (see, e.g., Nauck, M. A., et al., Diabetologia, 36:741-744 (1993); Gutniak, M., et al., New Eng. J. of Med., 326(20):1316-1322 (1992); Nauck, M. A., et al., J. Clin. Invest., 91:301-307 (1993); and Thorens, B., et al., Diabetes, 42:1219-1225 (1993)).
[00174] GLP-1(7-36) amide exerts a pronounced antidiabetogenic effect in insulin-dependent diabetics by stimulating insulin sensitivity and by enhancing glucose-induced insulin release at

physiological concentrations (Gutniak M., et al., New Eng. J. Med., 326:1316-1322 (1992)). Whe~ administered to non-insulin dependent diabetics, GLP-1(7-36) amide stimulates insulin release, lowers
glucagon secretion, inhibits gastric emptying and enhances glucose utilization (Nauck, 1993; Gutniak, 1992; Nauck, 1993). However, the use of GLP-l type molecules for prolonged therapy of diabetes has been complicated because the serum half-life of such peptides is quite short.
[00175] More particularly, GLP- 1 is a 30-amino acid peptide derived from proglucagon, a 160-amino acid prohormone. Actions of different prohormone convertases in the pancreas and intestine result in the production of glucagon and other ill-defined peptides, whereas cleavage of proglucagon results in the production of GLP-l and GLP-2 as well as two other peptides. The amino acid sequence of GLP-1 is 100% homologous in all mammals studied so far, implying a critical physiological role. GLP-1 (7-37) acid is C-terminally truncated and amidated to form GLP- 1 (7-36) N112. The biological effects and metabolic turnover of the free acid GLP-1 (7-37) OH, and the amide, GLP-1 (7-36) NH2, are indistinguishable. By convention, the numbering of the amino acids is based on the processed GLP- 1 (1-37) OH from proglucagon. The biologically active GLP-1 is the result of further processing: GLl'-1 .(7-
36) NH2. Thus the first amino acid of GLP-1 (7-37) OH or GLP-1 (7-36)NH2 is 7His.
[00176] In the gastrointestinal tract, GLP- 1 is produced by L-cells of intestinal, colonic and rectal mucosa, in response to stimulation by intraluminal glucose. The plasma half-life of active GLP-l is 5 minutes, and its metabolic clearance rate is around 12-13 minutes (Hoist, Gastroenterology 107(6): 1848-55 (1994)). The major protease involved in the metabolism of GLP-1 is dipeptidyl peptidase (DPP) IV (CD26) which cleaves the N-terminal His-Ala dipeptide, thus producing metabolites, GLP-l (9-37) OH or GLP-l (9-36) NH2 which are variously described as inactive, weak agonist or antagonists of GLP-l receptor. The GLP-l receptor (GLP-lR) is a G protein coupled receptor of 463 amino acid and is localized in pancreatic beta cells, in the lungs, and to a lesser extent in the brain, adipose tissue and kidneys. The stimulation of GLP-1R by GLP-1 (7-37) OH or GLP-1 (7-36)NH2 results in adenylate cyclase activation, cAMP synthesis, membrane depolarization, rise in intracellular calcium and increase in glucose-induced insulin secretion (Holz et al., J. Biol. Chem. 270(30): 17749-57 (1995)).
[00177] GLP-1 is a potent insulin secretagogue that is secreted from the intestinal mucosa in response to food intake. The profound incretin effect of GLP-1 is underscored by the fact that GLP-LR knockout mice are glucose-intolerant. The incretin response of i.v. infused GLP-1 is preserved in diabetic subjects, though the incretin response to oral glucose in these patients is compromised. GLP- 1 administration by infusion or sc injections controls fasting glucose levels in diabetic patients, and maintains the glucose threshold for insulin secretion (Gutniak et al., N. Engl. J. Med. 326:1316-22 (1992); Naock et al., Diabet. Med. 13:(9 Suppl 5):539-543 (1996); Nauck et al., J. Clin. Endocrinol. Metab. 76:912-917 (1993)). GLP-l has shown tremendous potential as a therapeutic agent capable of augmenting insulin secretion in a physiological manner, while avoiding hypoglycemia associated with sulfonylurca drugs.
[00178] Other important effects of GLP- 1 on glucose homeostasis are suppression of glucagon secretion and inhibition of gastric motility. GLP- 1 inhibitory actions on pancreatic alpha cell secretion of
glucagon leads to decreases in hepatic glucose production via reduction in gluconeogenesis anzj glycogenolysis. This antiglucagon effect of GLP-1 is preserved in diabetic patients.
[00179] The so-called ileal brake effect of GLP- 1, in which gastric motility and gastric secretion are inhibited, is effected via vagal efferent receptors or by direct action on intestinal smooth muscle. Reduction of gastric acid secretion by GLl'-l contributes to a lag phase in nutrient availability, thus obviating the need for rapid insulin response. In summary, the gastrointestinal effects of GLP-1 contribute significantly to delayed glucose and fatty acid absorption and modulate insulin secretion and glucose homeostasis.
[00180] GLP-1 has also been shown to induce beta cell specific genes, such as GLUT-i transporter, insulin (via the interaction of PDX-1 with insulin gene promoter), and hexokinase-1. Thus GLP-1 could potentially reverse glucose intolerance normally associated with aging, as demonstrated by rodent experiments. In addition, GLP-1 may contribute to beta cell neogenesis and increase beta cell mass, in addition to restoring beta cell function during states of beta cell insufficiency.
[00181] Central effects of GLP-1 include increases in satiety coupled with decreases in food int~ike, effected via the action of hypothalamic GLP-1R. A 48 hour continuous SC infusion of GLP-1 in type II diabetic subjects, decreased hunger and food intake and increased satiety. These anorectic effects were absent in GLP-lR knock out mice.
[OOl82lThus a GD' hybrid comprising an incretin family hormone module can provide the functions and uses associated with the incretin family module, e.g. exendin-4, GLPl, GLP2, as discussed, in addition to having a GD' function.
[00183] Any GLP-l peptide analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the GLP- 1 peptide analogs and derivatives have at least one hormonal activity of a native GLP-1 peptide. In certain embodiments, the GLP-1 peptide analogs are agonists of a receptor which a native GLP- I peptide is capable of specifically binding. Exemplary GLP- 1 peptide analogs and derivatives include those described in, e.g., WO 91/11457, which is hereby incorporated by reference.
[00184] GLP-1 analogs known in the art include:
(Table removed)

[00185] As known in the art, such GLP-1 analogs may preferably be amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.
[00186]Other GLP-1 analogs and derivatives are disclosed in U.S. Pat. No. 5,545,618 which is incorporated herein by reference. A exemplary group of GLP- I analogs and derivatives include those disclosed in U.S. Patent No. 6,747,006, which is herein incorporated by reference in its entirety. The use in the present invention of a molecule described in U.S. Pat. No. 5,188,666, which is expressly incorporated by reference, is also contemplated. Another group of molecules for use in the present invention includes compounds described in U.S. Pat. No. 5,512,549, which is expressly incorporated herein by reference. Another exemplary group of GLP- I compounds for use in the present invention is disclosed in WO 91/11457, which is herein incorporated by reference.
[00187] In another embodiment useful with GD' or novel GD' analogs are GLPl analogs with Trp-Cage tails (e.g. exendin C-terminal sequence with or without the tryptophan (or similar residue) (which can be optionally provided as described, e.g. by the presence of a tryptophan advantageously located in the honnone that is either naturally occurring, added as a substitution or as part of a linker.). These include:
GLPl (7-26)Gly8Ex-4(2 1-39):
HGEGTFTSDVSSYLEGQAAKLFIEWLKNGO PSSGAPPPS-NH2 (SEQ ID NO: 74);
GLP (7-33)(G8,E30,K33)[Ex4(28-39)I: HGEGTFTSDVSSYLEGQAAKEFIEWLKNGGPSSGAPPPSNH2 (SEQ ID NO: 75);
GLP1(7-33)(G8,K33MEX4(28-39)]: HGEGTFrSDVSSYLEGQAAKEFIAWLKNGGPSSGAPPPS-NH2 (SEQ ID NO: 76);
GLP1 (7-33)G8IIEx-4(28-39)]: HGEGTFTSDVSSYLEGQAAKEFIAWLVNGGPSSGAPPPS-NH2 (SEQ ID NO: 77); and
GLPl (7-35)G8[Ex-4(30-39)I: HGEGTFrSDVSSYLEGQAAKEFIAWLVKGGPSSGAPPPS-NH2 (SEQ ID NO: 78).
[001881 Component peptide hormones useful in the GD' hybrids of the present invention also include GLP-2 peptide hormones. Native GLP-2 peptide hormones, e.g., rat GLP-2 and its homologous including ox GLP-2, porcine GLP-2, degu GLP-2, bovine GLP-2, guinea pig GLP-2, hamster GLP-2, human GLP2, rainbow trout GLP-2, and chicken GLP-2, are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known GLP-2 peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.

[00189] Any GLP-2 peptide analog or derivative known in the art may be used in conjunction with the

present invention. In one embodiment, the GLP-2 peptide analogs and derivatives have at least onNd hormonal activity of a native GLP-2 peptide. In certain embodiments, the GLP-2 peptide analogs are

43


agonists of a receptor which a native GLP-2 peptide is capable of specifically binding. Exemplary GLP-2 peptide analogs and derivatives include those described in, e.g., U.S. Ser. No. 08/669,791 and PCT Application l'CT/CA97/00252, both of which are hereby incorporated by reference. Specific GLP-2 analogs known in the art include: rat or human GLl'-2 altered at position 2 to confer DPP-IV resistance by substituting a Gly for an Ala.


[00190] Component peptide hormones useful in the present invention also include oxyntomodulin (OXM) peptide hormones. Native OXM peptide hormones are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known OXM peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.

[00191] Any OXM peptide analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the OXM peptide analogs and derivatives have at least one hormonal activity of a native OXM peptide. In certain embodiments, the OXM peptide analogs are agonists of a receptor which a native OXM peptide is capable of specifically binding.

[00192] Component peptide hormones useful in the present invention also include exendin peptide hormones. Native exendin peptide honnones are known in art, as are functional peptide analogs and derivatives. Certain exemplary native peptides, peptide analogs and derivatives are described herein, however it should be recognized that any known exendin peptides that exhibit hormonal activity known in the art may be used in conjunction with the present invention.

[00193] Exendins are another family of peptides implicated in insulin secretion. Exendins are found in the saliva of the Gila-monster, a lizard endogenous to Arizona, and the Mexican Beaded Lizard. Exendin-3 is present in the saliva of Heloderma horridum, and exendin-4 is present in the saliva of Heloderma suspectum (Eng, J., et al., J. Biol. Chem., 265:20259-62, 1990; Eng., J., et al., J. Biol. Chem., 267:7402-05 (1992)). The exendins have some sequence similarity to several members of the glucagon-like peptide family, with the highest identity, 53%, being to GLP-l (Goke, et al., J. Biol. Chem., 268:1965(1-55 (1993)).

[00194] Exendin-4 binds the OLl'- 1 receptors on insulin-secreting TC 1 cells, at dispersed acinar cells from guinea pig pancreas, and at parietal cells from stomach; the peptide also stimulates somatostatin release and inhibits gastrin release in isolated stomachs (Goke, et al., J. Biol. Chem., 268:19650-55 (1993); Schepp, et al., Eur. J. Pharmacol., 69:183-91 (1994); Eissele, et al., Life Sci., 55:629-34 (1994)). Exendin-3 and exendin-4 were found to bind the GLP-1 receptors on, to stimulating cAMP production in, and amylase release from, pancreatic acinar cells (Malhotra, R., et al., Relulatory Peptides, 41:149-56 (1992); Raufman, et al., J. Biol. Chem., 267:21432-37 (1992); Singh, et al., Regul. Pept., 53:47-59 (1994)). The use of the insulinotropic activities of exendin-3 and exendin-4 for the treatment of diabetes mellitus and the prevention of hyperglycemia has been proposed (Eng, U.S. Pat. No. 5,424,286).

[00195]Truncated exendin peptides such as exendin[9-39], a carboxyamidated molecule, and fragments 3-39 through 9-39 have been reported to be potent and selective antagonists of GLP- 1 (Goke, et al., J. Biol. Chem., 268: 19650-55 (1993); Raufman, J. P., et al., J. Biol. Chem., 266:2897-902 (1991); Schepp, W., et al., Eur. J. Pharm., 269:183-91(1994); Montrose-Rafizadeh, et al., Diabetes, 45(Suppl. 2): 152A (1996)). Exendin[9-391 blocks endogenous GLP-l in vivo, resulting in reduced insulin secretion (Wang, et al., J. Clin. Invest., 95:417-21 (1995); D'Alessio, et al., J. Clin. Invest., 97:133-38 (1996)). The receptor apparently responsible for the insulinotropic effect of GLP- 1 has been cloned from rat pancreatic islet cells (Thorens, B., Proc. Natl. Acad. Sci. USA 89:8641-8645 (1992)). Exendins and exendin[9-39] bind to the cloned GLP-l receptor (rat pancreatic -cell GLl'-l receptor: Fehmann HC, et al., Peptides, 15 (3):
453-6 (1994); human GLP-1 receptor: Thorens B, et al., Diabetes, 42 (11): 1678-82 (1993)). In cells transfected with the cloned GLP-1 receptor, exendin-4 is an agonist, i.e., it increases cAMP, while exendin[9-39] is an antagonist, i.e., it blocks the stimulatory actions of exendin-4 and GLP- 1. Id.
[00196] More particularly, exendin-4 is a 39 amino acid C-terminal amidated peptide found in the saliva of the Gila Monster (Heloderma suspectum), with a 53% amino acid sequence identity to the GLP- 1 peptide sequence. See, e.g., Eng, I., et al. "Isolation and Characterization of Exendin, and Exendin-3 Analogue from Heloderma suspectum Venom," J. Bio. Chem., 267:11, p. 7402-7405 (1992), Young, A. A., et al., "Glucose-Lowering and Insulin-Sensitizing Actions of Exendin-4," Diabetes, Vol. 48, p. 1026-1034, May, 1999. In terms of its activity, exendin-4 is a highly specific agonist for the GLP- I receptor, and, like GLP-1, is able to stimulate insulin secretion. Therefore, like GLP-1, exendin-4 is regarded as an insulinotropic peptide.
[00197] However, unlike GLP-1, exendin-4 has a relatively long half-life in humans, because of its resistance to the dipeptidyl peptidase IV which rapidly degrades the GLP- 1 sequence in vivo. Furthermore, it has been shown that, as compared to GLP- 1, exendin-4 has a stronger capability to stimulate insulin secretion, and that a lower concentration of exendin-4 may be used to obtain such stimulating activity. See, e.g., U.S. Pat. No. 5,424,286, herein incorporated by reference. Therefore exendin-4 peptides or derivatives thereof (for examples of such derivatives, see, e.g., U.S. Pat. No. 6,528,486, herein incorporated by reference, and its corresponding international application WO
01/04156) have a greater potential utility for the treatment of conditions involving the dysregulation of insulin levels (e.g., conditions such as diabetes) than either insulin or GLP- 1. Thus a GD' hybrid comprising an exendin family hormone module can provide the functions and uses associated with the exendin family module, e.g. exendin-4, exendin tail, as discussed, in addition to having a GD' function.
[00198] Any exendin peptide analog or derivative known in the art may be used in conjunction with the present invention. In one embodiment, the exendin peptide analogs and derivatives have at least one hormonal activity of a native exendin peptide. In certain embodiments, the exendin peptide analogs are agonists of a receptor which a native exendin peptide is capable of specifically binding.
[00199] Exemplary exendin analogs include:
u, Phe-exendin-4
Leu, Ala, Phe-exendin-4
[002001 As known in the art, such exendin analogs are preferably amidated, but within the context of the present invention, may optionally be in the acid form unless otherwise specified.
[00201] Additional exemplary exendin analogs and derivatives are described in l'CT Application Serial No. PCT/U598/16387 filed Aug. 6, 1998, entitled "Novel Exendin Agonist Compounds," which claims the benefit of U.S. patent application Ser. No. 60/055,404, filed Aug. 8, 1997, both of which are herein incorporated by reference. Other exendin analogs and derivatives are described in PCT Application Serial No. PCTIUS98/24210, filed Nov. 13, 1998, entitled "Novel Exendin Agonist Compounds," which claims the benefit of U.S. Provisional Application No. 60/065,442 filed Nov. 14, 1997, both of which are herein incorporated by reference. Still other exendin analogs and derivatives are described in PCT Application Serial No. PCTJUS98/24273, filed Nov. 13, 1998, entitled "Novel Exendin Agonist Compounds," which claims the benefit of U.S. Provisional Application No. 60/066,029 filed Nov. 14, 1997, both of which are herein incorporated by reference. Still other exendin analogs and derivatives are described in PCT Application Serial No. PCT/U597/14199, filed Aug. 8, 1997, entitled "Methods for Regulating Gastrointestinal Activity," which is a continuation-in-part of U.S. patent application Ser. No. 08/694,954 filed Aug. 8, 1996, both of which are hereby incorporated by reference. Still other exendin analogs and derivatives are described in PCT Application Serial No. PCT/U598/00449, filed Jan. 7, 1998, entitled "Use of Exendins and Agonists Thereof for the Reduction of Food Intake," which claims priority to U.S. Provisional Application No. 60/034,905 filed Jan. 7, 1997, both of which are hereby incorporated by reference. Yet other exendin analogs and derivatives are described in US 2004/0209803 Al, filed December 19, 2003, entitled "Compositions for the Treatment and Prevention of Neuropathy," which is hereby incorporated by reference.
[00202]Catestatin Family. The catestatin fragment of chromogranin A is an endogenous inhibitor of nicotinic cholinergic transmission, functioning in negative feedback control of catecholamine secretion. We explored naturally occurring polymorphisms in the amino acid sequence of catestatin. Three human variants were identified: Gly364Ser, Pro37OLeu, and Arg374Gln.
[00203] Sequence variants in human catestatin (human chromogranin A352—372) and interspecies homologies in humans and other mammals are shown below. Amino acids at positions variant in human catestatin are shown in bold type. The typical dibasic proteolytic cleavage site at the carboxy-terminal side of catestatin is given in brackets, [RR]. For human chromogranin A, this [RR] site is Arg373Arg374. In further embodiments, variants absent either or both arginines are included. GD' hybrids containing catestatin family members, analogs, derivatives or fragments thereof, find use in the treatment methods of the invention. For example, such hybrids will find use in treating cardiovascular diseases and conditions as discussed herein, including hypertension and congestive heart failure and related conditionNd

Mouse

Rat

Cow

Pig

Horse

Chimp

Human

Wild-type

Variant

Variant

Variant
Combined with an active GD' hormone module, the compounds will find use in the critical care conditions described herein. Catestatin species variants include:


RSMRLSFRTRGYGFRDPGLQL[RR]

RSMRLSFRARGYGFRDPGLQL[RRJ

RSMRLSFRARGYGFRGPGLQL[RR}

RSMRLSFRAPAYGFRGPGLQL[RR]

RSMKLSFRARAYGFRGPGLQL[RR]

SSMKLSFRARAYGFRGPGPQL[RR]



SSMKLSFRARAYGFRGPGPQL[RR~

SSMKLSFRARAYSFRGPGPQL[RRJ

SSMKLSFRARAYGFRGPGLQL[RR]

SSMKLSFRARAYGFRGPGPQL[RQ]
CGA364—384 (SEQ ID NO: 79)

CGA367—387 (SEQ ID NO: 80)

CGA344-364 (SEQ ID NO: 81)

CGA343—363 (SEQ ID NO: 82)

CGA343—363 (SEQ ID NO: 83)

CGA3S4—374 (SEQ ID NO: 84)



CGA3S2—372 (SEQ ID NO: 85)

(SEQ ID NO: 86)

(SEQ ID NO: 87)

(SEQ ID NO: 88)


[OO2o4lNatriuretic Peptides. Natriuretic peptides are a family of hormones that consist of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP). They are synthesized and stored as 3 distinct precursor prohormones, which are the 126 amino acid AN?. 108 amino acid BNP, and 104 amino acid CNP. They are each encoded by separate genes and have distinct sites of synthesis and mechanisms of regulation. Parental natriuretic peptide sequences include:

(Table Removed)
[00ZO5IThe main site of synthesis of the AN? prohormone is the atrial myocyte where it is synthesized as a 151-amino acid preprohormone. Removal of a 25-amino acid signal peptide from its N terminal end

occurs in the endoplasmic reticulum, leaving a 126-amino acid AN? prohormone (ProAN?), the main storage form of AN? within the heart. The prohormone consists of 4 biologically active peptide segments: amino acids 1-30 (ProANF 1-30, also known as long acting Na stimulator), 31-67 (ProANF 31-67, also known as vessel dilator), 79-98 (ProANF 79-98, also known as potassium excreter), and 99-126 (ANE, also known as atrial natriuretic factor).
[00206] BN? was originally isolated from porcine brain but in humans it is synthesized and secreted from the left ventricle. Sequence analysis reveals that preproBN? consists of 134 residues and is cleaved to a 108-amino acid ProBN?. Cleavage of a 32-amino acid sequence from the C-terminal end of ProBNP results in human BNP (77-108), which is the physiologically active form in plasma.
[00207] CNP is the third member of the natriuretic peptide system and is primarily found in human vascular endothelial cells, kidney, and porcine brain. High concentrations of CNP are also found in huriian hypothalamus and midbrain. In humans, preproCN? is a 126-amino acid precursor processed into proCNP by cleavage of 23 residues from its N-terminal end. This 23-amino acid sequence serves as a signal peptide. The terminal 22 (105-126) amino acids are cleaved from proCN? to yield a biologically active form of CM'.
[002081 Urodilatin is a kidney-derived member of the natriuretic peptide family and is formed from the same AN? prohormone and consists of amino acids 95-126. Except for the 4 amino acid N terminal extension, it is identical to ANF (99-126). Urodilatin appears to be an important regulator of sodium and water handling in the kidney, as well as a mediator of sodium excretion in patients with congestive heart failure (CRlF).
[00209] Natriuretic peptides exert their biologic effects by binding to high-affinity receptors on the surface of target cells. Three subtypes of NPRs—N?R-A, N?R-B, and NPRC—have been isolated. Consequently, in one embodiment is provided a method to screen hybrids for natriuretic receptor binding and/or activation. Natriuretic peptides including prohormone variants can impart numerous natriuretic peptide hormone activities to hybrids of the invention. In other embodiments of interest are natriuretic antagonist hybrids. Natriuresis is the excretion of an excessively large amount of sodium into the urine. Natriuresis is similar to diuresis (the excretion of an unusually large quantity of urine), except that in natriuresis the urine is exceptionally salty. Natriuresis occurs with some diuretics and diseases (as of the adrenal) and can lead to the salt-losing syndrome characterized by dehydration, vomiting, low blood pressure, and the risk of sudden death. Exogenous administration of the 4 independent circulating peptides of the AN? prohormone (1-30, 31-67, 79-98, and 99-126) produce in vivo vasodilation, diuresis, suppression of the renin—angiotensin—aldosterone system and enhanced natriuresis and/or kaliuresis.
ProANF 1-30, ProANF 31-67 and ANF 99-126 each have natriuretic, blood pressure lowering anctNd
diuretic properties with ProANF 31-67 and ANF 99-126 having the greatest impact on blood pressure. There are varying effects of the AN? peptides on potassium homeostasis: ProANF 79-98 stimulates potassium excretion, whereas ProANF 31-67 spares potassium loss by inhibiting Na/K ATPase in the medullary collecting duct cells. Specific to ANF 99-126 is a dose-dependent inhibition of angiotensin II-mediated aldosterone secretion, whereas proANF 31-67 has the property of inducing natriuresis through generation of prostaglandin.


[00210] BNP produces similar biologic effects as ANE in normal humans. Infusions of BN? in normal men produced a 2-fold increase in sodium excretion, 50% reduction in plasma renin, angiotensin il and aldosterone secretion as well as a reduction in plasma volume.
[00211] CNP induces cardiovascular effects similar to the other natriuretic peptides but does not appear to mediate any renal effects. When CNP is infused in anesthetized dogs at equivalent doses of ANF, plasma cGMP rose with a concomitant reduction in mean arterial pressure, right atrial pressure and cardiac output, but glomerular filtration rate, renal blood flow and sodium excretion decreased.
[00212] Natriuretic peptides can provide therapeutic benefit in heart failure. Congestive heart failure (CHF) is associated with increases in vasopressin, endothelin, and with activation of the renin— angiotensin—aldosterone system, and sympathetic nervous systems, mediating vasoconstriction, sodium and water retention, and negative vascular and cardiac remodeling. These effects occur despite the elevated levels of the natriuretic peptides in patients with heart failure. In one embodiment of the invention are hybrids that provide increased or therapeutic serum levels of natriuretic peptide activity for treatment or prevention of cardiac related diseases and conditions, including CHF. Although ANF infusion in normal individuals can result in a sustained increase in sodium excretion and urine flow rates, in the heart failure patient a marked beneficial reduction in renal response can be obtained. BN? infusion markedly increases sodium excretion in patients with heart failure and exerts significant beneficial hemodynamic effects. As compared with ANP, the diuretic and natriuretic effects of BNP are significantly greater. BNP is cleared more slowly than AN? and exerts other effects including suppressing aldosterbne secretion and increasing serum levels of AN?. RN? peptides can also provide a beneficial decrease in pulmonary capillary wedge pressure, systemic vascular resistance, right atrial pressure and systolic blood pressure, with an increase in cardiac index in patients hospitalized for symptomatic CHF. In patients with decompensated heart failure, natriuretic peptide hybrids can provide a beneficial decrease in pulmonary capillary wedge pressure and an improved dyspnea score. (Dyspnea is an unpleasant sensation of difficulty in breathing, typically associated with early stages of cardiac heart failure.) The hybrids containing one, two or three natriuretic hormone functions provide methods of administration of pharmaceutically active compositions that are useful for both the prophylactic and therapeutic treatment of CRlF patients, preferably CHF patients that are decompensated, patients with chronic CRlF, and patients with hypertension. The natriuretic portion(s) of a hybrid is sufficient to provide a therapeutically effective

amount of a natriurertic peptide to such patient when administered in a therapeutically effective dose over a therapeutically effective period.
[002131 As discussed herein any of the family of therapeutically effective natriuretic peptides or their analogs can be used. Useful natriuretic peptides include, for example, atrial natriuretic peptide (ANP), brain natriuretic peptide (EN? or B-type natriuretic peptide) and C-type natriuretic peptide (CM'). Sequences of useful forms of natriuretic peptides are disclosed in U.S. Patent Publication 20010027181, which is incorporated herein by reference. Examples of ANPs include human AN? (Kangawa et al., BBRC 118:131(1984)) or that from various species, including pig and rat AN? (Kangawa et al., BBRC 121:585 (1984)). Such AN?s comprise 28 amino acids. Such AN?s may be administered as a peptide having a ring structure of AN? (formation of a disulfide bond based on Cys), and a C- terminal portion succeeding the ring structure. An example of such a peptide is a peptide having amino acid residues at the 7-position to the 28-position of AN? is provided in U.S. Patent Application Publication No.
20010027181. Another example is frog AN?. Specific examples of BN?s that can be used in the methods of the invention include human RN? (hBN?). Human RN? comprises 32 amino acids and involves the formation of a disulfide bond (Sudoh et al., BBRC 159:1420 (1989)) and U.S. Pat. Nos. 5,114,923, 5,674,710, 5,674,710, and 5,948,761, each of which is incorporated by reference. Various BNPs of origin other than human, including as pig RN? and rat RN?, are also known, and can be used. A further example is chicken BNP. Examples of CM's that can be used in the methods of the invention include pig CM'. Pig CM' comprises 22 amino acids and involves the formation of a disulfide bond, like the above- described AN? and RN? (Sudoh et al., BBRC 168:863 (1990)) (human and rat have the same amino acid sequence), chicken CM' (Arimura et al., BBRC 174:142 (1991)). Frog CNP (Yoshihara et al., BBRC 173:591 (1990) can also be used. As discussed herein, one skilled in the art can apply modifications, such as a deletion, substitution, addition or insertion, and/or chemical modification to amino acid residues in the amino acid sequence of a known natriuretic peptide as desired, by known methods. The resulting compound has the activity of acting on a receptor of the starting AN?, RN? or CM'. Analogs having this activity, therefore, are included in the hybrids for use in accordance with the methods of the present invention.
[00214] In another embodiment, the hybrids containing one or more natriuretic functions can be used in treating hypertension. In one embodiment a natriuretic hybrid will have no deleterious effect on heart rate and is not associated with arrhythmias. In one embodiment the hybrid will comprise at least one, two or three natriuretic peptide functions, for example, both AN? and BM' activity. One or more natriuretic hormone functions can be combined with any other hormone function or peptidic enhancer, as described herein. In another embodiment the natriuretic portion(s) is a more stable analog having an extended in vivo half-life when compared with that of a native natriuretic peptide. Analogs that prevent undesirable cleavage by endogenous enzymes such as NE? are also envisioned. The natriuretic containing hybrid~ are also further directed to hypertension reduction, diuresis inducement, natriuresis inducement, vascular
conduct dilatation or relaxation, natriuretic peptide receptors (such as NPR-A) binding, renin secretion suppression from the kidney, aldostrerone secretion suppression from the adrenal gland, treatment of cardiovascular diseases and disorders, reducing, stopping or reversing cardiac remodeling in congestive heart failure, treatment of renal diseases and disorders; treatment or prevention of ischemic stroke, and treatment of asthma. Hybrids can be administered to patients that would benefit from inducing natriuresis, diuresis and vasodilatation. Hybrids can be administered alone or in combination with one or more of the following types of compounds: ACE inhibitors, beta-blockers, diuretics, spironolactone, digoxin, anticoagulation and antiplatelet agents, and angiotensin receptor blockers. Additional diseases or conditions include renal disorders and diseases, asthma, hypertension and pulmonary hypertension. Hybrids are also useful to treat inflammatory-related diseases, erectile dysfunction and hypercholesterolemia.
Peotide Module Selection Considerations, Spacers. and Linking Groups.
[00215] The GD' hybrid polypeptides of the present invention generally comprise at least two bio-active peptide hormone modules of the invention, wherein at least one of the bio-active peptide hormone modules, typically a GD' module, exhibits at least one hormonal activity. Within the context of the present invention, at least one of the bio-active peptide hormone modules will be comprised from a GIl' peptide hormone, analog, derivative, fragment, or peptidic enhancer. The bio-active peptide hormone module that exhibits the at least one hormonal activity may be located at the N-terminal end of the hybrid polypeptide, the C-terminal end of the hybrid polypeptide, or in the event that the hybrid polypeptide comprises more than two bio-active peptide hormone modules, may be located in the internal portion of the hybrid polypeptide.
[00216] In certain embodiments, it may be preferable to locate the bio-active peptide hormone module exhibiting the at least one hormonal activity such that the C-terminal end of the bio-active peptide hormone module is amidated. Amidation of the C-tenninal end of the bio-active peptide hormone module may be accomplished by locating the module at the C-terminal end of the hybrid peptide, or by configuring the module in the C-terminal-to-N-terminal direction at the N-terminal end of the hybrid polypeptide. In both configurations, the C-terminal end of the bio-active peptide hormone module is available for amidation. Specific component peptide hormones where C-terminal amidation may preferably include amylin family peptide hormones, CCK, PYY, hGLP-I(7-36) and hGLP-2. Specific component peptide hormones where C-terminal amidation is not necessarily exemplary (stated otherwise, where elongation at the C-terminal end of the module is easily tolerated) include exendin-4, exendin-4(l-28), Gil', GLP-l(7-37), frog GLP-1(7-36), and frog GLP-2. However, if these component peptide hormones are located at the C-terminal end of the hybrid polypeptide, they may still be optionally amidated, and in fact may preferably be optionally amidated.
[00217] The bio-active peptide hormone modules may be covalently linked in any manner known in the art. Stable linkages may be used, or cleavable linkage may be used. In one embodiment, the carboxy of a first module may be directly linked to the amino of a second module. In another embodiment, linking groups may be used to attached modules. Further, if desired, spacers or turn inducers known in the art may be employed to stabilize the linkage. By way of example, where amidation of the C-terminal end of the N-terminally located bio-active peptide hormone module is not desired, the module may be attached to a second module directly, or using any appropriate linking group known in the art, such as, an alkyl; PEG; amino acid, e.g., Lys, Glu, beta-Ala; polyaminoacids, e.g., poly-his, poly-arg, poly-lys, poly-ala, Gly-LysArg (GKR) etc.; bifunctional linker (see, e.g., Pierce catalog, Rockford, Ii); aminocaproyl ("Aca"), betaalanyl, 8-amino-3,6-dioxaoctanoyl, or other cleavable and non-cleavable linker known in the art. Specifically described herein, as if each were explicitly drawn, are embodiments of specific hybrids in which the linker in each exemplified linker-containing hybrid is replaced by a Gly linker, particularly embodiments where the Gly linker is Gly-Gly-Gly. As an example, for exemplified YAEGTFISDYSIAMDKIRQQDFVNWLLAQK-LinkerKCNTATCVLGRLSQELHRLQTYPRTNTGSETF (SEQ ID NO: 92)(see tables herein) its Gly linker
species analog is also specifically intended and disclosed: this species is YAEGTFISDYSIAMDKIRQQDFVNWLLAQK-Gly-Gly-GlyKCNTATCVLGRLSQELHRLQTYPRTNTGSETF (SEQ ID NO: 92). In one embodiment a linker or spacer is 1 to 30 residues long, in another embodiment 2 to 30 residues, and in yet another 3-30 residues long, and any integer length from 2 to 30 inclusive; each integer unit is contemplated, e.g. 2, 3, 4, 5, 6, 7, etc. In one embodiment a Gly linker is used, and in a particular embodiment a three residue linker GlyGly-Gly.
[00218]In one embodiment as discussed herein the GD' is attached to the second bio-active hormone module in a C-terminus to C-terminus orientation. The C-terminus of a GD' is linked to the C-terminus of a second bio-active hormone module, optionally with a linker. Orthogonal chemistries can be used to ligate functionalized peptide modules, optionally with linkers as described herein. For example, native chemical ligation chemistries can be used. As shown below, where R is a good leaving group, a hybrid can be generated with a Cys-Lys linkage:
[00220] In a further example the hybrid can be prepared using functionalized modules:
[00221] to generate:
[00223] Of particular interest are GIP-exendin family hybrids that are linked C-terminus to C-terminus, keeping thefr respective receptor activation N-termini unhindered. The receptor activation regions of both molecules are pesent in such hybrids, for example using any of the GD' and the exendin and GLP1 family peptides and analogs, active fragments and derivatives herein. For example a dAla(2)-GIP( 1-30) analog can be linked to exendin-4, in a tail to tail manner, via suitable linking chemistry.
[00224] Further, as will be recognized by those of skill in the art, the peptides of the invention may be C-terminally amidated, or may exist as a free acid. In an exemplary embodiment the peptides of the invention are C-terminally amidated. Where amidation of the C-terminal end of N-terminally located bioactive peptide hormone module is desired, the module may again be attached to a second module using any appropriate linking group known in the art. More specifically, in the event that a bio-active peptide hormone module exhibiting at least one hormonal activity has been configured in the C-terminal-to-N-
terminal orientation, resulting in an amino to amino linkage, exemplary linking groups include dicarboxylic acids, alkyls, PEGs, and amino acids such as Lys, Cys, and Glu.
[00225] As mentioned above, the hybrid polypeptides may also preferably include spacer to further stablize the linkage of the bio-active peptide hormone modules. Any spacer or turn inducer known in the art may be used. By way of example, referred beta-turn mimetics include mimic A and mimic B illustrated herein, also Ala-Aib and Ala-Pro dipeptides. Their LUPAC names are Mimic A: N-(35,65,95)-2-oxo-3-amino- 1 -azabicyclo[4.3.0]-nonane-9-carboxylic acid. Mimic B: N-(35,65,9R)-2-


[00227] By agonist is meant a compound which elicits a biological activity of native human hormone. In a exemplary embodiment, the terms refer to a compound which elicits a biological effect in glucose lowering or other activity of native human GD'. Novel GD' analogs for example have activity in a glucose lowering assay, gastric secretion inhibition assay, dP/dt assay, blood pressure assay, insulin secretion assay, bone density assay, or plasma stability assay, preferably similar to or better than native human GD' and/or which binds specifically in a GD' receptor assay or in a competitive binding assay with labeled GD'. Preferably, the agonists will bind in such assays with an affinity of greater than 1 jIM, and more preferably with an affinity of greater than 1-5 nM. In another embodiment the agonist (or antagonist as the case may be) ICSO will be less than or about 100 micromolar, less than or about 50 micromolar, less than about 20 micromolar, and less than or about 10 micromolar. Such agonists may comprise a polypeptide having a GD' sequence with a Trp-cage motif (e.g. exendin tail PSSGAPS (SEQ ID NO: 93) or variant).
[00228] By "amino acid" and "amino acid residue" is meant natural amino acids, unnatural amino acids, and modified amino acid. Unless stated to the contrary, any reference to an amino acid, generally or specifically by name, includes reference to both the D and the L stereoisomers if their structure allow such stereoisomeric forms. Natural amino acids include alanine (Ala), arginine (Mg), asparagine (Asn), aspartic acid (Asp), cysteine (Cys), glutamine (GIn), glutamic acid (Glu), glycine (Gly), histidine (His), isoleucine (Ile), leucine (Leu), Lysine (Lys), methionine (Met), phenylalanine (Phe), proline (Pro), serine (Ser), threonine (Thr), tryptophan (Trp), tyrosine (Tyr) and valine (Val). Unnatural amino acids include, but are not limited to homolysine, homoarginine, azetidinecarboxylic acid, 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisbutyric acid, 2-aminopimelic acid, tertiary-butylglycine, 2,4-diaminoisobutyric acid, desmosine, 2,2'-diaminopimelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylasparagine, homoproline, hydroxylysine, allohydroxylysine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylalanine, Nmethylglycine, N-methylisoleucine, N-methylpentylglycine, N-methylvaline, naphthalanine, norvaline, norleucine, ornithine, pentylglycine, pipecolic acid, thioproline, sarcosine and citrulline. Additional unnatural amino acids include modified amino acid residues which are chemically blocked, reversibly or irreversibly, or chemically modified on their N-terminal amino group or their side chain groups, as for example, N-methylated D and L amino acids or residues wherein the side chain functional groups are chemically modified to another functional group. For example, modified amino acids include methionine sulfoxide; methionine sulfone; aspartic acid- (beta-methyl ester), a modified amino acid of aspartic acid; N-ethylglycine, a modified amino acid of glycine; or alanine carboxamide, a modified amino acid of alanine. Additional residues that can be incorporated are described in Sandberg et al., J. Med. Chem. 41:
[002291 By "Ahx" is meant 6-amino hexanoic acid. As used herein: "5 Apa" means 5 amino-pentanoyl "12 Ado" means 12-amino dodecanoyl, "PEG(8)" mean 3,6,-dioxyoctanoyl, and "PEG(13)" means
amino-4,7, l0-trioxa-13-tridecanamine succinimoyl. In addition are the following abbreviations: "ACN" or 'CH3CN" refers to acetonitrile. "Boc", "tRoc" or "Tboc" refers to t-butoxy carbonyl. "DCU" refers to N,N'-dicyclohexylcarbodiimide. "Fmoc" refers to fluorenylmethoxycarbonyl. "HIBTU" refers to 2-( lHbenzotriazol- 1 -yl)- 1,1 ,3,3,-tetramethyluronium hexaflurophosphate. "HOBt" refers to 1-hydroxybenzotriazole monohydrate. "Homol"' or "HiPro" refers to homoproline. "MeAla" or "Nnie" refers to N-methylalanine. "Naph" refers to naphthylalanine. "pG" or "pGly" refers to pentylglycine. "tRuG" refers to tertiary-butylglycine. "Thiol"' or "tPro" refers to thioproline. "3Hyp" refers to 3-hydroxyproline. "4Hyp" refers to 4-hydroxyproline. "NAG" refers to N-alkylglycine. "NAPG" refers to N-alkylpentylglycine. "Norval" refers to norvaline. "Norleu" refers to norleucine. "OctGly" refers to octyl-glycine in which the gl;ycine amino acid side group H is replaced with an eight carbon saturated aliphatic chain.
Further Exemplary Combinations and Specific Embodiments.
[00230] Exemplary combinations of bio-active peptide hormone modules to form the GD' hybrid polypeptides of the invention include combinations of two or more bio-active peptide hormone modules selected from: native peptide hormones, analogs and derivatives of peptide hormones that exhibit at least one hormonal activity, fragments of native peptide hormones that exhibit at least one hormonal activity, fragments of analogs and derivatives of peptides hormones that exhibit at least one hormonal activity, and peptidic enhancers, with the proviso that at least one module exhibit at least one hormonal activity.
[00231]The hybrid polypeptides of the invention will include at least two bio-active peptide hormone modules, wherein at least one module is comprised from a GD' peptide honnone, analog, derivative, fragment or peptidic enhancer. In one embodiment, at least two of the component peptide hormones are from different peptide hormone families, e.g., the amylin family, CCK, the leptin family, PPF, the proglucagon family, the natriuretic peptide family, and the exendin family. For example, a GD' hybrid can comprise a GD' portion, with or without a tail sequence, combined with a bio-active module that comprises two or more hormone modules (a non-GD' hormone hybrid) such as an exendin-amylin/sCT hybrid or a homone chimera such as an amylin-sCT chimera.
[00232]In certain embodiments, the hybrid polypeptides of the invention may comprise two or more modules that exhibit at least one hormonal activity. For instance, the hybrid polypeptide may comprise a fragment of a first peptide hormone or analog that exhibits at least one hormonal activity covalently, linked to a fragment of at least one additional peptide hormone analog. The additional fragment(s) may optionally exhibit at least one hormonal activity. The first peptide hormone may be the same or different from the additional peptide hormone(s), with the proviso that at least one of the additional peptide hormones are different from the first peptide hormone, and the first hormonal activity may be the same or different from the optional additional hormonal activity.
[00233] In other embodiments, the hybrid polypeptides of the invention may comprise one or more modules that exhibit at least one hormonal activity in combination with one or more peptidic enhancer modules. For instance, a fragment of a first peptide hormone that exhibits a at least one hormonal activity may be covalently linked to a peptidic enhancer, or a fragment of a first peptide hormone that exhibits at least one hormonal activity may be covalently linked to a second peptide hormone that exhibits at least one hormonal activity, which is in turn linked to a peptidic enhancer. Alternatively, a peptidic enhancer may be located between two peptide hormone modules as a stabilizing spacer. Again, the first peptide hormone may be the same or different from the second peptide hormone, and the first hormonal activity may be the same or different from the second hormonal activity.
[002341 In another embodiment, the hybrid polypeptides of the invention may comprise two, three, four, or more bio-active peptide hormone modules. Exemplary combinations include a module with a hormonal activity in combination with one, two, or three peptidic enhancers; two modules with a hormonal activity in combination with one or two peptidic enhancers; three modules with a hormonal activity in combination with one peptidic enhancer, etc.
[OO23SlThe component peptide hormones are preferably selected from amylin, adrenomedullin, calcitonin, calcitonin gene related peptide, intermedin, cholecystokinin, leptin peptide YY, glucagon-like peptide-1, glucagon-like peptide 2, oxyntomodulin, AN?, RN?, CM', urodilatin, GD', GLPl or exendin
[00236] More particularly, exemplary module combinations include those involving combinations of at least GD' and amylin (and/or sCT), RN?, CGRP, CT, CCK, leptin, PYY, GLP1, and exendin-4.
[00237] In exemplary embodiments, a first module comprising a GD' peptide is linked to a second bioactive peptide hormone module comprising an amylin peptide that exhibits at least one hormonal activity. In another embodiment, the second module is further linked to a third bio-active peptide hormone module comprising a calcitonin peptide that exhibits at least one hormonal activity. In yet another embodiment, the third module may be further linked to a fourth bio-active peptide hormone module comprising a peptidic enhancer selected from amylin peptides. In one embodiment, the first module may be located at the C-terminal end of the hybrid polypeptide. Alternatively, the first module may be located at the N-terminal end of the hybrid polypeptide. In certain embodiments, spacers or linkers such as betaAla or Gly may be inserted if desired to link the modules.
[00238] Exemplary exendin-4 peptides include: exendin-4, exendin-4(1-27), exendin-4(l-28), '4Leu,~Phe-exendin-4(l-28), and 5Ala,14Leu,~Phe-exendin-4(1-28). Also useful are exendin(7-15) and its Ser2 analog, HSEGTFTSD (SEQ ID NO. 94). Exemplary amylin peptides that exhibit at least one hormona' activity include amylin, amylin fragments such as amylin(1-17), amylin (1-16), amylin(1-15), and

ainylin( 1-7), and amylin analogs such as pramlintide, 2Ala-h-amylin, 27Ala-h-amylin, and fragments thereof. Exemplary calcitonin peptides that exhibit at least one hormonal activity sCT, sCT fragments such as sCT(8-l0), sCT(8-27), and, and calcitonin analogs such as '1"8Arg-sCT, '8Arg-sCT, '4Glu,18Mg-sCT, 14Glu,11'8Mg- sCT, and fragments thereof. Exemplary amylin peptidic enhancers include amylin(32-37), amylin(33-37), and amylin(34-37), and analogs thereof. Amylin/sCT combinations useful in connection with the present invention include those disclosed in PCT/U52005/00463 1 Amylin Family Agonist, Attorney Docket 18528.835, which is herein incorporated by reference. An amylin/sCT chimera particularly useful for creating hybrids of the invention with GD' is an amylin-sCT-amylin chimera, for example hAmylin(1-7)-1 "'8Arg-sCT(8-27)-Amylin(33-37), which has the sequence KCNTATCVLGRLSQELHRLQTYPRTNTGSNTY (SEQ ID NO: 95), and is preferably in its C-terminal amide form and analogs and derivatives thereof (described herein and in PCT/U52005/00463 1).
[00239] In one aspect, module combinations include those involving a first module comprising GD', a fragment of GD' that exhibits at least one hormonal activity, a Gil' analog or derivative that exhibits at least one hormonal activity, or a fragment of an GD' analog that exhibits at least one hormonal activity in combination with a second module comprising CCK, a fragment of CCK that exhibits at least one hormonal activity, a CCK analog or derivative that exhibits at least one hormonal activity, or a fragment of a CCK analog that exhibits at least one hormonal activity. Exemplary CCK compounds include: CCK8, and CCK-8(Phe(CH2503)). In one embodiment, the first module is located at the C-terminal end of the hybrid polypeptide and the second module is located at the N-terminal end of the hybrid polypeptide. Alternatively, the first module may be located at the N-terminal end of the hybrid polypeptide and second located at the C-terminal end of the hybrid polypeptide. In certain embodiments, spacers or linkers such as beta-Ala or Gly may be inserted if desired to attach the modules.
[00240] In one aspect, exemplary module combinations include those involving a first module comprising GD', a fragment of GD' that exhibits at least one hormonal activity, a GD' analog or derivative that exhibits at least one hormonal activity, or a fragment of an GD' analog that exhibits at least one hormonal activity in combination a second module comprising amylin, a fragment of amylin that exhibits at least one hormonal activity, an amylin analog or derivative that exhibits at least one hormonal activity, or a fragment of an amylin analog that exhibits at least one hormonal activity. The amylin module can be an Amylin/sCT chimera as disclosed herein. In one embodiment, the first module is located at the C-terminal end of the hybrid polypeptide and the peptidic enhancer is located at the N-terminal end of the hybrid polypeptide. Alternatively, the first module may be located at the N-terminal end of the hybrid polypeptide and second located at the C-terminal end of the hybrid polypeptide. In certain embodiments, spacers or linkers such as betaAla or Gly may be inserted if desired to attach the modules.
[00241] Yet other exemplary module combinations include those involving combinations of GD', amylir and calcitonin as tertiary and tetra-hybrid molecules. Exemplary combinations includ
GIP/amylin/calcitonin; GIP/amylin/calcitonin/amylin; amylin/calcitonin/GIP; and amylin/calcitonin/amylin/GIP combinations, with and without spacers or linking groups. Each module may independently be a peptidic enhancer or may exhibit a hormonal activity, depending on the desired properties of the hybrid polypeptide.
[00242] In another embodiment, one of the bio-active peptide hormone module(s) that exhibits at least one hormonal activity is GLP-l or an analog or fragment thereof, and a second bio-active peptide hormone module comprises GD'. In yet another such hybrid, the hybrid polypeptide comprises a third bio-active peptide hormone module. Exemplary third bio-active peptide hormone modules include amylin (including analogs, derivatives and fragments thereof) and amylin-sCT chimeras, PYY (including analogs, derivatives and fragments thereof) and CCK (including analogs, derivatives and fragments thereof).
[00243] Exemplary compounds of GIP-Neuromedin peptide hybrids include
YaGD'( I -30)-beta-Ala-beta-Ala-FN-38: YAEGTFISDYSIAMDKIHQQDFVNWLLAQK-beta-Ala-betaAla-FLFHYSKTQKLGKSNVVEELQSPFASQSRGYFLFRPRN-N112 (SEQ ID NO: 96);
YaGD'( 1 -30)-beta-Ala-beta-Ala-Neuromedin(U25): YAEGTFISDYSIAMDKLHQQDFVNWLLAQKbeta-Ala-beta-Ala-FR VDEEFQSPFASQSRGYFLFRPRN-NH2 (SEQ ID NO: 97); and
YaGD'( I -30)-beta-Ala-beta-Ala-Neuromedin(U-9): YAEGTFISDYSIAMDKIHQQDFVNWLLAQKbeta-Ala-beta-Ala-GYFLFRPRN-NH2 (SEQ ID NO: 98).
[00244] The beta-Ala-beta-Ala spacer is optional, and can be replaced with Gly-Gly-Gly, a mini-PEG group, or other linker known in the art, particularly those described herein.
[00245]Exemplary compounds of GD' and a natriuretic peptide include GD'-hBM' peptide hybrids, including YaGD'(1-30)-beta-Ala-beta-Ala-hBNP where IIBM' is
SPKMVQGSGCFGRKMDRISSSSGLGCKVLRRH (SEQ ID NO: 99) and YaGIP(1-30)-exendin(31-39)-beta-Ala-beta-Ala-hBN?, where hBN? is SPKMVQGSGCFGRKMDRISSSSGLGCKVLRRH (SEQ ID NO: 99). The beta-Ala-beta-Ala spacer is optional, and can be replaced with Gly-Gly-Gly, a mini-PEG group, or other linker known in the art, particularly those described herein.
[00246] Embodiments include:


(Table Removed)

[00247] As has been discussed throughout, in any of the embodiments herein, including the above hybrids, additional changes as discussed can be included. For example, in any of the embodiments herein, including the above hybrids, positions 1 (including the N-terminus), 2 or 3 can be modified to impart Dl'P-IV resistance. For example, specifically exemplified herein are each of the D-Ala2 analogs of each embodiment herein as well as of the above species.
GIl' and Peptidic Enhancer Hybrid.
[002481 In one hybrid embodiment GD' is C-terminally extended by a peptidic enhancer, e.g., a tail or C-terminal portion of another hormone, such as exendin or GLP- 1, which provide analogs that have enhanced in vivo efficacy. Exendin-4 (Figure 1) is a potent incretin mimetic that shares a 53% homology with GLP-l, and mirrors many of the key biological actions of GLP-I. Unlike GLP-1, exendin-4 is much more resistant to proteolytic cleavage to Dl'P-IV peptidase and NEP. This confers greater enhanced pharmacokinetics and improved in vivo efficacy in humans compared to GLP-1. Circular dichroism (CD) and NMR studies of Exendin-4 in aqueous media and in media containing organic cosolvents reveal that the C-terminal segment containing the sequence LFIEWLKNGGPSSGAPPPS (SEQ ID NO: 183) (residues 21-39) forms a unique hydrophobic Trp-cage cluster resulting from interactions of Pro37 with Phe22 and Pro38 with Trp25 (29-31). This Trp-cage cluster motif is the first example of a protein-like tertiary structure displayed by a peptide, and could be responsible for imparting greater metabolic stability by masking protease-sensitive sites in the molecule in vivo (32).
[002491 Accordingly, in one aspect, exemplary module combinations include those involving a first module comprising GD', a fragment of GD' that exhibits at least one hormonal activity, a GD' analog or derivative that exhibits at least one hormonal activity, or a fragment of an GD' analog that exhibits at least one hormonal activity in combination with a peptidic enhancer. Useful peptidic enhancers are the exendin-4 and frog GLP tails, as described herein, as well as PYY(25-3 6), PYY(30-36) and PYY(3 1 -36)~
In one embodiment, the first module is located at the C-terminal end of the hybrid polypeptide and The peptidic enhancer is located at the N-terminal end of the hybrid polypeptide. Alternatively, the first module may be located at the N-terminal end of the hybrid polypeptide and the peptidic enhancer may be located at the C-terminal end of the hybrid polypeptide. In certain embodiments, spacers or linkers such as betaAla or Gly may be inserted if desired to attach the modules.
[00250] A particular compound of interest is

YaEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS-NIi2 (Compound 0601GD'3794; which has a D-Ala at position 2, and is in amide form). Exemplary compounds having a modification at position 2 (i.e. residue 2) and their GD' Receptor binding and receptor activation activity include Y-Residue2-EGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS-NH2 (SEQ ID NO: 184) where Residue 2 is shown in the table (and see Figure 12):


(Table Removed)
[002.51] Further exemplary compounds having a modification at the N-terminus and their GD' Receptor binding and receptor activation activity, include N-TerminusYaEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS-NH2 where the N-terminus modfication is shown in the table (and see Figure 12):

(Table Removed)

[00255] In one embodiment the GD' hybrids or GD' portions thereof have one or more of the following modifications (for reference only to formula N-terminus-YaEGTFISDYSIAMDKIHQQDFVNWLLAQKLinker-PSSGAPPPS-NH2): dA1a2 to Abu, Ala, Gly, or Ser; Metl4 to Leu; Hisl8 to Ala, Arg, or Lys; Asp2I to Glu; Lys3O to Arg or His; an N-terminus as Gly(Oct); and/or a bAla-bAla or Gly-Gly-Gly linker. In a further embodiment one or more of such changes are made to compound 0601GD'3794.
[OOZS6IFurther exemplary modifications (compared to 0601GD'3794) which can be used in the GD' portions of the compounds of the invention, and exemplary compounds containing them, are shown in the table (and see Figure 12): (Table Removed)

bAla-PSSGAPPPS-NH2
(Table Removed)
[00257] Exemplary analogs also include amino acid modifications that eliminate or reduce oxidation. Such replacements include a deletion or replacement of methionine 14 with leucine and/or the tryptophan 25 with phenylalanine. Accordingly, specific exemplary analogs include a variant of each analog described herein by having one or more such replacements. As an example are compounds AAAA and RBBB which are the D-Ala2 and L-Ala2 analogs of 0601GD'3794 having a Phe for Trp replacement at position 25.
[00258] Further exemplary "tail" modifications (compare to 0601 GD'3 794) and exemplary compounds having them are depicted in the table below (and see Figure 12), along with receptor binding and activation data:
(Table Removed)
[00259] Figures 12A- 1 2AT depict further exemplary analogs and reference peptides of the invention. It is


intended that the various modifications and variants shown are to be used in the present invention, and

may be combined as discussed herein. For example, the terms exendin tail or exendin trp-cage motif includes any of the exendin tail variants depicted, which are useful as shield sequences (peptidic enhancers). Of further interest are the frog GLPI C-terminal extensions as shown in the figures, which are yet another example of a shield sequence that can be used in place of an exendin tail. For example, in one embodiment the Gil' compounds of the invention comprise a frog GLP1 "tail" sequence as a peptidic enhancer as described herein (and see Figure 12). Of particular interest are compounds 0601 GD'4 179, 0601GD'4233, 0601GD'4285, 0601GD'4 178, and 0601GD'4467.
[002601 In yet a further embodiment of any of the GD' modules herein, is a Serine2, Aspartic acid3 (i.e. 52,D3), double substitution at positions 2 and 3 at the N-terminus of a GD'. Thus it is expressly intended that for each of the species and formulas depeicted herein, that each 52,D3 analog is explicitly disclosed herein as if it had been written. As with the other DPP-IV resitance modifications described herein, the 52,D3 can be combined with one or two or more other modifications, for example a fatty acyl derivatization to decrease plasma (e.g. renal clearance) and/or increase duration of action, and/or with a C-terminal shield sequence, and/or with a GD' species amino acid substitution. Of course these GD' analogs can be used directly or as the GD' portion of a GD' hybrid.
[00261] Exemplary compounds (see Figure 12 for example) showing exceptional GD' Receptor binding activity at less than about 10 nm include 0601GD'3794 at 3.8 nM, 0601GD'4283 0.41 nM, 0601GD'4284 1.8 nM, 060 lGD'4285 0.58 nM, 0601GD'4288 0.28 nM, 0601GD'4289 0.58 nM, 0601GD'4290 0.35 nM, 0601GIP4147 at 7.4 nM, 0601GD'4178 1.5 nM, 0601GD'4293 0.12 nM, 0601GD'4292 0.17 nM, 0601GD'4238 4.2 nM, 0601GD'4179 3.3 nM, 0601GD'4294 0.92 nM, 0601GD'4252 5.6 nM, 0601GD'3794 3.8 nM, 0601 GD'4190 2.4 nM, 0601GD'4152 2.3 nM, 0601GD'4150 1.6 nM, 0601GD'4 153
0.53 nM, 0601GD'4149 1.5 nM, 0601GD'4176 4.8 nM, 0601GD'4177 2.9 nM, 0601GD'4213 7.9 nM, 0601GIP4215 4.5 nM, 0601GD'4263 0.66 nM, 0601GIP4278 0.24 nM, 0601GD'4264 0.4 nM, 0601GD'4279 2.1 nM, 0601GD'4233 0.1 nM, 0601G1l'4234 3.4 nM, 0601GD'4236 0.62 nM. These compounds also display GD' Receptor activation and appropriate receptor specificity (e.g., lack of binding to GLP1-R or glucagon receptor in absence of appropriate hybrid module).
[00262] As demonstrated herein, DPP-IV resistant hybrids of the invention have increased plasma stablity compared to native GD'. For example, 0601GD'3796 amide form is essentially 100% present after 5 hours in human plasma, in contrast to about 60% for GD'(l -42) acid form.
[00263]Embodiments of the invention further include the following. The following also provides in shorthand notation a description of each species and sub-genus derivable therefrom.
[002641 In one embodiment, the novel GD' analog or a GD' hybrid comprises a polypeptide comprising the formula D-L-C-S, wherein
D comprises a dipeptidyl peptidase IV resistant GD' N-terminal region,
L comprises a linker,
C comprises a GD' C-terminal region, and
S comprises a shield region; and
wherein L is optionally present and at least one of D or C are present, and wherein when C is present then C-S comprises a Trp-cage motif, or when C is absent then L-S further comprises a Trp-cage motif, and the polypeptide has GD' receptor binding and/or activating activity. When at least one of D or C is present, the at least one present D or C has GD' receptor activation and/or binding activity. In another embodiment both D and C are present. Either or both D and C can have GD' receptor activation and/or binding activity. In one embodiment the polypeptide can specifically bind a GD' receptor. Typically this binding will be at least two-fold greater than binding to another receptor such as an incretin receptor, e.g., GLP1R. The binding can be at least 5-, 10-, 50-, or even 100-fold greater than binding to another receptor such as an incretin receptor, e.g., GLP1R. In one embodiment the novel GD' analog comprises GD' agonist activity. In one embodiment the polypeptide can specifically activate a GD' receptor. Typically this activation will be at least two-fold greater than activation of another receptor such as an incretin receptor, e.g., GLP1R. The activation can be at least 3-, 4-, 5-, 10-, 50-, or even 100-fold greater than activation of another receptor such as an incretin receptor, e.g., GLP1R.
[00265] In another embodiment the polypeptide comprises at least one desired activity of a native GD'. When a desired activity of a novel GD' hybrid is greater than that of the native form, the activity can be at least two-fold greater. In further embodiments the desired activity is at least 3-, 4-, 5-, 10-, 50-, or even 100-fold greater than the activity compared to a native GD'. The activity may be receptor binding, receptor activation, receptor antagonism, glucose lowering, inhibition or reduction of gastric secretion, or any other activity, in vitro, ex vivo or in vivo, that may be known in the art or that is provided herein.
(00266] Tn further embodiments the D region of a novel GD' analog or hybrid comprises an 11 N-terminal amino acid sequence of a native Gil' or a 14 N-terminal amino acid sequence of native GD'. In other embodiments the D region comprises the N-terminal portion of a modified or substituted GD', e.g. see WO 00/58360, EP1171465 or published United States Patent Application 20030232761.
[00267] In further embodiments region C comprises C-terminal amino acids 19-26 of native GD', Cterminal amino acids 19-30 of native GD', C-terminal amino acids 19-39 of native GD' or C-terminal amino acids 19-42 of a native GD'. In one such embodiment region C can comprise amino acids 19-26, 19-30, 19-39 or 19-42 of human, mouse, porcine or rat GD'. In other embodiments the C region comprises the C-terminal portion of a modified or substituted GD', e.g. see published United States Patent Application 20030232761.
[00268] In one embodiment L comprises a sequence from native GD', including DKIH or DKIH. In a
further such embodiment the D-L-C portion of a novel GD' analog comprises amino acids 1-26, 1-30, 1-
39, or 1-42 of a native GD'. In one such embodiment regions D-L-C comprise amino acids 1-26, 1-30, 1-
39 or 1-42 of human, mouse, porcine or rat GD' (see for example Li et al., Regulatory Peptides 121(1-3)
pages 107-112 (2004)). In yet a further such embodiment the D-L-C portion of a novel GD' analog
comprises amino acids 1-26, 1-30, 1-39, or 1-42 of an N-terminally modified or substituted GD' (in any of
positions 1.4) that comprises DPP-IV resistance, e.g. see WO 00/58360, EPi 171465 or published United
States Patent Application 20030232761.
[00269] Novel GD' compounds of the formula D-L-C-S can comprise the sequence
YAEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 186),
(Tyr 1 -glucitol)AEGTFISDYSIAMDKJHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 187),
(Tyri -pyroglutamyl)AEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 188),
(Tyrl-glucitol)AEGTFISDYSIAMDKII{QQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 187),
(Tyri -9-fluorenylmethoxycarbonyl)AEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID
NO: 189),
(Tyrl-palmitate)AEGTFISDYSIAMDKil-IQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 190),
YSEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 191), or
YGEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 192).
[00270] In another embodiment novel GD' compounds comprise the sequence
YAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS (SEQ ID NO: 193),
(Tyri -glucitol) AEGTFISDYSIAMDKII-IQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS (SEQ
IDNO: 194),
(Tyr 1 -pyroglutamyl) AEGTFISDYSIAMDKII{QQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS

(SEQ ID NO: 195),
(Tyrl -glucitol) AEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQIDNO: 194),
(Tyri -9-fluorenylmethoxycarbonyl)
AEGTFISDYSIAMDKIJIQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS (SEQ ID NO: 196),
(Tyrl -palmitate) AEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKIIN1TQPSSGAPPPS (SEQ
IDNO: 197),
YSEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQ ID NO: 198), or
YGEGTFISDYSIAMDKIIIQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQ ID NO: 199).
[00271] Novel GD' compounds can also comprise the sequence
Y(D~Ala)EGTFISDYSIAMDKil-IQQDFVNWLLAQKPSSGAPPPS,

YAbuAEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 200), or YSarAEGTFISDYSIAMDKJHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 201).
[00272] In yet another embodiment novel GD' hybrids comprise the sequence
Y(D-Ala)EGTFISDYSIAMDKIiQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS,
YAbuAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQ ID NO:
202), or
YSarAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNlTQPSSGAPPPS (SEQ ID NO:
[00273]In one embodiment of novel Gil' analog or hybrid, D comprises the, sequence X1-X2-X3-X4-XS-X6-X7-XS-X9-X1O-Xl l-X12-X13-X14, wherein at least one of Xl, X2, and X3 is an amino acid substitution or modification providing DPP-IV resistance. In a further such embodiment DPP-IV resistance is provided by a modification comprising a peptide mimetic bond between X2 and X3. Such a bond can be provided as a reduced peptide bond of the formula 'P(CH2NH), for example as when X1-X2-. X3 is Tyrl-Ala'{'(CH2NH)-Glu. In a further embodiment any bond of the novel GD' analog is a reduced peptide bond of the formula '{'(CH2NH), particularly one that is identified as susceptible to protease or peptidase degradation. In a further embodiment the substitution or modification comprises a D-aniino acid substitution in Xl, X2 and/or X3 and/or glycation, alkylation, acetylation or acylation of the N-terminus. Positions XI-X14 can be independently selected. In other embodiments any or more of X12-X14 are optionally absent.
[00274] Of interest are novel GD' analogs or hybrids wherein X2 and X3 are independently Lys, Ser, 4-amino butyric amino acid, Aib, D-Ala, Sarcosine or Pro. The N-terminal modification can be selected from the group of glycation, alkylation, acetylation, alkyloxycarbonylation or arylalkoxycarbonylation.


[00275] In yet another embodiment Xl is Tyr, desamino-Tyr, D-Tyr, Ala, a D-amino acid, Tyr-glucitol, an N-methylated amino acid, and/or comprises an N:terminal glycation, alkylation, acetylation, alkyloxycarbonylation, arylalkoxycarbonylation or acylation. In a further embodiment when Xl is Tyr then the N-terminus of the tyrosine residue can be modified by alkylation, alkyloxycarbonylation, arylalkoxycarbonylation, sulphonylation, glycation, homoserine formation, pyroglutamic acid formation, acylation, methylation, t-butylation, t-butyloxycarbonylation, 4-methylbenzylation, benzyloxymethylation, benzyloxycarbonylation, 4-toluenesulphonylation, diphenylmethylation, 2-bromobenzyloxycarbonylation, 9-fluorenylmethyloxycarbonylation, acylation, formylation, acetylation, benzylation, benzoylation, phosphorylation, sulphation, glycolysation with pentoses, deoxyhexoses, glucosamines, or N-acetylglucosamines, farnesylation, biotinylation, palmitoylation, stearoylation. geranylgeranylation, glutathionylation, and modification with lipoic acid. In yet a further embodiment
N-terminus of the Xl residue can be modified by alkylation, alkyloxycarbonylation, arylalkoxycarbonylation, sulphonylation, glycation, homoserine formation, pyroglutamic acid formation, acylation, methylation, t-butylation, t-butyloxycarbonylation, 4-methylbenzylation, benzyloxymethylation, benzyloxycarbonylation, 4-toluenesulphonylation, diphenylmethylation, 2-bromobenzyloxycarbonylation, 9-fluorenylmethyloxycarbonylation, acylation, formylation, acetylation, benzylation, benzoylation, phosphorylation, sulphation, glycolysation with pentoses, deoxyhexoses, glucosamines, or N-acetylglucosamines, farnesylation, biotinylation, palmitoylation, stearoylation, geranylgeranylation, glutathionylation, and modification with lipoic acid.
[00276] In yet another embodiment X2 is Ala, Ala'P(CH2NH), Ser, D-amino acid, Lys, 4-amino butyric amino acid, Aib, D-Ala, Sarcosine, Pro, Gly, phosphorylated Ser, Val, Leu, lie, Thr, or an Nmethylated amino acid. In a further embodiment X2 is ~ conservative amino acid change from a native residue or from an X2 residue provided herein. In yet another embodiment X2 is any naturally occurring amino acid. In a still further embodiment X2 is any non-proteinogenic amino acid.
[00277] In yet another embodiment X3 is Glu, D-Glu, L-Pro, (N-Me)Glu, Pro, D-amino acid, Lys, Ser, 4-amino butyric amino acid, Aib, D-Ala, Sarcosine, Pro, or an N-methylated amino acid. In a further embodiment X3 is a conservative amino acid change from a native residue or from an X3 residue provided herein. In yet another embodiment X3 is any naturally occurring amino acid. In a still further embodiment X3 is any non-proteinogenic amino acid.
[00278] In yet another embodiment X4 is Gly or Ala. In a further embodiment X4 is a conservative amino acid change from a native residue or from an X4 residue provided herein.
[00279] In yet another embodiment XS is Thr or Ser.
[002801 In yet another embodiment X6 is Phe or Ala. In a further embodiment X6 is a conservative amino acid change from a native residue or from an X6 residue provided herein.
[00281] In yet another embodiment X7 is Ile or Ala. In a further embodiment X7 is a conservative amino acid change from a native residue or from an X7 residue provided herein.
[00282] In yet another embodiment X8 is Ser or Ala. In a further embodiment X8 is a conservative amino acid change from a native residue or from an X8 residue provided herein.


[00283] In yet another embodiment X9 is Asp or Ala. In a further embodiment X9 is a conservative amino acid change from a native residue or from an X9 residue provided herein.
[00284] In yet another embodiment X1O is Tyr or Ala. In a further embodiment XIO is a conservative amino acid change from a native residue or from an XlO residue provided herein.
[00285] In yet another embodiment Xli is Ser or Ala. In a further embodiment Xl 1 is a conservative amino acid change from a native residue or from an Xli residue provided herein.
[00286] In yet another embodiment X12 is Ile, Ala, Ser, or Lys. In a further embodiment X12 is a conservative amino acid change from a native residue or from an Xl 2 residue provided herein. In yet another embodiment X 12 is any naturally occurring amino acid. In a still further embodiment X12 is absent.
[00287] In yet another embodiment Xl 3 is Ala, Tyr, Glutamine, or Asp. In a further embodiment Xl 3 is a conservative amino acid change from a native residue or from an X13 residue provided herein. In yet another embodiment X13 is any naturally occurring amino acid. In a still further embodiment X13 is absent.
[00288] In yet another embodiment X14 is Met, Ala, or Leu. In a further embodiment X14 is a con~ervative amino acid change from a native residue or from an X 14 residue provided herein. In yet another embodiment X14 is any naturally occurring amino acid. In a still further embodiment X14 is absent.
[00289] In certain embodiments a modified D region of a novel Gil' analog and a hybrid comprises the sequence Tyr-Ala-Glu-Gly-Thr-Phe-Ile-Ser-Asp-Tyr-Ser-Ile-Tyr-Met (SEQ ID NO: 204).
[00290] In certain embodiments D comprises the sequence
Ala~Ala-Glu-Gly-Thr-l'he-lIe-Ser-Asp-Tyr-Ser-IIe-Ala-Met (SEQ ID NO: 205);
Tyr~Ala-Ala-Gly-Thr-Phe-Ile-Ser-A5p-Tyr-Ser-Ile-Ala-Met (SEQ ID NO: 206);
Tyr~Ala-Glu-Ala-Thr-Phe-Ile-Ser-A5p-Tyr-Ser-lle-Ala-Met (SEQ ID NO: 207);
Tyr~Ala~Glu-Gly-Ala-Phe-Ile-Ser-A5p-Tyr-Ser-Ile-Ala-Met (SEQ ID NO: 208);
Tyr~Ala-Glu-Gly-Thr-Ala-ile-Ser-A5p-Tyr-Ser-Ile-Ala-Met (SEQ ID NO: 209);
Tyr~Ala~Glu-Gly-Thr-Phe-Ala-Ser-ASp-Tyr-Ser-Ile-AlaMet (SEQ ID NO: 210);
Tyr~Ala~Glu-Gly-Thr-Phe-Ile-Ala-A5p-Tyr-Ser-lle-Ala-Met (SEQ ID NO: 211);
Tyr~Ala~Glu~Gly-Thr-Phe-Ile-Ser-Ala-Tyr-Ser-Ile-Ala-Met (SEQ ID NO: 212);
Tyr~Ala~Glu~Gly-Thr-Phe-Ile-Ser-A5p-Ala-Ser-Ile-Ala-Met (SEQ ID NO: 213);
Tyr~Ala~Glu~Gly~Thr-Phe-Ile-Ser-A5p-Tyr-Ala-Ile-Ala-Met (SEQ ID NO: 214);
Tyr~Ala~Glu~Gly-Thr-Phe-Ile-Ser-A5p-Tyr-Ser-Ala-Ala-Met (SEQ ID NO: 215); or
(SEQ ID NO: 216).
[00291] In still further embodiments D comprises the sequence
TyrSerGluGlyThrPhelleSerAspTyrSerlleAlaMet (SEQ ID NO: 217);
TyrGlyGluGlyThrPheIleSerAspTyrSerIleAlSMet (SEQ ID NO: 218); or
Tyr-DAIa-GluGlyThrPheIleSerASpTyrSerIleAlaMet.
[00292] In further embodiments D comprises the sequence

YAEGTFISDYSIAM (SEQ ID NO: 14), (TyrI-glucitol)AEGTFISDYSIAM (SEQ ID NO: 219), (TyrI:
pyroglutamyl)AEGTFISD (SEQ ID NO: 220), (Tyrl-glucitol)AEGTFISDYSIAM (SEQ ID NO: 219), (Tyr 1 -9-fluorenylmethoxycarbonyl)AEGTFISDYSIAM (SEQ ID NO: 221), (Tyri -
palmitate)AEGTFISDYSIAM (SEQ ID NO: 222), YSEGTFISDYSIAM (SEQ ID NO: 223), or YGEGTFISDYSIAM (SEQ ID NO: 224).
[00293] In yet other embodiments D comprises the sequence Y(D-Ala)EGTFISDYSIAM, YAbuAEGTFISDYSIAM (SEQ ID NO: 225), or YSarAEGTFISDYSIAM (SEQ ID NO: 226).
[002941 In a still further embodiment a novel GD' analog or hybrid region D may exhibit at least 60%, 65%, 70%, 80%, 85%, 90%, 95%, 98% or 100% sequence identity to a corresponding region of a native GD', for example amino acids 1-11, 1-12, 1-13 or 1-14 of a native GD', preferably a human GD', over the entire length of each corresponding sequence. Furthermore, a region D of a novel GD' analog may also exhibit at least 50%, 60%, 65%, 70%, 80%, 85%, 90%, 95% or even 100% sequence identity to a modified or substituted Gil', e.g. see WO 00/58360, EPI 171465 or published United States Patent Application 20030232761. Percent identity can be determined manually or by analysis with the AlignX module in Vector NTI (Invitrogen; Carlsbad CA) or the ClustaIW algorithm for global alignment. Native region D Gil' sequences include those derived from human, mouse, rat, porcine or bovine GD'. Native
GD' sequences include human GD'(1-42)
(YAEGTFISDYSIAMDKIIIQQDFVNWLLAQKGKKNDWKHN1TQ SEQ ID NO: 2), mouse GD'(l42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKHNrrQ; SEQ ID NO: 10), rat GD'(l
42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHNLTQ; SEQ ID NO: 11), pig GD'(1-
42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWKHNLTQ; SEQ ID NO: 12), or bovine GD'(142) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIIHN1TQ; SEQ ID NO: 13). In yet other embodiments region D comprises amino acids 1-11, 1-12, 1-13 or 1-14 of human, mouse, rat, pig or bovine GD', e.g. wherein region D comprises YAEGTFISDYS (SEQ ID NO: 227), YAEGTFISDYSI (SEQ ID NO: 228), YAEGTF'ISDYSIA (SEQ ID NO: 229) or YAEGTFISDYSIAM (SEQ ID NO: 14). In
still further embodiments a region D amino acid sequence further comprises a modified or substituted amino acid in addition to any one of positions Xl, X2 or X3.
[002951 In some embodiments of the novel GD' analog or hybrid L can be absent. In other embodiments L comprises
b) Aha-Aha,
c) Aha-Aha-Aha,
d) Amino alkanoic acid (C5-C12),
e) Glu-Lys-Glu-Lys (SEQ ID NO: 230),
f) Ala-Ala-Ala-Ala (SEQ ID NO: 231),
g) a linker peptide comprising 12 amino acid residues selected from the group consisting of amino acid residues, D-amino acids and non-proteinogenic amino acids,
h) Glu-Lys-Glu-Glu-Lys-Glu-Lys-Glu-Glu-Lys-Glu-Lys (SEQ ID NO: 232),
i) an omega-amino fatty acids (saturated and unsaturated) of omega-NH2-(CHx)n-COOH where n=l0 to 34; or
j) the sequence Xl 5-X I 6-X17-X 18 wherein each of X l5-X 18 is independently any naturally-occurring amino acid, non-proteinogenic amino acid, D-amino acid or is absent.
[00296] In further embodiments XiS is D, E, or a conservative amino acid change thereof or of a native XiS residue. In further embodiments X16 is K, G, E, or a conservative amino acid change thereof or of a native X 16 residue. In further embodiments X17 is I, Q, E, or a conservative amino acid change thereof or of a native Xl 7 residue. In further embodiments Xl 8 is H, R, A or a conservative amino acid change thereof or of a native Xl 8 residue. In novel GD' analogs L can comprise the sequence D-K-I-H or
D-K-I-R. In further embodiments L comprises a modified or substituted amino acid. In one such embodiment L comprises in a first position (e.g. X16) a (hetero)aryl (both optionally substituted) or 3:7C cycloalkyl, 1-lOC (hetero)alkylene, 2-bC (hetero)alkenylene, 2-IOC (hetero)alkynylene or phenyl, and in a second C-terminally adjacent position (e.g. X17) a 1-lOC (hetero)alkylene, 2-lOC (hetero)alkenylene, 2-lOC (hetero)alkynylene, or phenyl linked via a —CO- to the next residue in the amino acid backbone. In still further embodiments a region L amino acid sequence further comprises a modified or substituted amino acid.
[00297] In one embodiment of the invention region C comprises the sequence X19-X20-X21-X22-X23-X24-X25-X26-X27-X28-X29-X30, each position being independently selected, wherein
Xl 9 is Q or a conservative amino acid substitution thereof,
X20 is Q or a conservative amino acid substitution thereof,
X21 is D or a conservative amino acid substitution thereof,
X22 is F, Y or a conservative amino acid substitution thereof,
X23 is V, I, A, or a conservative amino acid substitution thereof,
X24 is N, Q or a conservative amino acid substitution thereof,
X25 is W, F, Y, napthylalanine or a conservative amino acid substitution thereof,
X26 is L, A or a conservative amino acid substitution thereof,
X27 is L, K, R, V, A, I or a conservative amino acid substitution thereof, or is absent
X28 is A, N, D, K, R, E or a conservative amino acid substitution thereof, or is absent
X29 is Q, G or a conservative amino acid substitution thereof or is absent, and
X30 is K, G, R, G, P. R or a conservative amino acid substitution thereof or is absent.
[00298] In a further aspect any one, two, or three of X26 to X30 is absent in region C.
[00299] In yet another embodiment a novel GD' analog region C comprises 8 to 24 C-terminal amino acids of a GD' (beginning at a position in the GD' corresponding to position Xl 9 (GIn) in human GD'), at least the first such 8 amino acids (e.g., comprising positions X19-X26, such as X19-27, X19-X28, X19-29), at least the first 12 such amino acids (e.g., comprising positions X19 to X30, such as X19-X31, X19-X32, X19-X33, X19-X34, X19-X35, X19-X36, X19-X37, X19-X38), at least the first 21 such amino acids (e.g. comprising positions X19 to X39, such as X19-X40, X19-X41) or at least 24 such amino acids. Notably in these embodiments X27-X30 may be independently present or absent. For example, region C can further comprise residues 31 to 39 of a GD' or a native GD', and X27toX30 may be optionally absent.
[00300] In a still further embodiment a novel GIl' analog or hybrid region C may exhibit at least 60%, 65%, 70%, 80%, 85%, 90%, 95%, 98% or 100% sequence identity to a corresponding region of a native GD', for example amino acids 19-30, 19-26, 19-26 or 19-42 of a native GD' over the entire length of each corresponding sequence. Furthermore, a region C of a novel GD' analog may also exhibit at least 50%, 60%, 65%, 70%, 80%, 85%, 90%, 95%, 98% or even 100% sequence identity to a modified or substituted GD', e.g. see WO 00/58360, EPi 171465 or published United States Patent Application 20030232761. Native region C GD' sequences include those derived from human, mouse, rat, porcine or bovine Gil'.
Native GD' sequences include human GIP( 1-42)
(YAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNJTQ; SEQ ID NO: 2), mouse Gil'(1-42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKI-INITQ; SEQ ID NO: 10), rat Gil'(1-
42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHNLTQ; SEQ ID NO: 11), pig Gil'(1-
42) (YAEGTFISDYSIAMDK1RQQDFVNWLLAQKGKKSDWKHNITQ; SEQ ID NO: 12), or bovine GD'(1-42) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIHN1TQ; SEQ ID NO: 13). In yet other embodiments region C comprises amino acids 19-30, 19-26, 19-26 or 19-42 of human, mouse, rat, pig or bovine GD', e.g. region C comprises GlnGlnAspPheValAsnTrpLetlLeuAlaGlnLyS (SEQ ID NO
233) or GlnGlnAspPheValAsnTrpLeuLeuAlaGlnArg (SEQ ID NO: 234). In still further embodiments a region C amino acid sequence further comprises a modified or substituted amino acid.
[00301] In one embodiment of the novel GD' analog or hybrid region S comprises the sequence X3 1-X32-X33 -X34-X35-X36-X37-X38-X39, each independently selected, wherein
X3 1 is Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alk'ylpentylglycine or Nalkylalanine, G, 5;
X32 is Ser, Pro, His, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alkylpentylglycine or Nalkylalanine;
X33 is Ser, Mg, Thr, Trp, Lys;
X34 is Gly, Ser;
X35 is Ala, Mg, Asp, Glu, Lys, Gly;
X36 is Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alkylpentylglycine or Nalkylalanine, A, absent;
X37 is Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alkylpentylglycine or Nalkylalanine, A, absent;
X38 is Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alkylpentylglycine or Nalkylalanine, Ala, Mg, Lys, His, or is absent; and
X39 is Ser, Thr, Tyr, Leu, Ala, Lys, His, Pro, Lys, Mg, Gly, or absent, wherein if an amino acid is absent then all subsequent positions are absent.
[00302] In a further embodiment at least one of X3 1-X39 contains a conservative substitution of an amino acid listed herein for the embodiments of region S. In further embodiments region S comprises ProSerSerGlyAlaProProProSer (SEQ ID NO: I), ProSerSerGlyAlaProProPro (SEQ ID NO: 235), ProSerSerGlyAlaProPro (SEQ ID NO: 236), ProSerSerGlyAlaPro (SEQ ID NO: 237), ProSerSerGlyAla (SEQ ID NO: 238), ProSerSerGly (SEQ ID NO: 239), or ProSerSer. In yet a further embodiment region S comprises a C-terminal amide. In still a further embodiment region S comprises a sequence wherein X37 is Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, N-alkylpentylglycine or N-alkylalanine that is capable of interacting with a Trp or Trp-like residue in region C to form a Trp-cage. In still another embodiment region S comprises a sequence wherein X3 1 is Pro, homoproline, 3Hyp, 4Hyp, thiopro line, N-alkylglycine, N-alkylpentylglycine or N-alkylalanine that is capable of interacting with a Trp or Trplike residue in region C to form a Trp-cage. In yet a further embodiment region S comprises a sequence wherein X3 1 and X37 are both Pro, homoproline, 3Hyp, 4Hyp, thioproline, N-alkylglycine, Nalkylpentylglycine or N-alkylalanine and that is capable of interacting with a Trp or Trp-like residue in
region C to form a Trp-cage. In some embodiments region S comprises a sequence of 5 to 14 amino acids, 5 to 10 amino acids, or 6 to 9 amino acids that is capable of forming a Trp cage with the region C.
[00303] In one embodiment the novel GD' analog or hybrid is modified. The novel GD' hybrid can comprise a modification by fatty acid addition at an epsilon amino group of at least one lysine residue.
[00304] In one embodiment a novel GD' analog or hybrid is a GIP-receptor agonist. The novel Gil' hybrid can potentiate cyclic AMP production. In another embodiment a novel GD' hybrid is a GD'-receptor antagonist.
[00305] In one embodiment a novel GD' analog or hybrid comprises a sequence of the formula D-L-C-S where D comprises a D-amino acid alanine at X2 and wherein D, L, C and S are as defined herein and is any of the other embodiments herein. For example, in a further such embodiment region C comprises amino acids 19-30 of a GD', particularly a human GD', even more particularly comprises the sequence QQDFVNWLLAQK (SEQ ID NO: 15). In a further such embodiment L is naturally occurring sequence X15-X18 from a native GD', particualry human GD'. In a further exemplary embodiment S is a sequence comprising PSSGAPPPS (SEQ ID NO: 1), PSSGAPPP (SEQ ID NO: 235), PSSGAPP (SEQ ID NO:
236), PSSGAP (SEQ ID NO: 237), PSSGA (SEQ ID NO: 238), or PSSG (SEQ ID NO: 239). In yet
another embodiment a novel GD' hybrid comprise the sequence:

Y(D~Ala)EGTFISDYSIAMDKiliQQDFVNWLLAQKPSSGAPPPS
Y(D~Ala)EGTFISDYSIAMDKIRQQDFVNWLLAQRPSSGAPPPS
Y(D~Ala)EGTFISDYSIAMDKlRQQDFVNWLLAQKPSSGAPPPS;
Y(D~Ala)EGTFISDYSIAMDKlRQQDFVNWLLAQKPSSGAPPPS or
Y(D~Ala)EGTFISDYSLAMDKIRQQDFVNWLLAQKPSSGAPPPS.
[00306] In a further example, an embodiment of such a novel D-Ala2-containing GD' analog or hybrid can exhibit at least 50% sequence identity to one of the above analogs and comprises a D-Ala at position X2. In additional embodiments, the embodiment can further comprise the sequence PSSGAPPPS (SEQ ID NO: 1) or KNGGKPSSGAPPPS (SEQ ID NO: 240).
[00307] In one embodiment of a novel GD' analog or hybrid region D comprises the formula X1-X2-X3-X4-X5-X6-X7-X8-X9-X1O-X1 I-X12-X13-X14 wherein Xl and X2 are absent. In a further embodiment region D comprises the formula X1~X2-X3-X4-X5-X6-X7-X8-X9-Xl0Xi 1-X12-X13-X14 wherein Xl and X2 are absent and at least one of X3, X4 or XS is an amino acid substitution or modification providing DPP-IV resistance, for example with a modification or substitution or reduced bond as defined herein. Other amino acid positions are as defined herein. Typically such embodiment of a novel Gi analog or hybrid has GD' antagonist activity.
[00308] In one embodiment D is absent. In another embodiment when D is abs.mt, a novel GD' analog can comprise L-C-S or C-S where region L or C comprises an N-terminal modification, substitution or modification providing protease resistance, particularly DPP-IV resistance. In yet a further embodiment is X18—X19-C-S wherein one or both of X18 and X19 are absent, such that for X18-X19-X20, X19-X20-X2 1, and X20-X2 1 -X22, an amino acid substitution or modification or N-terminal modification of at least one of X18, X19, X20, X21 or X22 provides DPP-IV resistance.
[003091 Additional embodiments include compounds comprising the sequence
Y(DAIa)EGTFISDYSIAMDKilIQQDFVNWLLAQKPSSGAPPPS, Y(pSer)EGTFlSDYSIAMDKJHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 241),
YA(N-MeGlu)EGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 242),
YPEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 243),
YVEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 244),
(D-Tyr)AEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS, or
YAPGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 245).
[003101 Further embodiments include compounds comprising the sequence
Y(DAIa)EGTFISDYSIAMDKII{QQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS,
Y(pSer)EGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKBNITQPSSGAPPPS (SEQ ID NO:
YA(N-MeGlu)EGTFISDYSIAMDKJHQQDFVNWLLAQKGKKNDWKHNlTQPSSGA?PPS (SEQ ID NO: 247),
YPEGTF1SDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQ ID NO: 248),

YVEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS (SEQ ID NO: 249),
(D-Tyr)AEGTFISDYSIAMDKLHQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS, or
YAPGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHN1TQPSSGAPPPS (SEQ ID NO: 250).
[00311] In a further embodiment any of the GD' polypeptides comprises an N-terminal His, D-histidine, desamino-histidine, 2-amino-histidine, beta-hydroxy-histidine, homohistidine, alpha-fluoromethylhistidine, alpha-methyl histidine. Additional embodiments include compounds comprising the sequence
HGEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 251) or HGEGTFISDYSIAMDKLHQQDFVNWLLAQKGKKNDWKHNITQPSSGAPPPS (SEQ ID NO: 252).
[003121 In one embodiment of a novel GD' analog or hybrid, region L and/or C comprises a modified or substituted amino acid. In certain embodiments the novel GD' analog comprises Cys, D-Cys, HomoCys or Penicillamine. In a further embodiment the linker L and/or region C comprises Cys, D-Cys, homoCys or penicillamine. These amino acids are inserted as a site for modification to add peg molecules, lipids or link to other SHi containing molecules such as other peptides or reactive molecules. In another embodiment at least one Cys, D-Cys, HomoCys or Penicillamine is modified with a lipid or a peg molecule or a reactive group. Typically 0, 1, 2 or 3 such residues are present.
[003131 In one such embodiment region L and/or C comprises at least one Cys, D-Cys, homocys or penicillamine residue. In one such embodiment of the novel GD' analog at least one of XIS to X18 or X3 1 to X40 is Cys, D-Cys, homocys or penicillamine. In a further such embodiment, no more than two of XiS to X18 or X31 to X40 is Cys, D-Cys, homocys or penicillamine. In a further embodiment no more than one of XIS to X18 or X31 to X40 is Cys, D-Cys, homocys or penicillamine. In a further such embodiment at least one of XIS to X18 or X3 I to X40 is Cys, D-Cys, homocys or penicillamine and at least one of said residues is modified by a lipid.
[003141 In an embodiment a novel GD' analog or hybrid comprises a pegylated amino acid. At least one PEG molecule can be attached to the polypeptide. In one such embodiment each peg molecule is attached to the compound at a D-Cys, homocys or penicillamine or Lys amino acid or to the carboxy terminal amino acid. In a further such embodiment the at least one PEG molecule is attached to an amino acid in region L and/or C. In a further embodiment a pegylated novel GD' analog has an elimination half-life of at least one hour. Further a novel GD' analog or hybrid can comprise 1, 2, or 3 peg molecules. In a further embodiment at least one of XiS to Xl 8 or X3 1 to X40 is Cys, D-Cys, homocys or penicillanune and at least one of said residues is attached to a peg molecule. Furthermore at least one, two or three of XIS to X18 or X31 to X40 is Cys, D-Cys, homocys or penicillamine and at least one, two or three of said residues is attached to a peg molecule.
[00315]A GD' compound of the present invention can be modified by attaching or coupling a reactive group. A GD' compound is thereby capable of covalently binding to a blood component through the reactive group. The reactive group typically will covalently bond with an amino group, a hydroxyl group, or a thiol group on a blood component, thereby covalently linking the GD' peptide to the blood component. Preferably, the reactive group will react with a thiol group on a blood component. More preferably, the reactive group will react with a thiol group on blood serum albumin. The reactive group may contain any of a number of chemically reactive entities that are capable of forming a covalent bond. Preferably, the reactive group will be capable of reacting with a thiol group on a blood component to form a disulfide bond.
[00316] Reactive groups that are capable of forming disulfide bonds with thiol groups include those having an activated disulfide bond or an 5-sulfonate. Reactive groups having an activated disulfide bond can be derived by coupling a GD' peptide cysteine (or cysteine analog) with an activating group, such as 2,2'-dithiodipyridine (DTDP), 2,2'-dithiobis(5-Nitropyridine) (N?YS), 5,5'-dithiobis(2-nitrobenzoic acid) (Ellman's reagent), or 6,6'-dithiodinicotinic acid. Reactive groups containing an activated disulfide bond are herein referred to as activated disulfide bond groups. In addition, an activated disulfide bond group can be derived by acylating a lysine side chain of a Gil' peptide with a mercapto-activated carboxylic acid. Another exemplary embodiment of the present invention is to utilize a reactive group that is capable of reacting with a thiol group on a blood component to form a thioether linkage. Preferably, such a reactive group will be derived by coupling a Gil' peptide with a chemically reactive entity from a maleimido-containing group, such as gamma-nialeimide- butyrylamide (GMBA), maleimide-benzoylsuccinimide (MBS), gamma-maleimido-butyryloxy succinimide ester (GMBS), and maleimidopropionic acid (MPA). These and other maleimide containing groups are herein referred to as maleimido groups. In an alternative embodiment of the present invention, the reactive group of a GD' compound will be capable of covalently bonding to a primary amine on a blood component to form an amide bond. Preferably, such reactive groups will be derived by coupling a Gil' peptide with N- hydroxysuccinimide (NHS) or Nhydroxysuifosuccinimide (sulfo-NHS) to form an NHS or sulfo-NHS ester. These succinimidyl groups may potentially react with alpha-amine groups on the N-termini of blood component proteins, provided that such amine groups are accessible or available to the reactive group. Preferably, these succinimidyl groups will react with the epsilon-amine of lysine in blood component proteins, since the epsilon-amine of lysine is the only amino acid side chain that reacts significantly with NHS esters. In yet another embodiment GD' compounds of the present invention contain reactive groups that are designed to covalently bond with thiol groups on blood components. Binding to thiol groups is exemplary over binding to amino groups, because thiol groups are less abundant in vivo than are amino groups. Fewer blood components are thereby targeted through binding to thiol groups compared to binding to amino groups, resulting in greater specificity of binding. Accordingly, the exemplary GD' compounds will contain GD' peptides modified with a maleimido group or more preferably, an 5-sulfonate or an activated disulfide bond group. While the GD' compounds of the present invention may bind to any of several blood components that contain a free thiol group, the GD' compounds preferably will covalently bond with the thiol group on serum albumin. Serum albumin is the most abundant blood protein, and contains a single thiol group, located at amino acid residue 34 in the protein (Cys34), which is highly conserved among species. The binding of GD' compounds to serum albumin not only provides specificity of binding, but also provides a reproducible formation of conjugates having a 1:1 binding of GD' compound to serum albumin. The reproducibility of this 1:1 ratio is desirable for use of a GD' cbmpound as a therapeutic, since reproducible conjugates of GD' compound and serum albumin will result upon administration of the GD' compound. Furthermore, the reproducibility of 1:1 conjugates of GD' compound and serum albumin is desirable for ex vivo or in vitro approaches to form conjugates for therapy. Conjugates can be form&
ex vivo by combining GD' compounds of the present invention with blood, allowing formation of the

conjugates, and then administering the conjugate-containing blood tc the host. Alternatively, GD' compound-serum albumin conjugates can also be formed in vitro, by combining Gil' compound with recombinant serum albumin to form conjugates which can be administered. The reproducibility of 1:1 conjugates of GD' compound and serum albumin provides for reproducible conjugates from ex vivo administration or in vitro batch to batch preparation.
[00317] In another embodiment provided are GD' compounds covalently attached to one or more molecules of polyethylene glycol (peg), or a derivative thereof wherein each peg is attached at a Cys or Lys amino acid or the carboxy terminus of the peptide, resulting in pegylated GD' compound with a half-life of at least one hour, at least 4, 6, 10, 15, 20 or 24. In one embodiment a pegylated GD' compound comprises any of the novel GD' compound sequences taught herein with a peg molecule covalently attached at 1, 2 or 3 residues.
[00318] In yet further contemplated embodiments the embodiments presented above or herein .are combined in any consistent combination. For example embodiments describing region D can be combined with embodiments describing region L to describe novel GD' analQg embodiments having the combined changes in region D and in region L. The analogs can be further combined with a second hormone module to form a GD' hybrid.
[00319] In another embodiment, the novel GD' analog or hybrid polypeptides of the invention are at least amino acids 25 amino acids in length. In other embodiments, the polypeptides may be at least 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, and each integer up to 54 amino acids in length (e.g. GD'(1-42) + long exendin tail (27-39)). Further, in one embodiment, the polypeptides of the invention include only natural L amino acid residues and/or modified natural L amino acid residues. Alternatively, in another embodiment, the polypeptides of the invention do not include unnatural amino acid residues.
[003201k yet another embodiment, the novel GD' analog or hybrid may exhibit at least 60%, 65%, 70%, 80%, 85%, 90%, 95% or 98% sequence identity to a native GD'(1-42), GD'(1-30), GD'(1-26), GD'(1-39), GD'(19-30), GD'(19-26), GIP(19-39), GD'(19-42), GD'(1-1 1), or GD'(1-14) over the entire length of each corresponding sequence. Such polypeptides of the invention may also exhibit at least 50%, 60%, 65%, 70%, 80%, 85%, 90%, 95% or 98% sequence identity to a modified or substituted GIP, e.g. see WO
00/58360, EPi 171465 or published United States Patent Application 20030232761. In yet another embodiment the D-L-C region of a novel GD' analog may exhibit at least 60%, 65%, 70%, 80%, 85%, 90%, 95% or 98% sequence identity to a native GD'(1-42), GD'(l-30), GLP(1-26), GD'(1-39), GIP(19-
30), GD'(19-26), GD'(19-39), GD'(19-42), GD'(1-1 1), or GD'(1-14) over the entire length of each corresponding sequence. Native GD' sequences include those derived from human GD'( 1-42)
(YAEGTFISDYS1AMDKLHQQDFVNWLLAQKGKKND~ITQ; SEQ ID NO: 2), human GD'(1
26)(YAEGTFISDYSIAMDKIRQQDFVNWL; SEQ ID NO: 253),
human GD'(l-30)(YAEGTHSDYSIAMDKIHQQDFVNWLLAQK; SEQ ID NO: 3),
human GD'(1-39)(YAEGTFISDYSIAMDKLHQQDFVNWLLAQKGKKNDWKHN; SEQ ID NO: 254), mouse GD'(1-42)(YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKHNITQ SEQ ID NO:
10), mouse GD'(l-26) (YAEGTFISDYSIAMDKIRQQDFVNWL; SEQ ID NO: 255), mouse GD'(1-30) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQR; SEQ ID NO: 256), mouse GD'(1-39) (YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKHN; SEQ ID NO: 257)), rat GD'(1-42) YAEGTFLSDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHNLTQ (SEQ ID NO: 11), rat GD'(1-26) YAEGTFISDYSIAMDKIRQQDFVNWL (SEQ ID NO: 258), rat GIP(1-30) YAEGTFISDYSIAMDKIRQQDFVNWLLAQK (SEQ ID NO: 259), rat GD'(1-39) YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHN (SEQ ID NO: 260), pig GIP(l-42) YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWKHN1TQ (SEQ ID NO: 12), pig GD'(l-26) YAEGTFISDYSIAMDKIRQQDFVNWL (SEQ ID NO: 261), pig GD'(l-30) YAEGTFISDYSIAMDKIRQQDFVNWLLAQK (SEQ ID NO: 262), pig GD'(1-39) YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWKHN (SEQ ID NO: 263), bovine GD'(l-42) YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIHNITQ (SEQ ID NO: 13), bovine GD'(l-26) YAEGTFISDYSIAMDKIRQQDFVNWL (SEQ ID NO: 264), bovine GD'(1-30) YAEGTFISDYSLAMDKIRQQDFVNWLLAQK (SEQ ID NO: 265), or bovine GD'(l-39) YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIHN (SEQ ID NO: 266).
[00321] In yet another embodiment, the S region of a novel GD' analog or hybrid of the invention may exhibit at least 50%, 60%, 65%, 70%, 80%, 85%, 90%, 95% or 98% sequence identity to a native Trpcage sequence, such as PSSGAPPPS (SEQ ID NO: 1).
[00322] In a further embodiment a novel GIl' analog or hybrid polypeptide of the invention comprises a sequence comprising at least 60%, 65%, 70%, 80%, 85%, 90%, 95% or 98% sequence identity to a novel GD' analog sequence disclosed herein. Such novel GD' analog or hybrid sequences, which are also useful for the sequence comparison, include:
YAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNITQPPSGA?PPS (SEQ ID NO: 267) (human GD'( 1-42) core),

YAEGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 186) (human GD'(1-30) core), YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKHNJTQPSSGAPPPS (SEQ ID NO:
268) (mouse GD'(1-42) core),

YAEGTFISDYSIAMDKIRQQDFVNWLLAQRPSSGAPPPS (SEQ ID NO: 269) (mouse GIP(1-30)
core), YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHNLTQPSSGAPPPS (SEQ ID NO:
270) (rat GD'( 1-42) core),
YAEGTFISDYSIAMDKIRQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 271) (rat GD'(l-30) core), YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWKHNITQPSSGAPPPS (SEQ ID NO: 272) (pig GIP( 1-42) core),
YAEGTFISDYSIAMDKIRQQDFVNWLLAQKPSSGAPPPS (SEQ ID NO: 273) (pig GD'(1-30) core),
YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIHNITQPPSGAPPPS (SEQ ID NO: 274)
(bovine GD'(142) core), or YAEGTFISDYSIAMDKIRQQDFVNWLLAQKPPSGAPPPS (SEQ ID NO:
275) (bovine GD'(1-30) core).
[00323] Further novel GD' analog or hybrid sequences, which are also used in the sequence comparisons mentioned above, include:
YAEGTFISDYSIAMDKIHQQDFVNWLKNGGPSSGAPPPS (SEQ ID NO: 276) (human GD'(1-26) core),
YAEGTFISDYSIAMDKIRQQDFVNWLKNGGPSSGAPPPS (SEQ ID NO: 277) (mouse GD'(1-26) core),
YAEGTFISDYSIAMDKIRQQDFVNWLKNGGPSSGAPPPS (SEQ ID NO: 278) (rat GD'(l-26) core), YAEGTFISDYSIAMDKIRQQDFVNWLKNGGPSSGAPPPS (SEQ ID NO: 279) (pig GD'(1-26) core), or YAEGTFISDYSIAMDKJRQQDFVNWLKNGGPPSGAPPPS (SEQ ID NO: 280) (bovine GD'(1-26) core).
[00324] Further novel GD' analog or hybrid sequences, which are also used in the sequence comparisons mentioned above, include:
YAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNPPSGAPPPS (SEQ ID NO: 281)
(human GIP( 1-39) core), YAEGTFISDYSIAMDKIRQQDFVNWLLAQRGKKSDWKHN?SSGAPPP~
(SEQ ID NO: 282) (mouse GD'(1-39) core),
YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKNDWKHNPSSGAPPPS (SEQ ID NO: 283) (rat
GIP(1-39) core), YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWKHNI'SSGAPPPS (SEQ ID

NO: 284) (pig GD'(1-39) core), or
YAEGTFISDYSIAMDKIRQQDFVNWLLAQKGKKSDWIHNPPSGAPPPS (SEQ ID NO: 285) (bovine
GD'(1-39) core).
Further Applicable Considerations and Intentions.
[00325] Within each of the combinations described herein, it is understood that reference to a component peptide hormone or module includes reference to analogs, derivatives, fragments, as well as peptidic enhancers related thereto.
[00326] As mentioned, the novel Gil' analogs are preferably C-terminally amidated, but need not be for the purposes of the instant invention. In other words, the C-terminus of these peptides, may have a free —OH or —NH2 group. These peptides may also have other post-translational modifications. One skilled in the art will appreciate that the novel GD' analog polypeptides of the present invention may also be constructed with an N-terminal methionine residue.
[00327] In yet other embodiments envisioned are variants of each of the sequences where the Gil' portion is modified by one, two or three modifications as described herein. Exemplary modifications are those at the first (including the terminal NH2), second or third N-terminal amino acid of GD' that impart DPP-IV resistance superior to that of native GD'. Of particular interest are GD' compounds as described herein having at least a D-Ala substitution at position 2.
[00328] More particularly, in one aspect, the present invention relates to novel GD' analog polypeptides including one or more amino acid sequence modifications. Such modifications include substitutions, insertions, and/or deletions, alone or in combination. In one aspect the GD' analog or hybrid polypeptide includes one or more modifications of a "non-essential" amino acid residue. In the context of the invention, a "non-essential" amino acid residue is a residue that can be altered, i.e., deleted or substituted, in the native human amino acid sequence without abolishing or substantially reducing the GD' or component peptide hormone receptor agonist activity of the GD' analog or hybrid.
[00329] Substitutions. In one embodiment, the GD' analog or hybrid polypeptides of the invention may have one or more substitutions in the amino acid sequence of native GD', GD'( 1-30), GD'( 1-14), GLl'( 1-26), GIP(l-39), GD'(19-26), GD'(19-30), GIP(19-39) or GD'(19-42) or a region 5, alone or in combination with one or more insertions or deletions. Preferably, the substitution does not abolish or substantially reduce the GD' agonist activity of the GD' analog polypeptide. In one aspect, the present invention relates to GD' analog polypeptides that have a single substitution, or consecutive or non-consecutive substitution of more than one amino acid residues in the amino acid sequence of native human GD'. Preferably, the GD' analog polypeptides of the invention include one, two, or three amino acid substitutions. In one embodiment the native GD' is human, rat, mouse, porcine or bovine.
[00330] Particulary useful substitutions include conservative amino acid substitutions. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain, or physicochemical characteristics (e.g.. electrostatic, hydrogen bonding isosteric, hydrophobic features). Families of amino acid residues having similar side chains are known
the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, methionine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). In yet other embodiments exemplary conservative substitutions are shown in the following table under the column "Exemplary Substitutions". In still other embodiments conserved substitutions are selected from the amino acids listed in the column labeled "Preferred Substitutions."

(Table Removed)
[00331] Optionally, a novel GD' analog or hybrid will have no more than one conservative amino acid substitution as compared to the sequence against which is being compared, alternatively no more than 2, 3, 4, 5, 6, 7, 8, 9, or 10 conservative amino acid substitution as compared to the sequence against which is being compared.
[00332] In another embodiment, the GD' analog or hybrid polypeptides of the invention may include substitutions of one or more unnatural and/or non-amino acids, e.g., amino acid mimetics, into the sequence of GD'. In a exemplary embodiment, the non-amino acids inserted into the sequence of GD' may be beta-turn mimetics or linker molecules, such as —NH-X-CO-, wherein X = (CH2)0 (where n can be 2-20) or ~ (where m = 1-5). Exemplary linker molecules include aminocaproyl ("Aca"), beta-alanyl, and 8-amino-3,6-dioxaoctanoyl. beta-turn mimetics are available commercially (RioQuadrant Inc, Quebec, Canada) and have been described in literature (Hanessian et al. Tetrahedron 12789-854 (1997); Gu et al., Tetrahedron Letters 44: 5863-6 (2003); Bourguet et al'.,
Bioorganic & Medicinal Chemistry Letters 13: 1561-4 (2003); Grieco et at., Tetrahedron Letters 43:
6297-9 (2002); Souers et at., Tetrahedron 57: 7431-48 (2001); Tsai et aL, Bioorganic & Medicinal Chemistry 7: 29-38 (1999); Virgilio et at., Tetrahedron 53: 6635-44 (1997)). Exemplary beta-turn mimetics include mimic A and mimic B illustrated herein. Their LUPAC names are Mimic A: N(35,65,95)-2-oxo-3-amino- 1 -azabicycloll4 .3 .0]-nonane-9-carboxylic acid. Mimic B: N-(35,65,9R)-2-oxo-3-amino-7-thia- 1 -azabicyclo[4.3.0)-nonane-9-carboxylic acid.
[00333] Exemplary GD' analog or hybrid polypeptides comprising amino acid sequence beta-turn mimetic substitutions include native human GD', wherein amino acids at positions x and x-i-l are substituted with beta-turn mimetics selected from the group consisting of mimic A and mimic B, wherein x is selected from the amino acids at amino acid positions 8 to 14 of native human GD'. (In addition to mimic A and B, Ala-Aib and Ala-Pro dipeptides are good turn inducers). These linkers are particularly useful to comprise region the region "L" of the D-L-C-S novel GD' analogs of the invention.
[00334jJ Deletions and Truncations. In another embodiment, the GD' analog or hybrid polypeptides of the invention may have one or more amino acid residues deleted from the amino acid sequence of native GD', or a region 5, alone or in combination with one or more insertions or substitutions. In one aspect, the GD' analog or hybrid polypeptides of the invention may have one or more amino acid residues deleted from the N-terminus or C-terminus of a native GJLP. In another embodiment, the GD' analog or hybrid polypeptides of the invention may have one or more amino acid residues deleted at amino acid positions 1 through 42 of a native GD', GD'(1-14), GD'(1-26), GD'(t-30), GD'(1-39), GIP(19-26), GD'(19-30), GD'(19-39) or GIP(19-42) or a region S. Such deletions may include more than one consecutive or non-consecutive deletions. in a exemplary embodiments no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 amino acids are deleted from a native GD', from GIP(1-30), GD'(1-14), GIP(l-26), GD'(1-39), GLP(19-30), GD'(19-26), GIP(19-39) or GD'(19-42) or from a region S as when region is exendin(3 1-39) or exendin(27-39) for example. In one embodiment the native GD' is human, rat, mouse, porcine or bovine.
[00335]Insertions. In another embodiment, the GD' analog or hybrid polypeptides of the invention may have one or more amino acid residues inserted into the amino acid sequence of native GD' from GD'( 1-30), GLP(1-14), GIP(l-26), GLP(1-39), GLP(19-30), GD'(19-26), GIP(19-39) or GIP(19-42) or region 5, alone or in combination with one or more deletions and/or substitutions. In one aspect, the present invention relates to GD' analog or hybrid polypeptides that have a single insertion, or consecutive or nonconsecutive insertions of more than one amino acid residues into the amino acid sequence of native GD', GLP(l-30), GIP(1-14), GIl'(1-26), GIP(l-39), GIP(19-30), GD'(19-26), GD'(19-39) or GIP(19-42), or region S, for example exendin(27-39) and exendin(3 1-39). In one embodiment the native GD' is human, rat, mouse, porcine or bovine-.
[00336] In another embodiment, the GD' analog or hybrid polypeptides of the invention may include insertions of one or more unnatural amino acids and/or non-amino acids into the sequence of GD', GD'( I -30), GD'(l-14), GD'(l-26), GD'(1-39), GD'(19-30), GD'(19-26), GD'(19-39) or GIP(19-42), or a region 5, for example exendin(27-39) and exendin(31-39). In a exemplary embodiment, the unnatural amino acids inserted into the sequence of GD', GD'(1-30), GD'(1-14), GD'(l-26), GD'(1-39), GIP(19-30), GD'(19-26), GD'( 19-39) or GD'( 1942) or region 5, for example exendin(27-39) and exendin(3 1-39) may be beta-turn mimetics or linker molecules. In further such embodiments the native Gil' can be human, rat, mouse, porcine or bovine. In some embodiments region S retains at least Proline (or proline analog) at X37 in order to interact with a Trp (or Trp analog) to favor Trp cage formation. In further exemplary embodiments a Pro is retained at position X3 I to also interact with Trp at X25. In the case of N-terminally truncated exendin-4 analogs (Trp-cages), it has been established that the helix is not significantly populated unless it is capped by either a Trp2SfPro3 1 hydrophobic staple or the complete formation of the Trp-cage. The latter, complete Trp-cage formation, serves as a very effective helix C-cap. There is also evidence for the contribution of the half-cage structure, with the Pro at X36, X37, X38 unit undocke&, in partially melted Trp-cage species.
[00337] Accordingly, while compounds are shown with optional linking groups, in one embodiment of the sequences herein, the linker is a Gly linker, for example Gly-Gly-Gly, or a betaAla linker, for example betaAla-betaAla; all of which are specifically envisioned. Linker molecules of particualr interest include aminocaproyl ("Aca"), beta-alanyl, and 8-amino-3,6-dioxaoctanoyl. Further in other embodiments a beta-turn mimetic is used, which includes mimic A: N-(35,65,95)-2-oxo-3-amino-1-azabicyclo[4.3.0]-nOnane-9-carboxylic acid, mimic B: N-(35,65,9R)-2-oxo-3-amino-7-thia- 1 -azabicyclo[4.3 .0]-nonane-9-carboxylic acid, and also Ala-Aib and Ala-Pro dipeptides.
[00338] In another embodiment, GD' analog or hybrid polypeptides of the invention may include insertions of polyamino acid sequences (e.g., poly-his, poly-arg, poly-lys, poly-ala. etc.) at either terminus of the polypeptide, known as "extensions" or "tails."
[00339] In some embodiments novel GD' analog or hybrid polypeptides comprising amino acid sequence insertions include an alanine substitution at each amino acid position along the length of native GD', GIP(l-30), GIP(l-14), GD'(I-26), GD'(l-39), GD'(19-30), GD'(19-26), GIP(19-39) or GIP(19-42), or region 5, for example exendin(27-39) and exendin(3 1-39).
[00340] Derivatives. The present invention also relates to derivatives of the GD' analogs and hybrid polypeptides. Such derivatives include GD' analog and hybrid polypeptides conjugated to one or more water soluble polymer molecules, such as polyethylene glycol ("PEG") or fatty acid chains of various lengths (e.g., stearyl, palmitoyl, octanoyl, etc.), or by the addition of polyamino acids, such as poly-his, poly-arg, poly-lys, and poly-ala. Modifications to the polypeptides can also include small molecule substituents, such as short alkyls and constrained alkyls (e.g., branched, cyclic, fused, adamantyl), arn~
aromatic groups. The water soluble polymer molecules will preferably have a molecular weight ranging from about 500 to about 20,000 Daltons.
[00341] Such polymer-conjugations and small molecule substituent modifications may occur singularly at the N- or C-terminus or at the side chains of amino acid residues within the sequence of the GD' analog and hybrid polypeptides. Alternatively, there may be multiple sites of derivatization along the GD' analog and hybrid polypeptide. Substitution of one or more amino acids with lysine, aspartic acid, glutamic acid, or cysteine may provide additional sites for derivatization. See, e.g., U.S. Patent Nos. 5,824,784 and 5,824,778. Preferably, the GD' analog and hybrid polypeptides may be conjugated to one, two, or three polymer molecules.
[00342] The water soluble polymer molecules are preferably linked to an amino, carboxyl, or thiol group, and may be linked by N or C termini, or at the side chains of lysine, aspartic acid, glutamic acid, or cysteine. Alternatively, the water soluble polymer molecules may be linked with diamine and dicarboxylic groups. In a exemplary embodiment, GD' analog and hybrid polypeptides of the invention are conjugated to one, two. or three PEG molecules through an epsilon amino group on a lysine amino acid.
[003431 GD' analog and hybrid polypeptide derivatives of the invention also include GD' analog and hybrid polypeptides with chemical alterations to one or more amino acid residues. Such chemical alterations include amidation, glycosylation, acylation, sulfation, phosphorylation, acetylation, and cyclization. The chemical alterations may occur singularly at the N- or C-terminus or at the side chains of amino acid residues within the sequence of the GD' analog and hybrid polypeptides. In one embodiment, the C-terminus of these peptides may have a free -OH or —NH2 group. In another embodiment, the N-terminal end may be capped with an isobutyloxycarbonyl group, an isopropyloxycarbonyl group, an nbutyloxycarbonyl group, an ethoxycarbonyl group, an isocaproyl group (isocap), an octanyl group, an octyl glycine group (G(Oct)), or an 8-aminooctanic acid group or a Fmoc group. In a exemplary embodiment, cyclization can be through the formation of disulfide bridges. Alternatively, there may be multiple sites of chemical alteration along the GD' analog and hybrid polypeptide.
[00344] A number of pseudopeptide bonds have been described that in general do not affect peptide structure and biological activity. One example of this approach is to substitute retro-inverso pseudopeptide bonds ("Biologically active retroinverso analogues of thymopentin", Sisto A. et al in Rivier, J. E. and Marshall, G. R. (eds) "Peptides, Chemistry, Structure and Biology", Escom, Leiden (1990), pp. 722-773) and Dalpozzo, et al. (1993), Int. J. Peptide Protein Res., 41:561-566, incorporated herein by reference). According to this modification, the amino acid sequences of the peptides may be identical to the sequences of a GD' described herein, except that one or more of the peptide bonds are replaced by a retroinverso pseudopeptide bond. Preferably the most N- terminal peptide bond is substituted, since such a substitution will confer resistance to proteolysis by exopeptidases acting on the N- terminus. Further modifications also can be made by replacing chemical groups of the amino acids with other chemicNd groups of similar structure. Another suitable pseudopeptide bond that is known to enhance stability to
enzymatic cleavage with no or little loss of biological activity is the reduced isostere pseudopeptide bond (Couder, et al. (1993), Int. J. Peptide Protein Res., 41:181-184, incorporated herein by reference in its entirety).
[00345]Thus, the amino acid sequences of these peptides may be identical to the sequences of a novel GD' analog and hybrid peptide, except that one or more of the peptide bonds are replaced by an isostere pseudopeptide bond. Preferably the most N-terminal peptide bond is substituted, since such a substitution would confer resistance to proteolysis by exopeptidases acting on the N-terminus. The synthesis of peptides with one or more reduced isostere pseudopeptide bonds is known in the art (Couder, et al. (1993), cited above). Other examples include the introduction of ketomethylene or methylsulfide bonds to replace peptide bonds.
[00346] In another embodiment the bond between the second and third residues that is a target for cleavage by DPP-IV is replaced to a peptidase-resistant bond as disclosed herein.
[00347] Peptoid derivatives of Gil' analog and hybrid peptides represent another class of peptide mimetics that retain the important structural determinants for biological activity, yet eliminate the peptide bonds, thereby conferring resistance to proteolysis (Simon, et al., Proc. NatI. Acad. Sci. USA, 89:9367-9371 (1992), incorporated herein by reference in its entirety). Peptoids are oligomers of N-substituted glycines. A number of N-alkyl groups have been described, each corresponding to the side chain of a natural amino acid (Simon, et al. (1992), cited above). Some or all of the amino acids of the GD' peptides may be replaced with the N-substituted glycine corresponding to the replaced amino acid.


[00348] In one embodiment the novel GD' analog or hybrid polypeptides include combinations of the above-described modifications, i.e., deletion, insertion, and substitution.
[00349] Also included within the scope of the invention are GD' analog or hybrid polypeptides of the formulas wherein the indicated amino acid residue is chemical modified or derivitized (e.g., through fatty acid derivitization, PEGylation, amidation, glycolization, etc.). Exemplary embodiments include deriviatization of a lysine residue, particualry at position 16 or 30. Also contemplated within the scope of the invention are D-amino acid residues of the indicated amino acids. In another embodiment, exemplary GD' analog or hybrid polypeptides include the polypeptides of the formulas with internal deletions, particularly in areas not corresponding to the active sites as described herein.
[00350] Exemplary GD' analog or hybrid polypeptides comprising substitutions of unnatural amino acids. Exemplary derivatives of the GD' analog or hybrid polypeptides of the invention include polymerconjugated GD' analog or hybrid polypeptides, wherein the GD' analog or hybrid polypeptide includes any of the above-described insertions, deletions, substitutions, or combinations thereof, and the polymer molecule is conjugated at a lysine residue.
[00351] Further specifically envisioned are D-Ala2 variants of each GD' sequence herein (e.g., see tables) In yet other embodiments envisioned are variants of each of the above sequences where the GD' portion
modified by one, two or three modifications as described herein. Exemplary modifications are those at the first, second or third N-terminal amino acid of GD' that impart DPP-IV resistance superior to that of native GD'. In yet a further embodiment the novel GD' compounds comprise a C-terminal amide.
[003521 In a further embodiment the novel GD' analog or a GD' hybrid comprises a half-life at least twice that of human GD'(1 -30)amide. Further the half-life can be at least 6 hours.
[00353] In another embodiment is a pharmaceutically acceptable salt of a novel GD' analog or hybrid. The novel GD' analogs and hybrids can be formulated in a composition comprising a pharmaceutically acceptable carrier.
[00354] In one embodiment an analog of exenatide with leucine substituted for Met at position 14 is used either as a hybrid component or in adjunct therapy with a GIl' hybrid:
HGEGTFTSDLSKQLEEEAVRLFIEWLKNGGPSSGAPl'PS-NH2 (SEQ ID NO: 286).
[00355] Preferably, the GD' analog or hybrid polypeptides of the invention retain at least about 25%, preferably about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% percent of the biological activity of native human GD' with regard to glucose lowering. In another embodiment, the GD' analog or hybrid polypeptides of the invention exhibit improved GD' agonist activity. Preferably, the Gil' analog or hybrid polypeptides of the invention exhibits at least about 110%, 125%, 130%, 140%, 150%, 200%, or more of the biological activity of native human GD'. Conversely, the novel Gil' analog or hybrids can be antagonists.
[00356] Exemplary Gil' analog or hybrid polypeptide are those having a potency which is equal to or greater than the potency of GD'(l-42) or GD'(1-30) in that same assay. Alternatively, exemplary GD' analog or hybrid polypeptides of the invention may exhibit improved ease of manufacture, stability, and/or ease of formulation, as compared to GIP( 1-42) or GD'( 1-30).
[00357] It is also contemplated that the novel GD' analogs of the invention, as well as GD' analogs, can be administered with agents such as small molecules or antibodies that are agonists or antagonists, as may be the case, for the peptide hormones and growth factors mentioned herein.
[00358]In further embodiments and uses of the GD' hybrids having the naturally-occurring C-terminal amino acid sequence of exendin-4, particularly PSSGAPPPS (SEQ ID NO: 1) sequence, described herein, specifically excluded are those having a modification at the "P' 1,' position as described in
W02004/103390.
[00359] Further examples of the analog and hybrid polypeptides of the present invention are provided in the Sequence Listing, Tables and in the Examples section.
Further Uses of Hybrid Polypeptides in the Treatment or Prevention Disease Conditions or Disorders.
[00360] Metabolic diseases and disorders take on many forms, including obesity, diabetes, dyslipidemia, insulin resistance, cellular apoptosis, etc. Obesity and its associated disorders are common and v&.

serious public health problems in the United States and throughout the world. Upper body obesity is the strongest risk factor known for type 2 diabetes mellitus, and is a strong risk factor for cardiovascular disease. Obesity is a recognized risk factor for hypertension, atherosclerosis, congestive heart failure, stroke, gallbladder disease, osteoarthritis, sleep apnea, reproductive disorders such as polycystic ovarian syndrome, cancers of the breast, prostate, and colon, and increased incidence of complications of general anesthesia (see, e.g., Kopelman, Nature 404: 635-43 (2000)). It reduces life-span and carries a serious risk of co-morbidities above, as well disorders such as infections, varicose veins, acanthosis nigricans, eczema, exercise intolerance, insulin resistance, hypertension hypercholesterolemia, cholelithiasis, orthopedic injury, and thromboembolic disease (Rissanen et al., Br. Med. J. 301: 835-7 (1990)). Obesity is also a risk factor for the group of conditions called insulin resistance syndrome, or "Syndrome X." Recent estimate for the medical cost of obesity and associated disorders is $150 billion worldwide. The pathogenesis of obesity is believed to be multifactorial but the basic problem is that in obese subjects nutrient availability and energy expenditure do not come into balance until there is excess adipose tissue. Obesity is currently a poorly treatable, chronic, essentially intractable metabolic disorder. A therapeutic drug useful in weight reduction of obese persons could have a profound beneficial effect on their health.
[00361] Diabetes is a disorder of carbohydrate metabolism characterized by hyperglycemia and glucosuria resulting from insufficient production or utilization of insulin. Diabetes severely affects the quality of life of large parts of the populations in developed countries. Insufficient production of insulin is characterized as type 1 diabetes and insufficient utilization of insulin is type 2 diabetes. However, it is now widely recognized that there are many distinct diabetes related diseases which have their onset long before patients are diagnosed as having overt diabetes. Also, the effects from the suboptimal control of glucose metabolism in diabetes gives rise to a wide spectrum of related lipid and cardiovascular disorders.
[003621 Dyslipidemia, or abnormal levels of lipoproteins in blood plasma, is a frequent occurrence among diabetics. Dyslipidemia is typically characterized by elevated plasma triglycerides, low HDL (High Density Lipoprotein) cholesterol, normal to elevated levels of LDL (Low Density Lipoprotein) cholesterol and increased levels of small dense, LDL (Low Density Lipoprotein) particles in the blood. Dyslipidemia is one of the main contributors to the increased incidence of coronary events and deaths among diabetic subjects. Epidemiological studies have confirmed this by showing a several-fold increase in coronary deaths among diabetic subjects when compared with non-diabetic subjects. Several lipoprotein abnormalities have been described among diabetic subjects.
[003631 Insulin resistance is the diminished ability of insulin to exert its biologically action across a broad range of concentrations. In insulin resistance, the body secretes abnormally high amounts of insulin to compensate for this defect and a state of impaired glucose tolerance develops. Failing to compensate for the defective insulin action, the plasma glucose concentration inevitable rises, resulting in the clinical state of diabetes. It is being recognized that insulin resistance and relative hyperinsulinemia have a contributory role in obesity, hypertension, atherosclerosis and type 2 diabetes. The association of insulin resistance
with obesity, hypertension and angina has been described as a syndrome, Syndrome X, having insulin resistance as the common pathogenic link.
[00364] Apoptosis is an active process of cellular self-destruction that is regulated by extrinsic and intrinsic signals occurring during normal development. It is well documented that apoptosis plays a key role in regulation of pancreatic endocrine beta cells. There is increasing evidence that in adult mammals the beta-cell mass is subject to dynamic changes to adapt insulin production for maintaining euglycemia in particular conditions, such as pregnancy and obesity. The control of beta cell mass depends on a subtle balance between cell proliferation, growth and programmed cell death (apoptosis). A disturbance of this balance may lead to impairment of glucose homeostasis. For example, it is noteworthy that glucose intolerance develops with aging when beta cell replication rates are reduced and human autopsy studies repeatedly showed a 40-60% reduction of beta cell mass in patients with non-insulin-dependent-diabetes mellitus compared with nondiabetic subjects. It is generally agreed that insulin resistance is an invariable accompaniment of obesity but that normoglycemia is maintained by compensatory hyperinsulinemia until the beta cells become unable to meet the increased demand for insulin, at which point type 2 diabetes begins.
[OO36SiAttempts to treat the multiple abnormalities associated with diabetes have prompted for the administration of several anti-diabetic medicaments in order to address these abnormalities in the different patients. However, the Gil' analogs, GD' hybrids and GIPR agonists as discussed herein find use, when administered at therapeutically effective amounts, either in monotherapy or in adjunct therapy, in treating or preventing these and other diseases and conditions discussed throughout.
[003661 Gastric inhibitory polypeptide (GD') and glucagon-like peptide 1 (GLP- 1) are gut peptide hormones that exert potent glucoregulatory action through their glucose-dependant stimulation of insulin secretion. Studies have shown that GD' and GLP- 1 act in concert to exert their incretin effects [1-3]. Consequently, these incretin hormones have attracted considerable interest as potential anti-diabetic agents with reduced risk for hypoglycemia. Whereas GLP- 1, GLP- 1 analogs and mimetics such as exenatide have been shown to be efficacious in controlling glucose levels in type 2 diabetic patients, the insulinotropic effect of GD' is significantly reduced in diabetic subjects, compared to normal individuals [4-6]. The preservation of insulinotropic action of GLP- 1 but not of GD' in the same diabetic subjects suggests that GD' signal transduction is impaired in type 2 diabetes. Reduced GD' receptor expression in pancreatic beta cells has been proposed to contribute to overall reduced incretin effects in diabetic subjects [7]. This hypothesis is supported by rodent studies showing decreased GD' receptor expression on beta-cells in diabetic fatty Zucker rats and reduced GD' incretin effect seen in first-degree relatives of patients with type 2 diabetes [8-9]. However, a recent study has shown significant increases in plasma insulin levels in type 2 diabetics to a bolus intravenous administration of GD', that is in marked contrast to weak increase in insulin secretion with continuous GD' infusion [10]. The similar relative beta-cell sensitivity towards GD' bolus in type 2 diabetic patients and healthy control subjects suggest that a specific GP receptor defect appears unlikely [101. Rather, the differences in insulin secretion after acute and durii~
continuous GD' administration indicate an impaired amplification of the late phase insulin response to glucose by GD' in type 2 diabetic patients, whereas the response in the early phase is almost preserved [11-121. While not to be bound by theory, it is believed that while GIP's incretin effect is attenuated during persistent hyperglycemia, there is potential for GD' or its analogs to act with a similar potency in diabetic patients as their action in nonnal subjects once glucose control is improved in these individuals.
[00367] Amylin Pharmaceuticals, Inc. has conducted three pivotal clinical studies to evaluate the effects of exenatide in patients with type 2 diabetes not achieving target blood glucose concentrations using inetformin alone, sulfonylureas alone, or using a combination of metformin and a sulfonylurea. All three studies met the primary glucose control endpoint as measured by HibAl c. The average reduction in HbA I c across the 'Phase 3 program in patients completing the studies on the highest dose of exenatide (10 ~sg twice daily) was approximately one percent. Additionally, approximately 40 percent of these patients achieved HbAlc measurements of 7 percent or less. The clinical data indicate that despite the efficacy of exenatide, some diabetic individuals fail to attain normal glucose concentrations.
[00368] In an embodiment of the invention, reduction of hyperglycemia (e.g., as by exenatide) in treated diabetic patients sets the stage for GD' intervention. Whereas the chronic hyperglycemic condition in type 2 diabetes patients attenuates s insulinotropic response, improved glycemic control resulting from exenatide treatment would restore responsiveness of the pancreatic beta-cell to GD' stimulation. Therefore adjunct therapy, e.g. co-administration, Gil' phybrids, of pharmacological doses of GD' or novel GD' analog or hybrids with exenatide (or other glucose lowering agents or agents or methods that reduce or inhibit gastric emptying) will lead to desired normoglycemia in diabetic patients or patients suffering from conditions associated with elevated glucose. Of note, GD' lacks the gastric emptying effect of GLP- 1 [13-141 that is a possible contributing factor to the incidence of nausea during GLP-1 treatment and which limits the peptide's therapeutic window [15]. Thus, it should~ permit the use of higher GD' dosing regimens.
[00369] Since currently prescribed anti-diabetic agents (metformin, sulfonyureas, TZDs, etc) are able to achieve various degrees of glycemic control, the combination of GD' or novel GD' analog or hybrids with any of these therapies should also elicit an improved response that leads to normalization of glu&ose levels.
[00370] Accordingly, in one embodiment the methods of the present invention are based on the notion that patients can be primed for GD' therapy through prior glucose lowering with other anti-diabetic agents, such as GLl'- 1, a GLl'- 1 analog or exendin-4 or other agents, e.g. metformin, sulfonyureas, thiazolidinediones (TZDs), pramlintide, insulin, acarbose, dipeptidyl peptidase (DPP-IV) inhibitors. DPPIV inhibitors are well-known and described for example in published application U5200500041 17, US Patent 6710040, and US Patent 6645995, which are incorporated herein by reference for their compounds. As example of a sulfonylureas (SEUs), which acts on the pancreatic tissue to produce insulin, is Glimepiride.
[00371] Adjunct therapy of a metabolically stable GD' analog or hybrid or novel GD' analog or hybrid with exenatide (or other antidiabetics) will provide for increased insulinotropic responses than either alone, in patients with conditions associated with elevated glucose, such as patients with type 2 diabetes. Such a treatment regimen leads to normalization of glucose concentrations, improvement in beta-cell function, and through their trophic activity on the I~ cells slows disease progression to obviate or lesson the need for insulin therapy.
[003721 Gil' hybrids of the invention can be useful for reducing food intake, reducing appetite, reducing caloric intake, inducing satiety, reducing nutrient availability, causing weight loss, affecting body composition, altering body energy content or energy expenditure, improving lipid profile (including reducing LDL cholesterol and triglyceride levels and/or changing HDL cholesterol levels), slowing gastrointestinal motility, delay gastric emptying, moderating the postprandial blood glucose excursions, preventing or inhibiting glucagon secretion, and decreasing blood pressure. In one embodiment such GD' hybrids contain an exendin, GLPI, amylin and/or sCT portion.
[00373] Thus, in certain embodiments, the hybrids of the invention are useful for treating or preventing conditions or disorders which can be alleviated by reducing nutrient availability comprising administering to said subject a therapeutically or prophylactically effective amount of a compound of the invention. Such conditions and disorders include, but are not limited to, eating disorders, insulin-resistance, obesity, abnormal postprandial hyperglycemia, diabetes of any kind, including Type I, Type II, and gestational diabetes, Metabolic Syndrome, Dumping Syndrome, hypertension, dyslipidemia, cardiovascular disease, hyperlipidemia, sleep apnea, cancer, pulmonary hypertension, cholecystitis, and osteoarthritis. In one embodiment such hybrids contain an exendin, GLPI, amylin and/or sCT portion.
[00374] Non-limiting examples of a cardiovascular condition or disease are hypertension, myocardial ischemia, and myocardial reperfusion. Compounds of the invention may also be useful in treating or preventing other conditions associated with obesity including stroke, cancer (e.g,. endometrial, breast, prostate, and colon cancer), gallbladder disease, sleep apnea, reduced fertility, and osteoarthritis, (see Lyznicki et at, Am. Fain. Phys. 63:2185, 2001). In other embodiments, compounds of the invention may be used to alter body composition for aesthetic reasons, to enhance one's physical capabilities, o to produce a leaner meat source. Hybrids are useful to change body composition by decreasing fat without significant decrease in muscle mass, thus producing a desirable loss of body fat while preserving lean body mass. In one embodiment such hybrids contain an exend in, GLP 1, amylin and/or sCT portion.
[003751 In another aspect of the invention, methods for treating or preventing obesity are provided, wherein the method comprises administering a therapeutically or prophylactically effective amount of a hybrid polypeptide to a subject in need thereof. In a exemplary embodiment, the subject is an obese or overweight subject. While "obesity" is generally defined as a body mass index over 30, for purposes of this disclosure, any subject, including those with a body mass index of less than 30, who needs or wishes to reduce body weight is included in the scope of "obese." Subjects who are insulin resistant, gluco
intolerant, or have any form of diabetes mellitus (e.g., type 1, 2 or gestational diabetes) can benefit from
these hybrids. In one embodiment such GD' hybrids contain an exendin, PYY, GLP 1, amylin and/or sCT portion.
[00376] In other aspects of the invention, methods of reducing food intake, reducing nutrient availability, causing weight loss, affecting body composition, and altering body energy content or increasing energy expenditure, treating diabetes mellitus, and improving lipid profile (including reducing LDL cholesterol and triglyceride levels and/or changing HDL cholesterol levels) are provided, wherein the methods comprise administering to a subject an effective amount of a hybrid polypeptide of the invention. In a exemplary embodiment, the methods of the invention are used to treat or prevent conditions or disorders which can be alleviated by reducing nutrient availability in a subject in need thereof, comprising administering to said subject a therapeutically or prophylactically effective amount of a hybrid polypeptide of the invention. Such conditions and disorders include, but are not limited to, hypertension, dyslipidemia, cardiovascular disease, eating disorders, insulin-resistance, obesity, and diabetes mellitus of any kind. In one embodiment such hybrids contain an exendin, PYY, GLP1, amylin and/or sCT portion.
[00377] In one embodiment in which a PPF or PYY family member comprises a GD'-hybrid component, and without intending to be limited by theory, it is believed that the effects of such peripherally-administered GD' hybrid polypeptides in the reduction of food intake, in the delay of gastric emptying, in the reduction of nutrient availability, and in the causation of weight loss are determined by interactions with one or more unique receptor classes in, or similar to, those in the PP family. More particularly, it appears that a receptor or receptors similar to the PYY-preferring (or Y7) receptors are involved.
[00378] Additional assays useful to the invention include those that can determine the effect of GD' hybrid compounds, particualry those containing an exendin, PPF, PYY, GLP1, amylin and/or sCT portion, on body composition. An exemplary assay can be one that involves utilization of a diet-induced obese (DIO) mouse model for metabolic disease. Prior to the treatment period, male C57BIJ6J mice can be fed a high-fat diet (#D 12331, 58% of calories from fat; Research Diets, Inc.,) for 6 weeks beginning at 4 weeks of age. During the study, the mice can continue to eat their high-fat diet. Water can be provided ad libitum throughout the study. One group of similarly-aged non-obese mice can be fed a low-fat diet (#D 12329, 11% of calories from fat) for purposes of comparing metabolic parameters to DIO groups.
[00379] DIO mice can be implanted with subcutaneous (SC) intrascapular osmotic pumps to deliver either vehicle (50% dimethylsulfoxide (DMSO) in water) or a compound of the invention. The pumps of the latter group can be set to deliver any amount, e.g., 1000 jzg/kg/d of a compound of the invention for 7-28 days. Body weights and food intake can be measured over regular intervals throughout the study periods. Respiratory quotient (RQ' defined as CO2 production 02 consumption) and metabolic rate can be determined using whole-animal indirect calorimetry (Oxymax, Columbus Instruments, Columbus, OH). The mice can be euthanized by isoflurane overdose, and an index of adiposity (bilateral epididymal fat pad weight) measured. Moreover, prior to determination of epididymal weight, body composition (lean mass, fat mass) for each mouse can be analyzed using a Dual Energy X-ray Absorptiometry (DEXA instrument per manufacturer's instructions (Lunar Piximus, GE Imaging System). In the methods of the

invention, GD' hybrids, particularly those comprising an exendin, PPF, PYY, GLP1, amylin and/or sCT portion having a potency in one of the assays described herein (preferably food intake, gastric emptying, pancreatic secretion, weight reduction or body composition assays) which is greater than the potency of a component peptide hormone in that same assay, can be identified.
[003801 In addition to the amelioration of hypertension in subjects in need thereof as a result of reduced food intake, weight loss, or treating obesity, compounds of the invention may be used to treat hypotension.
[00381] In another general aspect, hybrids of the invention may be used to inhibit the secretion of ghrelin. Accordingly, compounds of the invention may be utilize this mechanism to treat or prevent ghrelin related disorders such as Prader-Willi syndrome, diabetes of all types and its complications, obesity, hyperphagia, hyperlipidemia, or other disorders associated with hypernutrition. In one embodiment such hybrids contain an exendin, GLPI, amylin and/or sCT portion.
[00382] Compounds of the invention may also be useful for potentiating, inducing, enhancing or restoring glucose responsivity in pancreatic islets or cells. These actions may be useful for treating or preventing conditions associated with metabolic disorders such as those described above and in U.S. patent application no. U52004/0228846. Assays for determining such activity are known in the art. For example, in published U.S. patent application no. U52004/0228846 (incorporated by reference in its entirety), assays are described for islet isolation and culture as well as determining fetal islet maturation. In the examples of patent application U52004/0228846, intestine-derived hormone peptides including pancreatic polypeptide (Pl'), neuropeptide Y (NEY), neuropeptide K (M'K), PYY, secretin, glucagon-like peptide-l (GLl'-1) and bombesin were purchased from Sigma. Collagenase type XI was obtained from Sigma. RPMI 1640 culture medium and fetal bovine serum were obtained from Gibco. A radioimmunoassay kit containing anti-insulin antibody (['251]-RIA kit) was purchased from Linco, St Louis.
[00383]Post-partem rat islets were obtained from P-02 year old rats. Adult rat islets were obtained from 6-8 week old rats. Fetal rat islets were obtained as follows. Pregnant female rats were sacrificed on pregnancy day E21. Fetuses were removed from the uterus. 10-14 pancreata were dissected from each litter and washed twice in Hanks buffer. The pancreata were pooled, suspended in 6 ml I ing/mI collagenase (Type XI, Sigma) and incubated at 370 C for 8-10 minutes with constant shaking. The digestion was stopped by adding 10 volumes of ice-cold Hanks buffer followed by three washes with Hanks buffer. The islets were then purified by Ficoll gradient and cultured in 10% fetal bovine serum (FBS)/RPMI medium with or without addition of 1 .xM IBMX. At the end of five days, 20 islets were hand picked into each tube and assayed for static insulin release. Generally, islets were first washed with KR? buffer and then incubated with 1 ml of KR? buffer containing 3 mM (low) glucose for 30 minutes at 370 C. with constant shaking. After collecting the supernatant, the islets were then incubated with 17 mM
(high) glucose for one hour at 370 C. The insulin released from low or high glucose stimulation were
assayed by radioimmunoassay (RIA) using the [1~I]-RIA kit. E21 fetal islets were cultured for 5 days in the presence of 200 ng/mi PYY, PP. CCK, M'K, N?Y, Secretin, GLP-l or Bombesin.
[00384] An exemplary in vivo assay is also provided using the Zucker Diabetic Fatty (ZDF) male rat, an inbred (>F30 Generations) rat model that spontaneously expresses diabetes in all fa/fa males fed a standard rodent diet Purina 5008. In ZDF fa-fa males, hyperglycemia begins to develop at about seven weeks of age and glucose levels (fed) typically reach 500 mg[DL by 10 to 11 weeks of age. Insulin levels (fed) are high during the development of diabetes. However, by 19 weeks of age insulin drops to about the level of lean control titter mates. Triglyceride and cholesterol levels of obese rats are normally higher than those of leans. In the assay, three groups of 7-week old ZDF rats, with 6 rats per group, received the infusion treatment by ALZA pump for 14 days: 1) vehicle control, 2) and 3), PYY with two different doses, 100 pmol/kg/hr and 500 pmol/kg/hr respectively. Four measurements were taken before the infusion and after the infusion at day 7 and day 14:1) plasma glucose level, 2) plasma insulin level, and 3) plasma triglycerides (TG) level, as well as oral glucose tolerance (OGTT) test. Accordingly, these assays can be used with compounds of the invention to test for desired-activity.
[00385] Hybrids are also useful for the therapeutic and prophylactic treatment of neurological and nervous system disorders associated with neuronal loss or dysfunction, including, but not limited to, Parkinson's Disease, Alzheimer's Disease, Huntington's Disease, ALS, stroke, ADD, and neuropsychiatric syndromes, and to enhance or facilitate learning, memory and cognition in mammals. Particularly' useful in this regard are GD' hybrids containing an exendin or GLl'1 active portion, more specifically comprising at least the N-terminal 7-15 amino acids or analog thereof, for example HSEGTFTSD (SEQ ID NO. 94).

[00386] Other uses contemplated for the hybrid polypeptides include methods for reducing aluminum (Al) concentrations in the central nervous system (see U.S. Pat. 6,734,166, incorporated by reference in its entirety) for treating, preventing, or delay the onset of Alzheimer's disease. Assays for determining effects on Al are known in the art and can be found in US Pat 6,734,166 using diploid and Ts mice. These mice were individually housed in Nalgene® br~nd metabolism or polypropylene cages and given three days to adjust to the cages before experimentation. Mice had free access to food (LabDiet® NIH Rat and Moust/Auto 6F5K52, St. Louis, Mo.) and water during the experiment except for the 16 hours prior to euthanasia when no food was provided. Mice were given daily subcutaneous injections of either active compound or saline. Mice were sacrificed at the end of day 13 for one experiment and day 3 for another, and samples were collected. Mice brain samples were weighted in clean teflon liners and prepared for analysis by microwave digestion in low trace element grade nitric acid. Samples were then analyzed for Al content using Inductively Coupled Plasma Mass Spectrometry (Nuttall et al., Annals of Clinical and Laboratory Science 25, 3, 264-271(1995)). All tissue handling during analysis took place in a clean room environment utilizing HEPA air filtration systems to minimize background contamination.
[003871 Hybrids of the invention are useful for prevention and treatment of nephropathy, including hypertensive and diabetic nephropathy, and nephropathy associated with insulin resistance and metabol syndrome. Hybrids achieve these ends by, among other things, improving or preventing worsening of
hypertension, endothelial function, renal function, and glomerulosclerosis. In one embodiment, the invention provides a method for preventing or treating nephropathy, including hypertensive and diabetic nephropathy, or that related to insulin resistance, comprising administering a compound of the invention. Hybrids find further use for improving endothelial function in a patient having reduced vasodilatory capacity, or having glomerulosclerosis or any other reduction in glomerular flow. Such improvement in endothelial function serves both to reduce hypertension and to improve the function of the capillaries of the glomeruli. In additional embodiments, the molecules of the invention are useful to prevent progression of nephropathy to ESRD, to prevent, slow the progression of, treat or ameliorate proteinuria and/or glomerulosclerosis.
[00388] Hybrids are useful for reducing the risk of suffering from, preventing, or treating cardiac arrhythmias. Hybrids can provide anti-arrhythmic effects in patients with cardiac ischemia, cardiac ischemia-reperfusion, and congestive heart failure. For example, incretin GLP-1 has been found to reduce cardiac injury and enhance recovery in patients with these disorders. Incretins, including GLP-1, -are glucose-dependent insulinocropic hormones. GLP-1 and exendin effectively enhance peripheral glucose uptake without inducing dangerous hypoglycemia. They also strongly suppress glucagon secretion, independent of its insulinotropic action, and thereby powerfully reduce plasma free fatty acid (FFA) levels substantially more than can be accomplished with insulin. High FFA levels have been implicated as a major toxic mechanism during myocardial ischemia. In another embodiment hybrids are useful for preventing and treating cardiac arrhythmias that reliably reduce injury associated with reperfusion and ischemia, and enhance patient recovery. In yet a further embodiment hybrid treatment after acute stroke or hemorrhage, preferably intravenous administration, provides a means for optimizing insulin secretion, increasing brain anabolism, enhancing insulin effectiveness by suppressing glucagon, and maintaining euglycemia or mild hypoglycemia with no risk of severe hypoglycemia or other adverse side effects. In one embodiment such GD' hybrids contain a GLP1 or exendin portion.
[00389] In yet a further embodiment, hybrids that are capable of lowering insulin resistance or increasing insulin sensitivity are useful to treat polycystic ovary syndrome (PCOS). Administering hybrids of the invention can reduce or prevent insulin resistance in a subject suffering from PCOS. In yet another embodiment hybrids prevent the onset of type-2 diabetes in a subject suffering from PCOS. Further hybrids can restore regular menses, ovulation, or fertility in a subject suffering from PCOS. In one embodiment such GD' hybrids contain a GLP1 or an exendin portion for binding and activating a GLPl receptor.
[003901 By selection of hormone component modules the compounds of the invention can exhibit a broad range of biological activities, some related to their antisecretory and antimotility properties. The compounds may suppress gastrointestinal secretions by direct interaction with epithelial cells or, perhaps, by inhibiting secretion of hormones or neurotransmitters which stimulate intestinal secretion. Antisecretory properties include inhibition of gastric and/or pancreatic secretions and can be useful in th
treatment or prevention of diseases and disorders including gastritis, pancreatitis, Barrett's esophagus, and Gastroesophageal Reflux Disease, as well as conditions associated therewith including heartburn, heartburn accompanied by regurgitation of gastric/intestinal contents into the mouth or the lungs, difticulty in swallowing, coughing, intermittent wheezing and vocal cord inflammation (conditions associated with GERD), esophageal erosion, esophageal ulcer, esophageal stricture, Barrett's metaplasia (replacement of normal esophageal epithelium with abnormal epithelium), Barrett's espohageal adenocarcinoma, and pulmonary aspiration. In another embodiment GD' hybrids containing amylin and/or sCT portions can be useful for treating or preventing these diseases and conditions, such as Barrett's esophagus, Gastroesophageal Reflux Disease (GERD) and conditions associated therewith as disclosed herein. Such hybrids have particularly effective anti-secretory properties, such as inhibition of gastric acids, inhibition of bile acids, and inhibition of pancreatic enzymes. Moreover, such hybrids can have a gastroprotective effect, which renders them particularly useful in the treatment or prevention of intestinal diseases and conditions and of Barrett's esophagus, and/or GERD and related or associated conditions as described herein.
[00391] Compounds of the invention are useful in the treatment of any number of gastrointestinal disorders (see e.g., Harrison's Principles of Internal Medicine, McGraw-Hill Inco, New York, 12th Ed.) that are associated with excess intestinal electrolyte and water secretion as well as decreased absorption, e.g., infectious diarrhea, inflammatory diarrhea, short bowel syndrome, or the diarrhea which typically occurs following surgical procedures, e.g., ileostomy. Examples of infectious diarrhea include, without limitation, acute viral diarrhea, acute bacterial diarrhea (e.g., salmonella, campylobacter, and clostridium or due to protozoal infections), or traveller's diarrhea (e.g., Norwalk virus or rotavirus). Examples of inflammatory diarrhea include, without limitation, malabsorption syndrome, tropical sprue, chronic pancreatitis, Crohn's disease, diarrhea, and irritable bowel syndrome. It has also been discovered that the peptides of the invention can be used to treat an emergency or life-threatening situation involving a gastrointestinal disorder, e.g., after surgery or due to cholera.
[00392] Compounds of the invention may also be useful for treating or preventing intestinal, including gastrointestinal, damage as opposed to merely treating the symptoms associated with the intestinal damage (for example, diarrhea). Such damage to the intestine may be, or a result of, ulcerative colitis, inflammatory bowel disease, bowel atrophy, loss bowel mucosa, and/or loss of bowel mucosal function (see WO 03/105763, incorporated herein by reference in its entirety). Assays for such activity, as described in WO 03/105763, include 11 week old male HSD rats, ranging 250- 300 grams housed in a 12:12 light:dark cycle, and allowed ad libitum access to a standard rodent diet (Teklad LM 485, Madison, WI) and water. The animals were fasted for 24 hours before the experiment. A simple and reproducible rat model of chronic colonic inflammation has been previously described by Morris GP, et al., "Hapteninduced model of chronic inflammation and ulceration in the rat colon." Gastroenterology. 1989; 96:795-
803. It exhibits a relatively long duration of inflammation and ulceration, affording an opportunity to
study the pathophysiology of colonic inflammatory disease in a specifically controlled fashion, and to evaluate new treatments potentially applicable to inflammatory bowel disease in humans.
[00393] Rats were anesthetized with 3% isofluorane and placed on a regulated heating pad set at 370C. A gavage needle was inserted rectally into the colon 7 cm. The hapten trinitrobenzenesulfonic acid (TNBS) dissolved in 50% ethanol (v/v) was delivered into the lumen of the colon through the gavage needle at a dose of 30 mg/kg, in a total volume of 0 0.4-0.6 mL, as described in Mazelin, et al., "Protective role of vagal afferents in experimentally-induced colitis in rats." Juton Nerv Syst. 73:38-45 (1998). Control groups received saline solution (NaCl 0.9%) intracolonically. Four days after induction of colitis, the colon was resected from anesthetized rats, which were then euthanized by decapitation. Weights of excised colon and spleen were measured, and the colons photographed for scoring of gross morphologic damage. Inflammation was defined as regions of hyperemia and bowel wall thickening.
[00394] In another aspect, GD' hybrids can be useful for treating or preventing pancreatitis, pancreatic carcinoma, and gastritis, particularly in the treatment and prevention of pancreatitis in patients who have undergone endoscopic retrograde cholangiopancreatography (ERCP). Amylin and/or sCT containing Gil' hybrid agonists can have a suprisingly superior therapeutic effect when combined with somatostatin. Accordingly, in certain embodiments, methods for treating or Preventing pancreatitis comprise administering such hybrids and administering somatostatin and somatostatin agonists to a subject. Hybrid polypeptides of the invention may also be used to treat or prevent Barrett's esophageal adenocarcinoma or pancreatic tumors (e.g., inhibit the proliferation of pancreatic tumors). Methods of the invention include reducing the proliferation of tumor cells. The types of benign pancreatic tumor cells which may be treated in accordance with the present invention include serous cyst adenomas, microcystic tumors, and solid-cystic tumors. The method is also effective in reducing the proliferation of malignant pancreatic tumor cells such as carcinomas arising from the ducts, acini, or islets of the pancreas. Particualry useful GD' hybrids in this regard are those comprising a hormone module component of the PYY or PPF family. U.S. Pat. 5,574,010 (incorporated by reference in its entirety) provides exemplary assays for testing anti-proliferative properties. For example, the '010 patent provides that PANC-1 and MiaPaCa-2 are two human pancreatic adenocarcinoma cancer cell lines which are available commercially from suppliers such as American Type Culture Collection, ATCC (Rockville, Md.). The two tumor cells were grown in RPMI-1640 culture media supplemented with 10% fetal bovine serum, 29.2 mgfL of glutamine, 25 p.g gentamicin, 5 ml penicillin, streptomycin, and fungizone solution (JRH Biosciences, Lenexa, Kans.) at 37 degrees Celcius in a NAPCO water jacketed 5 % CO2 incubator. All cell lines were detached with 0.25 % trypsin (Clonetics, San Diego, Calif.) once to twice a week when a confluent monolayer of tumor cells was achieved. Cells were pelleted for 7 minutes at 500 g in a refrigerated centrifuge at 4 degrees Celcius, and resuspended in trypsin free fortified RPMI 1640 culture media. Viable cells were counted on a hemocytometer slide with trypan blue.
[00395] Ten thousand, 20,000, 40,000 and 80,000 cells of each type were added to 96 well microcultur plates (Costar, Cambridge, Mass.) in a total volume of 200 ul of culture media per well. Cells were

allowed to adhere for 24 hours prior to addition of the PYY or test peptide. Fresh culture media was exchanged prior to addition of peptides. In vitro incubation of pancreatic tumor cells with either PYY or test compound was continued for 6 hours and 36 hours in length. PYY was added to cells at doses of 250 pmol, 25 pmol, and 2.5 pmol per well (N =14). Test compound was added to cells cultures at doses of 400 pmol, 40 pmol, and 4 pmol per well. Control wells received 2 ul of 0.9% saline to mimic the volume and physical disturbance upon adhered tumor cells. Each 96 well plate contained 18 control wells to allow for comparison within each plate during experimentation. Ninety-six (96) well plates were repeated 6 times with varying concentrations of PYY and test compound in both the PANC-l and MiaPaCa-2 cells.
[00396] At the end of the incubation period, 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide, MTr tetrazolium bromide (Sigma, St. Louis, Mo.) was added to fresh culture media at 0.5 mg/mi. Culture media was exchanged and tumor cells were incubated for 4 hours with MT~ tetrazolium bromide at 370C. At the end of incubation, culture media was aspirated. Formazon crystal precipitates were dissolved in 200 ~±l of dimethyl sulfoxide (Sigma, St. Louis, Mo.). Quantitation of solubilized formazon was performed by obtaining absorption readings at 500 nm wavelength on an ELISA reader (Molecular Devices, Menlo Park, Calif.). The M'IT assay measures mitochondrial NADH dependent dehydrogenase activity, and it has been among the most sensitive and reliable method to quantitative in vitro chemotherapy responses of tumor cells. (Alley, M. C., et at., Cancer Res., 48:589-601, 1988; Carmichael, J., et at., Cancer Res., 47:936-942, 1987; McHale, A. P., et aL, Cancer Left., 41:315-321, 1988; and Saxton, R. E., et at., J. Clin. Laser Med. and Surg., 10(5):331-336, 1992.) Analysis of absorption readings at 550 nm were analyzed by grouping wells of the same test conditions and verifying differences occurring between control and the various peptide concentration treatments by one-way
[00397] An exemplary in vivo assay is also provided. The human pancreatic ductal adenocarcinoma Mia Paca-2 was examined for in vivo growth inhibition by peptide YY and test compound. Seventy thousand to 100,000 human Mia PaCa-2 cells were orthotopically transplanted into 48 male athymic mice. After one week, the animals were treated with either PYY or test compound at 200 pmollkg/hr via mini-osmotic pumps for four weeks. The paired cultures received saline. At sacrifice, both tumor size and mass were measured. Control mice had significant human cancer growth within the pancreas as evidenced by histologic sections. At 9 weeks, ninety percent (90%) of control mice had substantial metastatic disease. Tumor mass was decreased by 60.5 % in test treated mice and 27% in PYY treated mice.
[00398] In another general aspect, hybrids are useful for decreasing bone resorption, decreasing plasma calcium, and/or inducing an analgesic effect, particularly to treat bone disorders such as osteopenia and osteoporosis. In yet other embodiments, hybrids are useful to treat pain and painful neuropathy. In one embodiment such hybrids contain an exendin, GLPl, amylin and/or sCT portion. For example, a GD'-sCT or GD'-amylin/sCT hybrid compound of the invention can have a selectable property of a salmon calcitonin or amylin/sOT/Amylin chimera, such as decreasing bone loss and bone resorption or reducinr cartilage turnover (chondroprotection), and a property of a GD', such as plasma glucose lowering
(concomitant with an anti-catabolic aspect as described herein) and/or inhibiting bone resorption and maintaining or increasing bone density. A GD' hybrid with such selectable properties can enhance treatment of osteoporosis or conditions of high cartilage turnover, particularly in those who can also benefit from glycemic control, such as subjects with diabetes or under going critical care.
[00399] Gil' compounds, particularly GD' analogs, extended half-life GD' hybrids (e.g. DPP-IV cleavage resistant (such as a D-Ala2, N-Acetyl or N-pyroglutamyl analogs) optionally further comprising a peptidic enhancer such as a heterologous C-terminal tail, and GD' hybrids comprising other hormone modules known to provide beneficial cardiovascular effects, are useful to treat cardiovascular disease and related conditions. As demonstrated herein GD' compounds increase cardiac contractility (dp/dt), decrease blood pressure (for example by acute vasodilatation), decrease systolic pressure, decrease diastolic pressure, and can provide a direct beneficial action on cardiac cells. GD' compounds also improve cardiac function via metabolic actions, e.g. glucose lowering, insulin secretion, beta cell proliferation. By also providing direct effects on the cardiovascular system, the GD' compounds are surprisingly even more therapeutically beneficial.
[004001 Accordingly, provided herein are methods to treat, prevent or alleviate cardiovascular diseases and conditions by administering a therapeutically effective amount of a GD' compound, either alone or with another agent that provides cardiovascular benefit, to a patient in need of such treatment. As with the other conditions discussed throughout this specification, a GD' compound can be administered concurrently, sequentially or alternately with another agent.

[004011 Accordingly, in one embodiment the cardiovascular disease or condition is hypertension (including stage 1, stage 2 and stage 3 hypertension, diastolic or systolic), pulmonary hypertension, congestive heart failure, cardiac insufficiency, reduced stroke volume, cardiomyopathy (dilated, hypertrophic or restrictive), decreased cardiac contractility, pulmonary congestion associated with cardiovascular conditions, pulmonary and systemic edema, decreased cardiac output, abnormal left ventricular function, diastolic blood pressure abnormalities, renal failure associated with decreased cardiac contractility, increased cardiovascular risk (e.g. associated with elevated systolic pressure accompanied by normal diastolic pressure, associated with elevated diastolic pressure accompanied by normal systolic pressure, associated with elevated diastolic and systolic pressure, associated with elevated mean arterial blood pressure) and non-ischemic or ischemic heart tissue degeneration (such as from myocardial infarction). In one embodiment either or both the mortality or the morbidity associated with these diseases and conditions are reduced.
[00402] The patient in need of treatment includes those who are diabetic (e.g. suffering from diabetic cardiomyopathy), obese, undergoing intensive care, undergoing surgery, or a combination thereof, or who are otherwise normal. The patient may have had or be at risk of having such a disease or condition. For example, patients post myocardial infarction that are in need of preventing further heart failure can benefit from the methods herein.
100403] Preventing a disease or condition, e.g., a cardiovascular disease or condition, includes preventing the initiation of, delaying the initiation of, preventing the progression or advancement of, slowing the progression or advancement of, delaying the progression or advancement of, and reversing the progression of the disease or condition from an advanced to a less advanced stage.
[00404] in one embodiment the method provides treating or delaying the onset of such diseases or conditions. For example, impaired contractility can decrease stroke volume which in turn can precipitate congestive heart failure. Thus in one embodiment treating heart failure refers to treating any one or more of the conditions underlying heart failure, including, without limitation, decreased cardiac contractility, abnormal diastolic compliance, reduced stroke volume, high blood pressure, pulmonary congestion, and decreased cardiac output.
[00405] In one embodiment is provided a method for inducing an inotropic response, for reducing blood pressure, for reducing diastolic pressure, for reducing diastolic pressure, increasing vasodilation, or a any combination of the above, with or without a concomitant beneficial metabolic action of a GD' such as glucose lowering, insulin secretion, or beta cell proliferation, comprising administration of a therapeutically effective amount of a GD' compound to provide such desired beneficial action. These methods are useful for treating conditions or disorders that can be alleviated by an increase in cardiac contractility, a reduction in blood pressure, a reduction in diastolic pressure, a reduction in diastolic pressure, an increase in vasodilation, or a combination of the above, in patients in need of such benefits.
[00406] Inotropic compounds are compounds that induce inotropic effects (e.g., increase of force of contraction of the heart) have been recognized as being useful for the treatment of, for example, congestive heart failure. Congestive heart failure, which is one of the most common causes of death and disability in industrialized nations, has a mortality rate of about 50% at five years (Goodman and Gilman s The Pharmacological Basis of Therapeutics, 9th Ed. McGraw Hill, N.Y., pp. 809-838). Criteria, testing and guidelines as established by the American Heart Association (AHA) are suitable for diagnosing the cardiovascular diseases and conditions discussed herein.
[00407] GD' compounds display a desired positive inotropic effect without a substantial, concomitant increase in blood pressure. Such blood pressure changes in subjects experiencing heart failure or cardiovascular disease or condition could cause further deterioration in heart function. In fact as demonstrated herein, GD' compounds can function to reduce blood pressure or the rate of change in blood pressure.
[00408] Issues with available inotropic agents illustrates the need for, and desirability of, therapies that are inotropic, with rapid onset of action, with prolonged duration of action (including a persistent effect, with absence of tachyphylaxis), with low toxicity (a high ratio of toxic to therapeutic dose), with absent or low nausea effect, and with a convenient (non-intravenous) route of administration. GD' compounds can provide these benefits.

[00409] Further beneficial action of GD' compounds of the invention, particularly the Gill' hybrids comprising a DPl'-IV resistant GD' analog with a peptidic enhancer such as an exendin tail (e.g. Compound G), arises from a lack of or reduced anorectic effect and an absence of or relatively insignificant nausea effect, which can be important in the patient populations discussed throughout.
[00410] Also provided is a method for treating critically ill patients, exemplified as those sufficiently ill to warrant admission to an intensive care unit (ICU) and that find benefit from anti- or non-catabolic therapy, which comprises administering a therapeutically effective amount of a Gil' analog or hybrid to a patient in need of such treatment, alone or with another beneficial agent. Without being limited by theory, the methods are intended to benefit those patients in which the effect of Gil' agonist analogs and hybrids to stimulate insulin secretion and incur the benefits associated with intensive insulin therapy (GIK therapy; glucose-insulin-potassium), without the hazards and complexity associated with insulin/glucose infusion and without the side effects reportedly associated with the use of some glucagon-like peptide-1 agonists. Further, it is now observed that GD' can favorably affect a patient's metabolic state in addition to simply indirectly regulating glucose levels in response to digestion of food. Accordingly GD' compounds are useful to reduce the mortality and morbidity that occurs in critically ill patients.
[00411] As demonstrated herein GD' compounds provide beneficial metabolic effects such as glucose lowering, insulin secretion and/or beta cell proliferation. GD' compounds also improve cardiac function by increasing cardiac contractility (d?Idt), decreasing blood pressure (for example by acute vasodilatation), and decreasing systolic pressure, decreasing diastolic pressure, and providing a direct beneficial action on cardiac cells. Since many critically ill patients have or are at risk for or are complicated by cardiovascular diseases or conditions, these cardiovascular effects can provided additional benefit. By also providing direct effects on the cardiovascular system, the GD' compounds, surprisingly, have added therapeutic value.

[00412] Accordingly, provided herein are methods to treat, prevent or alleviate conditions and diseases of critical care by administering a therapeutically effective amount of a GD' compound, either alone or with another agent that provides desired benefits, to a critically ill patient in need of such treatment. As with the other conditions discussed throughout this specification, a GD' compound can be administered concurrently, sequentially or alternately with another agent.
[00413] The "intensive care unit" can be a part of a hospital where critically ill patients are treated, and of course may not officially bear the name "Intensive Care Unit". ICU also includes a nursing home a clinic, for example, a private clinic, or the like if the same or similar activities are performed there.
[00414]The term a "critically ill patient" can be a patient who has sustained or are at risk of sustaining acutely life-threatening single or multiple organ system failure due to disease or injury, a patient who is being operated and where complications supervene, and a patient who has been operated in a vital organ within the last week or has been subject to major surgery within the last week. A critically ill patient
be a patient who needs vital organ support (either mechanically such as with mechanical ventilation
dialysis etc., or pharmacologically such as with inotropes or vasopressors) without which they would not survive. ExpressiOns' "critical care", "intensive care" and "critically ill" are used interchangeably. Critically ill patients are those who generally experience an unstable metabolic state. This unstable metabolic state, e.g. catabolic state, can be a result of changes in substrate metabolism which may lead to relative deficiencies in some nutrients and/or increased oxidation (e.g. wasting) of both fat and muscle, undesirable accelerated protein breakdown, hyperglycemia and high concentrations of serum triglycerides and other lipids. Non-limiting examples of a critically ill patient is a patient in need of cardiac surgery, cerebral surgery, thoracic surgery, abdominal surgery, vascular surgery, or transplantation, or a patient suffering from cerebral trauma, respiratory insufficiency, critical illness polyneuropathy, multiple traumas or severe burns, or a patient being mechanically ventilated.
[00415] Accordingly, in one embodiment the critical care disease or condition is classified as medical, surgical (e.g. trauma) or coronary care, and further can be classified as sepsis and respiratory care. Of particular interest are those classifications, such as septic shock or myocardial infarction, for which QIK therapy would be warranted.
[00416] Catabolic change (e.g., loss of body weight) is an adverse risk factor for the critically ill patient. Although most admissions to the ICU receive intravenous lines that deliver calories in some form, a Canadian study of nutritional support in ICU reported that typically only 58% of daily requirements were supplied and only 26% received parenteral nutrition. Thus a method is provided in which a critically ill patient in need of prevention or alleviating catabolic effects, such as weigh loss, is administered a therapeutically effective amount of a GD' compound as provided herein to prevent or alleviate the catabolism. The critically ill patient in need of treatment includes patients who are non-diabetic patient (not diagnosed as having diabetes or prediabetes), diabetic, prediabetic, and/or obese. The patient may have had or be at risk of having the disease or conditions indicated.

[00417]The use of GD' compounds can minimize or avoid risks associated with GLP-1 in invoking a GIK-like benefit. Slowing of gastric emptying can complicate the delivery of oral medicines, for example, by altering their kinetics, and decrease nutrient uptake; GLP-1 agonists, amylin agonists, CCK agonists, and secretin agonists, for example, slow gastric emptying. As shown herein, within therapecxtic. ranges, doses of GD' compounds do not slow gastric emptying or show only a weak effect, and in any event much less so than GLPI and exendin 4. Typically such effect was blocked with a selective amylin antagonist, e.g. pramlintide, indicating that the effect was an indirect consequence of augmented beta-cell secretion of amylin, the latter being the most potent endogenous inhibitor of gastric emptying thus far identified. GD' administration is thus unassociated with a direct inhibition of gastric emptying. Importantly, it has been shown herein that GD' itself does not acutely inhibit food intake in mice. Neither does GD' cause weight loss in diet-induced-obese mice. Nor does the Gil' analog hybrid 060 1GD'3794. Further GD' was reported to exert a fat-sparing effect relative to GLP- 1 agonists which are associated with
a loss of body weight and adiposity. This fat-sparing effect is possibly attributable to an antilipolytir
effect in adipocytes or to such effects as promotion of lipoprotein lipase, amplification of insulin
signaling, and/or an increase of fatty acid incorporation into adipocyte lipid. Interestingly, 0601G1P3794 provided a slight decrease in percentage body fat with an increase in body protein without a concomitant reduction in body weight, indicating that body composition was affected. It is well recognized that loss of body energy stores predicts adverse outcomes in critical care whereas preservation of body energy stores promotes beneficial outcomes. The absence of or decreased anorexigenic effects, the absence of or decreased nausea and/or the absence of or reduction of weight loss, with or without the improvement in body composition, are advantageous in patients experiencing catabolic effects, such as critically ill patients. Although GD' receptor mRNA has been detected in heart tissue, hormone binding in heart has not been detected to date. However, as shown herein GD' binds to and activates cardiac myocytes and displays a positive inotropic effect in vivo. With regard to treatment of renal failure, particularly acute renal failure, in patients who remain oliguric despite attempts to enhance urine flow by approaches to establish urine output such as by the use of loop diuretics and osmotic diuretics, intervention by inotropic agents that increase cardiac contractility is generally attempted. Thus as discussed GD' compounds provide further benefit in patients with or at risk of compromised myocardium, and certain other conditions such as renal failure, independent of its insulinotropic action. Further the method includes administering to patients in need thereof a therapeutically effective amount of a GD' compound that provides a sparing of cardiac muscle, which otherwise tends to atrophy in catabolic states.
[00418] Accordingly, in one embodiment the critical care patient has a disease or condition of catabolic change associated with a critical illness, sepsis, post-traumatic, post-surgical, post-shock, comatose patients, stress-induced hyperglycemia (for example after a vascular event), stroke, myocardial infarction, acute mesenteric ischemia, respiratory distress, ventilator dependency, renal failure, congestive heart failure, edema, hibernating myocardium, cardiomyopathies (ischemic, diabetic), lowering of BM', ejection dysfunction, hypertension, polyneuropathy, isehemia/reperfusion injury (for example postthrombolytic therapy, post cardiac surgery), histoprotection of organ beds, myocardial infarction (mortality, function, symptomatology), acute coronary syndrome (stable/unstable angina, non-Q wave infarct, ECG positive), disturbances of conduction or rhythm, papillary dysfunction, and/or pulmonary edema. For example, in a one embodiment the method comprises attenuating, ameliorating or reducing such disease or conditions including pre- or post-surgical catabolic changes, comprising, administering to a patient in need thereof a GD' compound. The GD' compound is designed to provide a therapeutic benefit, for exmaple a benefit measured for example as a reduction in APACHE score, a reduction in mortality, a reduction in days in hospital, a reduction in need for readmission, a reduction in hospitalization costs.
[00419] In one embodiment is a method for the treatment of a critically ill patient in need thereof, to prevent or decrease the incidence of blood stream infection, sepsis or septic schock, to reduce morbidity associated with the critical care, to reduce mortality (e.g. in-hospital mortality) associated with the critical care, to prevent or decrease the incidence of prolonged inflammation, to prevent or decrease the incidence of acute renal failure and/or renal replacement therapy, to prevent or decrease the incidence of critical ca~
polyneuropathy, to reduce the use of antibiotics, to prevent or decrease the incidence of immune-mediated destruction of the beta cells, to reduce the likelihood of disturbance in markers of inflammation and/or inflammatory responses, to prevent or decrease the incidence of systemic inflammatory response syndrome (SIRS), to reduce the amount of red cell transfusion, to reduce stress-induced hyperglycemia, to protect from cholestasis, to reduce the need for invasive treatment, to prevent or decrease the incidence of endoneural edema, to decrease dialysis or hemofiltration, to reduce or eliminate a need of vital organ system support, to allow at least about one third of the caloric need through the normal enteral route, to reduce the risk or likelihood of multiple organ failure, to reduce the risk or likelihood of multiple organ failure associated with sepsis or septic shock, to reduce the use of mechanical ventilatory support, to reduce the likelihood of disturbed kidney function parameters, to reduce the likelihood of hyperbilirubinemia, or to treat, prevent or alleviate or reduce the incidence of one or more of the other critical care conditions mentioned herein, which comprises administering a therapeutically effective amount of a Gil' compound. In a further embodiment administration achieves a normo- or euglycemia, a lowering of blood glucose, insulin secretion, a cardiovascular benefit as discussed herein, a reduction of catabolic effect, or any combination thereof.
[00420] Critical care patients can include those suffering from respiratory distress in which the patient has difficulty breathing as a result of pulmonary dysfunction. The patient may exhibit varying degrees of hypoxemia that that may or may not be controlled with supplemental oxygen. Respiratory distress may occur in patients with impaired pulmonary function due to direct lung injury, such as from pneumonia, aspiration of gastric contents, pulmonary contusion, fat emboli, near-drowning, inhalation injury, high altitude and reperfusion pulmonary edema, or from indirect lung injury as from sepsis, severe trauma with shock and multiple transfusions, cardiopulmonary bypass, drug overdose, and acute pancreatitis. A critically ill patient may have a pulmonary disorders associated with chronic hypoxemia, which can result in raised pressure within the pulmonary circulation called pulmonary hypertension. A critically ill patient may have cor pulmonale, which is a failure of the right side of the heart caused by prolonged high blood pressure in the pulmonary artery and right ventricle of the heart. A critically ill patient may have acute respiratory distress syndrome (ARDS) or chronic obstructive pulmonary diseases (COPDs) which include emphysema and chronic bronchitis, which also cause respiratory distress.
[00421] In another embodiment the critical care patient is one who with a disturbed glucose metabolism such as insulin resistance but no overt diabetes, as well as a patient who for any reason cannot receive nutrition through the alimentary canal. Such patients include surgery patients, comatose patients, patients in shock, patients with gastrointestinal disease, patients with digestive hormone disease, and the like. In particular, obese patients, atherosclerotic patients, vascular disease patients, patients with gestational diabetes, patients with liver disease such as liver cirrhosis, patients with acromegaly, patients with glucorticoid excess such as cortisol treatment or Cushings disease, patients with activated counterregulatory hormones such as would occur after trauma, accidents and surgery and the like, patients with hypertriglyceridemia and patients with chronic pancreatitis can be readily and suitably nourishc~
according to the invention without subjecting the patient to hypo- or hyperglycemia, while reducing undesirable catabolic changes and/or providing cardiovascular benefit.
[00422] In one embodiment the administration of ~ GD' compound, alone or with other agents, reduces morbidity or mortality in critically ill patients (who require intensive care) or the time they stay in the ICU. A reduction in morbidity means reducing the likelihood that the critically ill patient will develop additional illnesses, conditions, or symptoms or reducing the severity of additional illnesses, conditions or symptoms. For example reducing morbidity can be achieved by decreasing the incidence of bacteremia or sepsis or complications associated with multiple organ failure.
[00423] These indications need not be confined to those with dysglycemia, nor require that euglycemia be achieved. Neither are they restricted to an insulinotropic mechanism, or those individuals capable of responding with increased insulin secretion. Although GD' resistance has been reported in type 2 diabetes, resistance to the insulinotropic effects of GD' is not expected to be a feature of critically ill patients, including those with acute stress induced dysglycemia. Normal glucose-dependent stimulation of insulin secretion will occur in the majority of critically ill patients, and in those patients resistant to GD' efficacy can be attained with higher GD' doses or with the use of the GD' analogs and hybrids disclosed herein.
[00424] Thus in one embodiment is provided a method for inducing an insulinotropic response, for providing an anti- or non-catabolic effect (e.g. reducing a catabolic effect) or providing any one of the cardiovascular benefits discussed herein, or a any combination of the above, comprising administration of a therapeutically effective amount of a GD' compound to provide such desired beneficial action. These methods are useful for treating critical care conditions or disorders that can be alleviated by such effects, preferably in conjunction with a reduction of catabolic effect, in critically ill patients in need of such benefits.
[004251 The GD' compound includes GD' and GD' analogs and hybrids, particularly novel GD' analogs as described herein, extended half-life GD' hybrids (e.g. DPP-IV cleavage resistant (such as a D-Ala2, NAcetyl or N-pyroglutamyl analogs) optionally further comprising a peptidic enhancer such as a heterologous C-terminal tail, and GD' hybrids comprising at least one hormone module known to provide beneficial benefit to a patient undergoing critical or intensive care. For example, GD' compounds of the invention particularly useful are the GD' hybrids comprising a DPP-IV resistant GD' analog with a peptidic enhancer such as an exendin tail (e.g. Compound G), or GD' hybrids comprising a GIl' analog and a amylin family or salmon calcitonin hormone module, which are additionally beneficial by their lack of or reduced anorectic effect while maintaining a desirable glucose lowering, insulinotropic and/or cardiovascular benefit.
[00426] In one embodiment administering a GD' compound reduces the problems associated with parenteral nourishment. Very often it is not possible to infuse a desired amount of glucose even to people with healthy metabolism without provoking hyperglycemia. This can be exacerbated in critical cart
patients. Insulin secretion during parenteral nourishment in the presence of a Gil' compound can be controlled such that the plasma glucose increase will be less than without the GD' compound. Therefore more glucose can be delivered over a 24-hour period than otherwise. The calorie deficit seen with parenterally nourished patients can thus be better satisfied. Accordingly, provided is a method for non-alimentary nutrition comprising administering by a parenteral route to a patient in need of parenteral nutrition, a nutritively effective amount of one or more nutrients selected from the group consisting of carbohydrates, ammo acids, lipids, free fatty acids, mono-or diglycerides, glycerol and any combination thereof; and a GD' compound, wherein the administration of the nutrient(s) produces a blood glucose level in the patient of from about 80 to 180 mg glucose per deciliter of blood, and the rate of administration is calculated to deliver up to about 1000 g of glucose or its equivalent per patient per day. In a further embodiment the patient receives at least about one third of the caloric need through the normal enteral route, at least about half of the caloric need through the normal enteral route, or at least about two third of the caloric need through the normal enteric route. When the nutrient source is carbohydrate, the source of carbohydrate can be present at a concentration of about a 2% to about a 50% by weight of glucose or its equivalent per liter. In a further embodiment enhance nutrient metabolism is achieved via the parenteral route in patients with a disturbed glucose metabolism, a surgery patient, a comatose patient, a patient in shock, a patient ~vith gastrointestinal disease, a patient with digestive hormone disease, an obese patient, an atherosclerotic patient, a patient with vascular disease, a patient with gestational diabetes, a patient with liver disease, a patient with liver cirrhosis, a patient with glucocorticoid excess, a patient with Cushings disease, a patient with activated counterregulatory hormones that occur after trauma or a disease, a patient with hypertriglyceridemia, or a patient with chronic pancreatitis, a nutritively effective amount of one or more nutrients or any combination thereof and one or more insulinotropic peptides. In yet a further embodiment the patient in need of enhanced parenteral nutrition via a non-alimentary route is a critical care patient.
[00427]In one embodiment for critical care use and parenteral nutrition enhancement use, a GD' compound is administered by continuous intravenous infusion to achieve blood glucose levels less than 200 mg/dl, or in the range of 80 to 150 mg/dl, or in the range of 80 to 110 mg/dl. For example, the GD' compound can be administered to maintain plasma glucose below the "renal threshold" of about 160 to 180 milligrams per deciliter. Patients not suffering from hyperglycemia can also be treated in view of the additional anti-catabolic and cardiovascular benefits provided by the GD' compounds.
[00428] As discussed herein, both acute and chronic administration by a range of routes is contemplated for critical care use and for enhancement of parenteral nutrition. In one embodiment for critical care use, a GD' compound is infused continuously at a rate of between about 0.1 and 100 pmollkg/min, between about 0.1 and 50 pmollkg/min, between about 0.5 and 30 pmollkg/min, between about 0.1 and 10 pmol/kg/min, between about 0.5 and 5 pmol/kg/min, or between about 1.0 and 3.0 pmolfkg/min. As discussed elsewhere herein, the GD' compound can also be provided via a sustained release formulatio~, e.g. comprising microspheres or a gel matrix, and/or administered via discrete or superimpositioning
dosages, via subcutaneous, nasal, intravenous or other routes. The GD' compound can be administered prior to, during and/or after commencement of critical care, such as surgery.
[00429] The therapeutically effective dose of the GD', GD' analog, novel GD' analog or GD' hybrid, or derivative thereof, will depend on a number of factors, including without limitation, the patient's sex, weight and age, the severity of inability to regulate blood glucose, the underlying cause(s) of inability to regulate blood glucose, whether glucose or another carbohydrate source is simultaneously administered, the route of administration and bioavailability, the persistence in the body, the formulation and the potency. It is within the skill of the ordinary physician to titrate the dose and rate of administration of a Gil' compound to achieve the desired clinical result.
Polypeptide Production and Purification.
[00430]The polypeptides described herein may be prepared using standard recombinant techniques or chemical peptide synthesis techniques known in the art, e.g., using an automated or semi-automated peptide synthesizer, or both.
[00431] The polypeptides of the invention can be synthesized in solution or on a solid support in accordance with conventional techniques. Such methods are described, for example, herein and in U.S. Patent No. 6,610,824 and U.S. Patent No. 5,686,411 and in patent application Serial No. 454,533 (filed December 6,1999), the entirety of which are incorporated herein by reference. Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, e.g., Stewart and Young, Solid Phase Peptide Synthesis, 2d. ed., Pierce Chemical Co. (1984); Tam et at., J. Am. Chem. Soc. 105: 6442 (1983); Merrifield, Science 232: 341-7 (1986); and Barany and Merrifield, The Peptides, Gross and Meienhofer, eds., Academic Press, New York, 1-284 (1979). Solid phase peptide synthesis may be carried out with an automatic peptide synthesizer (e.g., Model 430A, Applied Biosystems Inc., Foster City, California) using the NMPJHOBt (Option 1) system and tBoc or Fmoc chemistry (see, Applied Biosystems User's Manual for the ABI 430A Peptide Synthesizer, Version 1 .3B July 1, 1988, section 6, pp. 49-70, Applied Biosystems, Inc., Foster City, California) with capping. Peptides may also be assembled using an Advanced Chem Tech Synthesizer (Model MPS 350, Louisville, Kentucky). Peptides may be purified by RP-IIPLC (preparative and analytical) using, e.g., a Waters Delta Piep 3000 system and a C4, C8, or C18 preparative column (10 ~, 2.2x25 cm; Vydac, Hesperia, California). Polypeptides can be synthesized by convergent methods such as "native chemical ligation", and variations thereof, in which two or more peptide fragments with appropriate orthogonally reactive ends are ligated with native amide bond formation. The newly formed peptide can be further ligated to create even logner polypeptides. The individual starting peptides can be derivatized as desired or can be derivatized after a ligation step.
[00432]Peptides analogs were synthesized on a Pioneer continuous flow peptide synthesizer (Applied Biosystems) using PAL-PEG-PS resin (Applied Biosystems) with a loading of 0.2 mmollg (0.25 mnxole scale). Fmoc amino acid (4.0 eq, 1.0 mmol) residues were activated using 4.0 eq HBTU, 4.0 eq of HOBT,

8.0 eq DIEA and coupled to the resin for 1 hour. The Fmoc group was removed by treatment with 20% (v/v) piperidine in dimethylformamide. Final deprotection and cleavage of the peptide from the solid support was performed by treatment of the resin with reagent B (93% TFA, 3% phenol, 3% water and 1% triisopropylsilane) for 2-3 hours. The cleaved peptide was precipitated using tert-butyl methyl ether, pelleted by centrifugation and lyophilized. The pellet was re-dissolved in water (10-15 mL), filtered and purified via reverse phase HPLC using a C- 18 column and an acetonitrile/water gradient containing 0.1% TFA. The purified product was lyophilized and analyzed by ESI-LCIMS and analytical HPLC and were demonstrated to be pure (>98%). Mass results all agreed with calculated values.
[00433] Alternatively, peptides were assembled on a Symphony® peptide synthesizer (Protein Technologies, Inc., Woburn, MA) using Rink amide resin (Novabiochem, San Diego, CA) with a loading of 0.43-0.49 mmol/g at 0.050-0.100 mmol. Fmoc amino acid (Applied Biosystems, Inc. 5.0 eq. 0.250-.500 mmol) residues were dissolved at a concentration of 0.10 M in l-methyl-2-pyrrolidinone. All other reagents (HIBTU, HOBT and N,N-diisopropylethylamine) were prepared as 0.55 M dimethylformamide solutions. The Fmoc protected amino acids were then coupled to the resin-bound amino acid using, HBTU (2.0 eq, 0.100-0.200 mmol), HOBT (l.8eq, 0.090-0.18 mmol), N,N-diisopropylethylamine (2.4 eq, 0.120-0.240 mmol) for 2 hours. Following the last amino acid coupling, the peptide was deprotected using 20% (v/v) piperidine in dimethylformamide for 1 hour. Once peptide sequence is completed, the Symphony® peptide synthesizer is programmed to cleave the resin. Trifluoroacetic acid (TFA) cleavage of the peptide from resin was carried out using a reagent mixture composed of 93% TFA, 3% phenol, 3% water and 1% triisoprcpylsilane. The cleaved peptide was precipitated using tert-butyl methyl ether, pelleted by centifugation and lyophilized. The pellet was dissolved in acetic acid, lyophilized and then dissolved in water, filtered and purified via reverse phase HPLC using a C18 column and an acetonitrile/water gradient containing 0.1% TFA. Anaytical HPLC was used to assess purity of peptide and identity was confirmed by LC/MS and MALDI-MS.
[00434] The active protein can be readily synthesized and then screened in screening assays designed to identify reactive peptides.
[00435] The analog and hybrid polypeptides of the present invention may alternatively be produced by recombinant techniques well known in the art. See, e.g., Sambrook et at., Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor (1989). These GD' analog or hybrid polypeptides produced by recombinant technologies may be expressed from a polynucleotide. One skilled in the art will appreciate that the polynucleotides, including DNA and RNA, that encode such GD' analog or hybrid polypeptides may be obtained from the wild-type cDNA, e.g. Gil', GLP1, amylin, taking into consideration the degeneracy of codon usage, or may be engineered as desired. These polynucleotide sequences may incorporate codons facilitating transcription and translation of mRNA in microbial hosts. Such manufacturing sequences may readily be constructed according to the methods well known in the art. See, e.g., WO 83/04053. The polynucleotides above may also optionally encode an N-terminal methionyl residue. Non-peptide compounds useful in the present invention may be prepared by art-

known methods. For example, phosphate-containing amino acids and peptides containing such amino acids may be prepared using methods known in the art. See, e.g., Bartlett and Landen, Bioorg. Chem. 14:
356-77 (1986).
[004361 A variety of expression vector/host systems may be utilized to contain and express a GD' polypeptide coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid); or animal cell systems. Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), WI 38, BilK, HepG2, 3T3, RN, MDCK, A549, PCl2, K562 and 293 cells. Exemplary protocols for the recombinant expression of the protein are described herein.
[00437] As such, polynucleotide sequences provided by the invention are useful in generating new and useful viral and plasmid DNA vectors, new and useful transformed and transfected procaryotic and eucaryotic host cells (including bacterial, yeast, and mammalian cells grown in culture), and new and useful methods for cultured growth of such host cells capable of expression of the present GD' polypeptides. The polynucleotide sequences encoding GD' analogs or hybrids herein may be useful for gene therapy in instances where underproduction of Gil' or other component peptide hormone(s) of the hybrid would be alleviated, or the need for increased levels of such would be met.
[004381 The present invention also provides for processes for recombinant DNA production of the present GD' polypeptides. Provided is a process for producing the Gil' polypeptides from a host cell containing nucleic acids encoding such -GD' polypeptides comprising: (a) culturing said host cell containing polynucleotides encoding such GD' polypeptides under conditions facilitating the expression of such DNA molecule; and (b) obtaining such Gil' polypeptides.
[004391 Host cells may be prokaryotic or eukaryotic and include bacteria, mammalian cells (such as Chinese Hamster Ovary (CHO) cells, monkey cells, baby hamster kidney cells, cancer cells or other cells), yeast cells, and insect cells.
[00440] Mammalian host systems for the expression of the recombinant protein also are well known to those of skill in the art. Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post-translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphoiylation, lipidation and acylation. Post-translational processing, which cleaves a "prepro" form of the protein, may also be important for correct insertion, folding and/or function. Different host cells, such as CHO, HeLa, MDCK, 293, W138, and the like, have specific cellular
machinery and characteristic mechanisms for such post-translational activities, and may be chosen to ensure the correct modification and processing of the introduced foreign protein.
[00441] Alternatively, a yeast system may be employed to generate the GD' polypeptides of the present invention. The coding region of the GD' polypeptide cDNA is amplified by PCR. A DNA encoding the yeast pre-pro-alpha leader sequence is amplified from yeast genomic DNA in a PCR reaction using one primer containing nucleotides 1-20 of the alpha mating factor gene and another primer complementary to nucleotides 255-235 of this gene (Kurjan and Herskowitz, Cell, 30: 933-43 (1982)). The pre-pro-alpha leader coding sequence and GD' polypeptide coding sequence fragments are ligated into a plasmid containing the yeast alcohol dehydrogenase (ADH2) promoter, such that the promoter directs expression of a fusion protein consisting of the pre-pro-alpha factor fused to the mature GD' polypeptide. As taught by Rose and Broach, Meth. Enz. 185: 234-79, Goeddel ed., Academic Press, Inc., San Diego, California (1990), the vector further includes an ADH2 transcription terminator downstream of the cloning site, the yeast "2-micron" replication origin, the yeast leu-2d gene, the yeast REPi and REP2 genes, the E. coli beta-lactamase gene, and an E. coli origin of replication. The beta-lactamase and leu-2d genes provide for selection in bacteria and yeast, respectively. The leu-2d gene also facilitates increased copy number of the plasmid in yeast to induce higher levels of expression. The REPi and REP2 genes encode proteins involved, in regulation of the plasmid copy number.
[0044.2]The DNA construct described in the preceding paragraph is transformed into yeast cells using a known method, e.g., lithium acetate treatment (Steams et at., Meth. Enz. 185: 280-97 (1990)). The ADH2 promoter is induced upon exhaustion of glucose in the growth media (Price et at., Gene 55: 287 (1987)). The pre-pro-alpha sequence effects secretion of the fusion protein from the cells. Concomitantly, the yeast KEX2 protein cleaves the pre-pro sequence from the mature GD'-polypeptides (Bitter et aL, Proc. Natl. Acad. Sci. USA 81: 5330-4 (1984)).

[004431 GD' polypeptides of the invention may also be recombinantly expressed in yeast using a commercially available expression system, e.g., the Pichia Expression System (Invitrogen, San Diego, California), following the manufacturer's instructions. This system also relies on the pre-pro-alpha sequence to direct secretion, but transcription of the insert is driven by the alcohol oxidase (AOXl) promoter upon induction by methanol. The secreted GD' polypeptide is purified from the yeast growth medium by, e.g., the methods used to purify GD' polypeptide from bacterial and mammalian cell supernatants.
[00444] Alternatively, the cDNA encoding GD' polypeptides may be cloned into the baculovirus expression vector pVL 1393 (PharMingen, San Diego, California). This GIP-compound-encoding vector is then used according to the manufacturer's directions (PharMingen) to infect Spodoptera frugiperda cells in sF9 protein-free media and to produce recombinant protein. The protein is purified and concentrated from the media using a heparin-Sepharose column (Pharmacia, Piscataway, New Jersey) and sequential molecular sizing columns (Amicon, Beverly, Massachusetts), and resuspended in PBS. SDS-PAGE
analysis shows a single band and confirms the size of the protein, and Edman sequencing on a Proton 2090 Peptide Sequencer confirms its N-terminal sequence.
[00445] For example, the DNA sequence encoding the predicted mature GD' analog or hybrid polypeptide may be cloned into a plasmid containing a desired promoter and, optionally, a leader sequence (see, e.g., Better et al., Science 240: 104 1-3 (1988)). The sequence of this construct may be confirmed by automated sequencing. The plasmid is then transformed into E. coli, strain MC 1061, using standard procedures employing CaCI2 incubation and heat shock treatment of the bacteria (Sambrook et aL, sup ra). The transformed bacteria are grown in LB medium supplemented with carbenicillin, and production of the expressed protein is induced by growth in a suitable medium. If present, the leader sequence will affect secretion of the mature GD' analog or hybrid polypeptide and be cleaved during secretion. The secreted recombinant protein is purified from the bacterial culture media by the method described herein.
[00446] Alternatively, the GD' polypeptides of the invention may be expressed in an insect system. Insect systems for protein expression are well known to those of skill in the art. In one such system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. The GD' polypeptide coding sequence is cloned into a nonessential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of GD' analog or hybrid polypeptide will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein coat. The recombinant viruses are then used to infect S. frugiperda cells or Trichoplusia larvae in which GD' analog or hybrid polypeptide is expressed (Smith etal., I. Virol. 46: 584 (1983); Engelhard et at., Proc. Natl. Acad. Sci. USA 91: 3224-7 (1994)).
[00447] In another example, the DNA sequence encoding the GD' polypeptide may be amplified by PCR and cloned into an appropriate vector, for example, pGEX-3X (Pharmacia, Piscataway, New Jersey). The pGEX vector is designed to produce a fusion protein comprising glutathione-S-transferase (GST), encoded by the vector, and a protein encoded by a DNA fragment inserted into the vector's cloning site. The primers for the PCR may be generated to include, for example, an appropriate cleavage site. The recombinant fusion protein may then be cleaved from the GST portion of the fusion protein. The pOEX3X/GD' analog polypeptide construct is transformed into E. coli XL-l Blue cells (Stratagene, La Jolla, California), and individual transformants are isolated and grown at 370C in LB medium (supplemented with carbenicillin) to an optical density at wavelength 600 nm of 0.4, followed by further incubation for 4 hours in the presence of 0.5 mM Isopropyl beta-D-Thiogalactopyranoside (Sigma Chemical Co., St. Louis, Missouri). Plasmid DNA from individual transformants is purified and partially sequenced using an automated sequencer to confirm the presence of the desired GD' polypeptide-encoding gene insert in the proper orientation.
[00448] The fusion protein, expected to be produced as an insoluble inclusion body in the bacteria, may be purified as follows. Cells are harvested by centrifugation; washed in 0.15 M NaCI, 10 mM Tris, pH 8, 1 mM EDTA; and treated with 0.1 mg/mL lysozyme (Sigma Chemical Co.) for 15 mm. at room

temperature. The lysate is cleared by sonication, and cell debris is pelleted by centrifugation for 10 mm. at 12,OOOxg. The fusion protein-containing pellet is resuspended in 50 mM Tris, pH 8, and 10 mM EDTA, layered over 50% glycerol, and centrifuged for 30 mm. at 6000xg. The pellet is resuspended in standard phosphate buffered saline solution (PBS) free of Mg~~ and Ca~4. The fusion protein is further purified by fractionating the resuspended pellet in a denaturing SDS polyacrylamide gel (Sambrook et al., supra). The gel is soaked in 0.4 M KCI to visualize the protein, which is excised and electroeluted in gel-running buffer lacking SDS. If the GST/GD' polypeptide fusion protein is produced in bacteria as a soluble protein, it may be purified using the GST Purification Module (Pharmacia Biotech).
[00449] The fusion protein may be subjected to digestion to cleave the GST from the mature GD' analog or hybrid polypeptide. The digestion reaction (20-40 ~tg fusion protein, 20-30 units human thrombin (4000 U/mg (Sigma) in 0.5 mL PBS) is incubated 16-48 hrs. at room temperature and loaded on a denaturing SDS-PAGE gel to fractionate the reaction products. The gel is soaked in 0.4 M KCI to visualize the protein bands. The identity of the protein band corresponding to the expected molecular weight of the GIl' analog or hybrid polypeptide may be confinned by partial amino acid sequence analysis using an automated sequencer (Applied Biosystems Model 473A, Foster City, California).
[00450] In a particularly exemplary method of recombinant expression of the GD' polypeptides of the present invention, 293 cells may be co-transfected with plasmids containing the GD' analog or hybrid polypeptide cDNA in the pCMV vector (5' CMV promoter, 3' HGH poly A sequence) and pSV2neo (containing the neo resistance gene) by the calcium phosphate method. Preferably, the vectors should be linearized with Scal prior to transfection. Similarly, an alternative construct using a similar pCMV vector with the neo gene incorporated can be used. Stable cell lines are selected from single cell clones by limiting dilution in growth media containing 0.5 mg/mb G418 (neomycin-like antibiotic) for 10-14 days. Cell lines are screened for GD' analog or hybrid polypeptide expression by ELISA or Western blot, and high-expressing cell lines are expanded for large scale growth.
[00451] It is preferable that the transformed cells are used for long-term, high-yield protein production and as such stable expression is desirable. Once such cells are transformed with vectors that contain selectable markers along with the desired expression cassette, the cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media. The selectable marker is designed to confer resistance to selection, and its presence allows growth and recovery of cells that successfully express the introduced sequences. Resistant clumps of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell.
[00452] A number of selection systems may be used to recover the cells that have been transformed for recombinant protein production. Such selection systems include, but are not limited to, HSV thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase genes, in tk-, hgprt- or aprt- cells, respectively. Also, anti-metabolite resistance can be used as the basis of selection for dhfr, that confers resistance to methotrexate; gpt, that confers resistance to mycophenolic acid; neo, that confers resistance to the aminoglycoside, G418; also, that confers resistance to
,chlorsulfuron; and hygro, that confers resistance to hygromyc in. Additional selectable genes that may be useful include trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine. Markers that give a visual indication for identification of transformants include anthocyanins, beta-glucuronidase and its substrate, GUS, and luciferase and its substrate, luciferin.
[00453] Many of the GD' polypeptides of the present invention may be produced using a combination of both automated peptide synthesis and recombinant techniques. For example, a GD' polypeptide of the present invention may contain a combination of modifications including deletion, substitution, and insertion by PEGylation. Such a GD' polypeptide may be produced in stages. In the first stage, an intermediate GD' polypeptide containing the modifications of deletion, substitution, insertion, and any combination thereof, may be produced by recombinant techniques as described. Then after an optional purification step as described herein, the intermediate GD' polypeptide is PEGylated through chemical modification with an appropriate PEGylating reagent (e.g., from NeKtar Transforming Therapeutics, San Carlos, California) to yield the desired GD' polypeptide. One skilled in the art will appreciate that the above-described procedure may be generalized to apply to a GD' polypeptide containing a combination of modifications selected from deletion, substitution, insertion, derivation, and other means of modification well known in the art and contemplated by the present invention.
[00454] It may be desirable to purify the GD' polypeptides generated by the present invention. Peptide purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non-polypeptide fractions. Having separated the polypeptide from other proteins, the polypeptide of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity). Analytical methods particularly suited to the preparation of a pure peptide are ion-exchange chromatography, exclusion chromatography, polyacrylamide gel electrophoresis, and isoelectric focusing. A particularly efficient method of purifying peptides is reverse phase HPLC, followed by characterization of purified product by liquid chromatography/mass spectrometry (LC/MS) and Matrix-Assisted Laser Desorption Ionization (MALDI) mass spectrometry. Additional confirmation of purity is obtained by detennining amino acid analysis.
[00455] Certain aspects of the present invention concern the purification, and in particular embodiments, the substantial purification, of an encoded protein or peptide. The term "purified peptide" as used herein, is intended to refer to a composition, isolatable from other components, wherein the peptide is purified to any degree relative to its naturally obtainable state. A purified peptide therefore also refers to a peptide, free from the environment in which it may naturally occur.
[004561 Generally, "purified" will refer to a peptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term "substantially purified" is used, this designation will refer to a composition in

which the peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the peptides in the composition.
[00457] Various techniques suitable for use in peptide purification will be well known to those of skill in the art. These include, for example, precipitation with ammonium sulphate, PEG, antibodies, and the like; heat denaturation, followed by centrifugation; chromatography steps such as ion exchange, gel filtration, reverse phase, hydroxylapatite and affinity chromatography; isoelectric focusing; gel electrophoresis; and combinations of such and other techniques. As is generally known in the art, it is believed that the order of conducting the various purification steps may be changed, or that certain steps may be omitted, and still result in a suitable method for the preparation of a substantially purified protein or peptide.
[00458] There is no general requirement that the peptides always be provided in their most purified state. Indeed, it is contemplated that less substantially purified products will have utility in certain embodiments. Partial purification may be accomplished by using fewer purification steps in combinatIon, or by utilizing different forms of the same general purification scheme. For example, it is appreciated that a cation-exchange column chromatography performed, utilizing an HPLC apparatus, will generally result in a greater "-fold" purification than the same technique utilizing a low pressure chromatography system. Methods exhibiting a lower degree of relative purification may have advantages in total recovery of protein product, or in maintaining the activity of an expressed protein.
[00459] One may optionally purify and isolate such GD' polypeptides from other components obtained in the process. Methods for purifying a polypeptide can be found in U.S. Patent No. 5,849,883. These documents describe specific exemplary methods for the isolation and purification of G-CSF compositions that may be useful in isolating and purifying the GD' polypeptides of the present invention. Given the disclosure of these patents, it is evident that one of skill in the art would be well aware of numerous purification techniques that may be used to purify GD' polypeptides from a given source.
[00460] Also it is contemplated that a combination of anion exchange and immunoaffinity chromatography may be employed to produce purified GD' polypeptide compositions of the present invention.

Pharmaceutical Compositions.
[00461] The present invention also relates to pharmaceutical compositions comprising a therapeutically or prophylactically effective amount of at least one GD' polypeptide of the invention, or a pharmaceutically acceptable salt thereof, together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers useful in the delivery of the GD' polypeptides. Such compositions may include diluents of various buffer content (e.g., acetate, citrate, tartrate, phosphate, TRIS-HCI), pH and ionic strength; additives such as surfactants and solubilizing agents (e.g., sorb itan monooleate, lecithin, Pluronics, Tween 20 & 80, Polysorbate 20 & 80, propylene glycol, ethanol, PEG-40, sodium dodecyl sulfate), anti-oxidants (e.g., monothioglyercol, ascorbic acid, acetylcysteine, sulfurous acid salts (bisulfise and metabisulfite), preservatives (e.g., phenol, meta-cresol, benzyl alcohol, parabens (methyl,

propyl, butyl), benzalkonium chloride, chlorobutanol, thimersol, phenylmercuric salts, (acetate, borate, nitrate), and tonicity/bulking agents (glycerine, sodium chloride, mannitol, sucrose, trehalose, dextrose); incorporation of the material into particulate preparations of polymeric compounds, such as polylactic acid, polyglycolic acid, etc., or in association with liposomes. Such compositions will influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present GD' analog polypeptides. See, e.g., Remington's Pharmaceutical Sciences 1435-712, 18th ed., Mack Publishing Co., Easton, Pennsylvania (1990).
[00462] Optionally a GD' or a novel GIl' analog can be formulated with (or alternatively administered in adjunct therapy with or alternatively covalently linked or fused to) a second active agent. Such agents can comprise activity that will complement or enhance a GD' effect. In one embodiment such agents have glucose lowering or anti-diabetic activity. In another embodiment the agent inhibits or reduces gastric emptying. In other embodiments the agent can comprise any other desirable activity, such as increasing bone density, reducing food intake, or the like.
[00463] In general, the present GD' analog and hybrid polypeptides will be useful in the same way that GD' and/or the individual component polypeptides (in the case of a hybrid) are useful in view of their pharmacological properties. Generally, the GD' polypeptides are peripherally administered for the treatment or prevention of metabolic conditions and disorders. In particular, the compounds of the invention possess activity as glucose lowering agents, insulinotropic agents, reducing or inhibiting gastric secretion, effecting weight loss, reducing nutrient availability, reducing food intake, suppressing appetite, ameliorating mortality and morbidity in critical (intensive) care applications, providing improved memory and other neurological benefits, increasing or maintaining bone density, effecting cardiovascular benefits, providing cardioprotection, and effecting other therapeutic benefits as discussed herein. In another embodiment, a exemplary use is to administer such hybrid polypeptides for the treatment of diabetes or diabetes related conditions and disorders, obesity, and cardiovascular diseases and conditions. The present GD' polypeptides may be formulated for peripheral administration, including formulation for injection, oral administration, nasal administration, pulmonary administration, topical administration, or other types of administration as one skilled in the art will recognize. More particularly, administration of the pharmaceutical compositions according to the present invention may be via any common route so long as the target tissue is available via that route. In a exemplary embodiment, the pharmaceutical compositions may be introduced into the subject by any conventional peripheral method, e.g., by intravenous, intradermal, intramusclar, subcutaneous, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary (e.g., term release); by oral, sublingual, nasal, buccal, intratracheal, anal, vaginal, transmucosal, pulmonary or transdermal delivery, by suppository or by surgical implantation at a particular site. In one embodiment the administration is parenteral (including intravenous, intradermal, intraperitoneal, intramuscular and subcutaneous).
[004641The treatment may consist of a single dose or a plurality of doses over a period of time. Controlled continual release of the compositions of the present invention is also contemplated.
[00465] Supplementary active ingredients also can be incorporated into the compositions. For use by the physician, the compounds will be provided in dosage unit form containing an amount of a GIl' or novel GD' analog, with or without another therapeutic agent, for example, a glucose-lowering agent, a gastric emptying modulating agent, a lipid lowering agent, or a food intake inhibitor agent. Therapeutically effective amounts of a GD' or a novel GD' analog for example for use in the control of blood glucose or in the control of gastric emptying and in conditions in which gastric emptying is beneficially slowed or regulated are those that decrease post-prandial blood glucose levels, preferably to no more than about 8 or 9 mM or such that blood glucose levels are reduced as desired. In diabetic or glucose intolerant individuals, plasma glucose levels are higher than in normal individuals. In such individuals, beneficial reduction or "smoothing" of post-prandial blood glucose levels may be obtained. As will be recogni~ed by those in the field, an effective amount of therapeutic agent will vary with many factors including the patient's physical condition, the blood sugar level or level of inhibition of gastric emptying to be obtained, or the desired level of food intake reduction, and other factors. In some cases, it will be convenient to provide a GD' polypeptide and at least one other active agent, for example another food-intake-reducing, plasma glucose-lowering or plasma lipid-altering agent, such as an exendin or GLP1 or agonist thereof, amylin, an amylin agonist analog, a CCK or CCK agonist, or a leptin or leptin agonist or a small molecule cannabinoid CB 1 receptor antagonists, beta-hydroxysteroid dehydrogenase- 1 inhibitors, sibutramine and other drugs marketed for treatment of diabetes or obesity, in a single composition or solution for administration together. As has been discussed throughout, the GD' polypeptide may be a GD' hybrid comprising the at least one other such active agent. In other cases, it may be more advantageous to administer the additional agent separately from said GD' polypeptide.
[00466] In one embodiment a GD' polypeptide may be administered separately or together with one or more other compounds and compositions that exhibit a long term or short-term action to lower blood glucose, reduce or inhibit gastric emptying or reduce or inhibit gastric secretions or reduce nutrient availability, including, but not limited to other compounds and compositions that comprise an amylin or amylin analog agonist, salmon calcitonin, a cholecystokinin (CCK) or CCK agonist, a leptin (OB protein) or leptin agonist, an exendin or exendin analog agonist, or a GLP-1 or GLP-1 analog agonist. Suitable amylin agonists include, for example, [~'28'29l'ro-] human amylin (also known as "pramlintide," and described in U.S. Pat. Nos. 5,686,511 and 5,998,367). The CCK used is preferably CCK octapeptide (CCK-8), more preferably its sulfated form. Leptin is discussed in, for example, (Pelleymounter et al., Science 269: 540-3 (1995); Halaas et al., Science 269: 543-6 (1995); Campfield et al., Science 269: 546-9 (1995)). Suitable exendins include exendin-3 and exendin-4, and exendin agonist compounds include, for example, those described in PCT Publications WO 99/07404, WO 99/25727, and WO 99/25728. Suitable agents also include various anti-diabetic agents, e.g. metformin, sulfonyureas, TZDs. In another embodiment is provided a combination therapy of a GD' analog or hybrid, e.g. 0601GD'3794, with an
incretin mimetic, e.g., exenatide or liraglutide. In a further embodiment sub-therapeutic doses of the incretin mimetic are used, which provide a therapeutic benefit when combined with the GD' compound.
[00467] Accordingly, in one embodiment, particularly in patients subject to conditions associated with elevated glucose levels such as diabetic or glucose intolerant individuals, more particularly type 2 diabetic patients, where plasma glucose levels are higher than in normal individuals, GD' therapy in such individuals will benefit from a beneficial reduction or "smoothing" of post-prandial blood glucose levels prior to or concomitant with administration of a GD' or novel GD' analog.
[00468] In contrast to GLP- 1, GLP-1 analogs and mimetics such as exenatide which have been shown to be efficacious in controlling glucose levels in type 2 diabetic patients, the insulinotropic effect of GD' is significantly reduced in diabetic subjects compared to normal individuals. While not to be bound by theory, it is believed that while GD"s incretin effect is attenuated during persistent hyperglycemia, GD' or its analogs will act with a similar potency in diabetic patients as their action in normal subjects once glucose control is improved in these individuals. Thus according to the present invention GD' insensitivity can be reduced by achieving in a patient in need thereof, a glucose-lowering or a reduction or "smoothing" of post-prandial blood glucose levels by appropriate agents or means, prior to or concomitant with administration of a GD' or novel GD' analog that will enable or prolong even further glucose-lowering. In one embodiment the agent or means is provided at least one day prior to GD' administration. In another embodiment the agent or means is provided and a sufficient glucose-lowering is observed prior to GD' administration.
[00469] As mentioned herein suitable agents include exenatide, metformin, sulfonylureas, or combinations thereof. Primary glucose control endpoint can be measured by means known in the art to clinicians. One method is simply to determine blood glucose levels post-prandially. Another is to measure HbAlc levels, as is known in the art.
[00470] A particularly suitable target population is those patients who fail to attain normal glucose concentrations during treatment with a glucose-lowering agent (such as exenatide). A type of hemoglobin called hemoglobin Alc (HbAlc) forms when glucose attaches to hemoglobin. This happens only when blood glucose levels are high. The hemoglobin AIc level can be used to measure a subject's past average blood sugar, e.g. over two to three months. Normal HbAIc values for non-diabetics are approximately 4.0-6.2 percent. The American Diabetes Association recommends that it should be below 7 percent for diabetics, to minimize the complications from diabetes. Despite glucose-lowering therapies, such as exenatide, significant numbers of patients may still remain with elevated Hb lAc. Consequently, in one embodiment such subjects whose HbAlc level (despite treatment with a glucose-lowering agent) remains above normal, at least 7 percent, at least 8 percent, at least 9 percent or at least 10 percent, are suitable subjects for the Gil' therapy and novel adjunct therapy treatments of the invention. In yet another embodiment the HblAc level is greater than normal but no greater than 6.5%, no greater than 7%, no greater than 7.5%, no greater than 8.0%, no greater than 8.5%, no greater than 9.0%, no greater than 9.5%, or no greater than 10%. In yet another embodiment, while reduction of HblAc levels is not reduced to

iormal levels with monotherapy, it is preferably reduced at least 10 percent, at least 20 percent, at least 30, 40, 50, 60, 70, 80 or at least 90 percent from pre-treatment levels, prior to GD' administration or application of a novel adjunct therapy of the invention. In such patients, adjunct therapy with GD' is indicated according to the invention since reduction of hyperglycemia (e.g., as by exenatide) in treated patients, e.g. diabetic patients, poises the patient for reduced GD' insensitivity. Whereas the chronic hyperglycemic condition in type 2 diabetes patients attenuates GD" s insulinotropic response, improved glycemic control resulting from exenatide treatment, for example, would restore responsiveness- of the pancreatic beta-cell to GD' stimulation. Therefore GD' therapy or novel adjunct therapy (e.g. coadministration, GD' phybrid, Gil' fusion protein) of pharmacological doses of GD' or novel GD' analogs with exenatide (or other glucose lowering agents or agents or methods that lower glucose or that reduce or inhibit gastric emptying) will attain GD' sensitivity and lead to desired normoglycemia in diabetic patients or patients suffering from conditions associated with elevated glucose. Since Gil' lacks the gaslric emptying effect of GLP1, nausea may be avoided, thus permitting the use of higher GD' dosing regimens than for GLPl. In yet another embodiment, GD' therapy or novel adjunct therapy will reduce HblAc levels to at least normal levels, or at least 10 percent, at least 20 percent, at least 30, 40, 50, 60, 70, 80 or at least 90 percent from pre-treatment (pre GD' therapy or pre-novel-adjunct therapy) levels.
[00471] In yet a further embodiment, the GD' therapy or novel adjunct therapy can act at least additively, and preferably synergistically, to reduce the dose, dosing, amount, frequency or extent of treatment associated with another agent as mentioned herein. For example, such GD' or novel adjunct therapy can result in at least a 10, 20, 30, 40, 50, 60, 70, 80 or 90% reduction in the amount, dose, frequency of dosing, or length of treatment associated with a the other agent. The agent can be a glucose-lowering agent such a exendin-4, or any other agent, for diabetes or other conditions that benefit form the methods and compositions of the present invention.
[004721The GD' polypeptide of the invention may be prepared for administration as solutions of free base, or pharmacologically acceptable salts in water suitably mixed with surface active agents (e.g., sorbitan monooleate, polyoxyethylene sorbitain monolaurate (Tween 20), polyoxyethylene sorbitan monooleate (Tween 80), lecithin, polyoxyethylene-polyoxypropylene copolymers (Pluronics), hydroxypropylcellulose, ) or complexation agents (e.g., hydroxypropyl-b-cyclodextrmn, sulfobutyether-bcyclodextrin (Captisol), polyvinylpyrrolidone). Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Such products are readily prepared by procedures well known to those skilled in the art. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.

[00473] In one embodiment, the pharmaceutical compositions of the present invention are formulated so as to be suitable for parenteral administration, e.g., via injection or infusion. Preferably, the GD' polypeptide is suspended in an aqueous carrier, for example, in an buffer solution at a pH of about 3.0 to about 8.0, preferably at a pH of about 3.5 to about 7.4, about 3.5 to about 6.0, about 3.5 to about 5.0 or about 3.7 to about 4.7. Useful buffers include sodium acetate/acetic acid, sodium lactate/lactic acid, ascorbic acid, sodium citrate-citric acid, sodium bicarbonate/carbonic acid, sodium succinate/succinic acid, Histidine, Sodium benzoate/benzoic acid, and sodium phosphates, and Tris(hydroxymethyl)aminomehane. A form of repository or "depot" slow release preparation may be used so that therapeutically effective amounts of the preparation are delivered into the bloodstream over many hours or days following transdermal injection or delivery.
[004741 The pharmaceutical compositions suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form should be sterile and should be fluid to the extent that is easily syringable. It is also desirable for the GD' polypeptide of the invention to be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., sorbitol, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), dimethylacetamide, cremorphor EL, suitable mixtures thereof, and oils (e.g., soybean, sesame, castor, cottonseed, ethyl oleate, isopropyl myristate, glycofurol, corn). The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial an antifungal agents, for example, metacresol, benzyl alcohol, parabens (methyl, propyl, butyl), chlorobutanol, phenol, phenylmercuric salts (acetate, borate, nitrate), sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include tonicity agents (for example, sugars, sodium chloride). Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption (for example, aluminum monostearate and gelatin).
[00475] Sterile injectable solutions may be prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the exemplary methods of preparation are vacuum-drying and freeze-drying techniques that yield a powder of the active ingredient plus any additional desired~ ingredient from a previously sterile-filtered solution thereof.
[00476] Generally, a therapeutically or prophylactically effective amount of the present GD' polypeptides will be determined by the age, weight, and condition or severity of the diseases or metabolic conditions or

disorders of the recipient. See, e.g., Remington's Pharmaceutical Sciences 697-773. See also Wang and
-Hanson, Parenteral Formulations of Proteins and Peptides: Stability and Stabilizers, Journal of Parenteral Science and Technology, Technical Report No. 10, Supp. 42:2S (1988). Typically, a dosage of between about 0.001 pg/kg body weight/day to about 1000 pg/kg body weight/day, may be used, but more or less, as a skilled practitioner will recognize, may be used. Dosing may be one, two, three, four or more times daily, or less frequently, such as once a week, once a month, or once a quarter, depending on the formulation, and may be in conjunction with other compositions as described herein. It should be noted that the present invention is not limited to the dosages recited herein.
[004771 Appropriate dosages may be ascertained through the use of established assays for determining level of metabolic conditions or disorders in conjunction with relevant dose-response data. The final dosage regimen will be detennined by the attending physician, considering factors that modify the action of drugs, e.g., the drug's specific activity, severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. As studies are conducted, further information will emerge regarding appropriate dosage levels and duration of treatment for specific diseases and conditions.
[00478] An effective dose will typically be in the range of about 0.5 to 30 pg to about 5 mg/day, preferably about 10 to 30 pg to about 2 mg/day and more preferably about 5 to 100 pg to about 1 mg/day, most preferably about 5 pg to about 500 pg/day, for a 50 kg patient, administered in a single or divided doses and/or controlled continual release, of two, three, four or more administrations. Ine one embodiment the GD' compound is administered peripherally at a dose of about 0.5 pg to about 5 mg per day in single or divided doses or controlled continual release, or at about 0.01 pg/kg to about 500 pg/kg per dose, more preferably about 0.05 pg/kg to about 250 pg/kg, most preferably below about 50 pg/kg.
[00479] Accordingly, exemplary doses can be derived from the total amount of drug to be given a day and the number doses administered a day. For example, exemplary doses can range from about 0.125 pg/dose (0.5 pg given four times a day) to about 2 mg/dose (2 mg given once a day). Other dosages ca~ be between about 0.01 to about 100 pg/kg/dose. The exact dose to be administered may be determined by one of skill in the art and is dependent upon the potency of the particular compound, as well as upon the age, weight and condition of the individual. Administration should begin whenever the therapeutic benefit, e.g. suppression of nutrient availability, food intake, weight loss or control, blood glucose or plasma lipid lowering or modulation, is desired, for example, at the first sign of symptoms or shortly after diagnosis of obesity, diabetes mellitus, or insulin-resistance syndrome. In one embodiment the GD' compound is administered prophylatically. Administration may be by any route, e.g., injection, preferably subcutaneous or intramuscular, oral, nasal, transdermal, etc. Dosages for certain routes, for example oral administration, may be increased to account for decreased bioavailablity, for example, by about 5-100 fold.
[00480]In general, the GD' compounds may be formulated into a stable, safe pharmaceutical composition for administration to a patient. Phannaceutical formulations contemplated for use in the methods of the

invention may comprise approximately 0.01 to 20% (w/v), preferably 0.05 to 10%, of the GD' compound. The GD' compounds may be in an acetate, phosphate, citrate or glutamate buffer allowing a pH of the final composition of about 3.0 to about 7.0; containing carbohydrate or polyhydric alcohol as tonicity modifier and, optionally, approximately 0.005 to 5.0% (w/v) of a preservative selected from the group consisting of m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol. Such a preservative is generally included if the formulated peptide is to be included in a multiple use product.
In a particular embodiment of the present invention, a pharmaceutical formulation of the present invention may contain a range of concentrations of GD' compounds, e.g., between about 0.01% to about 98% w/w, or between about 1 to about 98% w/w, or preferably between 80% and 90% w/w, or preferably between about 0.01% to about 50% w/w, or more preferably between about 10% to about 25% w/w in this embodiment. A sufficient amount of water for injection may be used to obtain the desired concentration of solution. The pharmaceutical formulations described herein may be lyophilized. An exemplary formulation can be 1 mg/mL GD' compound in 10 mM sodium acetate buffer solution, pH 4.2, containing 9.3% sucrose as an osmolality modifier.
[00481] In one embodiment, where the pharmaceutical formulation is to be administered parenterally, the composition is formulation so as to deliver a dose of GD' polypeptide ranging from 0.1 pg/kg to 100 mg/kg body weight/day, preferably at doses ranging from 1 pg/kg to about 50 mg/kg body weight/day. Exemplary daily amounts may be in the range of a lower limit of 2, 5, 10, 20, 40, 60 or 80 to an upper limit of 80 100, 150, 200, or 250. Parenteral administration may be carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient.
[00482] The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration. The optimal pharmaceutical formulation will be determined by one of skill in the art depending on the route of administration and the desired dosage. See, e.g., Remingwn's Pharmaceutical Sciences, sup ra, pages 1435-1712. Such formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose may be calculated according to body weight, body surface areas or organ size. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein, as well as the pharmacokinetic data observed in animals or human clinical trials.
[00483] In one embodiment a formulation of the invention is a liquid, solid, or semi-solid depot, slow, or continuous release formulation capable of delivering an active ingredient of the invention (or multiple active ingredients as for use in the adjunct therapies mentioned herein) over a time period of at least one hour. The release can occur over a period of 24 hours to four months. Such slow or extended release formulations can, in some embodiments, comprise the active ingredient in a slow dissolving form or

formulation, such as a slow-dissolving peptide crystal (such as disclosed in, for example, US Patent No. 6,380,357), in a matrix, or in a coating such as, e.g., an enteric coating or slow-dissolving coating (e.g., coated granules of active ingredient). Slow release matrices are commonly a biodegradable polymer, non-biodegradable polymer, wax, fatty material, etc., and are known in the art (e.g., see U.S. Patent Nos. 6,368,630 and related patents, 6,379,704 and related patents). In addition, parenteral controlled release delivery can be achieved by forming polymeric microcapsules, matrices, solutions, implants and devices and administering them parenterally or by surgical means. These dosage forms would typically have a lower bioavailability due to entrapment of some of the peptide in the polymer matrix or device. (See e.g., US Pat. Nos. 6,379,704, 6,379,703, and 6,296,842).
[00484] As discussed herein one of ordinary skill in the art can readily determine frequency, timing and dose of a GD' or novel GD' analog of the invention for treatment, particularly for conditions associated with elevated glucose, and further when used in adjunct therapy. For example a dose-response relationship over time for the glucose-lowering effect of agent of interest can be first determined, followed by determining a dose-response relationship with the added GD' or novel GD' analog in relation to the doses selected for the other agent. In one example patients with type 2 diabetes are treated and assessed following randomized, subcutaneous injection of placebo, and various amounts of agent on separate days following an overnight fast. Injections are given immediately before ingestion of a standardized Sustacal® meal (7kcallkg) followed by collection of plasma glucose samples at frequent intervals during the subsequent 300 minutes. Glycemic response can be quantified as the time-weighted mean (±SE) change in plasma glucose concentration during the 5-hr period. An ED50 for the glucose lowering effect is determined, and appropriate dose of agent that lowers postprandial plasma glucose concentrations is selected. Subsequently, in a similar study, this dosage is administered to patients while administering varying doses of GD' or a novel GD' analog in order to determine an appropriate dose of GD' that will act in concert with the first agent to further lower or prolong glucose lowering.
[00485] Further. if desired, the timing of administration of the GD' dose relative to when or how long .the first agent was administered is examined in order to identify an optimal dosing regimen. For example, a dose of exenatide can be given on day 1, followed by administration of GD' on day 2, preferably once a glucose lowering response is observed resulting from the exenatide alone. In another embodiment, alternating agent dosing with GD' dosing -is provided. The agent may be administered every other period alternating with GD' administration. The period can be 1, 2, 3, 4, 5, 6, or 7 days, 1 , 2, 3, or 4 weeks, or I, 2, 3, 4, 5 or 6 months. The period can be varied, such as GD' administration 2 days after agent and agent administration 7 days after GD' treatment. Sustained release formulations or methods will extend the period in each case. Further, GD' or novel GD' analogs can be provided in adjunct therapy with (or after cessation of) beta cell neogenesis therapy or islet or beta cell transplantation, where beta cells are increased in number, thus providing a glucose-lowering post treatment. Consequently, in one embodiment such treatment, while not entirely creating a normoglycemia, nonetheless provides a patient
with a sufficiently lowered glucose level, that resistance to GD' is attenuated such that further or more normal glucose levels are achieved post-prandially.
[00486] In one embodiment the agent will have been administered or the method will have been applied at least twenty-four hours prior to commencing GD' administration. More specifically the intervening period can be 24, 36, 48, 60 or 72 hours to 4, 5, 6, 7 or more days, to 2, 3, 4, 5, 6 or more weeks, or any particular intervening time in hours or minutes within the above range. Alternatively, blood glucose levels can be monitored regularly or post-prandially as would be known in the art to measure the effect of the glucose-lowering agent, using methods well known in the art, during and/or following administration of the agent. The administration of GD' can then be performed either at a time when the subject shows a level of response e.g., blood glucose level, indicative of reduced resistance to GD', at which time or thereafter Gil' or a novel GD' analog can be administered.
[00487]As discussed herein, in another embodiment of adjunct therapy with a GD' or novel GD' analog, the therapy comprises an agent or method that slows nutrient to the duodenum (or other more distal nutrient-sensing GD'-secreting sites) that results in lowering glucose levels compared to the absence of the agent or method. In yet another embodiment adjunct therapy comprises an agent or method that slows gastric emptying. In addition to the viscous decelerants, such as described herein (e.g., guar and fiber), the agent includes pharmacologic decelerants of gastric emptying, and includes gut hormones for which gastric slowing is a physiologic event. Such agents include agonists of amylin (e.g. pramlintide), agonists of GLP1 and exendin (e.g. exenatide, NN221 1, ZP-10. liraglutide), agonists of CCK, agonists of PYY, agonists of secretin, agonists of GRP, agonists of neuromedins, and agonists of urocortin. In further embodiments are agents that directly or indirectly promote agonist signaling of the physiologic gastric decelerants. Such agents include secretagogues of endogenous gastric decelerants, and inhibitors of their degrading enzymes, including dipeptidyl dipeptidase inhibitors, or other inhibitors of their clearance, especially at the kidney. In other embodiments are therapeutic strategies that slow the appearance of nutrient stimulus at GD'-secreting cells. Examples include modulation of digestive functions through inhibition of digestive secretions (e.g. gastric acid, pancreatic enzymes, bile) or through inhibition of the digestive effect of such secretions (e.g. acid neutralization, enzyme inhibition, bile sequestration). Inhibiting digestive secretion can occur via agents that directly achieve this (e.g. somatostatin, amylin, calcitonins, PYY) or by agents that interfere with endogenous pro-secretory pathways (e.g. luminal CCKreleasing factor). In one embodiment the adjunct therapy comprises a method that achieves slower gastric emptying and lower nutrient uptake, e.g. diminishes nutrient drive at GD' secreting cells. In one such embodiment is gastric bypass surgery, for example Roux-En-Y Gastric Bypass Surgery, lap band, or physical devices that physically divert nutrient flow from the duodenum.
[00488] In yet other embodiments adjunct therapy comprises methods that stimulate secretion of, or slow degradation of, gastric decelerants. As mentioned guar gum ingestion is one exampl, for example 10 grams guar gum flour per meal. In Another agent is xanthum gum. Other decelerants of nutrient absorption include fiber, as for example provided in a high fiber diet, and as rough fiber in breads and

bran. Another agent is a glucosidase inhibitor, such as acarbose, which slows the rate at which glucose (and fructose) is generated from sucrose at gut brush border disaccaridases. Yet another agent is Miglitol, another a glucosidase inhibitor.
[00489] In yet another embodiment gastric emptying is achieved or mediated by administering a peptide or peptide agonist. Based on the rat, doses of the following peptides have been determined to slow gastric emptying, and are entirely suitable as agents for adjunct therapies of the invention: amylin (e.g., pramlintide doses of 60-600pg/day); GLPl or exendin agonist (e.g., exenatide dose range of 10-50pg/day); CCK and agonists (see Young et al. "Dose-responses for the slowing of gastric emptying in a rodent model by glucagon-like peptide (7-36)NH2, amylin, cholecystokinin, and other possible regulators of nutrient uptake." Metabolism 45 1-3 (1996); incorporated herein by reference). Other agents include Secretin and agonists, CGRP and agonists, Neuromedin agonists, Urocortin agonists, GRP and bombesin agonists, and PYY and agonists.
[004901k the embodiments herein, a method of defining doses can be based on a desired degree of slowing, which can be readily determined. For example, therapeutic doses of pramiintide (30, 60 9Opg; see Kong et al. "The effect of single doses of pramlintide on gastric emptying of two meals in men with IDDM." Diabetologia. 41577-583 (1998)) approximately doubled the half-emptying time of the stomach. Accordingly, in one embodiment adjunct therapy comprises an agent or method that increases the half-emptying time of the stomach by about 200%, however in other embodiments the t '/~ of stomach emptying can increased by more than about 25%, 50%, 75%, 100%, 200%, 300% and even 400% or more.
[00491] By example, an ED5O for exenatide slowing of gastric emptying has been reported at about 0.O5pglkg (e.g., approximately currently used clinical dose) (see Kolterman et al. "Dose-response for inhibition of glucagon secretion and gastric emptying by synthetic exendin-4 (AC2993) in subjects with type 2 diabetes." Diabetes 49 (suppl 1)A1 14 Abstract 460-P (2000)). In adjunct therapy with a Gil' or a novel GD' analog such slowing will provide a increased benefit to the subject. Consequently, in one embodiment agents or methods that provide an equivalent slowing of gastric emptying at pharmaceutically acceptable doses are suitable agents or methods. As discussed throughout, other embodiments of adjunct therapy comprise mammalian amylins, the (25,28,29)proline-human amylin analog (pramlintide), and salmon calcitonin, those being amongst the most potent. Additional suitable peptides and their ECSO (EC(50) nmollkg — sem (EC(50) in pg): Pramlintide, 0.09 _ 0.08 log (0.O7pg); Human amylin, 0.19 _ 0.11 log (0.lSpg); Rat amylin, 0.23 .0.08 log (0.l8pg); Salmon calcitonin, 0.28 _ 0.07 log (0.l9pg); Rat calcitonin, 0.94 _ 0.18 log (0.64pg); Rat CGRP, 2.13 _ 0.29 log (1 .62pg); GLP-1 (7-36)NH2, 2.76 _ 0.12 log (1.82pg); Secretin, 3.09 — 0.20 log (1.87pg); CCK-8, 12.8 _ 0.20 log (2.93pg); Gastrin releasing peptide, 49.9 _ 0.05 log (28.Spg) (see Gedulin et al. "Comparison of 21 peptides on inhibition of gastric emptying in conscious rats." Dig. Dis. Week. A742 (abstract 2967) (1996); This study reported the potency and effect of 21 peptides to modulate gastric emptying in doseresponse studies in conscious rats (n=—18 rats/peptide, rat weight .-200g). Peptide was injected
subcutaneously 5 mm before gavage with an acaloric dye-labeled methyl cellulose gel. Animals were sacrificed 20 mm later and the dye content of the stomach measured spectroscopically to assess emptying. Only 10 peptides were found to fully inhibit gastric emptying at doses up to 100ug/rat. Peptides found to be either weakiy active or inactive at doses of lOOpg were vasoactive intestinal peptide (VD'), gastric inhibitory peptide (GD'), pancreatic polypeptide, neuropeptide Y, -glucagon, insulin (if plasma glucose was maintained constant), gastrin, somatostatin, pituitary adenylate cyclase activating peptide (PACAP38), adrenomedullin and deamidated pramlintide.). In other embodiments of the invention the agent comprise a agonist of any of the peptides mentioned herein, such as an antibody or antibody fragment agonist or small molecule agonist.
[00492] As described throughout, such agents (or their combination) can be administered either separately or together with GD' or can comprise a chimeric molecule with a GD', e.g., either chemically linked or recombinant fusion. As described throughout (e.g., see times and periods mentioned herein), when administered separately GD' can be administered either at a designated time point after administration of the agent(s) or method, or it can be administered at or shortly after a desired effect has been obtained by prior administration of the agent or method, such effect associated with reducing the subject's GD' resistance.
[00493] It will be appreciated that the pharmaceutical compositions and treatment methods of the invention may be useful in fields of human medicine and veterinary medicine. Thus the subject to be treated may be a mammal, preferably human or other animal. For veterinary purposes, subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice, rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkeys, ducks and geese.
[00494] In addition, the present invention contemplates a kit comprising a GD' analog or hybrid polypeptide of the invention, components suitable for preparing said GD' compound polypeptide of-the invention for pharmaceutical application, and instructions for using GD' compound polypeptide and components for pharmaceutical application.
Additional References.
[00495] 1. Fehmann HC, Goke B, Goke R, Trautmann ME, Arnold R. Synergistic stimulatory effect of glucagon-like peptide-1 (7-36) amide and glucose-dependent insulin-releasing polypeptide on the endocrine rat pancreas.FEBS Lett. 1989 252:109-12
[00496] 2. Nauck MA, Bartels E, Orskov C, Ebert R, Creutzfeldt W. Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide- 1 -(7-3 6) amide infused at near-physiological insulinotropic hormone and glucose concentrations.J Chin Endocrinol Metab.
[00497] 3. Vilsboll T, Krarup T, Madsbad 5, HoIst JJ. Both GLP-1 and GD' are insulinotropic at basal and postprandial glucose levels and contribute nearly equally to the incretin effect of a meal in healthy subjects. Regul Pept. 2003;114:115-21
[00498] 4. Jones IR, Owens DR. Moody AJ, Luzio SD, Morris T, Hayes TM. The effects of glucose-
dependent insulinotropic polypeptide infused at physiological concentrations in normal subjects and type
2 (non-insulin-dependent) diabetic patients on glucose tolerance and beta-cell secretion. Diabetologia.
1987 30:707-12.
[00499] 5. Nauck MA, Heimesaat MM, Orskov C, HoIst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest. 1993 91:301-7.
[00500] 6. Elahi D, McAloon-Dyke M, Fukagawa NK, Meneilly GS, Sclater AL, Minaker KL, Habener JF, Andersen DK. The insulinotropic actions of glucose-dependent insulinotropic polypeptide (GD') and glucagon-like peptide-1 (7-37) in normal and diabetic subjects. Regul Pept. 1994 Apr 14;5 l(l):63-74.
[00501] 7. HoIst JI, Gromada J, Nauck MA. The pathogenesis of NIDDM involves a defective expression of the GD' receptor. Diabetologia. 1997 40:984-6.
[00502] 8. Lynn FC, Pamir N, Ng Eli, McIntosh CH, Kieffer TJ, Pederson RA. Defective glucose-dependent insulinotropic polypeptide receptor expression in diabetic fatty Zucker rats. Diabetes. 200150:1004-11
[00503] 9. Meier JJ, Hucking K, HoIst JJ, Deacon CF. Schmiegel WH, Nauck MA. Reduced insulinotropic effect of gastric inhibitory polypeptide in first-degree relatives of patients with type 2 diabetes. Diabetes. 2001 50:2497-504.
[00504] 10. Meier JJ, Gallwitz B, Kask B, Deacon CF, HoIst JJ, Schmidt WE, Nauck MA. Stimulation of insulin secretion by intravenous bolus injection and continuous infusion of gastric inhibitory polypeptide in patients with type 2 diabetes and healthy control subjects. Diabetes. 2004 53 Suppl 3:S220-
[00505] 11. Vilsboll T, Knop FK, Krarup T, Johansen A, Madsbad 5, Larsen 5, Hansen T, Pedersen 0, HoIst JJ. The pathophysiology of diabetes involves a defective amplification of the late-phase insulin response to glucose by glucose-dependent insulinotropic polypeptide-regardless of etiology and phenotype. J Clin Endocrinol Metab. 2003 88:4897-903.
[00506] 12. Vilsboll T, Krarup T, Madsbad 5, Holst JJ. Defective amplification of the late phase insulin response to glucose by GD' in obese Type il diabetic patients. Diabetologia. 2002 45:1111-9.
[00507] 13. Young AA, Gedulin BR, Rink TJ. Dose-responses for the slowing of gastric emptying in a rodent model by glucagon-like peptide (7-36) N}12, amylin, cholecystokinin, and other possible regulators of nutrient uptake. Metabolism. 1996 45:1-3.



[00508] 14. Meier JJ, Goetze 0, Anstipp J, Hagemann D, HoIst JJ, Schmidt WE, Gallwitz B, Nauck

MA. Gastric inhibitory polypeptide does not inhibit gastric emptying in humans. Am J Physiol Endocrinol Metab. 2004 286:E621-5.

[00509] 15. Kieffer TJ. Gastro-intestinal hormones GD' and GLP-l. Ann Endocrinol 200465:13-21. [00510] 16. Jones JR. Owens DR, Moody AJ, Luzio SD, Morris T, Hayes TM. The effects of glucose-dependent insulinotropic polypeptide infused at physiological concentrations in normal subjects and type 2 (non-insulin-dependent) diabetic patients on glucose tolerance and B-cell secretion. Diabetologia. 1987

30:707-12.

[00511] 17. Nauck MA, Heimesaat MM, Orskov C, Hoist JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest. 1993 91:301-7.

[00512] 18. Elahi D, McAloon-Dyke M, Fukagawa NK, Meneilly GS, Sclater AL, Minaker KL, Habener JF, Andersen DK. The insulinotropic actions of glucose-dependent insulinotropic polypeptide (GIl') and glucagon-like peptide-1 (7-3 7) in normal and diabetic subjects. Regul Pept. 1994 Apr 14;51(l):63-74.

[00513] 19. Holst JJ, Gromada J, Nauck MA. The pathogenesis of NIDDM involves a defective expression of the Gil' receptor. Diabetologia. 1997 40:984-6.

[005141 20. Young AA, Gedulin BR, Rink TJ. Dose-responses for the slowing of gastric emptying in a rodent model by glucagon-like peptide (7-36) NH2, amylin, cholecystokinin, and other possible regulators of nutrient uptake. Metabolism. 1996 45:1-3.

[00515] 21. Kieffer TJ. Gastro-intestinal hormones GD' and GLP-l. Ann Endocrinol 2004 65:13-21.

[00516] 22. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, WilIms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide 1 are rapidly degraded from -the N112-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995 44:1126-1131.
[00517] 23. Deacon CF. Johnsen AH, Hoist JJ. Degradation of glucagon-like-peptide- 1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J. Clin. Endocrinol. Metab. 1995 80:952-957.
[00518] 24. Kieffer TJ, McIntosh CH, Pederson RA. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology. 1995 Aug; I 36(8):3585-96.
[00519] 25. Plamboeck A, Holst JJ, Carr RD. Deacon CF. Neutral endopeptidase 24.11 and dipeptidyl peptidase IV are both involved in regulating the metabolic stability of glucagon-like peptide-l in vivo. Adv Exp Med Biol. 2003 ;524:303-12.
[005201 26. Hupe-Sodmann K, McGregor GP, Bridenbaugh R, Goke R, Goke B, Thole H, Zimmermann B, Voigt K. Characterisation of the processing by human neutral endopeptidase 24.11 of GLP- 1(7-36) amide and comparison of the substrate specificity of the enzyme for other glucagon-like peptides. Regul Pept. 1995 Aug 22;58(3): 149-56.
[00521] 27. Gault VA, Flatt PR, Harriott P. Mooney MH, Bailey CJ, O'Harte Fl'. Improved biological activity of Gly2- and Ser2-substituted analogues of glucose-dependent insulinotrophic polypeptide. J Endocrinol. 2003 Jan;176(1):133-41.
[00522] 28. Hinke SA, Gelling RW, Pederson RA, Manhart 5, Nian C, Demuth HU, McIntosh CH. Dipeptidyl peptidase IV-resistant [D-Ala(2)]glucose-dependent insulinotropic polypeptide (GD') improves glucose tolerance in normal and obese diabetic rats. Diabetes. 2002 Mar;5 l(3):652-6 1.
[005231 29. Hudson FM, Andersen NH. Exenatide: NMR/CD evaluation of the medium dependence of conformation and aggregation state. Biopolyiners. 2004 76:298-308.
[00524] 30. Andersen NH, Brodsky Y, Neidigh JW, Prickett KS. Medium-dependence of the secondary structure of exendin-4 and glucagon-like-peptide-1. Bioorg Med Chem. 2002 10:79-85.
[005251 31. Neidigh JW, Fesinmeyer RM, Prickett KS, Andersen NH. Exendin-4 and glucagoii-likepeptide-1: NMR structural comparisons in the solution and micelle-associated states. Biochemistry. 2001
40:13188-200.
[00526] 32. Thum A, Hupe-Sodmann K, Goke R, Voigt K, Goke B, McGregor GP. Endoproteolysis by isolated membrane peptidases reveal metabolic stability of glucagon-like peptide- I analogs, exendins-3 and -4. Exp Clin Endocrinol Diabetes. 2002 110:113-8.
[00527] 33. Fehmann HC, Goke B. Characterization of GD'(1-30) and GD'(1-42) as stimulators of proinsulin gene transcription. Peptides. 1995 16:1149-52. -
[Oos28lThroughout this application various publications are referenced. The disclosures of these publications in their entirety are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
[00529] To assist in understanding the present invention, the following Examples are included. The experiments - relating to this invention should not, of course, be construed as specifically limiting the invention and such variations of the invention, now known or later developed, which would be within the purview of one skilled in the art are considered to fall within the scope of the invention as described herein and hereinafter claimed.

EXAMPLES
[00530] The examples illustrate the preparation of the present GD' polypeptides, and the testing of these Gil' polypeptides of the invention in vitro and/or in vivo.

Example 1. Preparation of GD' Polvoentides
[00531]Peptides of the invention may be assembled on a Symphony peptide synthesizer (Protein Technologies, Inc.) using Rink amide resin (Novabiochem) with a loading of 0.43-0.49 mmol/g at 0.050-0.100 mmol or a pre-loaded Wang Resin (Fmoc-Tyr(tBu)-Wang resin) 0.63 mmol/g (Novabiochem). Fmoc amino acid (5.0 eq. 0.250-.500 mmol) residues are dissolved at a concentration of 0.10 M in 1-methyl-2-pyrrolidinone. All other reagents (HBTU, 1- hydroxybenzotriazole hydrate and N,NDiisopropylethylamine ) are prepared as 0.55 M dimethylformamide solutions. The Fmoc protected amino acids are then coupled to the resin-bound amino acid using, HBTU (2.0 eq, 0.100-0.200 mmol), 1-hydroxybenzotriazole hydrate (1.8 eq, 0.090-0.18 mmol), N,N-diisopropylethylamine (2.4 eq. 0.120-0.240 mmol) for 2 hours. Following the last amino acid coupling, the peptide is deprotected using 20% (v/v) piperidine in dimethylformamide for 1 hour. Once peptide sequence is complete, the Symphony peptide synthesizer is programmed to cleave the resin. Trifluoroacetic acid (TFA) cleavage of the peptide from resin is carried out using 93% TFA, 3% phenol, 3% water and 1% triisopropyisilane for 1 hour. The cleaved peptide is precipitated using tert-butyl methyl ether, pelleted by centrifugation and lyophilized. The pellet is re-dissolved in water (10-15 mL), filtered and purified via reverse phase HPLC using a C18 column and an acetonitrile/water gradient containing 0.1% TFA. The resulting peptides are purified to homogeneity by reverse phase HPLC and the purity is confirmed by LCMS.
[00532] A general procedure for N-capping the peptides of the invention with fatty acids (e.g., octanoic and stearic acids) is as follows: Peptide on rink amide resin (0.1 mmol) is suspended in NMP (5 mL). In a separate vial, IIBTU (0.3 mmol), HOBt (0.3 mmol) is dissolved in DMF (5 mL) followed by the addition of DIEA (0.6 mmol). This solution is added to the resin and this suspension is shaken for 2 hrs. The solvent is filtered and washed thoroughly with NMP (5 mLx4) and CH2CI2 (20 mL), dried and is subjected to the TFA cleavage for 1 hr. The yield of the desired peptide is ca. 40 mg after cleavage and purification.
[00533]PEG modification may be carried out in solution on a free epsilon-amino group of lysine or a terminal amino group of a purified peptide using commercially available activated PEG esters. The resulting PEGylated derivatives are purified to homogeneity by reverse phase I-D'LC and the purity is confirmed by LC/MS and MALDI-MS.
Example 2. Bindina Assays.
[00534] The GD' polypeptides of the invention may be tested in a variety of receptor, e.g. GD'R, GLP-IR, amylin receptor, binding assays using binding assay methodologies generally known to those skilled in the art. Such assays include those described herein.
[00535] Amylin binding assay: Evaluation of the binding of some exemplary compounds of the invention to amylin receptors may be carried out as follows in nuclueus accumbens membranes prepared from rat brain. Male Sprague-Dawley® rats (200-250) grams are sacrificed by decapitation. Brains are removed and place in cold phosphate-buffered saline (PBS). From the ventral surface, cuts are made rostral to the

hypothalamus, bounded laterally by the olfactory tracts and extending at a 45" angle medially from these tracts. This basal forebrain tissue, containing the nucleus accumbens and surrounding regions, is weighed and homogenized in ice-cold 20 mM HEPES buffer (20 mM HEPES acid, pH adjusted to 7.4 with NaOH at 230C). Membranes are washed three times in fresh buffer by centrifugation for 15 minutes at 48,000 x
g. The final membrane pellet is resuspended in 20 mM HEPES buffer containing 0.2 mM phenylmethylsulfonyl fluoride (PMSF).
[00536]To measure 1~I-amylin binding (see, Beaumont K et at. Can J Physiol Pharmacol. 1995 Jul; 73(7): 1025-9), membranes from 4 mg original wet weight of tissue are incubated with 1251 -amylin at 12-
16 pM in 20 mM HEPES buffer containing 0.5 mg/mi bacitracin, 0.5 mg/mi bovine serum albumin, and
0.2 mM PMSF. Solutions are incubated for 60 minutes at 20C. Incubations are terminated by filtration through GF/B glass fiber filters (Whatman Inc., Clifton, N.J.) that are presoaked for 4 hours in 0.3% poylethyleneimine in order to reduce nonspecific binding of radiolabeled peptides. Filters are washed immediately before filtration with 5 mi cold PBS, and immediately after filtration with 15 ml cold PBS. Filters are removed and radioactivity assessed in a gamma-counter at a counting efficiency of 77%. Competition curves are generated by measuring binding in the presence of 1012 to 10~ M unlabeled test compound and are analyzed by nonlinear regression using a 4-parameter logistic equation (Inplot program; GraphPAD Software, San Diego).
[00537] CGRP receptor binding assay: Evaluation of the binding of compounds of the invention to CGRP receptors are essentially as described for amylin except using membranes prepared from SK-N-MC cells, known to express CGRP receptors (Muff, R. et.at., Ann NY Acad. Sci. 1992: 657, 106-16). Binding assays are performed as described for amylin except using 13,500 cpm 1 251-hCGRP well or 21.7 pM/well (Amersham).
[005381 Adrenomedullin binding assay: Binding to the adrenomedullin receptor may be investigated using HUVECs that contain the adrenomedullin receptor (Kato J et.at., Eur J PhannacoL 1995, 289:383-5) using the Perkin Elmer AlphaScreenTM assay for cyclic AMP using an optimum of 25-30,000 cells per well. Elevation of cAMP levels is not large for HUVEC compared to CHO cells. As such, CHO cells may be chosen as a negative control since they do not express the adrenomedullin receptor if desired.
[00539] Calcitonin receptor binding assay: Binding to the calcitonin receptor may be investigated using CHO cells or T47D cells, which also express the calcitonin receptor (Muff R. et.at, Ann N Y Acad Sci. 1992, 657:106-16 and Kuestner R.E. et. al. Mol Pharmacol. 1994, 46:246-55), as known in the art.
[00540] Leptin binding assay: Two in vitro bioassays are routinely used to assess leptin binding and receptor activation (see e.g., White, et al., 1997. Proc.Natl. Acad. Sci. U. S. A. 94: 10657-10662). An alkaline phosphatase("AP")-leptin ("OB") fusion protein ("AP-OB") may be used to measure inhibition of leptin binding in the absence or presence of recombinant mouse leptin (positive control) or peptide, by COS-7 cells transfected with the long (signaling) form of the mouse OB receptor ("OB-RL"). Signal transduction assays may be done in GT1-7 cells cotransfected with AP reporter and OB-RL constructs.
Secreted alkaline phosphatase("SEAP") activity in response to stimulation with mouse leptin or peptide may be measured by chemiluminescence.
[00541] Y I receptor binding assay: Membranes are prepared from confluent cultures of SK-N-MC cells that endogenously expresses the neuropeptide Y1 receptors. Membranes are incubated with 60 pM [1251]- human Peptide YY (2200 Cilmmol, PerkinElmer Life Sciences), and with unlabeled test compound for 60 minutes at ambient temperature in a 96 well polystyrene plate. Then well contents are harvested onto a 96 well glass fiber plate using a Perkin Elmer plate harvestor. Dried glass fiber plates are combined with scintillant and counted on a Perkin Elmer scintillation counter.
[00542] Y2 receptor binding assay: Membranes are prepared from confluent cultures of SK-N-BE cells that endogenously expresses the neuropeptide Y2 receptors. Membranes are incubated with 30 l,M [1251]- human Peptide YY (2200 Cilmmol, PerkinElmer Life Sciences), and with unlabeled test compound for 60 minutes at ambient temperature in a 96 well polystyrene plate. Then well contents are harvested onto a 96 well glass fiber plate using a Perkin Elmer plate harvestor. Dried glass fiber plates are combined with scintillant and counted on a Perkin Elmer scintillation counter.
[005431 Y4 receptor binding assay: CHO-KI cells are transiently transfected with cDNA encoding neuropeptide Y4 gene, and then forty-eight hours later membranes are prepared from confluent cell cultures. Membranes are incubated with 18 pM [1251]- human Pancreatic Polypeptide (2200 Cilmmol, PerkinElmer Life Sciences), and with unlabeled test compound for 60 minutes at ambient temperature in a 96 well polystyrene plate. Then well contents are harvested onto a 96 well glass fiber plate using a Perkin Elmer plate harvestor. Dried glass fiber plates are combined with scintillant and counted on a Perkin Elmer scintillation counter.
[00544] YS receptor binding assay: CHO-Ki cells are transiently transfected with cDNA encoding neuropeptide YS gene, and then forty-eight hours later membranes are prepared from confluent cell cultures. Membranes are incubated with 44 pM [12511- human Peptide YY (2200 Cilmmol, PerkinElmer Life Sciences), and with unlabeled test compound for 60 minutes at ambient temperature in a 96 well polystyrene plate. Then well contents are harvested onto a 96 well glass fiber plate using a Perkin Elmer plate harvestor. Dried glass fiber plates are combined with scintillant and counted on a Perkin Elmer scintillation counter.
[00545] GLP- I receptor binding assay: GLP- 1 receptor binding activity and affinity may be measured using a binding displacement assay in which the receptor source is RINm5F cell membranes, and the ligand is [1251]GLP-1. Homogenized RINmSF cell membranes are incubated in 20 mM HEPES buffer with 40,000 cpm [1251]GLP-1 tracer, and varying concentrations of test compound for 2 hours at 230 C with constant mixing. Reaction mixtures are filtered through glass filter pads presoaked with 0.3% PEI solution and rinsed with ice-cold phosphate buffered saline. Bound counts are determined using a scintillation counter. Binding affinities are calculated using GraphPad Prism software (GraphPad Software, Inc., San Diego, CA).


Example 3: Mouse Food Intake Assay.
[00546] The GD' hybrid polypeptides of the invention may be tested for appetite suppression in the mouse food intake assay and for their effect on body weight gain in diet-induced obesity (DIO) mice. The experimental protocols for the screens are described herein.
[00547JFemale NIHISwiss mice (8-24 weeks old) are group housed with a 12:12 hour light:dark cycle with lights on at 0600. Water and a standard pelleted mouse chow diet are available ad libitum, except as noted. Animals are fasted starting at approximately 1500 hrs, 1 day prior to experiment. The morning of the experiment, animals are divided into experimental groups. In a typical study, n=4 cages with 3 mice/cage.
[00548] At time=0 mm, all animals are given an intraperitoneal injection of vehicle or compound, typically in an amount ranging from about 10 nmol/kg to 75 nmol/kg, and immediately given a preweighed amount (1O-lSg) of the standard chow. Food is removed and weighed, typically at 30, 60, aiid 120 minutes, to determine the amount of food consumed (Morley, Flood et aL, Am. J. Physiol. 267: Ri 78-Ri 84, 1994). Food intake is calculated by subtracting the weight of the food remaining at the e.g., 30, 60, 120, 180 and/or 240 minute time point, from the weight of the food provided initially at time=0. Significant treatment effects are identified by ANOVA (p [00549] Activity in the food intake assay and sequence of parent molecules used with GD' for the synthesis of hybrids herein include:

(Table Removed)
Examnle 4: Body Weight Gain in Fattened CS7B 1/6 (Diet-induced-obesity, or DIO) Mice.
[005501 Male C57BL/6 mice (4 weeks old at start of study) are fed high fat (HF, 58% of dietary kcal as fat) or low fat (LF, 11% of dietary kcal as fat) chow. After 4 weeks on chow, each mouse is implanted with an osmotic pump (AIzet # 2002) that subcutaneously delivers a predetermined dose of hybrid polypeptide continuously for two weeks. Body weight and food intake are measured weekly (Surwit et at., Metabolism—Clinical and Experimental, 44: 645-51, 1995). Effects of the test compound are expressed as the mean +/- sd of % body weight change (i.e., % change from starting weight) of at least 14 mice per treatment group (p.czo.05 ANOVA, Dunnett's test, Prism v. 2.01, GraphPad Software Inc., San Diego, California).
Example 5. Testing of GD' Hybrids Containinz a Heterolo~ous C-terminal Tail.
[00551] Circular dichroism (CD) and NMIR studies of Exendin-4 in aqueous media and in media containing organic cosolvents reveal that the C-terminal segment containing the sequence, LFIEWLKNGGPSSGAPPPS (SEQ ID NO: 183) (residue 2 1-39) forms a unique hydrophobic Trp-cage cluster resulting from interactions of Pro37 with Phe22 and Pro38 with Trp25 (14-16). Interestingly, there is no evidence of Trp-cage formation in water containing dodecylphosphocholine (DPC), a micellar state that mimics a biological membrane environment. NMR spectral data indicate rapid segmental motion of the eight C-terminal residues, presumably because the Trp-cage is destabilized due to energetically favorable association of the Trp residue with the phosphocholine head groups. This Trp-cage cluster motif is the first example of a protein-like tertiary structure displayed by a peptide, and could be responsible for imparting greater metabolic stability by masking protease-sensitive sites in the molecule in vivo (17).
[00552] Thus, GD' analog or hybrids were designed with the premise that these peptides could assume the 'I'rp cage structure reported for Exendin-4 by appending the Exendin tail sequence to the C-termini of the truncated GD' peptides GD'-( 1-30) and GIP-( 1-26). In addition, substitutions of the Tyr and Ala residues at the N-terminus of GD' were also made to confer resistance to DPP-IV peptidase degradation. These metabolically stable GD' analog or hybrids can be used as monotherapy or as an adjunct therapy with Exendin-4 (or other GLP- I agonists) or ant-diabetic drugs such as metformin, sulphonylureas, thiazolidinediones, pramiintide and insulin for treatment of type 2 diabetes.

[005531 Analogs can and were screened in GD' receptor binding and acute glucose lowering assays in NIH Swiss or diabetic db/db mice. See Figures 2-4. In vitro GLl'-1 and glucagon receptor binding counterscreens were carried out to assess receptor specificity. The enzymatic stability of these GD' analog or hybrids are also being tested by incubation of the peptides with kidney brush-border membrane extract, membrane extract from RINmSF cells and purified neutral endopeptidase 24.11, followed by analysis of the cleavage products by LC-MS/MS (data not shown).
[00554] Analogs can be and were characterized further to assess their anti-diabetic effects, and in particular, the glucose lowering action in normal mice of Compound G was found to be significantly more efficacious than full length GD' (Figures 3A and 3B). As seen in Figure 4, Compound G shows a pronounced and long-acting effect in lowering glucose in a diabetic mouse model, whereas the action of GD' at a 10 fold higher dose is modest and wanes over time. The glucose lowering profile of Compound G also differs from that of Exendin-4 (Compound K), which has a more rapid onset of action.
Example 6. Enhanced Glucose Lowerin2 Effects of GD' Analogs and Hybrids.
[00555] Glucose lowering effect in vivo of novel GD' analog or hybrids was determined. Figures 8A and
8b provide a result of one such study. Points represent mean ± sem. Peptide was injected D' at t=0
immediately following baseline sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60,
120, 180 and 240 minutes. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a
Johnson & Johnson Company, Milpitas, CA). *p [00556] Tested were: Compound Number A, human GD' acid form:
YAEGTFISDYSIAMDKilIQQDFVNWLLAQKGKKNDWKHNITQOH (SEQ ID NO: 2);
Compound No. I, D-Ala2 GD' acid form:
Y(D~Ala)EGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNITQOW and Compound No. G, D-AIa2GD'(l-30)-l'SSGAPPPS (SEQ ID NO: 1) amide form:
Y(D~Ala)EGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPSNH2.
100557] The data demonstrates that a D-alanine substitution at position 2 in the novel GD' analog or hybrids herein improves glucose lowering ability in vivo. Addition of D-Ala even to full length improves glucose lowering compared to unmodified GD': improved activity is seen in the first hour, but wanes over time. In contrast, a clearly superior and surprising profile is observed when both a protease resistant Nterminus and a Trp-cage C-terminal shield are present. For example, the profile an analog (see Compound G) comprising both aspects shows gradually increasing activity that peaks at t=l2Omin, and is more sustained than native GD' or its D-Ala2 version.

[005581 Figure 9 depicts glucose lowering effect of novel GD' analog or hybrids, particularly the effect of a Trp-cage. Points represent mean ± sem. Peptide was injected D' at t=0 immediately following baseline sample into 2-hour fasted NilhISwiss mice. Samples were taken at t=60, 120, 180 and 240mm. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA). p0.05 vs. vehicle control; ANOVA, Dunnett's test.
[00559] Tested were: Compound No. H, (D-Ala2)GD'( 1-30) amide form:
Y(D-Ala)EGTFISDYSIAMDKIHQQDFVNWLLAQK-NH2; and Compound No. G, (D-Ala2)GD'(l-30)-
PSSGAPPPS (SEQ ID NO: 1) amide form: Y(DAla)EGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPl'PS-NH2.
[00560] The data demonstrates that the presence of a C-terminal Trp-cage sequence provides a significant and surprising benefit to Gil' activity, particularly in truncated Gil' analog or hybrids.
[005611 Glucose lowering effect in vivo of various analogs and hybrids was determined and shown in -Figures IOA and lOB. In this example, Ac modification and a Pro3 substitution did not significantly enhance glucose lowering ability. Points represent mean ± sem. Peptide was injected D' at t=0 immediately following baseline sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60, 120, 180 and 240mm. Blood glucose was measured with a OneTouch® Ultra® (LifeScan, Inc., a Johnson & Johnson Company, Milpitas, CA). *p [00562] Tested were:
Cmpd. A, Human GD' acid form: YAEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKHNITQ (SEQ ID NO: 2);
Cmpd. B: (AcY)(D-Ala)GIP(1-30)-PSSGAPPPS (SEQ ID NO: 1) amide form:
Ac-Y(DAIa)EGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS-NH2;
Cmpd. C: (Ac-Y)GD' acid form:
AcY-AEGTFISDYSIAMDKIHQQDFVNWLLAQKGKKNDWKIINlTQPSSGAPPPS (SEQ ID NO:290);
Cmpd. D, Pro3GD'(l-30)-PSSGAPPPS (SEQ ID NO: 1) amide:
YAPGTFISDYSIAMDKIHQQDFVNWLLAQKPSSGAPPPS-NH2 (SEQ ID NO: 245);
Cmpd E, Pro3GIP(1-42) acid form: YAPGTFISDYSIAMDKLHQQDFVNWLLAQKGKKNDWKHN1TQ (SEQ ID NO: 291);
Crnpd F, Pro3GD'(1-30) amide:
'YAPGTFISDYSIAMDKIHQQDFVNWLLAQK-NH2 (SEQ ID NO: 292); and
Cmpd G, (D-Ala2)GD'(1-30)-PSSGAPPPS (SEQ ID NO: 1) amide (also referred to as 0601G1P3794):
Y(D-Ala)EGTFISDYSIAMDKIIIQQDFVNWLLAQKPSSGAPPPS-N112.
[00563] To compare the insulinotropic effect of various GD' analog hybrids in response to an intravenous glucose challenge, GD' analog hybrids were tested in an intra venous glucose tolerance test (IVGIT assay) at 100 pmol/kg/min infusion dose in rats. GD' compounds were selected that displayed significant glucose lowering in vivo. For example, while native GD' displayed no or little activity in basal glucose lowering (basal glucose lowering (% maximal decrease) at 0% and OGIT response of —10%), the analog 0601GW4042 and the analog hybrids 0601GW 3794 and 060 1GW 4178 dispiayed, respectively, a very transient —11%, -20% and delayed onset —20% for basal glucose lowering, and "ND" (not determined), —21% and —20% decrease in the OGTI' assay. The delayed onset with 0601GD' 4178 is consistent with the view that it binds to plasma proteins from which it is slowly released.
[00564]GD', 0601GD'3794 and 0601GD'4042 (dAla2-GD'(l-42) acid form) produced significant enhancement of the insulinotropic response to an intravenous glucose challenge (IVGTT assay), similar in magnitude to the effects of exenatide and GLP-1 (data not shown). 0601GD'4 178 (octylglycine-GIP(1-30)-Exendin-4-(3 1-39)), showed a diminished insulinotropic response, possibly due to its association with plasma proteins via the octylglycine group, which would however provide a compensating benefit of an even further extended duration of action. GD', 0601GD'3794 and 0601GD'4 178 produced a significant rise in insulin levels prior to the glucose challenge. All three analogs produced a significant lowering of the glucose excursion in response to the glucose challenge (data not shown). As described, fed, isoflurane anesthetized HSD male rats were intubated and cannulated via femoral artery and vein and allowed to stabilize for 1 hour. Following stabilization, an i.v. infusion of saline, GD', or GD' analog hybrid was started, (t=-30). At t=0, an i.v. bolus of 5.7mmol/kg D-glucose was administered over 2 minutes. Samples for glucose measurement and insulin concentrations were taken at various time points before and after (0 to 90 minutes) glucose infusion.
[00565] Additional GD' hybrids that will be active in the lVG'IT assay include:
0601 G1P4252 [dAla2JGD'-(1 -30)-tOctylglycine 34] Exendin-4 (31-39);
0601G1P4285 [dAla2, Leu14, Alal8, glu2l] GD'-(l-30)-Exendin-4 (3 1-39);
060 lGD'4233 [dAla2] GD'-(1-30)-fGLP-1 v2- (3 1-37); and
0601GD'4179 [dAla2, Leu14, beta-Ala3l, beta-Ala32] GD'-(1-32)-Exendin-4 (31-39).

Example 7. Peptide Stablility and Protease Resistance.
[00566] Res istance to proteases was determined according to the protocol of Hupe-Sodmann et al. (Peptides 18(5):625-632 (1997)) using the cell line RINmF5. This well-differentiated cell line is generally accepted as a pancreatic beta-cell model. The indicated GD' compounds (30p.M) were incubated with RNmSF cell membrane extracts at 370C, and degradation of peptides, in proportion to percentage of the parent peptide was monitored at different intervals for 6 hr using LC-MS. The data (see Figure 11) indicate that the [DAla2]-GD'(1-30)-PSSGAPPPS (SEQ ID NO: 1) GD' hybrid (Compound G (also designated 0601GD'3794); with C-terminally Trp-cage shield (e.g., exendin-4 tail)) is more stable than its parent molecule GD'(1-30) without the shield or the full length GD'(1-42). Accordingly the addition of a Trp-cage shield provides a surprisingly superior protease resistance compared to the absence of the cage or the presence of an alternate non-cage-forming sequence, such as the native GD' C-terminal sequence. Further the data indicates that a GD'(1-30) truncation can accommodate foreign sequences at its C-terminus and, in particular, can accommodate a Trp-cage shield sequence.
[00567] Analogs were also tested for stability in human plasma. Test compounds were dissolved water (or in very dilute DMSO as needed), added to human plasma, incubated for 370C for time-points (0, 1, 2, 3, 4, and 5 hrs), extracted with with methanol (1:4 ratio), and the supernatant analyze by LC/MS/MS. In the human plasma stability assays, "stable" was scored as greater than about 90% remaining at 5 hours of treatment to human plasma, "intermediately stable" was scored as greater than about 75% remaining at 2 hours and less than about 90% remaining at 5 hours, and "unstable" was scored as less than about 75% remaining at 2 hours. Unstable analogs and hybrids would generally be unsuited in the methods presented herein. Exemplary compounds were tested. Compounds scored as stable were 060 IGD'3794, 060 IGD' 4150 and 0601G1P4233. Compounds scored as intermediately stable were 0601GD'4 149, 0601GD'4 152, 0601GD'4153, 0601GD'4176, 0601GD'4177, 0601GD'4215, 0601GD'4284, and 0601GD'4289. Compounds scored as unstable were 0601GD'1540, 0601GD'4147, 06OlGil'4292, 0601GD'4293, and 060 lGD'4294. GD' Compound 0601GD'4178 which contains an octylglycine, was scored as unstable, however it is DPP-IV resistant, and binds to plasma proteins complicating its extraction and detection; it is nonetheless believed to be a stable compound for the purposes of the methods herein. The GD' analog hybrid 060 1GD'3794 was surprisingly more stable than exendin-4.
Example 8. Further Exemplarv GD'/amylin-sCT Hybrids.
[005681 Further exemplary GD' phybrids, amide forms, were prepared. (As used herein, lower case single amino acid code indicates a "D" amino acid. For example, YaE indicates a D-Alanine in position 2.) -


(Table Removed)
[00570] The hybrids effectively and selectively bound and activated the relevant receptors.
[00571] The compounds were also assayed in for food intake inhibition, lowering of blood glucose assay
and Oral Glucose Tolerance Test (OGTT) as described herein. In the food intake assays (Figures 1 3A, -14,
15 and 1 6A- 1 6B) points represent mean +1- sd of n—4 cages (3 mice/cage). Peptide was injected D' at t=0.
Food was introduced immediately after injection and amount consumed measured at t=30, 60, 120, and
180 minutes. ~p [00572] In Figure 1 3B bars represent mean +/- sd. Peptide was injected D' at t = -5 minutes into 4-hour fasted NIHISwiss female mice. Gavage (1.5g/kg) was given at t = 0. Sample was taken at t=30 minutesJ
p0.05 versus vehicle control; ANOVA, Dunnett's test. In Figure 17 points represent mean +/- sem.
Peptide was injected il' at t=) immediatley following baseline sample into 2-hour fasted NIH/Swiss mice. Samples were taken at t=60, t=120 and t=180. Blood glucose was measured with a ONETOUCH ULTRA (LifeScan, Inc., Milpitas CA). *p
Example 9. Direct Action of GD' Compounds on Cardiomyocytes.
[00573] Cyclic AMP (adenosine 3',5'-cyclic monophosphate) is a key second messenger in the G-Protein Coupled Receptor (GPCR) signaling pathway. The binding of a ligand to the receptor leads to G protein activation which in turns regulates Adenylyl Cyclase, the enzyme responsible for modulating intracellular levels of cAMP. Measurement of cAIvIP levels is widely used as an indicator of receptor function. Activation of cardiomyocyte Gs- and Gi-coupled receptors was determined as a measure of cAIMP production.
[00574] Cell Isolation. Neonatal cardiomyocytes were isolated using a commercially available system. The Cellutron Isolation kit (nc-603 1) (Cellutron, NJ) and protocol was used for cell isolation. Sprague Dawley neonatal rats (1-2 days old) were decapitated and the beating heart dissected out into digestion buffer (Cellutron Dl buffer) to dissociate cells. For 15 rat hearts, 8 digestions with 6mls of buffer (Cellutron D2 buffer) were sufficient. The resuspended cells incubated in buffer (Cellutron D3 buffer) for sufficient time to allow optimal dispersion and minimize the need for pipetting to break up clumps. Cells released from the first 2 digestions were discarded since a significant portion of these is red blood cells. Resuspended cells were filtered and allowed to pre-plate for 1 hour on plastic in an incubator in NS media (Cellutron, NJ) in order to remove fibroblasts. After 1 hour of pre-plating, floating cells are removed and the flask rinsed with NS media. The floating cells and the cells in the rinse media were combined and counted. Residual red blood cells, which are smaller that the cardiac myocytes, were not counted.
[00575] Cell Plating. At Day 0 of an experiment, the cells were isolated and plated in NS media at 8,000/384 well, 50,000/96 well or 500,000/12 well for 24 hours. At Day 1, the NS media was replaced with fresh NS media. It was observed that the recommended NW (Cellutron, NJ) media did not typic~lly support adequate cell growth or morphology.

[00576] cAMP Assay. GD' compounds were tested for cAMP induction on the isolated, cultured cardiomyocytes. Test-compound induced cAMP levels were determined using a commercially available CisBio (Bedford MA) HRTF® (homogeneous time-resolved fluorescence) cAMP dynamic assay in a Gs/Gi 384-well adherent format, essentially as described by the manufacture (see also Gabriel et al., High throughput screening technologies for direct cyclic AMP measurement, Assay & Drug Dev. Technol. 2:29 1-303 (2003) and Cenni et al., HTRF® cyclic AMP assay: new optimized cell-based assay for better investigation of Gi coupled receptors, in Smart assays for screening, Ilk Congress, Zurich (CH) (2003)). Dose response was determined.
[00577] For Gs assays the positive control was buM forskolin and negative control was buffer. For Gi assays the positive control was buffer and the negative control was lOuM forskolin. Test compound was

incubated with cells for 30 minutes at 37 degrees C in stimulation buffer ((200mls contains 198.3m1 IX HBSS, lml IM Hepes, 670u1 30% BSA titrated to pH 7.4 with iM NaOH). To measure cAMP produced, cells were lysed and cAMP assayed according to the manufacturers protocol. Exemplary results are shown in Figures 18A (GD': human GD'(1-42) acid form) and 18B (GD' hybrid Compound G). The Yaxis is a ratio of time resolved fluorescence measurements taken at 665nM divided by measurements taken at 620nM. In this experiment the ECSO was 11.7 nM for the GD' hybrid and 9.1 nM for GD'. GLPI and exendin-4 displayed very low to no significant activity (data not shown). Further, from three independent experiments of cardiomyocyte Gs-coupled receptor activation, the ECSO (nM) was 15.6 for GD', 0.7 for urocortin and 29.8 for the GD' hybrid Compound G. Urocortin is a known cardiac myocyte Gs-coupled receptor activator used as a positive control. GD' and the GD' hybrid typically gave receptor activation responses greater than 50%. Usdin et al. (Endocrinology 133:2861-2870 (1993)) reported cloning of a rat GD' receptor and its in situ hybridization to RNA in various rat tissues, including the heart, particularly the cardiac endothelium, which was consistent with labeling of the endothelium of major blood vessels. The results indicate that GD' compounds can provide a direct action on the heart. -
Example 10. Measurement of Circulatory System Effects in Conscious Rats by Telemetry After Administration of GD' Compounds.
[00578] Male Harlan Sprague Dawley rats housed at 22.8 ± 0.80C in a 12:12 hour light:dark cycle were used to study the effects of test compounds on the circulatory system through the use of telemetry. The experiments were performed during the light cycle. Telemetry allows for real-time hemodynamic readings including arterial blood pressure, heart rate and arterial dP/dt, via an implanted radio transmitter in conscious, non-anesthetized, unrestrained rats. In the present Example, rats were injected with either vehicle, 80 nmollkg GD' (human GD'(1-42) acid form), or 80 nmollkg of 0601GD'3794 by remote intravenous dosing. Remote intravenous dosing was achieved through in-dwelling vascular access ports (Access Technologies (Skokie, IL). The port is secured to the underlying muscle just below the skin between the scapulae. The catheter resides in the jugular vein. Data were collected for up to 60 minutes following injection.


[00579] As shown in Figures 1 9A-E the effect of the GD' hybrid to increase heart rate was transient relative to GD' (human GD'(1-42) acid form) (Figure 19B), that that the GD' hybrid decreased mean arterial pressure while GD' had no effect (Figure 19A), and that both compounds increased dP/dt similarly (Figure 19C). Figures 19D and 19E demonstrate that the GD' hybrid provides a sustained lowering of systolic and diastolic pressures compared to either vehicle or GD'.
[005801 From the data it can be seen that GD' displays a positive inotropic effect in intact rats without elevating arterial pressure. GD' also displayed a chronotropic (heart rate elevating) effect. The GD' hybrid displayed a prolonged inotropic effect (shown here as peak rate of increase in arterial pressure; dP/dt),

compared to GD', without causing cardiac acceleration, and invoked a decrease in arterial pressure. Accordingly, GD' hybrids can display vasodilation and perfusion benefits that are not associated with an increase in cardiac work. In addition the sustained lowering of either systolic pressure or diastolic pressure, including both, is a recognized beneficial cardiovascular effect linked to reducing hypertension and cardiovascular-associated morbidity and mortality events.
Example 11. Lack of Effect of GD' on Weight Loss.
[00581]The effect of GIP(l -42) and a GD' DPP-IV resistant analog/exendin tail hybrid (060 lGD'3794) were tested for effect on food intake and on weight loss in DIO mice as described herein. As shown in Figure 20A, it was observed that GD' did not acutely inhibit food intake in mice. In comparison, rat amylin at one tenth the dose caused a significant decrease in food intake within 30 mm, relative to control. A trend for GD' to decrease food intake at later time points (60, 120 mm) may have been an indirect effect due to its stimulation of amylin secretion. Points represent mean ± sd of n=4 cages (3 mice/cage). Peptide was injected D' at t=0. Food was introduced immediately after injection and amount consumed measured at t=30, 60, and 120 mm. * p [00582] As shown in Figure 21, Gil' did not cause weight loss in diet-induced-obese mice. In mice in which body weight had been increased by feed a high-fat diet (HF Saline group), GD' infused via mm-osmotic pump for 2 weeks had no effect on body weight change. In contrast, exenatide infused at a 30-fold lower rate reduced body weight change to that observed in mice fed a low fat diet (LF group). * P0.05; ANOVA vs HF Saline. This demonstrates an anti- or non-catabolic effect of GD'. In contrast, it was observed herein that A GD' hybrid having an appropriate choice of a second hormone module, such as an amylinlsCT/amylin chimera, did cause a reduction in food intake and weight loss.
[00583] While the present invention has been described in terms of exemplary examples and embodiments, it is understood that variations and modifications will occur to those skilled in the art.

Therefore, it is intended that the appended claims cover all such equivalent variations which comewithinthe scope of the invention as claimed.




What is claimed is:

1. A GIP hybrid polypeptide exhibiting at least one hormonal activity, said hybrid polypeptide comprising a first bio-active peptide hormone module covalently linked to at least one additional bio-active peptide hormone module; wherein:


the bio-active peptide hormone modules are independently selected from the group consisting of:
component peptide hormones, fragments of component peptide hormones that exhibit at least one hormonal activity of the component peptide hormones, analogs and derivatives of component pepfide hormones that exhibit at least one hormonal activity of the component peptide hormones, fragments of analogs and derivatives of component peptide hormones that exhibit at least one hormonal activity of the component peptide hormones, and peptidic enhancers;
the component peptide hormone of the first bio-active peptide hormone module is a Gil'
the component peptide homone of the at least one additional bio-active peptide bromone modules are independently selected from the group consisting of: amylin, adrenomedullin (ADM), calcitonin (CT), calcitonin gene related peptide (CGRP), intermedin, cholecystokinin ("CCK"), leptin, peptide YY (PYY), glucagon-like peptide- 1 (GLP- 1), glucagon-like peptide 2 (GLP-2), oxyntomodulin (OXM), a natriuretic peptide, a urocortin family peptide, a neuromedin family peptide, exendin-3, and exendin-4;
the peptidic enhancers are independently selected from the group consisting of: structural motifs of component peptide hormones that impart a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma protein binding, or other pharmacokinetic characteristic to the hybrid polypeptide, and structural motifs of analogs or derivatives of component peptide hormones that impart a desired chemical stability, conformational stability, metabolic stability, bioavailability, organ/tissue targeting, receptor interaction, protease inhibition, plasma proteinbinding, or other pharmacokinetic characteristic to the hybrid polypeptide; and
at least one of the bio-active peptide hormone modules exhibits at least one hormonal activity of a component peptide hormone.


2. The hybrid polypeptide of claim 1, wherein the peptidic enhancers are independently selected from the group consisting of: amylin(32-37), amylin(33-37), amylin(34-37), amylin(35-37), amylin(36-37), amylin(37), ADM(47-52), ADM(48-52), ADM(49-52), ADM(50-52), ADM(51-52), ADM(52), CT(27-32), CT(27-32), CT(28-32), CT(29-32), CT(30-32), CT(31-32), CT(32), CGRP(32-37), CGRP(33-37), CGRP(34-3 7), CGRP(35-37), CGRP(36-37), CGRP(37), intermedin (42-47), intermedin (43-47), intermedin (44-47), intermedin (45-47), intermedin (46-47), intermedin (47), PYY(25-36),

PYY(26-36), PYY(27-36), PYY(28-36), PYY(29-36), PYY(30-36), PYY(3 1-36), PYY(32-36), PYY(25-

35), PYY(26-35), PYY(27-35), PYY(28-35), PYY(29-35), PYY(30-35), PYY(31-35), PYY(32-35), frog

GLP-1(29-37), frog GLP-1(30-37), frog GLP-2(24-31), exendin-4(31-39), exendin-4(32-39), exendin4(33-39), exendin-4(34-39), exendin-4(35-39), exendin-4(36-39), exendin-4(37-39), exendin-4(38-39),

exendin-4(39), and analogs thereof.
3. The hybrid polypeptide of claim 1, wherein at least one of the first bio-active peptide hormone module or the at least one additional bio-active peptide hormone module is a component peptide hormone or fragment of a component peptide hormone that exhibits at least one hormonal activity of the component peptide hormone.
4. The hybrid polypeptide of claim 1, wherein at least one of the first bio-active peptide hormone module or the at least one additional bio-active peptide hormone module is an analog or derivative of a component peptide hormone that exhibits at least one hormonal activity or a fragment of an analog or derivative of a component peptide hormone that exhibits at least one hormonal activity of the component peptide hormone.

5. The hybrid polypeptide of claim 1, wherein at least one of the first bio-active peptide hormone module or the at least one additional bio-active peptide hormone module is peptidic enhancer.

6. The hybrid polypeptide of claim 1, wherein the component peptide hormones of the at least one additional bio-active peptide hormone modules are independently selected from the group consisting of: amylin, calcitonin, CCK, PYY, and exendin-4.
7. The hybrid polypeptide of claim 1, wherein the at least one bio-active peptide hormone module that exhibits at least one hormonal activity is located at the N-terminal portion of the hybrid polypeptide.

8. The hybrid polypeptide of claim 7, wherein the at least one bio-active peptide hormone module that exhibits at least one hormonal activity located at the N-terminal portion of the hybrid polypeptide is configured in the C-terminal to N-terminal orientation.
9. The hybrid polypeptide of claim 8, wherein the N-terminal end of the hybrid polypeptide is amidated.
10. The hybrid polypeptide of claim 1, wherein the at least one bio-active peptide hormone module that exhibits at least one hormonal activity is located at the C-terminal portion of the hybrid polypeptide.
11. The hybrid polypeptide of claim 10, wherein the C-terminal end of the hybrid polypeptide is amidated or acylated
12. The hybrid polypeptide of claim 1, wherein the C-terminal end of one bio-active peptide hormone module is directly attached to the N-terminal end of another bio-active peptide hormone module to form the covalent attachment.
13. The hybrid polypeptide of claim 1, wherein the bio-active peptide hormone modules are covalently attached using one or more linking groups independently selected from the group consisting of:
alkyls; dicarboxylic acids PEGs; amino acids; polyaminoacids; bifunctional linkers; aminocaproyl (Aca), Gly, beta-alanyl, 8-amino-3,6-dioxaoctanoyl, and Gly-Lys-Arg (GKR).
14. The hybrid polypeptide of claim 1, wherein the at least one additional bio-active peptide hormone module is independently selected from the group consisting of: GLP1, a fragment of GLP 1 that exhibits at least one hormonal activity, a GLPI analog or derivative that exhibits at least one hormonal activity, or a fragment of a GLP1 analog that exhibits at least one hormonal activity, exendin-4, a fragment of exendin-4 that exhibits at least one hormonal activity, an exendin-4 analog or derivative that exhibits at least one hormonal activity, or a fragment of an exendin-4 analog that exhibits at least one hormonal activity, amylin, a fragment of amylin that exhibits at least one hormonal activity, an amylin analog or derivative that exhibits at least one hormonal activity, or a fragment of an amylin analog that exhibits at least one hormonal activity, CCK, a fragment of CCK that exhibits at least one hormonal activity, a CCK analog or derivative that exhibits at least one hormonal activity, a fragment of a CCK analog that exhibits at least one hormonal activity, CT, a fragment of CT that exhibits at least one hormonal activity, a CT analog or derivative that exhibits at least one hormonal activity, a fragment of a CT analog that exhibits at least one hormonal activity, a peptidic enhancer, and combinations thereof.


15. The GIP hybrid of claim I having a GIP hybrid sequence selected from Figure 12.
16. The GIP hybrid of claim 1 having a sequence Y(DAla)EGTFISDYSIAMDKLHQQDFVNWLLAQKpsSGApppS (SEQ ID NO: 1).



17. A GIP hybrid of claim 16 that retains the D-Ala at position 2 and a heterologous C-terminal peptidic enhancer and has at least 65% identity to sequence Y(DAla)EGTFISDYSIAMDKJHQQDFVNWLLAQKPSSGApppS (SEQ ID NO: I).



18. A Gil' hybrid of claim 1 wherein the hybrid comprises a polypeptide comprising the formula D-L-C-S, wherein, D comprises a dipeptidyl peptidase IV resistant Gil' N-terminal region, L comprises a linker, C comprises a Gil' C-terminal region, and S comprises a shield region; and wherein L is optionally present and at least one of D or C are present, and wherein when C is present then C-S comprises a Trp-cage motif, or when C is absent then L-S further comprises a Trp-cage or exendin tail motif, and the polypeptide has Gil' receptor binding and/or activating activity.
19. A method to treat or prevent diabetes, prediabetes or hyperglycemia in a patient in need thereof comprising, administering to the patient a therapeutically effective amount of a Gil' hybrid of claim 1.
20. The method of claim 19 wherein the patient is obese.
21. A method to treat or prevent cardiovascular disease or condition, in a patient in need thereof comprising, administering to the patient a therapeutically effective amount of a Gil' hybrid 9f claim 1.


22. The method of claim 21, wherein the patient is non-diabetic.

23. The method of claim 21 wherein the patient is need of and benefits by an increase in cardiac contractility, a decrease blood pressure, acute vasodilatation, a decrease in systolic pressure, a decrease in diastolic pressure, plasma glucose lowering, insulin secretion, a slowing of gastric emptying, beta cell proliferation, weight loss, weight maintenance, a reduction in catabolic effects or any combination thereof.


24. The method of claim 21 wherein the Gil' hybrid provides to the patient an increase in cardiac contractility, a decrease blood pressure, acute vasodilatation, a decrease in systolic pressure, a decrease in diastolic pressure, plasma glucose lowering, insulin secretion, a slowing of gastric emptying, a reduction in catabolic effects or any combination thereof.
25. The method of claim 21 wherein the Gil' hybrid acts directly on cardiac cells.
26. The method of claim 21 wherein the cardiovascular disease or condition is hypertension (including stage 1, stage 2 and stage 3 hypertension, diastolic or systolic), pulmonary hypertension, congestive heart failure, cardiac insufficiency, reduced stroke volume, cardiomyopathy (dilated, hypertrophic or restrictive), decreased cardiac contractility, pulmonary congestion associated with cardiovascular conditions, pulmonary and systemic edema, decreased cardiac output, abnormal left ventricular function, diastolic blood pressure abnormalities, renal failure associated with decreased cardiac contractility, increased cardiovascular risk, myocardial infarction, and non-ischemic or ischemic heart tissue degeneration.

27. The method of claim 26 wherein the increased cardiovascular risk is associated with elevated systolic pressure accompanied by normal diastolic pressure, associated with elevated diastolic pressure accompanied by normal systolic pressure, associated with elevated diastolic and systolic pressure, associated with elevated mean arterial blood pressure or any combination thereof.
28. The method of claim 21 wherein the Gil' hybrid provides a benefit of reducing elevated systolic pressure accompanied by normal diastolic pressure, of reducing the elevated diastolic pressure accompanied by normal systolic pressure, by reducing elevated diastolic and systolic pressure, by reducing elevated mean arterial blood pressure or any combination thereof.

29. The method of claim 21 wherein the treating or preventing is selected from preventing the initiation of, delaying the initiation of, preventing the progression or advancement of, slowing the progression or advancement of, delaying the progression or advancement of, and reversing the progression of the disease or condition from an advanced to a less advanced stage.
30. The method of claim 21 wherein the Gil' hybrid comprises an amylin family hormone module.

31. The method of claim 21 wherein the Gil' hybrid comprises an amylin family hormone module that comprises an amylin/sCT/amylin chimera.
32. The method of claim 21 wherein the Gil' hybrid comprises a DPP-IV resistant Gil' analog.
33. The method of claim 21 wherein the Gil' hybrid comprises a DPP-IV resistant Gil' analog and an amylin family hormone module that comprises an amylin/sCT/amylin chimera, optionally joined with linker.
34. A method to treat a patient undergoing critical care, comprising administering to the patient in need thereof a therapeutically effective amount of a Gil' hybrid of claim 1.
35. The method of claim 34, wherein the patient is non-diabetic.
36. The method of claim 34 wherein the critical care is for a disease or condition of catabolic change associated with a critical illness, sepsis, post-traumatic, post-surgical, post-shock, comatose patients, stress-induced hyperglycemia, stroke, myocardial infarction, acute mesenteric ischemia, respiratory distress, ventilator dependency, renal failure, congestive heart failure, edema, hibernating myocardium, cardiomyopathies, lowering of BNP, ejection dysfunction, hypertension, polyneuropathy, ischemia/reperfusion injury, histoprotection of organ beds, myocardial infarction, acute coronary syndrome, disturbances of conduction or rhythm, papillary dysfunction, and/or pulmonary edema.
37. The method of claim 34 wherein the patient is undergoing surgery.
38. The method of claim 34 wherein the Gil' provides a reduction in APACIJIE score, a reduction in mortality, a reduction in days in hospital, a reduction in need for readmission, a reduction in hospitalization costs or any combination thereof.

39. The method of claim 34 wherein the critically ill patient in need of treatment is non-diabetic, diabetic, prediabetic, and/or obese.
40. The method of claim 34 wherein the Gil' hybrid comprises an amylin family hormone module.
41. The method of claim 34 wherein the Gil' hybrid comprises an amylin family hormone module that comprises an amylin/sCT/amylin chimera.
42. The method of claim 34 wherein the GIP hybrid comprises a DPP-IV resistant Gil' analog.
The method of claim 34 wherein the Gil' hybrid comprises a DPP-1V resistant Gil' analog and an amylin family hormone module that comprises an amylin/sCT/amylin chimera, optionally joined with linker.
44. The method of claim 19 wherein the Gil' hybrid comprises an amylin family hormone module.
45. The method of claim 19 wherein the GIP hybrid comprises an amylin family hormone module that comprises an amylin/sCT/amylin chimera.

The method of claim 19 wherein the Gil' hybrid comprises a DPP-IV resistant Gil' analog.

47. The method of claim 19 wherein the Gil' hybrid comprises a DPP-IV resistant Gil' analog and an amylin family hormone module that comprises an amylin/sCT/amylin chimera, optionally joined with linker.



Documents:

6776-delnp-2007-abstract.pdf

6776-delnp-2007-Assignment-(10-04-2013).pdf

6776-delnp-2007-Claims-(10-01-2013).pdf

6776-delnp-2007-Claims-(17-12-2013).pdf

6776-delnp-2007-claims.pdf

6776-delnp-2007-Correspondence Others-(10-01-2013).pdf

6776-delnp-2007-Correspondence Others-(10-04-2013).pdf

6776-delnp-2007-Correspondence Others-(11-07-2012).pdf

6776-delnp-2007-Correspondence Others-(17-12-2013).pdf

6776-delnp-2007-Correspondence Others-(28-01-2013).pdf

6776-delnp-2007-Correspondence-Others-(11-10-2012).pdf

6776-DELNP-2007-Correspondence-Others-(22-02-2011).pdf

6776-DELNP-2007-Correspondence-Others-(28-07-2009).pdf

6776-delnp-2007-correspondence-others.pdf

6776-delnp-2007-description (complete).pdf

6776-delnp-2007-Drawings-(10-01-2013).pdf

6776-delnp-2007-drawings.pdf

6776-delnp-2007-Form-1-(10-04-2013).pdf

6776-delnp-2007-form-1.pdf

6776-delnp-2007-Form-2-(10-01-2013).pdf

6776-delnp-2007-Form-2-(10-04-2013).pdf

6776-delnp-2007-Form-2-(28-01-2013).pdf

6776-delnp-2007-form-2.pdf

6776-delnp-2007-Form-3-(11-07-2012).pdf

6776-DELNP-2007-Form-3-(22-02-2011).pdf

6776-DELNP-2007-Form-3-(28-07-2009).pdf

6776-delnp-2007-form-3.pdf

6776-delnp-2007-form-5.pdf

6776-delnp-2007-GPA-(10-04-2013).pdf

6776-delnp-2007-pct-210.pdf

6776-delnp-2007-pct-237.pdf

6776-delnp-2007-pct-304.pdf

6776-delnp-2007-pct-306.pdf

6776-delnp-2007-pct-373.pdf

6776-delnp-2007-Petition-137-(11-10-2012).pdf


Patent Number 258367
Indian Patent Application Number 6776/DELNP/2007
PG Journal Number 01/2014
Publication Date 03-Jan-2014
Grant Date 02-Jan-2014
Date of Filing 31-Aug-2007
Name of Patentee AMYLIN PHARMACEUTICALS LLC.
Applicant Address 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
Inventors:
# Inventor's Name Inventor's Address
1 LEVY ODILE ESTHER C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
2 HANLEY MICHAEL R C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
3 JODKA CAROLYN M C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
4 LEWIS DIANA Y C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
5 SOARES CHRISTOPHER J C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
6 GHOSH SOUMITRA S C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
7 D'SOUZA LAWRENCE J C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
8 PARKES DAVID G C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
9 MACK CHRISTINE M C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
10 SRIVASTAVA VED C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
11 LEVY ODILE ESTHER C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
12 HANLEY MICHAEL R C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
13 JODKA CAROLYN M C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
14 LEWIS DIANA Y C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
15 SOARES CHRISTOPHER J C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
16 GHOSH SOUMITRA S C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
17 D'SOUZA LAWRENCE J C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
18 PARKES DAVID G C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
19 MACK CHRISTINE M C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
20 SRIVASTAVA VED C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
21 GHOSH SOUMITRA S C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
22 D'SOUZA LAWRENCE J C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
23 PARKES DAVID G C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
24 MACK CHRISTINE M C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
25 SRIVASTAVA VED C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
26 ERICKSON MARY C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
27 JANSSEN SAMUEL C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
28 BARON ALAIN D C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
29 YOUNG ANDREW A C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
30 PITTNER RICHARD A C/O AMYLIN PHARMACEUTICALS INC.,OF 9360 TOWNE CENTRE DRIVE, SAN DIEGO, CALIFORNIA 92121 USA
PCT International Classification Number C07K 14/575
PCT International Application Number PCT/US2006/005020
PCT International Filing date 2006-02-10
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/709,320 2005-08-17 U.S.A.
2 60/652,662 2005-02-11 U.S.A.
3 60/653,433 2005-02-11 U.S.A.
4 60/707,369 2005-08-11 U.S.A.
5 60/651,647 2005-02-11 U.S.A.