US20120165503A1 - Oxyntomodulin analogs - Google Patents

Oxyntomodulin analogs Download PDF

Info

Publication number
US20120165503A1
US20120165503A1 US13/202,524 US200913202524A US2012165503A1 US 20120165503 A1 US20120165503 A1 US 20120165503A1 US 200913202524 A US200913202524 A US 200913202524A US 2012165503 A1 US2012165503 A1 US 2012165503A1
Authority
US
United States
Prior art keywords
seq
peptide
residue
amino
peptide analog
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/202,524
Other languages
English (en)
Inventor
Paul E. Carrington
George Eiermann
Donald Marsh
Joseph Metzger
Alessandro Pocai
Ranabir Sinha Roy
Bianchi Bianchi
Paolo Ingallinella
Antonello Pessi
Alessia Santoprete
Elena Capito
Richard DiMarchi
Brian Ward
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MSD Italia SRL
Indiana University Research and Technology Corp
Merck Sharp and Dohme LLC
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US13/202,524 priority Critical patent/US20120165503A1/en
Assigned to INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION reassignment INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DIMARCHI, RICHARD, WARD, BRIAN
Assigned to MERCK SHARP & DOHME CORP. reassignment MERCK SHARP & DOHME CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MARSH, DONALD J., ROY, RANABIR SINHA, CARRINGTON, PAUL E., EIERMANN, GEORGE J., METZGER, JOSEPH J., POCAI, ALESSANDRO
Assigned to ISTITUTO DI RICERCHE DI BIOLOGIA P. ANGELETTI S.R.L. reassignment ISTITUTO DI RICERCHE DI BIOLOGIA P. ANGELETTI S.R.L. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CAPITO, ELENA, BIANCHI, ELISABETTA, INGALLINELLA, PAOLO, PESSI, ANTONELLO, SANTOPRETE, ALESSIA
Publication of US20120165503A1 publication Critical patent/US20120165503A1/en
Assigned to MSD ITALIA S.R.L. reassignment MSD ITALIA S.R.L. MERGER AND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ISTITUTO DI RECERCHE DI BIOLOGIA MOLECOLARE P. ANGELETTI S.R.L., MSD ITALIA S.R.L.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to peptide analogs of oxyntomodulin (OXM, glucagon-37), which have been modified to be resistant to cleavage and inactivation by dipeptidyl peptidase IV (DPP-IV) and to increase in vivo half-life of the peptide analog while enabling the peptide analog to act as a dual GLP-1/glucagon receptor (GCGR) agonist, and the use of such peptide analogs for treatment of metabolic disorders such as diabetes and obesity.
  • OXM oxyntomodulin
  • DPP-IV dipeptidyl peptidase IV
  • GCGR GLP-1/glucagon receptor
  • the hormone oxyntomodulin (OXM, glucagon-37) is a posttranslational product of preproglucagon processing in the intestine and central nervous system (CNS) and is secreted from L-cells in the gut in response to food intake.
  • OXM has been implicated in the regulation of food intake and energy expenditure (Jarrouse et al., Endocrinol. 115: 102-105 (1984); Schjoldager et al., Eur. J. Clin. Invest., 18: 499-503 (1988)).
  • Central or peripheral administration of OXM in rats causes a decrease in short term food intake with minimal effects on gastric emptying (Dakin et al.
  • OXM peripheral administration of OXM dose-dependently inhibited both fast-induced and dark phase food intake, but unlike GLP-1, had no effect on gastric emptying. OXM also reduced levels of fasting ghrelin and increased c-fos immunoreactivity, in the arcuate nucleus (ARC). Repeated seven-day IP administration of OXM caused a reduction in the rate of body weight gain and adiposity in rats (See Dakin et al. Endocrinology, 145: 2687-2695 (2004)).
  • OXM is a weak agonist at the GLP-1 receptor, when used in pharmacological concentrations (See Baggio et al., Gastroenterol. 127: 546-58 (2004)).
  • OXM was also found to ameliorate glucose intolerance in mice fed a high fat diet (Dakin et al., Am. J. Physiol. Endocrinol, Metab. 294: E142-E147 (2008) and increase the intrinsic heart rate in mice independent of the GLP-1 receptor (Sowden et al., Am. J. Physiol. Regal. Integr. Comp. Physiol. 292: R962-R970 (2007). OXM has also been shown to differentially affect GLP-1 receptor beta-arrestin recruitment and signaling through Galpha (Jorgensen et al., J. Pharma. Exp. Therapeut.
  • OXM has a very short half-life and is rapidly inactivated by the cell surface dipeptidyl peptidase IV (DPP-IV) (Zhu et al., J. Biol. Chem. 278: 22418-22423 (2002).
  • DPP-IV inhibitors are weight-neutral in the clinic, suggesting that supraphysiological levels of OXM (900-1000 pM) may be required to achieve weight loss in humans.
  • OXM peptide analogs for inducing weight loss in humans has been the object of Published International Application Nos. WO03/022304, WO2004/062685, and WO2006/134340.
  • OXM therefore shows potential as a treatment for metabolic disorders such as diabetes and obesity.
  • OXM analogs that can be safely and efficaciously administered for the treatment of metabolic diseases, such as diabetes and obesity. It would be further desirable if analogs or derivatives were developed that were modified by conjugation to moieties that would improve stability and pharmacokinetics, more particularly modifications that confer resistance to DPP-IV cleavage. It would further be desirable to provide OXM analogs that are capable of acting as dual GLP-1 receptor/glucagon receptor agonists.
  • the present invention provides oxyntomodulin (OXM, glucagon-37) peptide analogs, which have been modified to be resistant to cleavage and inactivation by dipeptidyl peptidase IV (DPP-IV) and to increase in vivo half-life of the peptide analog while enabling the peptide analog to act as a dual GLP-1/glucagon receptor (GCGR) agonist, and the use of such peptide analogs for treatment of metabolic disorders such as diabetes and obesity.
  • OXM oxyntomodulin
  • DPP-IV dipeptidyl peptidase IV
  • GCGR GLP-1/glucagon receptor
  • the analogs disclosed herein reduce food intake and body weight, increase metabolic rate, mediate glucose-dependent insulin secretion (GDIS) from pancreatic islets and improve glucose tolerance, thereby providing a treatment option for individuals afflicted with a metabolic disorder such as metabolic syndrome, obesity, diabetes, metabolic syndrome X, hyperglycemia, impaired fasting glucose, dyslipidemia, atherosclerosis, and other prediabetic states.
  • GDIS glucose-dependent insulin secretion
  • the present invention provides a peptide comprising the amino acid sequence of a human oxyntomodulin (OXM) shown in SEQ ID NO:1 wherein the second amino acid from the N-terminus is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV the peptide includes a lipid or cholesterol moiety covalently linked to the peptide; the peptide optionally includes one to three amino acid substitutions in addition to the substitution at position 2; and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide is capable of acting as a dual GLP-1 receptor and glucagon receptor agonist and has a serum half-life greater than the serum half-life of the human OXM, and pharmaceutically acceptable salts thereof.
  • the amino acid substituted for the second amino acid from the N-terminus is selected from the group consisting of D-s
  • a peptide comprising the amino acid sequence of a human oxyntomodulin (OXM) shown in SEQ ID NO:1 wherein the second amino acid from the N-terminus is substituted with an amino acid selected from the group consisting of D-serine and ⁇ -aminoisobutyric acid; the C-terminus of the peptide includes a cysteine residue in which the thiol group of the cysteine residue is covalently linked to a cholesterol moiety by a hydrophilic linker; the peptide optionally includes one to three amino acid substitutions in addition to the substitution at position 2; and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide acts as a dual GLP-1 receptor and glucagon receptor agonist and has a serum half-life greater than the serum half-life of the human OXM, and pharmaceutically acceptable salts thereof.
  • OXM human oxyntomodulin
  • a peptide comprising the amino acid sequence of a human oxyntomodulin (OXM) shown in SEQ ID NO:1 wherein the second amino acid from the N-terminus is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV; the peptide includes a lipid or cholesterol moiety covalently linked to the peptide; the peptide includes one or more amino acid substitutions at amino acid positions selected from the group consisting of positions 17, 18, and 27; and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide acts as a dual GLP-1 receptor and glucagon receptor agonist and has a serum half-life greater than the serum half-life of the human OXM, and pharmaceutically acceptable salts thereof.
  • OXM human oxyntomodulin
  • the one or more amino acid substitutions selected from the group consisting of glutamic acid for the arginine at position 17, alanine for the arginine at position 18, norleucine or O-methyl-L-homoserine for the methionine at position 27.
  • a peptide comprising the amino acid sequence of a human oxyntomodulin (OXM) shown in SEQ ID NO:1 wherein the second amino acid from the N-terminus is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV; the peptide includes a lipid or cholesterol moiety covalently linked to the peptide; the peptide optionally includes one to three amino acid substitutions in addition to the substitution at position 2; the peptide lacks the amino acid sequence RNRNNIA (SEQ ID NO:104); and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide acts as a dual GLP-1 receptor and glucagon receptor agonist and has a serum half-life greater than the serum half-life of the human OXM, and pharmaceutically acceptable salts thereof.
  • OXM human oxyntomodulin
  • the amino acid substitution at position 2 is an amino acid selected from the group consisting of D-serine and ⁇ -aminoisobutyric acid and the cholesterol moiety is covalently linked to the thiol group of a cysteine by a hydrophilic linker and the cysteine residue is covalently linked to the C-terminus of the peptide by a peptide bond.
  • the peptide further includes one or more amino acid substitutions at amino acid positions selected from the group consisting of positions 17, 18, and 27.
  • the peptide further includes one or more amino acid substitutions selected from the group consisting of arginine at position 17 to glutamic acid, arginine at position 18 to alanine, methionine at position 27 to norleucine or O-methyl-L-homoserine.
  • the peptide includes a cholesterol moiety covalently linked to the thiol group of a cysteine residue at the C-terminus of the peptide.
  • the cholesterol moiety is covalently linked to the thiol group by a hydrophilic linker.
  • the hydrophilic linker can be a peptide or polymer such as an ethoxy polymer that includes one to ten ethoxy units.
  • the hydrophilic linker is an ethoxy polymer that includes four ethoxy units.
  • the peptide includes a lipid moiety covalently linked to the ⁇ -amino group of a lysine residue covalently linked to the C-terminus of the peptide, which in particular embodiments is a palmitoyl.
  • the lysine residue is linked to the C-terminus of the peptide by means of a linker moiety.
  • the linker moiety includes one or more gamma-glutamic acid residues and in other aspects, the linker moiety is 1-amino-4,7,10-tioxa-13-tridecanamine succinimic acid.
  • OXM peptide analog selected from the peptide analogs presented in Table 1 and Table 2 and the use of one more of the OXM peptide analogs presented in Table 1 and Table 2 in the manufacture of a medicament for the treatment of a metabolic disorder.
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is amino acid sequence RNRNNIA (SEQ ID NO:104) or a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • Z 2 is a cysteine residue, which in particular embodiments, can be covalently linked by its thiol group to a cholesterol moiety or a lipid moiety.
  • the cholesterol moiety or lipid moiety is covalently linked to the thiol group of the cysteine by a hydrophilic linker.
  • the hydrophilic linker is an ethoxy polymer that includes one to twenty-four ethoxy units or in more specific aspects, the hydrophilic linker is an ethoxy polymer that includes four ethoxy units.
  • Z 2 is a lysine residue, which in particular embodiments, the ⁇ -amino group of the lysine is covalently linked to a cholesterol moiety or a lipid moiety, either directly or by means of a hydrophilic linker.
  • the lipid moiety is a palmitoyl.
  • L 1 is a linker moiety.
  • the linker moiety is a hydrophilic linker moiety.
  • the linker moiety comprises one or more gamma-glutamic acid residues and in other aspects, the linker moiety is 1-amino-4,7,10-tioxa-13-tridecanamine succinimic acid.
  • the peptide comprises a structure selected from OXM70 (SEQ ID NO:12); OXM110 (SEQ ID NO:19); OXM115 (SEQ ID NO:21); OXM177 (SEQ ID NO:24); OXM212 (SEQ ID NO:27); OXM213 (SEQ ID NO:28); OXM216 (SEQ ID NO:29); OXM290 (SEQ ID NO:46); OXM301 (SEQ ID NO:51); or OXM325 (SEQ ID NO:65).
  • the peptide comprises a structure selected from OXM237 (SEQ ID NO:31); OXM238 (SEQ ID NO:32); OXM259 (SEQ ID NO:33); OXM260 (SEQ ID NO:34); OXM261 (SEQ ID NO:35); OXM262 (SEQ ID NO:36); OXM263 (SEQ ID NO:37); OXM264 (SEQ ID NO:38); OXM265 (SEQ ID NO:39); OXM266 (SEQ ID NO:40); OXM267 (SEQ ID NO:41); OXM268 (SEQ ID NO:42); OXM306 (SEQ ID NO:43); OXM307 (SEQ ID NO:44); and OXM308 (SEQ ID NO:45).
  • OXM237 SEQ ID NO:31
  • OXM238 SEQ ID NO:32
  • OXM259 SEQ ID NO:33
  • OXM260 SEQ ID NO:
  • the peptide comprises a structure selected from OXM291 (SEQ ID NO:47); OXM292 (SEQ ID NO:48); OXM293 (SEQ ID NO:49); OXM294 (SEQ ID NO:50); OXM302 (SEQ ID NO:52); OXM303 (SEQ ID NO:53); OXM304 (SEQ ID NO:54); OXM305 (SEQ ID NO:55); OXM311 (SEQ ID NO:56); OXM312 (SEQ ID NO:57); OXM314 (SEQ ID NO:58); OXM313 (SEQ ID NO:59); OXM317 (SEQ ID NO:60); OXM318 (SEQ ID NO:61); OXM319 (SEQ ID NO:62); OXM323 (SEQ ID NO:64); OXM327 (SEQ ID NO:
  • the peptide when the peptide lacks RNRNNIA (SEQ ID NO:104), the peptide comprises the structure selected from OXM345 (SEQ ID NO:69); OXM355 (SEQ ID NO:70); OXM357 (SEQ ID NO:71); OXM359 (SEQ ID NO:72); OXM361 (SEQ ID NO:73); OXM373 (SEQ ID NO:74); OXM374 (SEQ ID NO:75); OXM380 (SEQ ID NO:76); OXM381 (SEQ ID NO:77); OXM383 (SEQ ID NO:78); OXM388 (SEQ ID NO:79); OXM392 (SEQ ID NO:80); OXM395 (SEQ ID NO:81); OXM398 (SEQ ID NO:82); OXM399 (SEQ ID NO:83); OXM400 (SEQ ID NO:84); OXM401 (SEQ ID NO:85); OXM404
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • Z 2 is a cysteine residue, which in particular embodiments, can be covalently linked by its thiol group to a cholesterol moiety or a lipid moiety.
  • the cholesterol moiety or lipid moiety is covalently linked to the thiol group of the cysteine by a hydrophilic linker.
  • the hydrophilic linker is an ethoxy polymer that includes one to twenty-four ethoxy units or in more specific aspects, the hydrophilic linker is an ethoxy polymer that includes four ethoxy units.
  • Z 2 is a lysine residue, which in particular embodiments, the ⁇ -amino group of the lysine is covalently linked to a cholesterol moiety or a lipid moiety, either directly or by means of a hydrophilic linker.
  • the lipid moiety is a palmitoyl.
  • L 1 is a linker moiety.
  • the linker moiety is a hydrophilic linker moiety.
  • the linker moiety comprises one or more gamma-glutamic acid residues and in other aspects, the linker moiety is 1-amino-4,7,10-tioxa-13-tridecanamine succinimic acid.
  • the peptide comprises a structure selected from OXM291 (SEQ ID NO:47); OXM292 (SEQ ID NO:48); OXM293 (SEQ ID NO:49); OXM294 (SEQ ID NO:50); OXM302 (SEQ ID NO:52); OXM303 (SEQ ID NO:53); OXM304 (SEQ ID NO:54); OXM305 (SEQ ID NO:55); OXM311 (SEQ ID NO:56); OXM312 (SEQ ID NO:57); OXM314 (SEQ ID NO:58); OXM313 (SEQ ID NO:59); OXM317 (SEQ ID NO:60); OXM318 (SEQ ID NO:61); OXM319 (SEQ ID NO:62); OXM323 (SEQ ID NO:64); OXM327 (SEQ ID NO:66); or OXM329 (SEQ ID NO:67).
  • OXM291 SEQ ID NO:
  • the peptide comprises the structure selected from OXM345 (SEQ ID NO:69); OXM355 (SEQ ID NO:70); OXM357 (SEQ ID NO:71); OXM359 (SEQ ID NO:72); OXM361 (SEQ ID NO:73); OXM373 (SEQ ID NO:74); OXM374 (SEQ ID NO:75); OXM380 (SEQ ID NO:76); OXM381 (SEQ ID NO:77); OXM383 (SEQ ID NO:78); OXM388 (SEQ ID NO:79); OXM392 (SEQ ID NO:80); OXM395 (SEQ ID NO:81); OXM398 (SEQ ID NO:82); OXM399 (SEQ ID NO:83); OXM400 (SEQ ID NO:84); OXM401 (SEQ ID NO:85); OXM404 (SEQ ID NO:86); OXM406 (SEQ ID NO:69); OX
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • Z 2 is a cysteine residue, which in particular embodiments, can be covalently linked by its thiol group to a cholesterol moiety or a lipid moiety.
  • the cholesterol moiety or lipid moiety is covalently linked to the thiol group of the cysteine by a hydrophilic linker.
  • the hydrophilic linker is an ethoxy polymer that includes one to twenty-four ethoxy units or in more specific aspects, the hydrophilic linker is an ethoxy polymer that includes four ethoxy units.
  • Z 2 is a lysine residue, which in particular embodiments, the ⁇ -amino group of the lysine is covalently linked to a cholesterol moiety or a lipid moiety, either directly or by means of a hydrophilic linker.
  • the lipid moiety is a palmitoyl.
  • L 1 is a linker moiety.
  • the linker moiety is a hydrophilic linker moiety.
  • the linker moiety comprises one or more gamma-glutamic acid residues and in other aspects, the linker moiety is 1-amino-4,7,10-tioxa-13-tridecanamine succinimic acid.
  • the peptide comprises a structure selected from OXM290 (SEQ ID NO:46); OXM301 (SEQ ID NO:51); OXM321 (SEQ ID NO:63); OXM325 (SEQ ID NO:65); or OXM330 (SEQ ID NO:68).
  • the peptide comprises a structure selected from OXM70 (SEQ ID NO:12); OXM110 (SEQ ID NO:19); OXM115 (SEQ ID NO:21); OXM177 (SEQ ID NO:24); OXM212 (SEQ ID NO:27); OXM213 (SEQ ID NO:28); or OXM216 (SEQ ID NO:29).
  • the peptide comprises a structure selected from OXM237 (SEQ ID NO:31); OXM238 (SEQ ID NO:32); OXM259 (SEQ ID NO:33); OXM260 (SEQ ID NO:34); OXM261 (SEQ ID NO:35); OXM262 (SEQ ID NO:36); OXM263 (SEQ ID NO:37); OXM264 (SEQ ID NO:38); OXM265 (SEQ ID NO:39); OXM266 (SEQ ID NO:40); OXM267 (SEQ ID NO:41); OXM268 (SEQ ID NO:42); OXM306 (SEQ ID NO:43); OXM307 (SEQ ID NO:44); and OXM308 (SEQ ID NO:45).
  • OXM237 SEQ ID NO:31
  • OXM238 SEQ ID NO:32
  • OXM259 SEQ ID NO:33
  • OXM260 SEQ ID NO:
  • peptide analogs that are a dual GLP-1/glucagon receptor (GCGR) agonist and have a pI of less than 6.0.
  • Peptide analogs comprising a cholesterol or fatty acid moiety and have a pI of less than 6 have been found to have a reduced capacity for stimulating mast cell degranulation as determined by a in vitro counterscreening assay using the human mast cell line LAD2.
  • a peptide analog comprising the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNTK (SEQ ID NO:109) in which the second amino acid from the N-terminus of the peptide is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV;
  • the peptide includes a lipid or cholesterol moiety covalently linked to the peptide by a spacer comprising one or more gamma-glutamic acid residues;
  • the peptide optionally includes one to three amino acid substitutions in addition to the substitution at position 2; and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide analog has a pI of less than 6.0 and is a dual GLP-1 receptor agonist and glucagon receptor agonist, and pharmaceutically acceptable salts thereof.
  • peptide analogs has
  • the amino acid substituted for the second amino acid from the N-terminus of the peptide analog is selected from the group consisting of D-serine, ⁇ -aminoisobutyric acid, and 1-amino-1-cyclobutane carboxylic acid.
  • the peptide analog includes a cholesterol moiety covalently linked to the thiol group of a cysteine residue that is covalently linked to the 8-amino group of the lysine residue at the C-terminus of the peptide analog by a spacer comprising one or more gamma glutamic acid residues.
  • the cholesterol moiety is covalently linked to the thiol group by a hydrophilic linker, which in particular embodiments is an ethoxy polymer that includes one to twelve ethoxy units for example, an ethoxy polymer that includes four ethoxy units.
  • the peptide analog includes a lipid moiety covalently linked to the ⁇ -amino group of a lysine residue: in particular embodiments the lipid moiety is a fatty acid such as a palmitoyl, myristoyl, or stearoyl moiety. In a further embodiment, the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue by one or more gamma-glutamic acid residues. In a further embodiment, the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue at the C-terminus by one or more gamma-glutamic acid residues.
  • the lipid moiety is covalently linked to the 8-amino group of the lysine residue by one or more gamma-glutamic acid residues and the lysine residue linked to the lysine residue at the C-terminus by one or more gamma-glutamic acid residues.
  • the peptide analog further includes one or more amino acid substitutions at amino acid positions selected from the group consisting of positions 10, 12, 16, 17, 18, and 27.
  • the peptide analog includes one or more amino acid substitutions selected from the group consisting of lysine for the tyrosine at position 10, serine for the lysine at position 12, glutamic acid or ⁇ -aminoisobutyric acid for the serine at position 16, glutamic acid for the arginine at position 17, alanine for the arginine at position 18, lysine for the glutamine at position 20, and norleucine or O-methyl-L-homoserine for the methionine at position 27.
  • the tyrosine at position 10 in the peptide analog is replaced with a lysine and the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue by one or more gamma-glutamic acid residues.
  • the glutamine at position 20 in the peptide analog is replaced with lysine and the lipid moiety is covalently linked to the ⁇ -amino group of the lysine by one or more gamma-glutamic acid residues.
  • the peptide analog further includes one or more gamma-glutamic acid residues covalently linked to the C-terminus.
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe) residue; X 2 is a tyrosine,
  • peptide analogs have a pI between 4 and 6, or a pI of about 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, or 5.9, or a pI of about 5.4 to 5.5.
  • M is a cysteine residue covalently linked to a cholesterol moiety with a hydrophilic linker and the cysteine residue is linked to the C-terminus of P.
  • the hydrophilic linker is an ethoxy polymer that includes one to twenty-four ethoxy units, which in particular aspects can include for example, four ethoxy units.
  • M is a lysine residue covalently linked to a lipid moiety by a spacer comprising one or more gamma glutamic acid residues and the lysine residue is linked to the C-terminus of P or M is a lysine residue covalently linked to a lipid moiety by a spacer comprising one or more gamma glutamic acid residues and the lysine residue is at position X 2 or X 7 of P.
  • the lipid moiety is a palmitoyl, myristoyl, or stearoyl moiety.
  • the peptide analog is OXM317 (SEQ ID NO:60); OXM318 (SEQ ID NO:61); OXM319 (SEQ ID NO:62); OXM323 (SEQ ID NO:64); OXM327 (SEQ ID NO:66); or OXM329 (SEQ ID NO:67).
  • the peptide analog is OXM345 (SEQ ID NO:69); OXM355 (SEQ ID NO:70); OXM357 (SEQ ID NO:71); OXM359 (SEQ ID NO:72); OXM361 (SEQ ID NO:73); OXM373 (SEQ ID NO:74); OXM374 (SEQ ID NO:75); OXM380 (SEQ ID NO:76); OXM381 (SEQ ID NO:77); OXM383 (SEQ ID NO:78); OXM388 (SEQ ID NO:79); OXM392 (SEQ ID NO:80); OXM395 (SEQ ID NO:81); OXM398 (SEQ ID NO:82); OXM399 (SEQ ID NO:83); OXM400 (SEQ ID NO:84); OXM401 (SEQ ID NO:85); OXM404 (SEQ ID NO:86); OXM406 (SEQ ID NO:87); OXM345
  • a GLP-1 agonist is a peptide, small molecule, or chemical compound that binds to the GLP-1 receptor and stimulates the same biological activity as does GLP-1.
  • an agonist for the GLP-1 receptor binds to the receptor with at least 1% of the affinity as native GLP-1.
  • an agonist for the GLP-1 receptor binds to the receptor with an equal or greater affinity than native GLP-1.
  • a glucagon agonist is a peptide, small molecule, or chemical compound that binds to the glucagon receptor and stimulates the same biological activity as does glucagon.
  • an agonist for the glucagon receptor binds to the receptor with at least 1% of the affinity as native glucagon.
  • an agonist for the glucagon receptor binds to the receptor with an equal or greater affinity than native glucagon.
  • a “dual GLP1/glucagon receptor agonist” is “GLP1 receptor/glucagon receptor co-agonist molecule”, which exhibit activity at both the glucagon receptor and the GLP1 receptor.
  • a dual GLP1 receptor agonist and glucagon receptor agonist exhibits activity at the glucagon receptor of at least about 10% relative to native glucagon and also exhibits activity at the GLP-1 receptor of at least about 10% relative to native GLP-1.
  • FIG. 1 shows the results of an ex vivo assay comparing glycogenolysis of native OXM to OXM (Q3E),
  • the Figure shows that native OXM induces glycogenolysis in a dose dependent fashion and induces full glycogenolysis at 1.5 nM and has an approximate EC 50 of 0.5 nM whereas OXM-Q3E induced only about 58% at 300 nM, consistent with its poor GCGR agonist potency.
  • FIG. 2 shows the results of an in vivo GCG Receptor occupancy experiment showing that glucagon (GCG) at 1.5 mpk gave 84% GCGR occupancy and 3 mpk OXM gave 31% GCGR occupancy but that OXM-Q3E gave 0% GCGR occupancy.
  • GCG glucagon
  • FIGS. 3A and 3B show the blood glucose levels and glucose infusion rates, respectively, in response to OXM and OXM-Q3E.
  • FIG. 4 shows the results of an intraperitoneal glucose tolerance test (IPGTT) in lean mice for subcutaneous (s.c.) administration of OXM and OXM-Q3E.
  • IPGTT intraperitoneal glucose tolerance test
  • FIG. 5A shows ex vivo measurement of glycogenolysis in perfused liver in the presence of OXM70, OXM110, OXM177, OXM115, and OXM216.
  • FIG. 58 shows GGCR occupancy following subcutaneous (s.c.) or intravenous (i.v.) administration of the OXM70, OXM110, OXM177, OXM115, and OXM216 compared to native OXM in a competition assay in wild-type mice.
  • FIG. 6 summarizes the acute in vivo efficacy of OXM70 on reducing food intake and body weight in established DIO mice.
  • Food intake was measured about two hours and 18 hours later.
  • FIG. 7 summarizes the acute in vivo efficacy of OXM110 and OXM177 on reducing food intake and body weight in established DIO mice.
  • Food intake was measured about two hours and 18 hours later.
  • FIG. 8 summarizes the acute in vivo efficacy of OXM216, OXM115, and OXM177 on reducing food intake and body weight in established DIO mice.
  • Food intake was measured about two hours and 18 hours later.
  • FIGS. 9A-D shows the pharmacological end points of a chronic body weight and food intake study in DIO mice for OXM peptide analogs OXM110, OXM177, and OXM115.
  • FIG. 9A shows the cumulative change in food intake.
  • FIG. 9B shows the cumulative change in body weight.
  • FIG. 9C shows basal glucose levels over several days of the study.
  • FIG. 9D shows IPGTT assay on day 13 of the study.
  • FIG. 10 shows the results of an intraperitoneal glucose tolerance test (IPGTT) in lean mice for subcutaneous administration of OXM70.
  • IPGTT intraperitoneal glucose tolerance test
  • FIG. 11 shows the results of an intraperitoneal glucose tolerance test (IPGTT) in lean mice for subcutaneous administration of OXM110.
  • IPGTT intraperitoneal glucose tolerance test
  • FIG. 12 shows the results of an intraperitoneal glucose tolerance test (IPGTT) in lean mice for subcutaneous administration of OXM177.
  • IPGTT intraperitoneal glucose tolerance test
  • FIG. 13 shows the results of an intraperitoneal glucose tolerance test (IPGTT) in lean mice for subcutaneous administration of OXM115.
  • IPGTT intraperitoneal glucose tolerance test
  • FIG. 14A shows the metabolic rate effects of OXM115 in DIO Mice over time.
  • FIG. 14B shows the percent change in metabolic rate caused by OXM115 in the DIO Mice.
  • FIG. 15 summarizes the acute in vivo efficacy of several +/+ peptides on reducing food intake and body weight in established DIO mice.
  • FIG. 16 shows single dose of several +/+ peptides on food intake on reducing food intake and body weight in established DIO mice.
  • OXM-based therapy has the potential to favorably impact both obesity and, the as yet to be carefully characterized, concomitant effects on improving diabetes.
  • Weight loss efficacy and reduction in food intake upon peripheral administration of OXM has been well validated in humans (Wynne et al., Diabetes 54: 2390-2395 (2005)). More recently, OXM has been shown to increase metabolic rate and specifically activity-related energy expenditure in obese subjects (Wynne et al., Int. J. Obes. 30: 1729-1736 (2006), advance online publication, Apr. 18, 2006; doi:10.1038/sj.ijo.0803344). OXM has also been shown to reduce body weight in humans (See, for example, Published International Application Nos.
  • OXM (and GLP-1) has a very short half-life and is rapidly inactivated by the cell surface dipeptidyl peptidase IV (DPP-IV). Mutations can be incorporated into the OXM peptide to render the peptide resistant to DPP-IV cleavage; however, many of these mutations have also been found to inactivate the native peptide or adversely affect the ability of the peptide to interact with the glucagon receptor (GCGR) or GLP-1 receptor (GLP-1R) (See Published International Application No. WO2007/100535, which is incorporated herein in its entirety). OXM (and GLP-1) is also rapidly cleared by the kidneys.
  • DPP-IV cell surface dipeptidyl peptidase IV
  • the OXM peptide analogs disclosed herein solve these problems.
  • the limited in vivo stability of native OXM and GLP-1 due to cleavage by DPP-IV and rapid renal clearance, which necessitates frequent dosing (t.i.d. s.c. injections for OXM, continuous infusion for GLP-1) at high doses in humans has been solved by mutating the site of DPP-IV cleavage (the penultimate residue at the N-terminus) and lipidating the OXM peptide to increase in vivo half-life (t 1/2 ).
  • the t 1/2 of the lipidated (such as acylated or conjugated with a cholesterol moiety) OXM peptide analogs in humans will be suitable for at least once daily administration.
  • the OXM peptide analogs disclosed herein are more convenient for therapeutic purposes than native OXM, which requires t.i.d. dosing.
  • the tolerability profile of polypeptides of the current invention is anticipated to be similar to that of native OXM in humans, which may be superior to that of conventional GLP-1 mimetics like exenatide and liraglutide.
  • the chronic efficacy of the OXM analogs disclosed herein may therefore be better than conventional GLP-1 mimetics, such as Byetta® (Amylin Pharmaceuticals), due to minimal nausea and vomiting, which have typically been dose limiting for the GLP-1 mimetics. Unlike the latter, no dose-titrations to mitigate nausea may be required for polypeptides of this invention.
  • OXM is potent agonist of the glucagon receptor
  • the expectation would be that chronic administration would lead to impaired glucose control (hyperglycemia).
  • the OXM analogs disclosed herein which incorporate a balanced GLP-1 receptor and GCGR co-agonism, gives enhanced reductions in food intake and body weight with chronic administration and results in improved glucose tolerance.
  • OXM peptide analogs that have dual GLP-1 receptor (GLP-1R) and glucagon receptor (GCGR) agonist activity are indicated herein as (+/+).
  • OXM peptide analogs that are GLP-1 agonists only are indicated herein as (+/0).
  • OXM analogs disclosed herein can be used for is the treatment of obesity and/or diabetes. Secondary indications are metabolic syndrome, hyperglycemia, impaired fasting glucose, and other prediabetic states. Further indications include all indications for GLP-1 such as irritable bowel syndrome and other absorptive diseases of the gut, ischemia, stroke, and neurological disorders including anxiety, impaired cognition, and Alzheimer's disease.
  • OXM analogs disclosed herein are anticipated to exhibit at least comparable if not superior efficacy and a better safety profile than current anti-obesity agents such as orlistat (Xenical® (Roche), a lipase inhibitor) and sibutramine (Meridia® (Abbott Laboratories), a seratonin/norepinephrine re-uptake inhibitor), for which GI intolerance (diarrhea, flatulence) and hypertension are common side effects, respectively.
  • orlistat Xenical® (Roche)
  • sibutramine Meridia® (Abbott Laboratories)
  • GI intolerance diarrhea, flatulence
  • the OXM peptide analogs optionally includes a protecting group covalently joined to the N-terminal amino group.
  • a protecting group covalently joined to the N-terminal amino group of the peptide reduces the reactivity of the amino terminus under in vivo conditions.
  • Amino protecting groups include —C 1-10 alkyl, —C 1-10 substituted alkyl, —C 2-10 alkenyl, —C 2-10 substituted alkenyl, aryl, —C 1-6 alkyl aryl, —C(O)—(CH 2 ) 1-6 —COOH, —C(O)—C 1-6 alkyl, —C(O)-aryl, —C(O)—O—C 1-6 alkyl, or —C(O)—O-aryl.
  • the amino terminus protecting group is selected from the group consisting of acetyl, propyl, succinyl, benzyl, benzyloxycarbonyl, and t-butyloxycarbonyl.
  • Deamination of the N-terminal amino acid is another modification that is contemplated for reducing the reactivity of the amino terminus under in vivo conditions.
  • the OXM peptide analogs may be modified to have a protecting group covalently joined to the C-terminal carboxy group, which reduces the reactivity of the carboxy terminus under in vivo conditions.
  • carboxylic acid groups of the peptide may be provided in the form of a salt of a pharmacologically-acceptable cation or esterified to form a C 1-6 ester, or converted to an amide of formula NRR 2 wherein R and R 2 are each independently H or C 1-6 alkyl, or combined to form a heterocyclic ring, such as a 5- or 6-membered ring.
  • the carboxy terminus protecting group is preferably attached to the ⁇ -carbonyl group of the last amino acid.
  • Carboxy terminus protecting groups include, but are not limited to, amide, methylamide, and ethylamide.
  • Amino groups of the peptide, whether N-terminal or side chain, may be in the form of a pharmacologically-acceptable acid addition salt, such as the HCl, HBr, acetic, benzoic, toluene sulfonic, maleic, tartaric, and other organic salts, or may be modified to C 1-6 alkyl or dialkyl amino or further converted to an amide.
  • the OXM peptide analogs that are capable of acting as dual GLP-1 and glucagon agonists comprise the amino acid sequence of a human oxyntomodulin (OXM) shown in SEQ ID NO:1 wherein the second amino acid from the N-terminus is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV; a lipid or cholesterol moiety covalently linked to the OXM peptide analog; optionally one to three amino acid substitutions in addition to the substitution at position 2; and optionally has a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide.
  • OXM human oxyntomodulin
  • the peptide analogs further lack the amino acid sequence RNRNNIA (SEQ ID NO:104).
  • OXM peptide analogs having the above structure will act as a dual GLP-1 receptor and glucagon receptor agonist and have a serum half-life greater than the serum half-life of native human OXM.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine or ⁇ -aminoisobutyric acid (aib) residue
  • X 2 is an arginine (R) or glutamic acid (E) residue
  • X 3 is an arginine (R) or alanine (A) residue
  • X 4 is a methionine (M), norleucine (Nle), or O-methyl-L-homoserine (o) residue
  • M methionine
  • Nle norleucine
  • o O-methyl-L-homoserine
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is amino acid sequence RNRNNIA (SEQ ID NO:104) or a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine or ⁇ -aminoisobutyric acid (aib) residue
  • X 2 is an arginine (R) or glutamic acid (E) residue
  • X 3 is an arginine (R) or alanine (A) residue
  • X 4 is a methionine (M), norleucine (Nle), or O-methyl-L-homoserine (o) residue
  • M methionine
  • Nle norleucine
  • o O-methyl-L-homoserine
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid (Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is an arginine (R)
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is amino acid sequence RNRNNIA (SEQ ID NO:104) or a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is amino acid sequence RNRNNIA (SEQ ID NO:104) or a linker moiety;
  • Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and
  • P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Ser (S) or Lys (K
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the OXM peptide analog can be represented by the structure
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe); X 2 is Tyr or Lys; X 3 is
  • Z 1 is an optionally present protecting group that, if present, is joined to the N-terminal amino group, L 1 is optional but when present is amino acid sequence RNRNNIA (SEQ ID NO:104) or a linker moiety; Z 2 is optional but when present is a lysine (K) residue, a lysine residue covalently linked to a lipid moiety, a lysine residue covalently linked to a cholesterol moiety, a cysteine (C) residue, a cysteine residue covalently linked to a lipid moiety; a cysteine residue covalently linked to a cholesterol moiety; and P or Z 2 optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group; and pharmaceutically acceptable salts thereof.
  • L 1 -Z 2 can be linked to the C-terminal amino acid of P or to an internal amino acid of P.
  • the lipid or cholesterol moiety can be directly or indirectly linked to the side chain of an amino acid.
  • the lipid or cholesterol moiety is linked to the amino acid side-chain by means of a linker moiety.
  • the lipid or cholesterol is directly linked to the side chain of an amino acid which is then linked to the OXM peptide analog by a linker moiety (L 1 ).
  • residues 16 and 20 are Glu and Lys, respectively, the side chains of said residues can participate in a lactam bridge between said Glu and Lys residues.
  • the linker moiety is a hydrophilic linker.
  • the chemical structure of the linker moiety is not critical, however, since it serves primarily as a spacer. However, in certain embodiments, the linker moiety may itself provide improved properties to the OXM peptide analogs.
  • the linker moiety is covalently linked to the C-terminal amino acid of the peptide (P) and Z 2 is covalently linked to the linker moiety.
  • the linker moiety is covalently linked to an internal amino acid of P and Z 2 is covalently linked to the linker moiety.
  • X 9 can include one or more gamma glutamic acid residues.
  • the OXM peptide analogs include diastereomers as well as their racemic and resolved enantiomerically pure forms.
  • the amino acids are in the L-form with particular amino acids in D-form.
  • Table 1 shows the structures for a representative number of OXM peptide analogs based on the full-length OXM molecule that have activity at both the glucagon receptor and the GLP-1 receptor (+/+).
  • OXM peptide analogs OXM29, 208, 209 and 229 are peptide analogs precursors (thiolated peptides) that were used for the conjugation reaction with the bromo-cholesterol moieties.
  • OXM peptide analogs OXM36 and OXM115 were prepared from OXM229, which includes the native human OXM peptide but with a D-serine at amino acid position 2 and a cysteine residue in peptide linkage at the C-terminus and in which the C-terminal carboxy group has been amidated.
  • OXM36 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNRNNIAC 4 —CONH 2 (SEQ ID NO:8) wherein C 4 is a cysteine residue in which cholest-5-en-3-yl ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ acetate is covalently linked thereto as shown below.
  • OXM115 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNRNNIAC 7 —CONH 2 (SEQ ID NO:21) wherein the C 7 is a cholest-5-en-3-yl 1- ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ -2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oate (or Cys(Oxa4-cholesterol)) as shown below.
  • C 7 differs from C 4 in that it has a hydrophilic spacer formed by four ethylene glycol repeats (ethoxy units) between the cholesterol group and the cysteine moiety.
  • the C7 group increases solubility of the resulting peptide-cholesterol conjugate.
  • the peptide-C 7 conjugate shows a less prominent serum shift than peptide-C 4 conjugates. This was found to be important for maintaining an optimal balanced activity on the two receptors GLP-1R and Glucagon receptor. The results shown in the Examples illustrate this aspect.
  • OXM peptide analogs OXM70 and OXM216 were prepared from OXM29, which includes the native human OXM peptide but with ⁇ -aminoisobutyric acid at amino acid position 2 and with a cysteine residue in peptide linkage at the C-terminus and in which the C-terminal carboxy group has been amidated.
  • OXM70 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTKRNRNNIAC 4 —CONH 2 (SEQ ID NO:12) wherein C 4 is cholest-5-en-3-yl ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ acetate as shown for OXM36.
  • OXM216 has the structure H ⁇ QGTETSDYSKYLDSRRAQDFVQWLMNTKRNRNNIAC 7 —CONH 2 (SEQ ID NO:29) wherein the C 7 is a cholest-5-en-3-yl 1- ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ -2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oate (or Cys(Oxa4-cholesterol)) as for OXM115.
  • OXM peptide analog OXM212 was prepared from OXM208, which includes the native human OXM peptide but with ⁇ -aminoisobutyric acid at amino acid position 2, norleucine in place of the methionine residue at position 27, with a cysteine residue in peptide linkage at the C-terminus and in which the C-terminal carboxy group has been amidated.
  • OXM212 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLnNTKRNRNNIAC 4 —CONH 2 (SEQ ID NO:26) wherein C 4 is cholest-5-en-3-yl ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ acetate as shown for OXM36.
  • OXM peptide analog OXM213 was prepared from OXM209, which includes the native human OXM peptide but with ⁇ -aminoisobutyric acid at amino acid position 2, O-methyl-L-homoserine in place of the methionine residue at position 27, with a cysteine residue in peptide linkage at the C-terminus and in which the C-terminal carboxy group has been amidated.
  • OXM213 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLYNTKRNRNNIAC 4 —CONH 2 (SEQ ID NO:28) wherein C 4 is cholest-5-en-3-yl ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ acetate as shown for OXM36.
  • OXM237 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLmNTKRNRNNIAC 7 —CONH 2 (SEQ ID NO:31). It was designed to contain D-Ser (s) at position 2, Met(O) at position 27, and a Cys(Oxa 4 -cholesterol) (C 7 ) at position site 38.
  • OXM238 was designed to contain D-Ser (s) at position 2, Met(O) at position 27, and a Cys(acetamide) (C 5 ) at position site 38.
  • This peptide has the same peptide sequence of OXM237 and is a control peptide with no cholesterol but with the cysteine thiol blocked with acetamide.
  • OXM259-OXM268 and OXM306-308 (as shown in Table 1) have the amino acid sequence of OXM but wherein the amino acid at position 2 has been substituted with a non-natural amino acid to identify amino acid substitutions that confer resistance to DPP4 catalytic activity.
  • OXM peptide analogs that lack the amino acid sequence RNRNNIA can also display full agonistic activity on the GLP-1 and the Glucagon receptors.
  • These OXM peptide analogs along with some additional peptides based upon the full-sized OXM peptide are shown in Table 2.
  • These truncated OXM peptide analogs are variants of the Glucagon K (GcgK) genus of peptides that were disclosed in commonly owned International Published Application No. WO2007100535.
  • the GcgK peptides are peptides where the OXM sequence has been truncated at the C terminus to eliminate the seven amino acid residues at the C-terminus.
  • the resulting peptide is that one of glucagon with an extra lysine residue at the C-terminus.
  • This GcgK peptide is compatible with a +/+ pattern in which the peptides show subnanomolar potency on both the GLP-1R and the GCGR.
  • native OXM shows nanomolar potency on the two receptors.
  • An additional advantage of the GcgK peptide is that the two basic arginine residues at the C-terminus have been eliminated. This was important in the construction of lipidated (acylated or cholesterylated) peptide analogs having dual GLP-1/GCGR agonist activity.
  • lipidated peptide analogs with a pI above 7 based on the OXM sequence stimulate mast cell degranulation while lipidated peptide analogs with a pI of about 5 appear not to stimulate mast degranulation (See Example 11).
  • the pI for lipidated based peptide analogs should be around 5. Therefore, in particular aspects the lipidated peptide analogs have a pI less than 6.0. In further aspects, the lipidated peptide analogs have a pI between 4.5 and 6.0. In a further aspect, the lipidated peptide analogs have a pI of about 5.4.
  • Reduction of pI was done using one or more of the following three different strategies: 1) substitution of residues in the peptide, 2) introduction of a carboxylate group at the C terminus, and 3) introduction of negative residues such as one or more gamma-carboxy-glutamic acid residues as spacers.
  • the acyl or cholesterol group is covalently linked to the peptide by means of a spacer that comprises two gamma-carboxy-glutamic acid residues.
  • these peptide analogs include those peptide analogs shown in Table 2.
  • peptides with a pI of about 5.4 include but are not limited to OXM345, OXM380, OXM381, OXM392, OXM395, OXM398, OXM399, OXM400, and OXM401.
  • the acyl or cholesterol group is covalently linked to an internal amino acid in the peptide analogs by means of a spacer that comprises one or two gamma-carboxy-glutamic acid residues and the peptide further includes one or more gamma-carboxy-glutamic acid residue covalently to the lysine residue at the C-terminus.
  • these peptide analogs include but are not limited to OXM407, OXM408, OXM414, and OXM418.
  • the acyl or cholesterol group is covalently linked to the C-terminus of the peptide by means of a spacer that comprises one gamma-carboxy-glutamic acid residue and the peptide further includes one or more amino acid substitutions to reduce the pI of the peptide.
  • OXM374 is an example of such a peptide analog.
  • a peptide analog comprising the amino acid sequence HSQGTFTSDYSKYLDSRRAQDFVQWLMNTK (SEQ ID NO:109) in which the second amino acid from the N-terminus of the peptide is substituted with an amino acid that renders the peptide resistant to cleavage and inactivation by dipeptidyl peptidase IV;
  • the peptide includes a lipid or cholesterol moiety covalently linked to the peptide by a spacer comprising one or more gamma-glutamic acid residues;
  • the peptide optionally includes one to three amino acid substitutions in addition to the substitution at position 2; and the peptide optionally includes a protecting group that, if present, is joined to the C-terminal carboxy group of the peptide; wherein the peptide analog has a pI of less than 6.0 and is a dual GLP-1 receptor agonist and glucagon receptor agonist, and pharmaceutically acceptable salts thereof.
  • peptide analogs has
  • the amino acid substituted for the second amino acid from the N-terminus of the peptide analog is selected from the group consisting of D-serine, ⁇ -aminoisobutyric acid, and 1-amino-1-cyclobutane carboxylic acid.
  • the peptide analog includes a cholesterol moiety covalently linked to the thiol group of a cysteine residue that is covalently linked to the ⁇ -amino group of the lysine residue at the C-terminus of the peptide analog by a spacer comprising one or more gamma glutamic acid residues.
  • the cholesterol moiety is covalently linked to the thiol group by a hydrophilic linker, which in particular embodiments is an ethoxy polymer that includes one to twelve ethoxy units for example, an ethoxy polymer that includes four ethoxy units.
  • the peptide analog includes a lipid moiety covalently linked to the ⁇ -amino group of a lysine residue: in particular embodiments the lipid moiety is a fatty acid such as a palmitoyl, myristoyl, or stearoyl moiety. In a further embodiment, the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue by one or more gamma-glutamic acid residues. In a further embodiment, the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue at the C-terminus by one or more gamma-glutamic acid residues.
  • the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue by one or more gamma-glutamic acid residues and the lysine residue linked to the lysine residue at the C-terminus by one or more gamma-glutamic acid residues.
  • the peptide analog further includes one or more amino acid substitutions at amino acid positions selected from the group consisting of positions 10, 12, 16, 17, 18, and 27.
  • the peptide analog includes one or more amino acid substitutions selected from the group consisting of lysine for the tyrosine at position 10, serine for the lysine at position 12, glutamic acid or ⁇ -aminoisobutyric acid for the serine at position 16, glutamic acid for the arginine at position 17, alanine for the arginine at position 18, lysine for the glutamine at position 20, and norleucine or O-methyl-L-homoserine for the methionine at position 27.
  • the tyrosine at position 10 in the peptide analog is replaced with a lysine and the lipid moiety is covalently linked to the ⁇ -amino group of the lysine residue by one or more gamma-glutamic acid residues.
  • the glutamine at position 20 in the peptide analog is replaced with lysine and the lipid moiety is covalently linked to the ⁇ -amino group of the lysine by one or more gamma-glutamic acid residues.
  • the peptide analog further includes one or more gamma-glutamic acid residues covalently linked to the C-terminus.
  • P is a peptide having the amino acid sequence
  • X 1 is a D-serine, ⁇ -aminoisobutyric acid (aib), 1-Amino-1-cyclobutane carboxylic acid (Acb) residue, 1-Amino-1-cyclohexane carboxylic acid (Acx); alpha-aminobutyric acid (Abu); D-alpha-aminobutyric acid (D-Abu); Aminovaleric acid Nva); beta-cyclopropyl-alanine (Cpa); propargylglycine (Prg); Allylglycine (Alg); 2-Amino-2-cyclohexyl-propanoic acid (2-Cha); D-tertbutylglycine (D-tbg); Vinylglycine (Vg); 1-Amino-1-cyclopropane carboxylic acid (Acp); or 1-Amino-1-cyclopentane carboxylic acid (Acpe) residue; X 2 is a tyrosine,
  • peptide analogs have a pI between 4 and 6, or a pI of about 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, or 5.9, or a pI of about 5.4 to 5.5.
  • M is a cysteine residue covalently linked to a cholesterol moiety with a hydrophilic linker and the cysteine residue is linked to the C-terminus of P.
  • the hydrophilic linker is an ethoxy polymer that includes one to twenty-four ethoxy units, which in particular aspects can include for example, four ethoxy units.
  • M is a lysine residue covalently linked to a lipid moiety by a spacer comprising one or more gamma glutamic acid residues and the lysine residue is linked to the C-terminus of P or M is a lysine residue covalently linked to a lipid moiety by a spacer comprising one or more gamma glutamic acid residues and the lysine residue is at position X 2 or X 7 of P.
  • the lipid moiety is a palmitoyl, myristoyl, or stearoyl moiety. Examples of these peptide analogs are shown in Table 2.
  • OXM Peptide Analogs SEQ ID NO. Name Structure 46 OXM290 H s QGTFTSDYSKYLDS EA AQDFVQWLMNTKRNRNNIA C -CONH 2 47 OXM291 H s QGTFTSDYSKYLDSRRAQDFVQWLMNTK C -CONH 2 48 OXM291 H s QGTFTSDYSKYLDS EA AQDFVQWLMNTK C -CONH 2 49 OXM293 H s QGTFTSDYSKYLDS E RAQDFVQWLMNTK C -CONH 2 50 OXM294 H s QGTFTSDYSKYLDSR A AQDFVQWLMNTK C -CONH 2 51 OXM301 H s QGTFTSDYSKYLDS EA AQDPVQWLMNTKRNRNNIA C 7 -CONH 2 52 OXM302 H s QGTFTSDYSKYLDSRRAQDF
  • C 9 S- ⁇ 1-[46-(cholest-5-en-3-yloxy)-3,43,46-trioxo-7,10,13,16,19,22,25,28,31,34,37,40-dodecaoxa-4,44-diazahexatetracont-1-yl]-2,5-dioxopyrrolidin-3-yl ⁇ -L-cysteine or Cys (mal-oxa 12 -cholesterol)
  • C 10 S-[42-(cholest-5-en-3-yloxy)-2,42-dioxo-6,9,12,15,18,21,24,27,30,33,36,39-dodecaoxa-3-azadotetracont-1-yl]-L-cysteine or Cys (oxa 12 -cholesterol)
  • C 11 S-[78-(cholest-5-en-3-yloxy)-2,78-dioxo-6,9,12,15
  • OXM290 is the precursor for OXM301 and OXM291 to 294 are precursors for the remainder of the peptide analogs shown in Table 2.
  • OXM301 is an analogue having D-Ser in amino acid position 2 for DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arm residues, and a C(Oxa 4 -cholesterol) group (C 7 ) for improved pharmacokinetic properties in vivo.
  • the substitutions at Arg 17 and Arg 18 were made to increase in vivo stability, since these residues are primary proteolytic cleavage sites.
  • OXM302 is a peptide analog that lacks the amino acid sequence RNRNNIA (SEQ ID NO:96) at the C-terminus (See commonly owned International Published Application No. WO2007/100535, which is incorporated herein in its entirety). Thus, OXM302 is one amino acid longer than the glucagon peptide by one lysine residue at the C-terminus.
  • OXM302 which has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK 7 —CONH 2 (SEQ ID NO:52) there is a D-Ser in amino acid position 2 for DPPIV resistance and a C(Oxa4-cholesterol) group (C 7 ) for improved pharmacokinetic properties in vivo.
  • OXM303 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTKC 7 —CONH 2 (SEQ ID NO:53).
  • the D-Ser in amino acid position 2 provides DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arg 18 residues improves stability and a C(Oxa4-cholesterol) group (C 7 ) improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM304 has the structure HsQGTFTSDYSKYLDSERAQDFVQWLMNTKC 7 —CONH 2 (SEQ ID NO:54).
  • the D-Ser in position 2 provides DPPIV resistance, substitution of Glu for the Arg 17 residue improves stability and a C(Oxa4-cholesterol) group (C 7 ) improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM305 has the structure HsQGTFTSDYSKYLDSERAQDFVQWLMNTKC 7 —CONH 2 (SEQ ID NO:55).
  • the D-Ser in position 2 provides DPPIV resistance, substitution of Ala for the Arg 18 residue improves stability and a C(Oxa4-cholesterol) group (C 7 ) improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM311 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK-K(Palmitoyl)-CONH 2 (SEQ ID NO:56).
  • the D-Ser in amino acid position 2 provides DPPIV resistance and the K(palmitoyl) group improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM312 has the structure HsQGTETSDYSKYLDSEAAQDFVQWLMNTKK(Palmitoyl)-CONH 2 (SEQ ID NO:57).
  • the D-Ser in amino acid position 2 provides DPPIV resistance, substitution of Gln and Ala, respectively, for the Arg 17 and Arg 18 residues improves stability and the K(palmitoyl) group improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM314 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK-Ttds-K(Palmitoyl)-CONH 2 (SEQ ID NO:58).
  • the D-Ser in amino acid position 2 provides DPPIV resistance and the K(palmitoyl) group improves the peptide analog's pharmacokinetic properties in vivo.
  • the Ttds (1-amino-4,7,10-trioxa-13-tridecanamine succinimic acid) is a linker that acts as a flexible and hydrophilic spacer between the peptide sequence and the K(palmitoyl) group.
  • OXM313 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTK-Ttds-K(Palmitoyl)-CONH 2 (SEQ ID NO:59).
  • the D-Ser in amino acid position 2 provides DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arg 18 residues improves stability and the Ttds-K(palmitoyl) group improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM317 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E-C 4 —CONH 2 (SEQ ID NO:60).
  • the peptide there is a D-Ser in amino acid position 2 for DPPIV resistance, and a gamma glutamic residue linked to cholest-5-en-3-yl ⁇ [(2R)-3-amino-2-(amino)-3-oxopropyl]thio ⁇ acetate (C 4 ) for improves pharmacokinetic properties in vivo.
  • the gamma glutamic residue ( ⁇ E) is a linker that acts as a flexible spacer between the peptide sequence and the C 4 group.
  • OXM318 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTK- ⁇ E-C 4 —CONH 2 (SEQ ID NO:61).
  • the D-Ser in amino acid position 2 provides DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arg 18 residues improves stability and the ⁇ E —C 4 group improves the peptide analog's pharmacokinetic properties in vivo.
  • OXM319 has the structure H ⁇ QGTFTSDYSKYLDSEAAQDFVQWLMNTKC 7 —CONH 2 (SEQ ID NO:62).
  • the peptide there is a Aib in amino acid position 2 for DPPIV resistance, and the C(Oxa 4 -cholesterol) group (C 7 ) improves the peptide analogs pharmacokinetic properties in vivo.
  • OXM321 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTKRNRNNIA- ⁇ E-C 4 —CONH 2 (SEQ ID NO:63). It is an analogue having D-Ser in amino acid position 2 for DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arg 18 residues and a C (-cholesterol) group (C 4 ) improves pharmacokinetic properties in vivo. The substitutions at Arg 17 and Arg 18 were made to increase in vivo stability, since these residues are primary proteolytic cleavage sites.
  • the gamma glutamic residue ( ⁇ E) is a linker that acts as a flexible spacer between the peptide sequence and the C 4 group.
  • OXM323 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTKR-C 7 —CONH 2 (SEQ ID NO:64).
  • the D-Ser in position 2 provides DPPIV resistance
  • substitution of Glu and Ala for Arg 17 and Arg 18 residue provides a peptide with improved stability
  • the C(Oxa 4 -cholesterol) group (C 7 ) provides a peptide with improved pharmacokinetic properties in vivo.
  • OXM325 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTKRNRNNIAC 7 —CO 2 H (SEQ ID NO:65). It is an analogue having D-Ser in amino acid position 2 for DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arg 18 residues, and a C(Oxa 4 -cholesterol) group (C 7 ) improves pharmacokinetic properties in viva. The substitutions at Arg 17 and Arg 18 were made to increase in vivo stability, since these residues are primary proteolytic cleavage sites.
  • OXM327 has the structure H ⁇ AQGTFTSDYSKYLDSERAQDFVQWLMNTKC 7 —CONH 2 (SEQ ID NO:66).
  • the Aib in position 2 provides DPPIV resistance
  • substitution of Glu for the Arg 17 residue provides a peptide with improved stability
  • the C(Oxa 4 -cholesterol) group (C 7 ) provides improved pharmacokinetic properties in vivo.
  • OXM329 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E-C 4 —CO 2 H (SEQ ID NO:67).
  • the Aib in position 2 provides DPPIV resistance and a C(Oxa4-cholesterol) group (C 7 ) provides a peptide with improved pharmacokinetic properties in vivo.
  • the gamma glutamic residue ( ⁇ E) is a linker that acts as a flexible spacer between the peptide sequence and the C 4 group.
  • OXM330 has the structure HsQGTFTSDYSKYLDSEAAQDFVQWLMNTKRNRNNIA- ⁇ E-K(Pam)-CONH 2 (SEQ ID NO:68). It is an analogue having D-Ser in amino acid position 2 for DPPIV resistance, substitution of Glu and Ala, respectively, for the Arg 17 and Arm residues and a -K(palmitoyl) group improves the peptide analog's pharmacokinetic properties in vivo.
  • the gamma glutamic residue ( ⁇ E) is a linker that acts as a flexible spacer between the peptide sequence and the K(palmitoyl) group. The substitutions at Arg 17 and Arg 18 were made to increase in vivo stability, since these residues are primary proteolytic cleavage sites.
  • OXM345 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLIVINTK- ⁇ E- ⁇ E-C 4 —COOH (SEQ ID NO:69). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacers between the peptide sequence, and the cholesterol group, one Cys(cholesterol) at position 33 (C 4 ).
  • OXM355 has the structure HsQGTFTSDYSSYLDSRRAQDFVQWLMNTK- ⁇ E-C 4 —COOH (SEQ ID NO:70). It was designed to contain D-Ser (s) at position 2, a Lys 12 Ser substitution, one gamma-glutamic acid at position 31 as spacer between the peptide sequence, and the cholesterol one Cys(cholesterol) at position 32 (C 4 ). A serine in position 12 is present in the GLP-1 peptide sequence. Therefore, it is expected to have at least compatible potency at the GLP-1R. The substitution Lys 12 Ser eliminates a positive charge which results in a pI of about 5.
  • OXM357 has the structure HsQGTFTSDYSKYLDSRAAQDFVQWLMNTK- ⁇ E-C 4 —COOH (SEQ ID NO:71). It was designed to contain D-Ser (s) at position 2, one gamma-glutamic acid at position 31 as spacer between the peptide sequence, and the cholesterol one Cys(cholesterol) (C 4 ) at position 32. OXM357 differs from OXM345 in having only one ⁇ -Glu while OXM345 has two ⁇ -Glu.
  • OXM359 has the structure HsQGTFTSDYSSYLDSRRAQDFVQWLMNTK- ⁇ E-C 7 —COOH (SEQ ID NO:72). It was designed to contain D-Ser (s) at position 2, a Lys 12 Ser substitution, one gamma-glutamic acid at position 31 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) at position 32 (C 7 ). Thus, OXM359 differs from OXM355 in having C 7 (cholesterol group with the tetraethylene glycol spacer) instead of C 4 (cholesterol without an ethylene glycol spacer).
  • OXM361 has the structure HsQGTFTSDYSKYLDSRAAQDFVQWLMNTK- ⁇ E-C 7 —COOH (SEQ ID NO:73). It was designed to contain D-Ser (s) at position 2, an Arg 18 Ala substitution, one gamma-glutamic acid at position 31 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) at position 32 (C 7 in the table). The Arg 18 Ala substitution eliminates a positive charge which is important for the tuning of the optimal pI value to 5.
  • OXM373 has the structure HsQGTFTSDYSKYLDERRAQDFVQWLMNTK- ⁇ E-C 4 —COOH (SEQ ID NO:74). It was designed to contain D-Ser (s) at position 2, a Ser 16 Glu substitution, one gamma-glutamic acid at position 31 as spacers between the peptide sequence, and the cholesterol, one Cys(cholesterol) at position 32 (C 4 ). The Glu at position 16 is present in the exendin-4 peptide so this substitution is expected to be at least compatible with GLP-1R activation.
  • OXM374 has the structure HsQGTFTSDYSKYLDERRAQDFVQWLMNTK- ⁇ E-C 7 —COOH (SEQ ID NO:75). It was designed to contain D-Ser (s) at position 2, a Ser 16 Glu substitution, one gamma-glutamic acid at position 31 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) at position 32 (C 7 ). Thus, the difference between OXM373 and 374 is due to the presence of the tetraethylene glycol spacer attached to the cholesterol group in OXM374.
  • OXM380 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 7 —COOH (SEQ ID NO:76). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) (C 7 ) at position 33.
  • OXM380 has the same peptide sequence of OXM345 but differs in the Oxa 4 spacer linked to the cholesterol group.
  • OXM381 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 9 —COOH (SEQ ID NO:77). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(maleimide-Oxa 12 -cholesterol) (C 9 ) at position 33.
  • OXM381 has the same peptide sequence of OXM345 and OXM380 differing in the maleimide-Oxa 12 spacer linked to the cholesterol. The structure of C 9 is shown below.
  • OXM383 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 7 —COOH (SEQ ID NO:78). It was designed to contain Aib (u) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) (C 7 ) at position 33. OXM383 differs from OXM380 only with respect to the amino acid at position 2.
  • OXM388 has the structure H-Acb-QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E-C ⁇ E-C 7 —COOH (SEQ ID NO:79). It was designed to contain Acb at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 4 -cholesterol) (C 7 ) at position 33. OXM388 differs from OXM380 and 383 only with respect to the amino acid at position 2.
  • OXM392 has the structure HsQGTFTSDYSKYLD E RRA K DFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (lactam bridge between E and K ) (SEQ ID NO:80). It was designed to contain Ser (s) at position 2, Ser 16 Glu and Gln 20 Lys substitutions wherein the Glum and Lys 20 are linked with a lactam bridge on the side chains, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33.
  • the lactam bridge is between position i (16) and i+4 (20) and is meant to stabilize the alpha-helical conformation of the peptide and to stabilize the peptide against proteolytic degradation.
  • the C 10 group differs from the C 9 group for the thioether bond connecting the cysteine thiol group to the Oxa 12 -cholesterol.
  • C 9 there is a maleimide thioether bond while in C 10 there is an acetamide thioether.
  • the structure of C 10 is shown below.
  • OXM395 has the structure H ⁇ QGTFTSDYSKYLD E RRA K DFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (lactam bridge between E and K ) (SEQ ID NO:81). It was designed to contain Aib ( ⁇ ) at position 2, Ser 16 Glu and a Gln 20 Lys substitutions wherein the Glu 16 and Lys 20 are linked with a lactam bridge between the side chains, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33.
  • the lactam bridge is between position 16(i) and 20 (i+4) and is meant to stabilize the peptide's alpha-helical conformation and to stabilize the peptide against proteolytic degradation.
  • OXM398 has the structure H-Acb-QGTFTSDYSKYLD E RRA K DFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (lactam bridge between E and (SEQ ID NO:82). It was designed to contain Acb at position 2, Ser 16 Glu and a Gln 20 Lys substitutions wherein the Glu 16 and Lys 20 are linked with a lactam bridge between the side chains, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33.
  • the lactam bridge is between position 16 (i) and 20 (i+4) and is meant to stabilize the alpha-helical conformation of the peptide and to stabilize the peptide against proteolytic degradation.
  • OXM392, 395 and 398 differ only in the amino acid that is at position 2.
  • OXM399 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (SEQ ID NO:83). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33. OXM399 differs from OXM392 only in that it lacks the lactam bridge present in OXM392.
  • OXM400 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (SEQ ID NO:84). It was designed to contain Aib (a) at position 2, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33. OXM400 differs from OXM395 only in that it lacks the lactam bridge present in OXM395.
  • OXM401 has the structure H-Acb-QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 10 —COOH (SEQ ID NO:85). It was designed to contain Acb at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33. OXM401 differs from OXM398 only in that it lacks the lactam bridge present in OXM398. OXM399, OXM40, and 401 differ from each other in the amino acid at position 2.
  • OXM404 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-K( ⁇ E-palmitoyl)-CONH 2 (SEQ ID NO:86). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the palmitoyl group, one Lys( ⁇ E-palmitoyl) at position 33.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM406 has the structure HsQGTFTSDYSKYLDERRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 10 —CONH 2 (SEQ ID NO:87). It was designed to contain D-Ser (s) at position 2, a Ser 16 Glu substitution, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM407 has the structure HsQGTFTSDYSKYLDSRRAK( ⁇ E-palmitoyl)DFVQWLMNTK- ⁇ E ⁇ E-CONH 2 (SEQ ID NO:88). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the palmitoyl group, one Lys( ⁇ E-palmitoyl) at position 33.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM1408 has the structure HsQGTFTSDYSKYLDSRRAK( ⁇ E- ⁇ E-palmitoyl)DFVQWLMNTK- ⁇ E-CONH 2 (SEQ ID NO:89). It was designed to contain D-Ser (s) at position 2, one gamma-glutamic acid at positions 31, and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 20.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM409 has the structure HsQGTFTSDK( ⁇ E- ⁇ E-palmitoyl)SKYLDSRRADEVQWLMNTK-CONH 2 (SEQ ID NO:89). It was designed to contain D-Ser (s) at position 2 and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM410 has the structure HsQGTFTSDYSKYLDERRAK( ⁇ E- ⁇ E-palmitoyl)DFVQWLMNTK-CONH 2 (SEQ ID NO:91). It was designed to contain D-Ser (s) at position 2, a Ser 16 Glu substitution, and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 20.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM411 has the structure HsQGTFTSDK( ⁇ E- ⁇ E-palmitoyl)SKYLDERRAQDFVQWLMNTK-CONH 2 (SEQ ID NO:92). It was designed to contain D-Ser (s) at position 2, a Ser 16 Glu substitution and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM412 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 11 —COOH (SEQ ID NO:93). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 24 -cholesterol) (C 11 ) at position 33. OXM412 differs from OXM399 for the length of the Oxa spacer linked to the cholesterol group. The structure of C 11 is shown below.
  • OXM413 has the structure H ⁇ DGTFTSDYSKYLDSRRAQDFVK(DOTA)WLmNTK- ⁇ E- ⁇ E-C 10 —CONH 2 (SEQ ID NO:94). It was designed as a peptide for imaging to target GLP-1R in vivo.
  • the sequence contains Aib at position 2, an Asp at position 3 to give selectivity on the GLP-1R, a Lys(DOTA) at position 24, a Met(O) at position 27, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -cholesterol) (C 10 ) at position 33.
  • the peptide in this embodiment is amidated at the C-terminus.
  • the structure of Fmoc-Lys(DOTA)-OH is shown below.
  • OXM414 has the structure HsQGTFTSDK( ⁇ E-palmitoyl)SKYLDERRAQDFVQWLMNTK-E-CONH 2 (SEQ ID NO:95). It was designed to contain D-Ser (s) at position 2, two ⁇ -glutamic acids at positions 31, a Ser 16 Glu substitution and a palmitoyl group linked to the side chain amino group of a lysine [Lys(palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM415 has the structure HsQGTFTSDK(palmitoyl)SKYLDERRAQDFVQWLMNTK- ⁇ E- ⁇ E-CONH 2 (SEQ ID NO:96). It was designed to contain D-Ser (s) at position 2, one ⁇ -glutamic acid at positions 31, a Ser 16 Glu substitution and a palmitoyl group linked to one ⁇ -glutamic acid residue and to the side chain amino group of a lysine [Lys( ⁇ E-palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM416 has the structure H ⁇ QGTFTSDK( ⁇ E- ⁇ E-palmitoyl)SKYLDERRAQDFVQWLMNTK-CONH 2 (SEQ ID NO:97). It was designed to contain Aib (a) at position 2, a Ser 16 Glu substitution and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM417 has the structure H-Acb-QGTFTSDK( ⁇ E- ⁇ E-palmitoyl)SKYLDERRAQDFVQWLMNTK-CONH 2 (SEQ ID NO:98). It was designed to contain Acb at position 2, a Ser 16 Glu substitution and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 10. The peptide in this embodiment is amidated at the C-terminus.
  • OXM418 has the structure HsQGTFTSDK( ⁇ E- ⁇ E-palmitoyl)SKYLD ⁇ RRAQDFVQWLMNTK- ⁇ E-CONH 2 (SEQ ID NO:99). It was designed to contain D-Ser (s) at position 2, one gamma-glutamic acid at positions 31, a Ser 16 Aib (a) substitution and a palmitoyl group linked to two ⁇ -glutamic acid residues and to the side chain amino group of a lysine [Lys( ⁇ E- ⁇ E-palmitoyl)] at position 10.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM419 has the structure H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-K( ⁇ E-palmitoyl)-CONH 2 (SEQ ID NO:100). It was designed to contain Aib (a) at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the palmitoyl group, one Lys( ⁇ E-palmitoyl) at position 33. The peptide in this embodiment is amidated at the C-terminus.
  • OXM420 has the structure H-Acb-QGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-K( ⁇ E-palmitoyl)-CONH 2 (SEQ ID NO:101). It was designed to contain Acb at position 2, two gamma-glutamic acid at positions 31 and 32 as spacer between the peptide sequence, and the palmitoyl group, one Lys( ⁇ E-palmitoyl) at position 33.
  • the peptide in this embodiment is amidated at the C-terminus.
  • OXM421 has the structure HsQGTFTSDYSKYLDSRRAQDFVQWLMNTK- ⁇ E- ⁇ E-C 12 —COOH (SEQ ID NO:102). It was designed to contain D-Ser (s) at position 2, two gamma-glutamic acid residues at positions 31 and 32 as spacer between the peptide sequence, and the cholesterol, one Cys(Oxa 12 -O-cholesterol) (C 12 ) at position 33.
  • the Cys(Oxa 12 -O-cholesterol) differs from Cys(Oxa 12 -cholesterol) in having the cholesterol linked through an ether bond to the Oxa 12 spacer.
  • the ether bond can confer more stability towards the ester bond present in the Cys(Oxa 12 -cholesterol) or C 11 group.
  • the structure of Cys(oxa 12 -O-cholesterol) is shown below.
  • compositions comprising a therapeutically effective amount of one or more of the OXM analogs disclosed herein for the treatment of a metabolic disorder in an individual.
  • Such disorders include, but are not limited to, obesity, metabolic syndrome or syndrome X, type II diabetes, complications of diabetes such as retinopathy, hypertension, dyslipidemias, cardiovascular disease, gallstones, osteoarthritis, and certain forms of cancers.
  • the obesity-related disorders herein are associated with, caused by, or result from obesity.
  • “Obesity” is a condition in which there is an excess of body fat. The operational definition of obesity is based on the Body Mass Index (BMI), calculated as body weight per height in meters squared (kg/m2). “Obesity” refers to a condition whereby an otherwise healthy subject has a Body Mass Index (BMI) greater than or equal to 30 kg/m2, or a condition whereby a subject with at least one co-morbidity has BMI greater than or equal to 27 kg/m2.
  • An “obese subject” is an otherwise healthy subject with a Body Mass Index (BMI) greater than or equal to 30 kg/m2 or a subject with at least one co-morbidity with a BMI greater than or equal to 27 kg/m2.
  • a “subject at risk for obesity” is an otherwise healthy subject with a BMI of 25 kg/m2 to less than 30 kg/m2 or a subject with at least one co-morbidity with a BMI of 25 kg/m2 to less than 27 kg/m2.
  • “obesity” refers to a condition whereby a subject with at least one obesity-induced or obesity-related co-morbidity that requires weight reduction or that would be improved by weight reduction, has a BMI greater than or equal to 25 kg/m2.
  • an “obese subject” refers to a subject with at least one obesity-induced or obesity-related co-morbidity that requires weight reduction or that would be improved by weight reduction, with a BMI greater than or equal to 25 kg/m2.
  • a “subject at risk of obesity” is a subject with a BMI of greater than 23 kg/m2 to less than 25 kg/m2.
  • obesity is meant to encompass all of the above definitions of obesity.
  • Obesity-induced or obesity-related co-morbidities include, but are not limited to, diabetes, non-insulin dependent diabetes mellitus-type 2, impaired glucose tolerance, impaired fasting glucose, insulin resistance syndrome, dyslipidemia, hypertension, hyperuricacidemia, gout, coronary artery disease, myocardial infarction, angina pectoris, sleep apnea syndrome, Pickwickian syndrome, fatty liver; cerebral infarction, cerebral thrombosis, transient ischemic attack, orthopedic disorders, arthritis deformans, lumbodynia, emmeniopathy, and infertility.
  • co-morbidities include: hypertension, hyperlipidemia, dyslipidemia, glucose intolerance, cardiovascular disease, sleep apnea, diabetes mellitus, and other obesity-related conditions.
  • Treatment refers to the administration of the compounds of the present invention to reduce or maintain the body weight of an obese subject.
  • One outcome of treatment may be reducing the body weight of an obese subject relative to that subject's body weight immediately before the administration of the compounds of the present invention.
  • Another outcome of treatment may be preventing body weight regain of body weight previously lost as a result of diet, exercise, or pharmacotherapy.
  • Another outcome of treatment may be decreasing the occurrence of and/or the severity of obesity-related diseases.
  • the treatment may suitably result in a reduction in food or calorie intake by the subject, including a reduction in total food intake, or a reduction of intake of specific components of the diet such as carbohydrates or fats; and/or the inhibition of nutrient absorption; and/or the inhibition of the reduction of metabolic rate; and in weight reduction in patients in need thereof.
  • the treatment may also result in an alteration of metabolic rate, such as an increase in metabolic rate, rather than or in addition to an inhibition of the reduction of metabolic rate; and/or in minimization of the metabolic resistance that normally results from weight loss.
  • Prevention refers to the administration of the compounds of the present invention to reduce or maintain the body weight of a subject at risk of obesity.
  • One outcome of prevention may be reducing the body weight of a subject at risk of obesity relative to that subject's body weight immediately before the administration of the compounds of the present invention.
  • Another outcome of prevention may be preventing body weight regain of body weight previously lost as a result of diet, exercise, or pharmacotherapy.
  • Another outcome of prevention may be preventing obesity from occurring if the treatment is administered prior to the onset of obesity in a subject at risk of obesity.
  • Another outcome of prevention may be decreasing the occurrence and/or severity of obesity-related disorders if the treatment is administered prior to the onset of obesity in a subject at risk of obesity.
  • Such treatment may prevent the occurrence, progression or severity of obesity-related disorders, such as, but not limited to, arteriosclerosis, Type II diabetes, polycystic ovarian disease, cardiovascular diseases, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, and cholelithiasis.
  • the obesity-related disorders herein are associated with, caused by, or result from obesity.
  • obesity-related disorders include overeating and bulimia, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dyslipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovarian disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, GE-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia.
  • obesity-related disorders are metabolic syndrome, also known as syndrome X, insulin resistance syndrome, sexual and reproductive dysfunction, such as infertility, hypogonadism in males and hirsutism in females, gastrointestinal motility disorders, such as obesity-related gastro-esophageal reflux, respiratory disorders, such as obesity-hypoventilation syndrome (Pickwickian syndrome), cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, hyperuricaemia, lower back pain, gallbladder disease, gout, and kidney cancer.
  • the compounds of the present invention are also useful for reducing the risk of secondary outcomes of obesity, such as reducing the risk of left ventricular hypertrophy.
  • diabetes includes both insulin-dependent diabetes mellitus (IDDM, also known as type I diabetes) and non-insulin-dependent diabetes mellitus (NIDDM, also known as Type II diabetes).
  • IDDM insulin-dependent diabetes mellitus
  • NIDDM non-insulin-dependent diabetes mellitus
  • Type I diabetes or insulin-dependent diabetes
  • Type II diabetes is the result of an absolute deficiency of insulin, the hormone which regulates glucose utilization.
  • Type II diabetes, or insulin-independent diabetes i.e., non-insulin-dependent diabetes mellitus
  • Most of the Type II diabetics are also obese.
  • the compounds of the present invention are useful for treating both Type I and Type II diabetes.
  • the compounds are especially effective for treating Type II diabetes.
  • the compounds of the present invention are also useful for treating and/or preventing gestational diabetes mellitus.
  • U.S. Pat. No. 6,852,690 which is incorporated herein in its entirety, discloses methods for enhancing metabolism of nutrients comprising administering to a non-diabetic patient a formulation comprising a nutritively effective amount of one or more nutrients or any combination thereof and one or more insulinotropic peptides.
  • the OXM peptide analogs disclosed herein are insulinotropic and can be administered to patients with a disturbed glucose metabolism such as insulin resistance but no overt diabetes, as well as patients 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.
  • 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 nourished according to the invention without subjecting the patient to hypo- or hyperglycemia.
  • the administration to such a patient aims to provide a therapy to as rapidly as possible deliver the nutritional and caloric requirements to the patient while maintaining his plasma glucose below the so-called renal threshold of about 160 to 180 milligrams per deciliter of glucose in the blood.
  • renal threshold of about 160 to 180 milligrams per deciliter of glucose in the blood.
  • normal patients not having glucose levels just below the renal threshold can also be treated according to the invention as described above, patients with disturbed glucose metabolism such as hyperglycemic patients whose plasma glucose level is just above the renal threshold also find the therapy suitable for their condition.
  • such patients who have a degree of hyperglycemia below the renal threshold at intermittent intervals can receive a combination treatment of nutrients plus insulinotropic peptides according to any of the following regimens.
  • Normal patients not suffering from such hyperglycemia can also be treated using the peptide analogs disclosed herein.
  • compositions may be used in a pharmaceutical composition when combined with a pharmaceutically acceptable carrier.
  • Such compositions comprise a therapeutically-effective amount of one or more of the OXM analogs disclosed herein and a pharmaceutically acceptable carrier.
  • Such a composition may also be comprised of (in addition to the OXM analog disclosed herein and a carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
  • Compositions comprising the OXM analogs disclosed herein can be administered, if desired, in the form of salts provided the salts are pharmaceutically acceptable. Salts may be prepared using standard procedures known to those skilled in the art of synthetic organic chemistry.
  • compositions comprising formula I are also useful for treating or preventing obesity and obesity-related disorders in cats and dogs.
  • mamal includes companion animals such as cats and dogs.
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glutamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such as arg
  • pharmaceutically acceptable salt further includes all acceptable salts such as acetate, lactobionate, benzenesulfonate, laurate, benzoate, malate, bicarbonate, maleate, bisulfate, mandelate, bitartrate, mesylate, borate, methylbromide, bromide, methylnitrate, calcium edetate, methylsulfate, camsylate, mutate, carbonate, napsylate, chloride, nitrate, clavulanate, N-methylglucamine, citrate, ammonium salt, dihydrochloride, oleate, edetate, oxalate, edisylate, pamoate (embonate), estolate, palmitate, esylate, pantothenate, fumarate, phosphate/diphosphate, gluceptate, polygalacturonate, gluconate, salicylate, glutamate, stearate, glycolly
  • the term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s), approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals and, more particularly, in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered and includes, but is not limited to such sterile liquids as water and oils. The characteristics of the carrier will depend on the route of administration.
  • OXM analogs disclosed herein may be in multimers (for example, heterodimers or homodimers) or complexes with itself or other peptides.
  • pharmaceutical compositions of the invention may comprise one or more OXM analogs disclosed herein in such multimeric or complexed form.
  • the term “therapeutically effective amount” means the total amount of each active component of the pharmaceutical composition or method that is sufficient to show a meaningful patient benefit, i.e., treatment, healing, prevention or amelioration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions.
  • a meaningful patient benefit i.e., treatment, healing, prevention or amelioration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions.
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially, or simultaneously.
  • the pharmacological composition can comprise one or more OXM analogs disclosed herein; one or more OXM analogs disclosed herein and one or more other agents for treating a metabolic disorder; or the pharmacological composition comprising the one or more OXM analogs disclosed herein can be used concurrently with a pharmacological composition comprising an agent for treating a metabolic disorder.
  • Such disorders include, but are not limited to, obesity, metabolic syndrome or syndrome X, type II diabetes, complications of diabetes, hypertension, dyslipidemias, cardiovascular disease, gallstones, osteoarthritis, and certain forms of cancers.
  • the agent includes, but are not limited to, cannabinoid (CB1) receptor antagonists, glucagon like peptide 1 (GLP-1) receptor agonists, lipase inhibitors, leptin, tetrahydrolipstatin, 2-4-dinitrophenol, acarbose, sibutramine, phentamine, fat absorption blockers, simvastatin, mevastatin, ezetimibe, atorvastatin, sitagliptin, metformin, orlistat, Qnexa, topiramate, naltrexone, bupriopion, phentermine, losartan, losartan with hydrochlorothiazide, and the like.
  • CBD1 cannabinoid
  • GLP-1 glucagon like peptide 1
  • Suitable agents of use in combination with a compound of the present invention include, but are not limited to:
  • anti-diabetic agents such as (1) PPAR ⁇ agonists such as glitazones (e.g. ciglitazone; darglitazone; englitazone; isaglitazone (MCC-555); pioglitazone (ACTOS); rosiglitazone (AVANDIA); troglitazone; rivoglitazone, BRL49653; CLX-0921; 5-BTZD, GW-0207, LG-100641, R483, and LY-300512, and the like and compounds disclosed in WO97/10813, 97/27857, 97/28115, 97/28137, 97/27847, 03/000685, and 03/027112 and SPPARMS (selective PPAR gamma modulators) such as T131 (Amgen), FK614 (Fujisawa), netoglitazone, and metaglidasen; (2) biguanides such as buformin; metformin
  • WO 99/16758 WO 99/19313, WO 99/20614, WO 99/38850, WO 00/23415, WO 00/23417, WO 00/23445, WO 00/50414, WO 01/00579, WO 01/79150, WO 02/062799, WO 03/033481, WO 03/033450, WO 03/033453; and (14) other insulin sensitizing drugs; (15) VPAC2 receptor agonists; (16) GLK modulators, such as PSN105, RO 281675, RO 274375 and those disclosed in WO 03/015774, WO 03/000262, WO
  • NS-220/R1593 Nippon Shinyaku/Roche
  • ST1929 Sigma Tau
  • MC3001/MC3004 MaxoCore Pharmaceuticals, gemcabene calcium, other fibric acid derivatives, such as Atromid®, Lopid®, and Tricor®, and those disclosed in U.S. Pat. No.
  • FXR receptor modulators such as GW 4064 (GlaxoSmithkline), SR 103912, QRX401, LN-6691 (Lion Bioscience), and those disclosed in WO 02/064125, WO 04/045511, and the like;
  • LXR receptor modulators such as GW 3965 (GlaxoSmithkline), T9013137, and XTCO179628 (X-Ceptor Therapeutics/Sanyo), and those disclosed in WO 03/031408, WO 03/063796, WO 04/072041, and the like
  • lipoprotein synthesis inhibitors such as niacin
  • PPAR ⁇ partial agonists such as those disclosed in WO 03/024395
  • bile acid reabsorption inhibitors such as BARI 1453, SC435, PHA384640, S8921, AZD7706, and the like; and
  • anti-hypertensive agents such as (1) diuretics, such as thiazides, including chlorthalidone, chlorthiazide, dichlorophenamide, hydroflumethiazide, indapamide, and hydrochlorothiazide; loop diuretics, such as bumetanide, ethacrynic acid, furosemide, and torsemide; potassium sparing agents, such as amiloride, and triamterene; and aldosterone antagonists, such as spironolactone, epirenone, and the like; (2) beta-adrenergic blockers such as acebutolol, atenolol, betaxolol, bevantolol, bisoprolol, bopindolol, carteolol, carvedilol, celiprolol, esmolol, indenolol, metaprolol, nadolol, nebivolol
  • anti-obesity agents such as (1) 5HT (serotonin) transporter inhibitors, such as paroxetine, fluoxetine, fenfluramine, fluvoxamine, sertraline, and imipramine, and those disclosed in WO 03/00663, as well as serotonin/noradrenaline re uptake inhibitors such as sibutramine (MERIDIA/REDUCTIL) and dopamine uptake inhibitor/Norepenephrine uptake inhibitors such as radafaxine hydrochloride, 353162 (GlaxoSmithkline), and the like; (2) NE (norepinephrine) transporter inhibitors, such as GW 320659, despiramine, talsupram, and nomifensine; (3) CB1 (cannabinoid-1 receptor) antagonist/inverse agonists, such as rimonabant (ACCOMPLIA Sanofi Synthelabo), SR-147778 (Sanofi Synthelabo), AVE
  • MCH1R melanin-concentrating hormone 1 receptor
  • T-226296 Takeda
  • T71 Takeda/Amgen
  • AMGN-608450 AMGN-503796
  • Amgen 856464
  • A798 Abbott
  • ATC0175/AR224349 Arena Pharmaceuticals
  • GW803430 GaxoSmithkine
  • NBI-1A Neurorocrine Biosciences
  • NGX-1 Neurogen
  • SNP-7941 Synaptic
  • SNAP9847 Synaptic
  • T-226293 Schering Plough
  • TPI-1361-17 Saitama Medical School/University of California Irvine
  • NPY1 neuropeptide Y Y1
  • BMS205749, BIBP3226, J-115814, BIBO 3304, LY-357897, CP-671906, and GI-264879A and those disclosed in U.S. Pat. No.
  • NPY5 neuropeptide Y Y5
  • E-6999 Esteve
  • GW-587081X GW-548118X
  • FR 235,208 FR226928, FR 240662, FR252384
  • 1229U91 GI-264879A
  • CGP71683A C-75
  • LY366377 LY366377, PD-160170, SR-120562A, SR-120819A, S2367 (Shionogi), JCF-104, and H409/22; and those compounds
  • WO 97/19682 WO 97/20820, WO 97/20821, WO 97/20822, WO 97/20823, WO 98/27063, WO 00/107409, WO 00/185714, WO 00/185730, WO 00/64880, WO 00/68197, WO 00/69849, WO 01/09120, WO 01/14376, WO 01/85714, WO 01/85730, WO 01/07409, WO 01/02379, WO 01/02379, WO 01/23388, WO 01/23389, WO 01/44201, WO 01/62737, WO 01/62738, WO 01/09120, WO 02/20488, WO 02/22592, WO 02/48152, WO 02/49648, WO 02/051806, WO 02/094789, WO 03/009845, WO 03/014083, WO 03/0228
  • leptin such as recombinant human leptin (PEG-OB, Hoffman La Roche) and recombinant methionyl human leptin (Amgen);
  • leptin derivatives such as those disclosed in U.S. Pat. Nos.
  • opioid antagonists such as nalmefene (Revex®), 3-methoxynaltrexone, naloxone, and naltrexone; and those disclosed in WO 00/21509; (13) orexin antagonists, such as SB-334867-A (GlaxoSmithkline); and those disclosed in WO 01/96302, 01/68609, 02/44172, 02/51232, 02/51838, 02/089800, 02/090355, 03/023561, 03/032991, 03/037847, 04/004733, 04/026866, 04/041791, 04/085403, and
  • CNTF ciliary neurotrophic factors
  • GI-181771 Gaxo-SmithKline
  • SR146131 Sanofi Synthelabo
  • butabindide butabindide
  • PD170,292, PD 149164 Pfizer
  • CNTF derivatives such as axokine (Regeneron); and those disclosed in WO 94/09134, WO 98/22128, and WO 99/43813
  • GHS growth hormone secretagogue receptor
  • GHS growth hormone secretagogue receptor
  • GLP-1 glucagon-like peptide 1 agonists
  • Topiramate Topimax®
  • phytopharm compound 57 CP 644,673
  • ACC2 acetyl-CoA carboxylase-2
  • ⁇ 3 beta adrenergic receptor 3) agonists, such as rafebergron/AD9677/TAK677 (Dainippon/Takeda), CL-316,243, SB 418790, BRL-37344, L-796568, BMS-196085, BRL-35135A, CGP12177A, BTA-243, GRC1087 (Glenmark Pharmaceuticals)
  • GW 427353 solabegron hydrochloride
  • Trecadrine Zeneca D7114, N-5984 (Nisshin Kyorin)
  • glucocorticoid receptor antagonists such as CP472555 (Pfizer), KB 3305, and those disclosed in WO 04/000869, WO 04/075864, and the like; (37) 11 ⁇ HSD-1 (11-beta hydroxy steroid dehydrogenase type 1) inhibitors, such as BVT 3498 (AMG 331), BVT 2733, 3-(1-adamantyl)-4-ethyl-5-(ethylthio)-4H-1,2,4-triazole, 3-(1-adamantyl)-5-(3,4,5-trimethoxyphenyl)-4-methyl-4H-1,2,4-triazole, 3-adamantanyl-4,5,6,7,8,9,10,11,12,3a-decahydro-1,2,4-triazolo[4,3-a][11]annulene, and those compounds disclosed in WO 01
  • Specific compounds that can be used in combination with the OXM analogs disclosed herein include specific CB1 antagonists/inverse agonists include those described in WO03/077847, including: N-[3-(4-chlorophenyl)-2(S)-phenyl-1(S)-methylpropyl]-2-(4-trifluoromethyl-2-pyrimidyloxy)-2-methylpropanamide, N-[3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-(5-trifluoromethyl-2-pyridyloxy)-2-methylpropanamide, N-[3-(4-chlorophenyl)-2-(5-chloro-3-pyridyl)-1-methylpropyl]-2-(5-tri fluoromethyl-2-pyridyloxy)-2-methylpropanamide, and pharmaceutically acceptable salts thereof; as well as those in WO05/000809, which includes the following: 3- ⁇
  • NPY5 antagonists that can be used in combination with the OXM analogs disclosed herein include: 3-oxo-N-(5-phenyl-2-pyrazinyl)-spiro[isobenzofuran-1(3H), 4′-piperidine]-1′-carboxamide, 3-oxo-N-(7-trifluoromethylpyrido[3,2-b]pyridin-2-yl)spiro-[isobenzofuran-1(3H), 4′-piperidine]-1′-carboxamide, N-[5-(3-fluorophenyl)-2-pyrimidinyl]-3-oxospiro-[isobenzofuran-1(3H), 4′-piperidine]-1′-carboxamide, trans-3′-oxo-N-(5-phenyl-2-pyrimidinyl)spiro[cyclohexane-1,1′(3′H)-isobenzofuran]-4-carboxamide, trans-3
  • Specific ACC-1/2 inhibitors that can be used in combination with the OXM analogs disclosed herein include: 1′-[(4,8-dimethoxyquinolin-2-yl)carbonyl]-6-(1H-tetrazol-5-yl)spiro[chroman-2,4′-piperidin]-4-one; (5- ⁇ 1′-[(4,8-dimethoxyquinolin-2-yl)carbonyl]-4-oxospiro[chroman-2,4′-piperidin]-6-yl)-2H-tetrazol-2-yl)methyl pivalate; 5- ⁇ 1′-[(8-cyclopropyl-4-methoxyquinolin-2-yl)carbonyl]-4-oxospiro[chroman-2,4′-piperidin]-6-yl ⁇ nicotinic acid; 1′-(8-methoxy-4-morpholin-4-yl-2-naphthoyl)-6-(1H-tetrazol-5
  • MCH1R antagonist compounds that can be used in combination with the OXM analogs disclosed herein include: 1- ⁇ 4-[(1-ethylazetidin-3-yl)oxy]phenyl ⁇ -4-[(4-fluorobenzyl)oxy]pyridin-2(1H)-one, 4-[(4-fluorobenzyl)oxy]-1- ⁇ 4-[(1-isopropylazetidin-3-yl)oxy]phenyl ⁇ pyridin-2(1H)-one, 1-[4-(azetidin-3-yloxy)phenyl]-4-[(5-chloropyridin-2-yl)methoxy]pyridin-2(1H)-one, 4-[(5-chloropyridin-2-yl)methoxy]-1- ⁇ 4-[(1-ethylazetidin-3-yl)oxy]phenyl ⁇ pyridin-2(1H)-one, 4-[(5-chloropyridin-2
  • a specific DPP-IV inhibitor that can be used in combination with the OXM analogs disclosed herein is 7-[(3R)-3-amino-4-(2,4,5-trifluorophenyl)butanoyl]-3-(trifluoromethyl)-5,6,7,8-tetrahydro-1,2,4-triazolo[4,3-a]pyrazine, or a pharmaceutically acceptable salt thereof.
  • H3 (histamine H3) antagonists/inverse agonists that can be used in combination with the OXM analogs disclosed herein include: those described in WO05/077905, including: 3- ⁇ 4-[(1-cyclobutyl-4-piperidinyl)oxy]phenyl)-2-ethylpyrido[2,3-d]-pyrimidin-4(3H)-one, 3-(4-[(1-cyclobutyl-4-piperidinyl)oxy]phenyl ⁇ -2-methylpyrido[4,3-d]pyrimidin-4(3H)-one, 2-ethyl-3-(4- ⁇ 3-[(3S)-3-methylpiperidin-1-yl]propoxy ⁇ phenyl)pyrido[2,3-d]pyrimidin-4(3H)-one 2-methyl-3-(4- ⁇ 3-[(3S)-3-methylpiperidin-1-yl]propoxy ⁇ phenyl)pyrido[4,3-d]pyrimidin
  • CCK1R agonists of use in combination with the OXM analogs disclosed herein include: 3-(4- ⁇ [1-(3-ethoxyphenyl)-2-(4-methylphenyl)-1H-imidazol-4-yl]carbonyl ⁇ -1-piperazinyl)-1-naphthoic acid; 3-(4- ⁇ [1-(3-ethoxyphenyl)-2-(2-fluoro-4-methylphenyl)-1H-imidazol-4-yl]carbonyl ⁇ -1-piperazinyl)-1-naphthoic acid; 3-(4- ⁇ [1-(3-ethoxyphenyl)-2-(4-fluorophenyl)-1H-imidazol-4-yl]carbonyl ⁇ -1-piperazinyl)-1-naphthoic acid; 3-(4- ⁇ [1-(3-ethoxyphenyl)-2-(2,4-difluorophenyl)-1H-
  • MC4R agonists of use in combination with the OXM analogs disclosed herein include: 1) (5S)-1′- ⁇ [(3R,4R)-1-tert-butyl-3-(2,3,4-trifluorophenyl)piperidin-4-yl]carbonyl ⁇ -3-chloro-2-methyl-5-[1-methyl-1-(1-methyl-1H-1,2,4-triazol-5-yl)ethyl]-5H-spiro[furo[3,4-b]pyridine-7,4′-piperidine]; 2) (5R)-1′- ⁇ [(3R,4R)-1-tert-butyl-3-(2,3,4-trifluorophenyl)-piperidin-4-yl]carbonyl ⁇ -3-chloro-2-methyl-5-[1-methyl-1-(1-methyl-1H-1,2,4-thiazol-5-yl)ethyl]-5H-spiro[furo[3,4-b]pyridine-7,4′
  • GLP-1 incretin hormone glucagon-like peptide 1
  • Methods of administrating the pharmacological compositions comprising the one or more OXM analogs disclosed herein to an individual include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compositions can be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (for example, oral mucosa, rectal and intestinal mucosa, and the like), ocular, and the like and can be administered together with other biologically-active agents. Administration can be systemic or local.
  • Intraventricular injection may be facilitated by an intraventricular catheter attached to a reservoir (for example, an Ommaya reservoir).
  • Pulmonary administration may also be employed by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. It may also be desirable to administer the one or more OXM analogs disclosed herein locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, by injection, by means of a catheter, by means of a suppository, or by means of an implant.
  • the OXM analogs disclosed herein may be delivered in a vesicle, in particular a liposome.
  • a liposome the OXM analogs disclosed herein are combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids which exist in aggregated form as micelles, insoluble monolayers, liquid crystals, or lamellar layers in aqueous solution.
  • Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. No. 4,837,028 and U.S. Pat. No. 4,737,323.
  • the OXM analogs disclosed herein can be delivered in a controlled release system including, but not limited to: a delivery pump (See, for example, Saudek, et al., New Engl. J. Med.
  • the controlled release system can be placed in proximity of the therapeutic target (for example, the brain), thus requiring only a fraction of the systemic dose. See, for example, Goodson, In: Medical Applications of Controlled Release, 1984. (CRC Press, Bocca Raton, Fla.).
  • compositions comprising one or more of the OXM analogs disclosed herein which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and may be determined by standard clinical techniques by those of average skill within the art. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • dose to be employed in the formulation will also depend on the route of administration, and the overall seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances, Ultimately, the attending physician will decide the amount of the composition with which to treat each individual patient. Initially, the attending physician will administer low doses of the composition and observe the patient's response.
  • the daily dose range lie within the range of from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 50 mg per kg, and most preferably 0.1 to 10 mg per kg, in single or divided doses. On the other hand, it may be necessary to use dosages outside these limits in some cases.
  • suitable dosage ranges for intravenous administration of the compositions comprising the one or more OXM analogs disclosed herein are generally about 5-500 micrograms ( ⁇ g) of active compound per kilogram (Kg) body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient. Ultimately the attending physician will decide on the appropriate duration of therapy using compositions comprising the one or more OXM analogs disclosed herein of the present invention. Dosage will also vary according to the age, weight and response of the individual patient.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions and OXM analogs disclosed herein.
  • Optionally associated with such container(s) may be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Oxyntomodulin (OXM) analogs were essentially as follows.
  • the OXM analogs shown in Table 2 below were synthesized by solid phase using Fmoc/t-Bu chemistry on a peptide multisynthesizer APEX 396 (Advanced Chemtech) using a 40-well reaction block. Each peptide was synthesized in a single well.
  • peptide amides For peptide amides, 0.1 g of an aminomethylated polystirene LL (100-200 mesh, 0.41 mmol/g) (Novabiochem) resin derivatized with a modified Rink linker p-[(R,S)- ⁇ -[9H-Fluoren-9-yl-methoxyformamido]-2,4-dimethoxybenzyl]-phenoxyacetic acid (Rink, H., 1987 , Tetrahedron Lett. 28:3787-3789; Bernatowicz, M. S. et al., 1989 , Tetrahedron Lett. 30:4645-4667) was used.
  • All the amino acids were dissolved at a 0.5 M concentration in a solution of 0.5M HOBt (Hydroxybenzotriazole) in DMF.
  • the acylation reactions were performed for 45 minutes with six-fold excess of activated amino acid over the resin free amino groups.
  • the amino acids were activated with equimolar amounts of HBTU (2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) and a two-fold molar excess of DIEA (N,N-diisopropylethylamine).
  • the peptides were synthesized by solid phase using Fmoc/t-Bu chemistry on a Pioneer Peptide Synthesizer (Applied Biosystems).
  • the side chain protecting groups were: OMpe (O-3-Methyl-pent-3-yl) for Asp; tert-butyl for Glu, Ser, D-Ser, Thr and Tyr; trityl for Asn, Cys, Gln, and H; and, tert-butoxy-carbonyl for Lys, Trp; and, 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl for Arg.
  • OMpe O-3-Methyl-pent-3-yl
  • tert-butyl for Glu, Ser, D-Ser, Thr and Tyr
  • trityl for Asn, Cys, Gln, and H
  • tert-butoxy-carbonyl for Lys, Trp
  • 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl for Arg.
  • Lysine-Palmitoyl was manually acylated by reaction with equimolar amounts of HBTU (2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) and a 2-fold molar excess of DIEA (N,N-diisopropylethylamine).
  • HBTU 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate
  • DIEA N,N-diisopropylethylamine
  • the dry peptide-resins were individually treated with 20 mL of the cleavage mixture, 88% trifluoroacetic acid (TFA), 5% phenol, 2% triisopropylsilane and 5% water (Sole, N. A. and G. Barany, 1992 , J. Org, Chem. 57:5399-5403) for 1.5 hours at room temperature. Each resin was filtered and the solution was added to cold methyl-t-butyl ether in order to precipitate the peptide. After centrifugation, the peptide pellets were washed with fresh cold methyl-t-butyl ether to remove the organic scavengers. The process was repeated twice. Final pellets were dried, resuspended in H 2 O, 20% acetonitrile, and lyophilized.
  • the crude peptides were purified by reverse-phase HPLC using preparative Waters RCM Delta-PakTM C ⁇ 4 cartridges (40 ⁇ 200 mm, 15 ⁇ m) and using as eluents (A) 0.1% TFA in water and (B) 0.1% TFA in acetonitrile.
  • the following gradient of eluent B was used: 20%-20% over five minutes and 20%-35% over 20 minutes for the OXM229 (the peptide thiolated precursor of OXM36 and OXM115), OXM29 (the thiolated precursor of OXM70 and OXM216), OXM208 (the peptide thiolated precursor of OXM212), and OXM209 (the peptide thiolated precursor of OXM213).
  • peptides OXM110 and 177 the following gradient of eluent B was used: 32%-32% over five minutes and 32%-42% over 20 minutes flow rate 80 mL/min.
  • Analytical HPLC was performed on a Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45 C.°, using H 2 O, 0.1% TFA (A) and CH 3 CN, 0.1% TFA (B) as solvents and the following linear gradient: 20%-20% B (in five minutes)-35% B (in 20 minutes)-80% B (in two minutes), flow 1 mL/min.
  • the purified peptide was characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • Oxyntomodulin (OXM) cholesterylated analogs OXM36, OXM70, OXM115, OXM212, OXM213, and OXM216 was as follows.
  • the reactions were run under conditions that permit the formation of a thioether bond.
  • the cholesterylaled OXM peptides were then isolated using reverse-phase HPLC and characterized on a Micromass LCZ platform.
  • the analogs OXM36, 70, 212, and 213 were synthesized from the thiol containing OXM peptide precursor OXM229, OXM208, and 209, respectively, by reaction with the bromo derivative, cholest-5-en-3-yl bromoacetate having the structure
  • peptide precursor were dissolved in one mL of DMSO (conc. 30 mg/mL) and a one molar excess of cholest-5-en-3-yl bromoacetate dissolved in THF (conc. 20 mg/mL) was added. Then 1% by volume of DIPEA (N,N-diisopropyl-ethylamine) was added to the mixture; after 30 minutes of incubation, the peptide solution was purified by RP-HPLC and characterized on a Micromass LCZ platform.
  • DIPEA N,N-diisopropyl-ethylamine
  • the peptides OXM115 and 216 were synthesized from the thiol containing OXM peptide precursor OXM229 and OXM29 to produce analogs by reaction with the bromo derivative, cholest-5-en-3-yl]-bromo-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oate
  • peptide precursor were dissolved in 1 mL of DMSO (conc. 30 mg/mL) and a one molar excess of cholest-5-en-3-yl]-bromo-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oate dissolved in THF (conc. 20 mg/mL) was added. Then 3% by volume of DIPEA (N,N-diisopropyl-ethylamine) was added to the mixture; after 30 minutes of incubation, the peptide solution was purified by RP-HPLC and characterized on a Micromass LCZ platform.
  • DIPEA N,N-diisopropyl-ethylamine
  • PEGylation reactions were run under conditions permitting thioester bond formation.
  • the PEGylated OXM peptide was then isolated using reverse-phase HPLC or ion exchange chromatography and size exclusion chromatography (SEC).
  • SEC size exclusion chromatography
  • PEGylated OXM analogs were characterized using RP-HPLC, HPLC-SEC and MALDI-Tof Mass Spectrometry.
  • OXM33, 34, 35, 36 and 54 peptides were synthesized from the thiol-containing OXM peptide precursor OXM229 to produce analogs with PEG covalently attached via a thioether bond.
  • control peptide OXM54 was prepared by incubating the thiol containing peptide precursor with 10 eq. of iodoacetamide in 0.1 M TrisHCl pH 7.5, 6M guanidinium chloride. After 30 minutes incubation the peptide was purified by RP-HPLC and characterized by electrospray mass spectrometry.
  • OXM peptide analogs that that display full agonistic activity on the GLP-1 and the Glucagon receptors and shown in Table 2 were synthesized as follows.
  • the peptides OXM290, 291, 292, 293 and 294 that are precursors of OXM301, OXM302, OXM303, OXM304 and OXM305 were synthesized by solid phase using Fmoc/t-Bu chemistry on a peptide multisynthesizer Simphony Protein Technologies Inc.
  • peptide amides For peptide amides, 0.5 g of a resin Aminomethylated polystirene LL (100-200 mesh, 0.41 mmol/g) (Novabiochem) resin derivatized with a modified Rink linker p-[(R,S)- ⁇ -[9H-Fluoren-9-yl-methoxyformamido]-2,4-dimethoxybenzyl]-phenoxyacetic acid (Rink, Tetrahedron Lett. 28:3787-3789 (1987); Bematowiez et al., Tetrahedron Lett. 30:4645-4667 (1989)) was used.
  • All the amino acids were dissolved at a 0.5 M concentration in a solution of 0.5 M HOBt (Hydroxybenzotriazole) in DMF.
  • the acylation reactions were performed for 60 minutes with 8-fold excess of activated amino acid over the resin free amino groups.
  • the amino acids were activated with equimolar amounts of HBTU (2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate), solution 0.5 M in DMF, and a 2-fold molar excess of DIEA (N,N-diisopropylethylamine), solution 2 M in NMP.
  • HBTU 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate
  • DIEA N,N-diisopropylethylamine
  • the side chain protecting groups were: OMpe (O-3-Methyl-pent-3-yl) for Asp; tert-butyl for Glu, Ser, D-Ser, Thr and Tyr; trityl for Asn, Cys, Gln and H; tert-butoxy-carbonyl for Lys, Trp; and, 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl for Arg; Boc-His(Trt)-OH was used in the synthesis.
  • the dry peptide-resins were individually treated with 25 mL of the cleavage mixture, 82.5% trifluoroacetic acid (TFA), 5% phenol, 5% thioanisole, 2.5% Ethandithiole, and 5% water for 1.5 hours at room temperature. Each resin was filtered and the volume of the solution was reduced then added to cold methyl-t-butyl ether in order to precipitate the peptide. After centrifugation, the peptide pellets were washed with fresh cold methyl-t-butyl ether to remove the organic scavengers. The process was repeated twice. Final pellets were dried, resuspended in H 2 O, 20% acetonitrile, and lyophilized.
  • TFA trifluoroacetic acid
  • the crude peptides were purified by reverse-phase HPLC using preparative Waters RCM Delta-PakTM C ⁇ 4 cartridges (40 ⁇ 200 mm, 15 ⁇ m) and using as eluents (A) 0.1% TFA in water and (B) 0.1% TFA in acetonitrile.
  • the following gradient of eluent B was used: 22%-22% over 5 min and 22%-32% over 20 min for OXM301 precursor, 22%-22% over 5 min and 22%-35% over 20 min for OXM302 precursor, 25%-25% over 5 min and 25%-40% over 20 min for OXM303 precursor, 25%-25% over 5 min and 25%-35% over 20 min for OXM304 precursor and OXM305 precursor, flow rate 80 mL/min, wavelength 214 nm.
  • Analytical HPLC was performed on a Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents and the following linear gradient: 25%-25% B (in 5 min)-40% B (in 20 min)-80% B (in 2 minutes), flow 1 mL/min.
  • the purified peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • the final peptides were characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH 3 CN, 0.1% TFA (B) as solvents and the following linear gradient: 40%-40% B (in 5 min)-70% B (in 20 min)-80% B (in 2 min), flow 1 in mL/min.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • OXM237-OXM308 and OXM345-OXM414 were synthesized by solid phase using Fmoc/t-Bu chemistry on a peptide multisynthesizer Simphony Protein Technologies Inc.
  • the amino acids were activated with equimolar amounts of HBTU (2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate), solution 0.5 M in DMF, and a 2-fold molar excess of DIEA (N,N-diisopropylethylamine), solution 2M in NMP.
  • HBTU 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate
  • DIEA N,N-diisopropylethylamine
  • the side chain protecting groups were: Ompe (O-3-Methyl-pent-3-yl) for Asp; tert-butyl for Gln, Ser, D-Ser, Thr and Tyr; trityl for Asn, Cys, Gln and H; tert-butoxy-carbonyl for Lys, Trp; and, 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl for Arg; Boc-His(Trt)-OH was used in the synthesis.
  • Glu16 and Lys20 were incorporated as Glu(Oall) and Lys(Alloc).
  • the resins were dried, the protecting groups of Glu(Oall) and Lys(Alloc) were removed and the lactam bridge was formed by incubating the resin with 5 molar excess of HBTU and 10 molar excess of DIPEA.
  • lipidated peptides such as OXM404, 407, 408, 410, 411, 414, 415, 416, 417, 418, 419, 420 the lysine to be derivatized on the side chain was incorporated as Lys(Alloc).
  • Alloc protecting group was removed and the synthesis was completed by condensation of the ⁇ -carboxyglutamic acid residues and the palmitic acid using HBTU and DIPEA as activators.
  • the dry peptide-resins were individually treated with 25 mL of the cleavage mixture, 82.5% trifluoroacetic acid (TFA), 5% phenol, 5% thioanisole, 2.5% ethandithiole and 5% water for 1.5 hours at room temperature. Each resin was filtered and the volume of the solution was reduced then added to cold methyl-t-butyl ether in order to precipitate the peptide. After centrifugation, the peptide pellets were washed with fresh cold methyl-t-butyl ether to remove the organic scavengers. The process was repeated twice. Final pellets were dried, resuspended in H 2 O, 20% acetonitrile, and lyophilized.
  • TFA trifluoroacetic acid
  • the crude peptides were purified by reverse-phase HPLC using preparative Waters RCM Delta-PakTM C 4 cartridges (40 ⁇ 200 mm, 15 ⁇ m) and using as eluents (A) 0.1% TFA in water and (B) 0.1% TFA in acetonitrile.
  • Analytical HPLC was performed on a Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the purified peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • the peptide OXM238 was synthesized from a thiol containing OXM peptide precursor to produce derivatives with the acetamide covalently attached via the thioethers group of the cysteine residue at the C-terminus.
  • derivatization of the precursor 50 mg of peptide precursor were dissolved in Tris HCl buffer 0.25 M, EDTA 2 mM, Urea 6M pH 8.3 (conc. 30 mg/mL) and a 10 molar excess of iodoacetamide dissolved in DMSO (conc. 30 mg/mL) was added. After 30 minutes the reaction is complete. The solution is acidified with acetic acid and purified on preparative HPLC.
  • Synthesis of C 4 conjugates was as follows.
  • the peptides OXM345, OXM355, OXM357, OXM373 were synthesized from the thiol containing OXM peptide precursors respectively to produce derivatives with the cholesterol group covalently attached via the thioethers group of the cysteine at the C-terminus.
  • derivatization of the precursor 40 mg of peptide precursor were dissolved in DMSO (conc. 30 mg/mL) and a 2.1 molar excess of cholest-5-en-3-yl 1-bromo-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oate dissolved in THF (conc.
  • the final peptides were characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass.
  • the final peptides were characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • Synthesis of C 9 conjugates was as follows.
  • the peptide OXM381 was synthesized from the thiol containing OXM peptide precursor to produce derivatives with the Oxa12-cholesterol covalently attached via the maleimide thioether bond to the cysteine residue at the C-terminus.
  • derivatization of the precursor 25 mg of peptide precursor were dissolved in DMSO (conc.
  • Synthesis of C 10 conjugates was as follows.
  • the peptides OXM392, 395, 398, 399, 400, and 401 were synthesized from the thiol containing OXM peptide precursors to produce derivatives with the Oxa12-cholesterol covalently attached via a thioether bond to the cysteine residue at the C-terminus.
  • derivatization of the precursor 25 mg of peptide precursor were dissolved in DMSO (conc.
  • the final peptide was characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • Synthesis of C 11 conjugates was as follows.
  • the peptides OXM412 was synthesized from the thiol containing OXM peptide precursor to produce derivatives with the Oxa24-cholesterol covalently attached via a thioether bond to the cysteine residue at the C-terminus.
  • derivatization of the precursor 25 mg of peptide precursor were dissolved in DMSO (conc.
  • DIPEA N,N-diisopropyl-ethylamine
  • the final peptide was characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TFA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • Synthesis of C 12 conjugates was as follows.
  • the peptides OXM421 was synthesized from the thiol containing OXM peptide precursor to produce derivatives with the Oxa12-O-cholesterol covalently attached via a thioether bond to the cysteine residue at the C-terminus.
  • derivatization of the precursor 25 mg of peptide precursor were dissolved in DMSO (conc.
  • the final peptide was characterized on an Alliance Waters Chromatograph, with a ACE C-4 (300 A), 3 um column, 150 ⁇ 4.6 mm, (CPS analitica p/n ACE-213-1546), at 45° C., using H 2 O, 0.1% TEA (A) and CH3CN, 0.1% TFA (B) as solvents.
  • the peptides were characterized by electrospray mass spectrometry on a Micromass LCZ platform.
  • Table 3 shows the structures for the native OXM and OXM analogs that are disclosed herein.
  • OXM Peptide Analogs SEQ ID NO. Name Structure 1 OXM HSQGTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA (native) 2 OXM8 HS E GTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA (Q3E) 3 OXM9 HS D GTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA (Q3D) 4 OXM29 H ⁇ QGTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA C -CONH 2 5 OXM33 H s QGTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA C 1 -CONH 2 6 OXM34 H s QGTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA C 2 -CONH 2 7 OXM35 H s QGTFTSDYSKYLDSRRAQDFVQWLMNTKRNNIA C 3
  • C 9 S- ⁇ 1-[46-(cholest-5-en-3-yloxy)-3,43,46-trioxo-7,10,13,16,19,22,25,28,31,34,37,40-dodecaoxa-4,44-diazahexatetracont-1-yl]-2,5-dioxopyrrolidin-3-yl ⁇ -L-cysteine or Cys (mal-oxa 12 -cholesterol)
  • C 10 S-[42-(cholest-5-en-3-yloxy)-2,42-dioxo-6,9,12,15,18,21,24,27,30,33,36,39-dodecaoxa-3-azadotetracont-1-yl]-L-cysteine or Cys (oxa1 2 -cholesterol)
  • C 11 S-[78-(cholest-5-en-3-yloxy)-2,78-dioxo- 6,9,12,
  • GLP-1R/GCGR co-agonists to reduce body weight and food intake and improve glycemic control is demonstrated.
  • CHO cells stably expressing mouse or human GLP-1R or GCGR were grown in Iscove's Modified Dulbecco's Medium (IMDM), 10% FBS, 1 mM L-glutamine, penicillin-streptomycin (100 ⁇ /mL) and 750 ⁇ g G418/mL for three-four days before harvesting using Enzymefree Dissociation Media (EFDM, Specialty Media).
  • IMDM Iscove's Modified Dulbecco's Medium
  • FBS FBS
  • 1 mM L-glutamine penicillin-streptomycin
  • EFDM Enzymefree Dissociation Media
  • OXM and OXM-Q3E were diluted in assay buffer and were incubated with cells in the absence or presence of 10% mouse plasma, respectively, for 30 min at room temperature.
  • the assay was terminated with the addition of the LANCE kit Detection buffer as per the manufacturer's instructions.
  • the plates were held an additional hour at room temperature, then increasing cAMP levels were detected by a decrease in TR-FRET signal as measured in an EnVision counter (PerkinElmer) in comparison to a standard curve of cAMP as per the manufacturer's instructions. Data were analyzed using the linear and nonlinear regression analysis software, GraphPad Prism.
  • Percent inhibition values for each treatment were generated from the AUC data normalized to the saline-challenged controls. Areas under the curve (AUC) for glucose (AUC glucose) was calculated using the trapezoidal method. Statistics. Statistical analysis was done using unpaired 2-tailed Student's t test. P values ⁇ 0.05 were reported as significant.
  • 31 P NMR spectroscopy was performed initially to examine the ATP and inorganic phosphate (Pi) levels to assess hepatic viability.
  • a 13 C NMR-visible pool of glycogen was then created by the addition of the gluconeogenic substrate [2- 13 C] Pyruvate and ammonium chloride ( ⁇ 7 mM and 1 mM, respectively) to the perfusate, and the amount of 13 C glycogen contained in the liver was monitored in real time via the C1 resonance of the glucosyl units in glycogen.
  • GDIS Glucose dependent insulin secretion
  • Insulin was measured in aliquots of the incubation buffer by enzymelinked immunosorbent assay with a commercial kit (ALPCO diagnostics, Windham, N.H.). Animal study protocols were reviewed and approved by the Merck Research Laboratories Institutional Animal Care and Use Committee in Rahway, N.J.
  • 13 C-NMR spectroscopy was used to non-invasively monitor glycogen and glucose levels in response to acute treatment with novel oxyntomodulin analogs in real time.
  • Mice expressing human GCGR were anesthetized (Nembutal IP, 50 mg/kg) at approximately the middle of the dark cycle.
  • the portal vein was then cannulated and tied off and the liver was excised.
  • the liver was then placed into a 20 mm NMR tube and the initial Krebs solution was exchanged for a Krebs-BSA perfusate, which was recirculated.
  • 31 P NMR spectroscopy was performed initially to examine the ATP and inorganic phosphate (Pi) levels in the liver which can be used to assess hepatic viability.
  • a 13 C NMR-visible pool of glycogen was then created by the addition of the gluconeogenic substrate [2- 13 C] Pyruvate NH 4 Cl and the amount of glycogen contained in the liver was monitored in real time via the C1 resonance of the glucosyl units in glycogen. After approximately 45 minutes, OXM or an OXM peptide analog was infused and the subsequent response of glycogen levels was used to assess human GCGR activation. The area of the C1 resonance of the glucosyl units in glycogen is plotted over time in FIG. 1 for livers that received either the OXM peptide analogs, 50 pM glucagon, or media.
  • native OXM induces glycogenolysis in a dose dependent fashion and induces full glycogenolysis at 1.5 nM and has an approximate EC 50 of 0.5 nM, in comparison, OXM-Q3E induced only about 58% at 300 nM, consistent with its poor GCGR agonist potency.
  • mice Given the ability of OXM to stimulate a robust glycogenolysis response, we further examined the in vivo GCGR receptor occupancy of OXM and OXM-Q3E in a competition assay in wild-type mice.
  • Three cohorts of mice were dosed with either vehicle (total counts), vehicle and cold glucagon (non-specific binding), or OXM-Q3E subcutaneously. About 15 minutes post dose, 125 I-glucagon was administered intra-venously and about 15 minutes later, the liver was collected and total radioactivity assayed. As shown in FIG. 2 , glucagon (GCG) at 1.5 mpk gave 84% GCGR occupancy, 3 mpk OXM gave 31% GCGR occupancy, and OXM-Q3E gave 0% GCGR occupancy.
  • vehicle total counts
  • vehicle and cold glucagon non-specific binding
  • OXM-Q3E subcutaneously.
  • 125 I-glucagon was administered intra-venously and about 15 minutes later, the liver was collected
  • OXM, OXM-Q3E Using the matched peptides, OXM, OXM-Q3E, we investigated both acute and chronic pharmacological effects of co-agonism of both receptors in a chronic hyperglycemic clamp and a chronic food intake (FI) and body weight (BW) study (Table 5) and in an acute intraperitoneal glucose tolerance test (IPGTT, FIG. 3 ). While both peptides effectively improved glucose tolerance, OXM-Q3E out-performed native OXM in reducing glucose excursion in both lean and diet induced obesity (DIO) mouse models.
  • DIO diet induced obesity
  • the native peptide proved superior at reducing food intake and body weight ( ⁇ BW: ⁇ 6% for OXM vs ⁇ 2.4% for OXM-Q3E at 5 mg/kg) in DIO mice, an effect attributed to co-agonism of the GCGR.
  • native OXM did not cause increased glucose excursion.
  • DIO mice (16 wk on high fat diet) were anesthetized with xylazine and ketamine and catheterized through the right internal jugular vein 3 days before the in vivo studies. 25% glucose was infused and adjusted for the duration of the experiment to maintain hyperglycemia. Due to the short half-life of the peptides (both are rapidly inactivated by dipeptidyl peptidase IV and renally cleared (Zhu et al., J. Biol. Chem. 278: 22418-23, (2003))), vehicle, OXM ( ⁇ 16 ⁇ g/kg/min) and OXM-Q3E ( ⁇ 16 ⁇ g/kg/min) were infused intravenously during the last 60 minutes of the study.
  • the venous catheter was used for infusion, and blood samples were collected from the tail vein. Each animal was monitored for food intake and weight gain after surgery to ensure complete recovery.
  • Hyperglycemic clamps were performed in conscious, unrestrained, catheterized mice as previously described (Pocai et al., Nature. 43: 1026-31, (2005)). Briefly, plasma glucose was measured by OneTouch glucometer every 10 minutes during the 2 hour period. Twenty-five percent glucose (D-glucose, Sigma) was infused i.v. and the rate was adjusted periodically to clamp the plasma glucose levels ( ⁇ 20 mM).
  • OXM ⁇ 16 ⁇ g/kg/min
  • OXM-Q3E 16 ⁇ g/kg/min
  • vehicle sterile saline
  • FIGS. 3A and 3B The results of the hyperglycemic clamp assay are shown in FIGS. 3A and 3B .
  • Blood glucose levels were maintained at about 20 mmol/L during the clamp.
  • GIR glucose infusion rate
  • vehicle-treated mice GIR0-60 55 ⁇ 4 vs 6 ⁇ 2 mg/kg/min; OXM-Q3E vs vehicle; p ⁇ 0.05.
  • FIG. 4 shows that both native and OXM peptide anlogs reduce glucose excursion.
  • a comparison of the minimal effective dose indicates that OXM-Q3E is about 10-fold more effective at reducing glucose than native peptide (in viva EC 50 0.055 nM and 0.38 nM respectively).
  • Native OXM has an in viva half-life (t 1/2 ) of about 8 to 12 minutes in humans, unsuitable for sustained pharmacological therapy. Therefore, we investigated the addition of bulky substituents to the peptide to reduce renal clearance.
  • FIG. 5A shows ex vivo measurement of glycogenolysis in perfused liver in the presence of OXM70, OXM110, OXM177, OXM115, and OXM216.
  • FIG. 5B shows GGCR occupancy following subcutaneous (s.c.) or intravenous (i.v.) administration of the OXM70, OXM110, OXM177, OXM115, and OXM216 compared to native OXM in a competition assay in wild-type mice.
  • FIG. 6 summarizes the acute in vivo efficacy of OXM70 on reducing food intake and body weight in established DIO mice.
  • FIG. 7 summarizes the acute in vivo efficacy of OXM110 and OXM177 on reducing food intake and body weight in established DIO mice.
  • FIG. 8 summarizes the acute in vivo efficacy of OXM216, OXM115, and OXM177 on reducing food intake and body weight in established DIO mice.
  • FIGS. 9A and 9B chronic treatment with the lipidated analogs significantly reduced body weight and food intake in a dose dependent manner throughout the duration of the study.
  • FIG. 9C shows the basal glucose over the time period of the study in response to the various peptides.
  • the total cycle time was set for 20 minutes, consisting of a reference reading with a 70 second settling time and a 10 second measure period as well as 60 second settle time and 10 second measure time for each of the 16 cages. Results shown in FIGS. 14A and B show that administration of OXM115 dose dependently increased the acute metabolic rate in DIO mice.
  • FIG. 15 summarizes the acute in vivo efficacy of OXM303, OXM304, and OXM305 on reducing food intake and body weight in established DIO mice and shows OXM305 as being particularly efficacious at reducing food intake and reducing body weight.
  • FIG. 16 summarizes the acute in vivo efficacy of OXM395, OXM399, OXM400, and OXM401 on reducing food intake and body weight in established DIO mice.
  • OXM401 is a 0/+ control peptide having the sequence HAibEGTFTSDYSKYLDSRRAQDFVQWLmNTK- ⁇ E ⁇ E-C(Oxa 12 Chol) (SEQ ID NO:99).
  • This example shows that lipidated peptides with a pI above 7 based on the OXM sequence stimulate mast cell degranulation while lipidated peptides with a pI of about 5 appears not to stimulate mast degranulation.
  • LAD2 cells 50,000 cells/well, 96-well plate
  • the degranulation of LAD2 cells was determined by quantification of the ⁇ -hexosaminidase released into the supernatants and in cell lysates after hydrolysis of 4-methylumbelliferyl-N-acetyl- ⁇ -D-glucosaminide. Results are expressed as percentage of ⁇ -hexosaminidase release, calculated as percent of total content.
  • Table 1 summarizes the degranulation potency (EC 50 ) of OXM analogues in LAD2 cells. The results suggest that to reduce the likelihood of stimulating degranulation of mast cells, the pI for lipidated based analogues should be around 5.
  • OXM >10 OXM115 0.01-0.04 OXM238 1.5 OXM303 0.1 OXM305 0.1 OXM392 0.3 OXM399 0.3 OXM400 >10 OXM401 >10 OXM404 >10 OXM408 >10 OXM409 >10 OXM410 >10 OXM411 >10
  • This example presents tables showing the in vitro potencies of various peptide analogs at the human GLP-1 receptor (hGLP-1R) and the human glucagon receptor (hGCGR).
  • the in vitro potencies shown in Table 10 or Table 11 were determined from the results of assays that were similar to either the cell-based cAMP assays described herein above (Table 10) or the cell-based cAMP assays described in International Published Application No. WO2008101017, which is incorporated herein in its entirety, in particular the cell-based cAMP assays described therein (Table 11).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Obesity (AREA)
  • Child & Adolescent Psychology (AREA)
  • Emergency Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Physical Water Treatments (AREA)
  • Medicinal Preparation (AREA)
US13/202,524 2009-02-19 2009-12-18 Oxyntomodulin analogs Abandoned US20120165503A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/202,524 US20120165503A1 (en) 2009-02-19 2009-12-18 Oxyntomodulin analogs

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/US2009/034448 WO2010096052A1 (en) 2009-02-19 2009-02-19 Oxyntomodulin analogs
USPCTUS2009034448 2009-02-19
PCT/US2009/068678 WO2010096142A1 (en) 2009-02-19 2009-12-18 Oxyntomodulin analogs
US13/202,524 US20120165503A1 (en) 2009-02-19 2009-12-18 Oxyntomodulin analogs

Publications (1)

Publication Number Publication Date
US20120165503A1 true US20120165503A1 (en) 2012-06-28

Family

ID=42634127

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/202,524 Abandoned US20120165503A1 (en) 2009-02-19 2009-12-18 Oxyntomodulin analogs
US14/686,760 Active US9593155B2 (en) 2009-02-19 2015-04-14 Oxyntomodulin analogs

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/686,760 Active US9593155B2 (en) 2009-02-19 2015-04-14 Oxyntomodulin analogs

Country Status (24)

Country Link
US (2) US20120165503A1 (pl)
EP (1) EP2398483B1 (pl)
JP (1) JP5756961B2 (pl)
KR (1) KR101634139B1 (pl)
CN (1) CN102573872B (pl)
AU (1) AU2009340439B2 (pl)
BR (1) BRPI0924307B8 (pl)
CA (1) CA2754350C (pl)
CL (1) CL2011002036A1 (pl)
CY (1) CY1116917T1 (pl)
DK (1) DK2398483T3 (pl)
ES (1) ES2524477T3 (pl)
HK (1) HK1165333A1 (pl)
HR (1) HRP20141215T1 (pl)
IL (1) IL214729A (pl)
MX (1) MX2011008717A (pl)
PE (1) PE20120621A1 (pl)
PL (1) PL2398483T3 (pl)
PT (1) PT2398483E (pl)
RU (1) RU2542362C2 (pl)
SG (1) SG173791A1 (pl)
SI (1) SI2398483T1 (pl)
WO (2) WO2010096052A1 (pl)
ZA (1) ZA201106051B (pl)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120329708A1 (en) * 2011-06-22 2012-12-27 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
WO2014049610A2 (en) 2012-09-26 2014-04-03 Cadila Healthcare Limited Peptides as gip, glp-1 and glucagon receptors triple-agonist
US8969294B2 (en) 2008-06-17 2015-03-03 Istituto Di Recerche Di Biologia Molecolare P. Angeletti S.R.L. Glucagon/GLP-1 receptor co-agonists
EP2916860A4 (en) * 2012-11-06 2016-06-22 Hanmi Pharm Ind Co Ltd COMPOSITION FOR THE TREATMENT OF DIABETES OR DIABESITY COMPRISING AN OXYNTOMODULIN ANALOGUE
WO2017074798A3 (en) * 2015-10-27 2017-05-26 Merck Sharp & Dohme Corp. Long-acting co-agonists of the glucagon and glp-1 receptors
US9724420B2 (en) 2012-11-06 2017-08-08 Hanmi Pharm. Co., Ltd. Liquid formulation of protein conjugate comprising an oxyntomodulin derivative covalently linked to a non-peptidyl polymer to an immunoglobulin FC region
US9765131B2 (en) 2011-06-10 2017-09-19 Hanmi Science Co., Ltd. Oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
US9790263B2 (en) 2009-06-16 2017-10-17 Indiana University Research And Technology Corporation GIP receptor-active glucagon compounds
US9901621B2 (en) 2012-07-25 2018-02-27 Hanmi Pharm. Co., Ltd. Composition for treating hyperlipidemia comprising oxyntomodulin derivative
US10233230B2 (en) 2014-09-16 2019-03-19 Hanmi Pharm. Co., Ltd. Use of a long acting GLP-1/glucagon receptor dual agonist for the treatment of non-alcoholic fatty liver disease
US10513550B2 (en) 2014-12-30 2019-12-24 Hanmi Pharm Co., Ltd Glucagon derivatives
US10696725B2 (en) 2015-06-30 2020-06-30 Hanmi Pharm. Co., Ltd. Glucagon derivative and a composition comprising a long acting conjugate of the same
US10981967B2 (en) 2015-12-31 2021-04-20 Hanmi Pharm. Co., Ltd. Long-acting conjugate of triple glucagon/GLP-1/GIP receptor agonist
US11135271B2 (en) 2014-12-30 2021-10-05 Hanmi Pharm. Co., Ltd. Glucagon derivatives with improved stability
US11142559B2 (en) 2016-06-29 2021-10-12 Hanmi Pharm. Co., Ltd. Glucagon derivative, conjugate thereof, composition comprising same, and therapeutic use thereof
US11872283B2 (en) 2011-06-17 2024-01-16 Hanmi Science Co., Ltd Conjugate comprising oxyntomodulin and an immunoglobulin fragment, and use thereof

Families Citing this family (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1959986B1 (en) 2005-11-07 2014-07-23 Indiana University Research and Technology Corporation Glucagon analogs exhibiting physiological solubility and stability
AU2008205229B2 (en) 2007-01-05 2014-03-27 Indiana University Research And Technology Corporation Glucagon analogs exhibiting enhanced solubility in physiological pH buffers
NZ578948A (en) 2007-02-15 2012-03-30 Univ Indiana Res & Tech Corp Glucagon/glp-1 receptor co-agonists
MX2010004298A (es) 2007-10-30 2010-05-03 Univ Indiana Res & Tech Corp Compuestos que exhiben actividad antagonista de glucagon y agonista de glp-1.
US8981047B2 (en) 2007-10-30 2015-03-17 Indiana University Research And Technology Corporation Glucagon antagonists
EP2952202B1 (en) 2008-06-17 2017-10-18 Indiana University Research and Technology Corporation Gip-based mixed agonists for treatment of metabolic disorders and obesity
CN104447980A (zh) 2008-06-17 2015-03-25 印第安纳大学研究及科技有限公司 在生理pH缓冲液中具有增强的溶解性和稳定性的胰高血糖素类似物
SG172291A1 (en) 2008-12-19 2011-07-28 Univ Indiana Res & Tech Corp Amide based glucagon superfamily peptide prodrugs
WO2010096052A1 (en) 2009-02-19 2010-08-26 Merck Sharp & Dohme Corp. Oxyntomodulin analogs
DK2454282T3 (en) 2009-07-13 2015-05-04 Zealand Pharma As acetylated glucagonanaloger
EP2512503A4 (en) 2009-12-18 2013-08-21 Univ Indiana Res & Tech Corp COAGONISTS OF GLUCAGON / GLP-1 RECEPTOR
AR079344A1 (es) 2009-12-22 2012-01-18 Lilly Co Eli Analogo peptidico de oxintomodulina, composicion farmaceutica que lo comprende y uso para preparar un medicamento util para tratar diabetes no insulinodependiente y/u obesidad
AR079345A1 (es) 2009-12-22 2012-01-18 Lilly Co Eli Analogo peptidico de oxintomodulina
CA2788304A1 (en) 2010-01-27 2011-08-04 Indiana University Research And Technology Corporation Glucagon antagonist - gip agonist conjugates and compositions for the treatment of metabolic disorders and obesity
WO2011134471A1 (en) 2010-04-27 2011-11-03 Zealand Pharma A/S Peptide conjugates of glp-1 receptor agonists and gastrin and their use
MX2012013005A (es) 2010-05-13 2013-02-26 Univ Indiana Res & Tech Corp Peptidos de la superfamilia de glucagon que presentan actividad del receptor acoplado a proteinas g.
KR20130062931A (ko) 2010-05-13 2013-06-13 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 핵 호르몬 수용체 활성을 나타내는 글루카곤 슈퍼패밀리 펩티드
JP2013540102A (ja) 2010-06-24 2013-10-31 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーション アミド結合を介して修飾されたグルカゴンスーパーファミリーのペプチドプロドラッグ
NZ612297A (en) 2010-12-22 2015-10-30 Univ Indiana Res & Tech Corp Glucagon analogs exhibiting gip receptor activity
WO2012150503A2 (en) * 2011-05-03 2012-11-08 Zealand Pharma A/S Glu-glp-1 dual agonist signaling-selective compounds
WO2012153196A2 (en) * 2011-05-10 2012-11-15 Zealand Pharma A/S Glu-glp-1 dual agonist signaling-selective compounds
US9309301B2 (en) 2011-06-22 2016-04-12 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
EP2729493B1 (en) 2011-07-04 2020-06-10 IP2IPO Innovations Limited Novel compounds and their effects on feeding behaviour
WO2013064669A1 (en) 2011-11-03 2013-05-10 Zealand Pharma A/S Glp-1 receptor agonist peptide gastrin conjugates
MX2014003579A (es) * 2011-11-17 2015-04-10 Univ Indiana Res & Tech Corp Peptidos de la superfamilia de glucagon que presentan actividad del receptor de glucocorticoide.
CN104583234A (zh) 2012-06-14 2015-04-29 赛诺菲 毒蜥外泌肽-4肽类似物
AR091477A1 (es) 2012-06-21 2015-02-04 Univ Indiana Res & Tech Corp Analogos de glucagon que presentan actividad de receptor de gip
BR112015001451B1 (pt) 2012-07-23 2022-03-29 Zealand Pharma A/S Composto, construção de ácido nucleico, vetor de expressão, célula hospedeira, composição farmacêutica, utilização de um composto ou de seu sal ou solvato farmaceuticamente aceitável
TWI608013B (zh) 2012-09-17 2017-12-11 西蘭製藥公司 升糖素類似物
UA116217C2 (uk) 2012-10-09 2018-02-26 Санофі Пептидна сполука як подвійний агоніст рецепторів glp1-1 та глюкагону
CN104968674A (zh) * 2012-12-19 2015-10-07 诺和诺德股份有限公司 具有胆固醇流出活性的新颖的glp-1受体激动剂
JP2016506401A (ja) 2012-12-21 2016-03-03 サノフイ 二重glp1/gipまたは三方glp1/gip/グルカゴンアゴニストとしてのエキセンジン−4誘導体
AU2014234400B2 (en) 2013-03-21 2017-11-16 Sanofi-Aventis Deutschland Gmbh Synthesis of hydantoin containing peptide products
AU2014234314B2 (en) 2013-03-21 2018-02-15 Sanofi-Aventis Deutschland Gmbh Synthesis of cyclic imide containing peptide products
WO2014161835A1 (en) 2013-04-03 2014-10-09 Sanofi Modified blood glucose regulating proteins with altered pharmacological activity profile and preparation thereof
JP6822839B2 (ja) * 2013-09-13 2021-01-27 ザ・スクリップス・リサーチ・インスティテュート 修飾された治療剤、及びその組成物
US9988429B2 (en) 2013-10-17 2018-06-05 Zealand Pharma A/S Glucagon analogues
BR112016008115B1 (pt) 2013-10-17 2024-03-12 Boehringer Ingelheim International Gmbh Análogos de glucagon acilados
CN105849122B (zh) 2013-11-06 2021-04-30 西兰制药公司 Gip-glp-1双重激动剂化合物及方法
AU2014345570B2 (en) 2013-11-06 2019-01-24 Zealand Pharma A/S Glucagon-GLP-1-GIP triple agonist compounds
WO2015086728A1 (en) 2013-12-13 2015-06-18 Sanofi Exendin-4 peptide analogues as dual glp-1/gip receptor agonists
TW201609799A (zh) 2013-12-13 2016-03-16 賽諾菲公司 雙重glp-1/gip受體促效劑
WO2015086731A1 (en) 2013-12-13 2015-06-18 Sanofi Exendin-4 peptide analogues as dual glp-1/glucagon receptor agonists
WO2015086733A1 (en) 2013-12-13 2015-06-18 Sanofi Dual glp-1/glucagon receptor agonists
TW201609796A (zh) 2013-12-13 2016-03-16 賽諾菲公司 非醯化之艾塞那肽-4(exendin-4)胜肽類似物
EP3082797A4 (en) 2013-12-18 2017-12-13 The California Institute for Biomedical Research Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
EP3107560A1 (en) 2014-02-18 2016-12-28 Novo Nordisk A/S Stable glucagon analogues and use for treatment of hypoglycaemia
TW201625669A (zh) 2014-04-07 2016-07-16 賽諾菲公司 衍生自艾塞那肽-4(Exendin-4)之肽類雙重GLP-1/升糖素受體促效劑
TW201625670A (zh) 2014-04-07 2016-07-16 賽諾菲公司 衍生自exendin-4之雙重glp-1/升糖素受體促效劑
TW201625668A (zh) 2014-04-07 2016-07-16 賽諾菲公司 作為胜肽性雙重glp-1/昇糖素受體激動劑之艾塞那肽-4衍生物
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
CN104926934B (zh) * 2014-09-23 2016-11-09 蒋先兴 胃泌酸调节素类似物
MA55068A (fr) 2014-10-24 2022-01-05 Merck Sharp & Dohme Co-agonistes des récepteurs du glucagon et du glp-1
AU2015340586B2 (en) 2014-10-29 2020-04-30 Zealand Pharma A/S GIP agonist compounds and methods
PL3283507T3 (pl) 2015-04-16 2020-05-18 Zealand Pharma A/S Acylowany analog glukagonu
AR105319A1 (es) 2015-06-05 2017-09-27 Sanofi Sa Profármacos que comprenden un conjugado agonista dual de glp-1 / glucagón conector ácido hialurónico
WO2016198628A1 (en) 2015-06-12 2016-12-15 Sanofi Non-acylated exendin-4 derivatives as dual glp-1/glucagon receptor agonists
WO2016198624A1 (en) 2015-06-12 2016-12-15 Sanofi Exendin-4 derivatives as trigonal glp-1/glucagon/gip receptor agonists
AR105284A1 (es) 2015-07-10 2017-09-20 Sanofi Sa Derivados de exendina-4 como agonistas peptídicos duales específicos de los receptores de glp-1 / glucagón
US10800826B2 (en) 2015-10-05 2020-10-13 Merck Sharp & Dohme Corp. Antibody peptide conjugates that have agonist activity at both the glucagon and glucagon-like peptide 1 receptors
EP3359194A4 (en) 2015-10-06 2019-04-24 Merck Sharp & Dohme Corp. ANTIBODY-ACTIVE CONJUGATE FOR ANTI-INFLAMMATORY APPLICATIONS
US10493125B2 (en) 2015-12-09 2019-12-03 Merck Sharp & Dohme Corp. Co-agonists of the glucagon and GLP-1 receptors
WO2017132103A2 (en) 2016-01-29 2017-08-03 Merck Sharp & Dohme Corp. Phosphonate linkers and their use to facilitate cellular retention of compounds
AR110299A1 (es) 2016-12-02 2019-03-13 Sanofi Sa Conjugados que comprenden un agonista dual de glp-1 / glucagón, un conector y ácido hialurónico
PE20191083A1 (es) 2016-12-09 2019-08-20 Zealand Pharma As Agonistas duales acilados de glp-1/glp-2
CN108299553B (zh) * 2017-01-13 2021-07-16 博瑞生物医药(苏州)股份有限公司 胃泌酸调节素修饰物
CN106986924A (zh) * 2017-03-23 2017-07-28 中国药科大学 胃泌酸调节素(oxm)类似物及其应用
BR112020003319A2 (pt) * 2017-08-16 2020-08-25 Dong-A St Co., Ltd. análogo de peptídeo de oxintomodulina acilada
WO2019060660A1 (en) 2017-09-25 2019-03-28 Merck Sharp & Dohme Corp. CO-AGONISTS WITH EXTENDED ACTION OF GLUCAGON AND GLP-1 RECEPTORS
CN115304666A (zh) * 2017-11-24 2022-11-08 浙江道尔生物科技有限公司 一种治疗代谢疾病的胰高血糖素类似物
CN115109166A (zh) * 2017-11-24 2022-09-27 浙江道尔生物科技有限公司 一种治疗代谢疾病的多结构域活性蛋白
CN111171135B (zh) * 2018-11-12 2023-04-07 天津药物研究院有限公司 具有双重受体激动作用的胰高血糖素衍生肽及其用途
JP2022516439A (ja) 2018-12-21 2022-02-28 ジエンス ヘンルイ メデイシンカンパニー リミテッド 二特異性タンパク質
US11471512B2 (en) 2019-03-01 2022-10-18 Merck Sharp & Dohme Llc Pharmaceutical compositions of a peptide
CN110684082B (zh) * 2019-10-08 2021-12-10 江苏诺泰澳赛诺生物制药股份有限公司 Gip和glp-1双激动多肽化合物及药学上可接受的盐与用途
EP3842449A1 (en) 2019-12-23 2021-06-30 Merck Sharp & Dohme Corp. Stapled olefin co-agonists of the glucagon and glp-1 receptors
EP3842061A1 (en) 2019-12-23 2021-06-30 Merck Sharp & Dohme Corp. Stapled triazole co-agonists of the glucagon and glp-1 receptors
EP3842060A1 (en) 2019-12-23 2021-06-30 Merck Sharp & Dohme Corp. Stapled lactam co-agonists of the glucagon and glp-1 receptors
WO2021227989A1 (zh) * 2020-05-09 2021-11-18 天津药物研究院有限公司 具有双受体激动作用的多肽衍生物及其用途
CN112898405B (zh) * 2021-01-22 2023-02-28 深圳市图微安创科技开发有限公司 多肽化合物及其在预防或治疗糖尿病或糖尿病并发症中的应用

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4275152A (en) * 1977-02-03 1981-06-23 Eastman Kodak Company Hydrolysis of protein-bound cholesterol esters
WO2003020201A2 (en) * 2001-08-28 2003-03-13 Eli Lilly And Company Pre-mixes of glp-1 and basal insulin
AU2002353784B2 (en) 2001-09-24 2008-04-10 Oregon Health And Science University Assessment of neurons in the arcuate nucleus to screen for agents that modify feeding behavior
WO2003057235A2 (en) * 2002-01-10 2003-07-17 Imperial College Innovations Ltd Modification of feeding behavior
EP1575490A4 (en) * 2002-06-04 2007-08-08 Lilly Co Eli MODIFIED ANALOGUES OF GLUCAGON-LIKE PEPTIDE-1 (GLP-1)
US7364875B2 (en) * 2003-10-30 2008-04-29 Cresent Innovations, Inc. Method for producing medical and commercial grade poly-gamma-glutamic acid of high molecular weight
US20080318837A1 (en) * 2003-12-26 2008-12-25 Nastech Pharmaceutical Company Inc. Pharmaceutical Formation For Increased Epithelial Permeability of Glucose-Regulating Peptide
JP2008543816A (ja) * 2005-06-13 2008-12-04 インペリアル イノベーションズ リミテッド 新規化合物および該化合物が摂食行動に及ぼす効果
CA2638800A1 (en) 2006-02-22 2007-09-07 Merck & Co., Inc. Oxyntomodulin derivatives
WO2008030558A2 (en) * 2006-09-08 2008-03-13 Ambrx, Inc. Modified human plasma polypeptide or fc scaffolds and their uses
JP5231732B2 (ja) 2006-10-26 2013-07-10 株式会社東芝 冷却装置、およびこれを備えた電子機器
NZ578948A (en) * 2007-02-15 2012-03-30 Univ Indiana Res & Tech Corp Glucagon/glp-1 receptor co-agonists
MX2010013454A (es) 2008-06-17 2011-01-21 Tion Indiana University Res And Technology Corpora Co-agonistas de receptor de glucagon/glp-1.
SG172291A1 (en) 2008-12-19 2011-07-28 Univ Indiana Res & Tech Corp Amide based glucagon superfamily peptide prodrugs
WO2010096052A1 (en) 2009-02-19 2010-08-26 Merck Sharp & Dohme Corp. Oxyntomodulin analogs

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8969294B2 (en) 2008-06-17 2015-03-03 Istituto Di Recerche Di Biologia Molecolare P. Angeletti S.R.L. Glucagon/GLP-1 receptor co-agonists
US9790263B2 (en) 2009-06-16 2017-10-17 Indiana University Research And Technology Corporation GIP receptor-active glucagon compounds
US9765131B2 (en) 2011-06-10 2017-09-19 Hanmi Science Co., Ltd. Oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
US10442848B2 (en) 2011-06-10 2019-10-15 Hanmi Science Co., Ltd. Oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
US11872283B2 (en) 2011-06-17 2024-01-16 Hanmi Science Co., Ltd Conjugate comprising oxyntomodulin and an immunoglobulin fragment, and use thereof
US9758562B2 (en) * 2011-06-22 2017-09-12 Indiana University and Technology Corporation Glucagon/GLP-1 receptor co-agonists
US10174093B2 (en) 2011-06-22 2019-01-08 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
US10730923B2 (en) 2011-06-22 2020-08-04 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
US20120329708A1 (en) * 2011-06-22 2012-12-27 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
US9156902B2 (en) * 2011-06-22 2015-10-13 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
US20150376256A1 (en) * 2011-06-22 2015-12-31 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
US9901621B2 (en) 2012-07-25 2018-02-27 Hanmi Pharm. Co., Ltd. Composition for treating hyperlipidemia comprising oxyntomodulin derivative
US10493132B2 (en) 2012-07-25 2019-12-03 Hanmi Pharm. Co., Ltd. Composition for treating hyperlipidemia comprising oxyntomodulin derivative
WO2014049610A2 (en) 2012-09-26 2014-04-03 Cadila Healthcare Limited Peptides as gip, glp-1 and glucagon receptors triple-agonist
EP3679944A1 (en) * 2012-11-06 2020-07-15 Hanmi Pharm. Co., Ltd. A composition for treating diabetes comprising oxyntomodulin analog
US11071785B2 (en) 2012-11-06 2021-07-27 Hanmi Pharm. Co., Ltd. Liquid formulation of long-lasting protein conjugate comprising the oxyntomodulin and an immunoglobulin Fc region
US10279041B2 (en) 2012-11-06 2019-05-07 Hanmi Pharm Co. Ltd. Liquid formulation of long-lasting protein conjugate comprising the oxyntomodulin and an immunoglobulin fragment
EP2916860A4 (en) * 2012-11-06 2016-06-22 Hanmi Pharm Ind Co Ltd COMPOSITION FOR THE TREATMENT OF DIABETES OR DIABESITY COMPRISING AN OXYNTOMODULIN ANALOGUE
US9724420B2 (en) 2012-11-06 2017-08-08 Hanmi Pharm. Co., Ltd. Liquid formulation of protein conjugate comprising an oxyntomodulin derivative covalently linked to a non-peptidyl polymer to an immunoglobulin FC region
US10550168B2 (en) 2012-11-06 2020-02-04 Hanmi Pharm. Co., Ltd. Composition for treating diabetes or diabesity comprising oxyntomodulin analog
US10435459B2 (en) 2014-09-16 2019-10-08 Hanmi Pharm. Co., Ltd. Use of a long acting GLP-1/glucagon receptor dual agonist for the treatment of non-alcoholic fatty liver disease
US10233230B2 (en) 2014-09-16 2019-03-19 Hanmi Pharm. Co., Ltd. Use of a long acting GLP-1/glucagon receptor dual agonist for the treatment of non-alcoholic fatty liver disease
US12018060B2 (en) 2014-12-30 2024-06-25 Hanmi Pharm Co., Ltd. Glucagon derivatives
US10513550B2 (en) 2014-12-30 2019-12-24 Hanmi Pharm Co., Ltd Glucagon derivatives
US11135271B2 (en) 2014-12-30 2021-10-05 Hanmi Pharm. Co., Ltd. Glucagon derivatives with improved stability
US11254724B2 (en) 2014-12-30 2022-02-22 Hanmi Pharm. Co., Ltd. Glucagon derivatives
US10696725B2 (en) 2015-06-30 2020-06-30 Hanmi Pharm. Co., Ltd. Glucagon derivative and a composition comprising a long acting conjugate of the same
US11261227B2 (en) 2015-06-30 2022-03-01 Hanmi Pharm. Co., Ltd. Glucagon derivative and a composition comprising a long acting conjugate of the same
US11667688B2 (en) 2015-06-30 2023-06-06 Hanmi Pharm. Co., Ltd. Glucagon derivative and a composition comprising a long acting conjugate of the same
US11492385B2 (en) 2015-10-27 2022-11-08 Merck Sharp & Dohme Llc Long-acting co-agonists of the glucagon and GLP-1 receptors
US10793615B2 (en) 2015-10-27 2020-10-06 Merck Sharp & Dohme Corp. Long-acting co-agonists of the glucagon and GLP-1 receptors
WO2017074798A3 (en) * 2015-10-27 2017-05-26 Merck Sharp & Dohme Corp. Long-acting co-agonists of the glucagon and glp-1 receptors
US10981967B2 (en) 2015-12-31 2021-04-20 Hanmi Pharm. Co., Ltd. Long-acting conjugate of triple glucagon/GLP-1/GIP receptor agonist
US11332508B2 (en) 2015-12-31 2022-05-17 Hanmi Pharm. Co., Ltd. Triple glucagon/GLP-1/GIP receptor agonist
US11142559B2 (en) 2016-06-29 2021-10-12 Hanmi Pharm. Co., Ltd. Glucagon derivative, conjugate thereof, composition comprising same, and therapeutic use thereof

Also Published As

Publication number Publication date
BRPI0924307A8 (pt) 2017-12-12
SI2398483T1 (sl) 2015-01-30
ZA201106051B (en) 2014-07-30
HK1165333A1 (en) 2012-10-05
EP2398483B1 (en) 2014-09-24
CA2754350C (en) 2019-07-09
CY1116917T1 (el) 2017-04-05
AU2009340439B2 (en) 2015-07-16
PL2398483T3 (pl) 2015-03-31
IL214729A (en) 2016-06-30
RU2542362C2 (ru) 2015-02-20
SG173791A1 (en) 2011-10-28
EP2398483A4 (en) 2013-03-06
CN102573872A (zh) 2012-07-11
CN102573872B (zh) 2016-03-23
CA2754350A1 (en) 2010-08-26
ES2524477T3 (es) 2014-12-09
IL214729A0 (en) 2011-11-30
JP2012518035A (ja) 2012-08-09
KR20120068755A (ko) 2012-06-27
DK2398483T3 (da) 2014-10-27
MX2011008717A (es) 2012-09-28
CL2011002036A1 (es) 2012-02-03
EP2398483A1 (en) 2011-12-28
WO2010096142A1 (en) 2010-08-26
BRPI0924307B1 (pt) 2020-05-05
RU2011134596A (ru) 2013-03-27
HRP20141215T1 (hr) 2015-02-13
JP5756961B2 (ja) 2015-07-29
KR101634139B1 (ko) 2016-06-28
PT2398483E (pt) 2014-12-04
PE20120621A1 (es) 2012-06-08
US20150307580A1 (en) 2015-10-29
WO2010096052A1 (en) 2010-08-26
AU2009340439A1 (en) 2011-09-15
BRPI0924307A2 (pl) 2017-08-22
US9593155B2 (en) 2017-03-14
BRPI0924307B8 (pt) 2021-05-25

Similar Documents

Publication Publication Date Title
US9593155B2 (en) Oxyntomodulin analogs
EP1999143B1 (en) Neuromedin u receptor agonists and uses thereof
US20110301079A1 (en) Neuromedin u receptor agonists and uses thereof
US20090104210A1 (en) Peptide compounds for treating obesity and insulin resistance
WO2011005611A1 (en) Neuromedin u receptor agonists and uses thereof
US11773150B2 (en) Antibody peptide conjugates that have agonist activity at both the glucagon and glucagon-like peptide 1 receptors
WO2010138343A1 (en) Neuromedin u receptor agonists
BRPI1001307B1 (pt) injeção de oxigênio em bacia de flotação do processo de remoção de substâncias poluentes em cursos d'água
US20100143383A1 (en) Oxgr1 agonists for use in therapies for metabolic disorders
CN104684907A (zh) 抗糖尿病的三环化合物

Legal Events

Date Code Title Description
AS Assignment

Owner name: INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DIMARCHI, RICHARD;WARD, BRIAN;REEL/FRAME:027806/0676

Effective date: 20120225

AS Assignment

Owner name: ISTITUTO DI RICERCHE DI BIOLOGIA P. ANGELETTI S.R.

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BIANCHI, ELISABETTA;INGALLINELLA, PAOLO;PESSI, ANTONELLO;AND OTHERS;SIGNING DATES FROM 20120120 TO 20120130;REEL/FRAME:027948/0413

Owner name: MERCK SHARP & DOHME CORP., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EIERMANN, GEORGE J.;METZGER, JOSEPH J.;POCAI, ALESSANDRO;AND OTHERS;SIGNING DATES FROM 20111208 TO 20111221;REEL/FRAME:027947/0481

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE

AS Assignment

Owner name: MSD ITALIA S.R.L., ITALY

Free format text: MERGER AND CHANGE OF NAME;ASSIGNORS:ISTITUTO DI RECERCHE DI BIOLOGIA MOLECOLARE P. ANGELETTI S.R.L.;MSD ITALIA S.R.L.;REEL/FRAME:038663/0032

Effective date: 20121113