US20160108098A1 - Therapeutic peptides - Google Patents

Therapeutic peptides Download PDF

Info

Publication number
US20160108098A1
US20160108098A1 US14/888,085 US201414888085A US2016108098A1 US 20160108098 A1 US20160108098 A1 US 20160108098A1 US 201414888085 A US201414888085 A US 201414888085A US 2016108098 A1 US2016108098 A1 US 2016108098A1
Authority
US
United States
Prior art keywords
seq
argtyr
xaa
polypeptide
amu
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
US14/888,085
Other languages
English (en)
Inventor
Steven Thomas Dock
Yulin Wu
Ved P Srivastava
Robert Neil Hunter, III
Andrew James Carpenter
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.)
GlaxoSmithKline Intellectual Property Development Ltd
Original Assignee
GlaxoSmithKline Intellectual Property Development Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by GlaxoSmithKline Intellectual Property Development Ltd filed Critical GlaxoSmithKline Intellectual Property Development Ltd
Priority to US14/888,085 priority Critical patent/US20160108098A1/en
Assigned to GLAXOSMITHKLLNE INTELLECTUAL PROPERTY (NO.2) LIMITED reassignment GLAXOSMITHKLLNE INTELLECTUAL PROPERTY (NO.2) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SRIVASTAVA, Ved P., CARPENTER, ANDREW JAMES, DOCK, STEVEN THOMAS, HUNTER, III, ROBERT NEIL, WU, YULIN
Assigned to GLAXOSMITHKLINE INTELLECTUAL PROPERTY DEVEOLPMENT LIMITED reassignment GLAXOSMITHKLINE INTELLECTUAL PROPERTY DEVEOLPMENT LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED
Publication of US20160108098A1 publication Critical patent/US20160108098A1/en
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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K14/57545Neuropeptide Y
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones

Definitions

  • This invention relates to therapeutic peptides useful in the treatment of obesity and metabolic disorders. More specifically, the invention relates to novel analogs of Peptide YY (PYY) and their use.
  • PYY Peptide YY
  • PYY belongs to the PP-fold family of peptides together with pancreatic polypeptide and neuropeptide Y, which have a role in controlling appetite. See, for example, Schwartz et al. (2002) Nature: 418(6898):595-7. PYY is secreted as a 36 amino acid, straight chain polypeptide and then cleaved by dipeptidyl peptidase IV to produce PYY(3-36). Fasting and post-prandial concentrations of PYY in morbidly obese individuals after gastric bypass surgery are suggested as playing a role in their dramatic weight loss. See, for example, le Roux (2006) Ann Surg. 243(1):108-14.
  • PYY(3-36) Peripheral infusion of PYY(3-36) has been shown to increase energy expenditure and fat oxidation rates in obese and lean subjects. See, for example, Batterham et al. (2003) N Engl J Med. 349(10):941-8, and Sloth et al. (2007) Am J Physiol Endocrinol Metab.: 293(2):E604-9.
  • Administration of a PYY(3-36) nasal spray reduced daily caloric intake of obese individuals by 2713 kJ, resulting in a weight loss of 0.6 kg over a six-day study period. See, for example, Gantz et al. (2007) J Clin Endocrinol Metab. 92(5):1754-7.
  • the present invention relates to novel analogs of PYY that have an improved therapeutic profile when compared to native human PYY. These novel PYY analogs are useful in the treatment of obesity, diabetes, and other disorders.
  • the invention provides a polypeptide comprising the amino acid sequence:
  • Xaa 1 is Ala, His, or Ser
  • Xaa 2 is Glu or Lys
  • Xaa 3 is Pro or Ala
  • Xaa 4 is Leu or Trp
  • Xaa 5 is Asn, Ala, or Thr;
  • Xaa 6 is Arg or Lys
  • Xaa 7 is Ser, Asp, or Ala
  • Xaa 8 is His or Lys
  • Xaa 9 is Leu or lie
  • Xaa 10 is Val or Leu.
  • the invention provides a polypeptide selected from the group consisting of:
  • the invention provides a nucleic acid molecule encoding a polypeptide of the invention
  • the invention includes an expression vector comprising a nucleic acid molecule encoding a polypeptide of the invention.
  • the invention encompasses a host cell containing an expression vector comprising a nucleic acid molecule encoding a polypeptide of the invention.
  • the invention provides a pharmaceutical combination comprising a novel PYY polypeptide of the invention and exendin-4.
  • the invention provides a pharmaceutical combination comprising a novel PYY polypeptide of the invention and GLP-1.
  • the invention provides a pharmaceutical composition comprising a novel PYY polypeptide of the invention and one or more pharmaceutically acceptable excipients.
  • the invention encompasses a method of treating a metabolic disorder or obesity, the method comprising administering a novel PYY polypeptide or pharmaceutical combination of the invention to a subject in need thereof.
  • the invention also provides the use of a polypeptide or pharmaceutical combination of the invention in the preparation of a medicament for use in the treatment of obesity.
  • the invention provides a polypeptide or pharmaceutical combination of the invention for use in the treatment of a metabolic disorder or obesity.
  • FIG. 1 shows the effects of the peptide shown in Example 5 (Analog #5), PYY(3-36)NH 2 (PYY3-36), and exendin-4 singly and in combination on changes in body weight in diet-induced obese (DIO) Long Evans (LE) rats.
  • FIG. 2 shows the effects of the peptide shown in Example 5 (Analog #5), PYY(3-36)NH 2 (PYY3-36), and exendin-4 singly and in combination on body composition changes in DIO LE rats.
  • the invention provides novel analogs of PYY that have an improved therapeutic profile when compared to native human PYY.
  • the novel PYY analogs of the invention show improved effects on food intake when compared with the native PYY sequence.
  • novel PYY analogs comprise the amino acid sequence:
  • Xaa 1 is Ala, His, or Ser
  • Xaa 2 is Glu or Lys
  • Xaa 3 is Pro or Ala
  • Xaa 4 is Leu or Trp
  • Xaa 5 is Asn, Ala, or Thr;
  • Xaa 6 is Arg or Lys
  • Xaa 7 is Ser, Asp, or Ala
  • Xaa 8 is His or Lys
  • Xaa 9 is Leu or lie
  • Xaa 10 is Val or Leu.
  • novel polypeptides of the invention show a statistically significant increase in the reduction of food intake in either a lean and/or diet-induced obesity animal model when compared with human PYY(3-36).
  • the polypeptides of the invention reduce the intake of food in a lean and/or diet-induced obesity animal model by at least 20%, at least 30%, or at least 40%. More preferably, the polypeptides reduce the intake of food in a lean and/or diet-induced obesity animal model by at least 50%.
  • the invention provides a polypeptide selected from the group consisting of:
  • polypeptides of the invention may have either a carboxamide or carboxylic acid at the end of the amino acid chain.
  • the invention encompasses salts of the recited polypeptides, including pharmaceutically acceptable salts.
  • salts include, but are not limited to, including inorganic and organic acids and bases, including but not limited to, sulfuric, citric, maleic, acetic, oxalic, hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfite, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (
  • salts formed with free amino groups such as, for example, hydrochloric, phosphoric, acetic, trifluoroacetic, oxalic, and tartaric acids.
  • salts that may form with free carboxy groups such as, for example sodium, potassium, ammonium, sodium, lithium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, and procaine salts.
  • polypeptides of the invention may be prepared using standard recombinant expression or chemical peptide synthesis techniques known in the art. See, for example, Chan, Weng C., and Peter D. White, eds. Fmoc Solid Phase Peptide Synthesis: A Practical Approach . New York: Oxford UP, 2000, and Howl, John, ed. Peptide Synthesis and Applications ( Methods in Molecular Biology ). Totowa, N.J.: Humana, 2005.
  • compositions and pharmaceutical combinations of the invention are useful for the treatment of metabolic disorders including, for example, hyperglycemia, impaired glucose tolerance, beta cell deficiency, diabetes (including type 1 diabetes, type 2 diabetes, and gestational diabetes), non-alcoholic steatotic liver disease, steatosis of the liver, polycystic ovarian syndrome, hyperlipidemia, and Metabolic Syndrome.
  • the compositions and pharmaceutical combinations may be used for treating obesity or diseases characterized by overeating and for the suppression of appetite.
  • the methods comprise administering to a subject a therapeutically effective amount of a composition of the invention to a subject in need thereof, preferably a human subject.
  • compositions and combinations of the invention include, but are not limited to, insulin resistance, insulin deficiency, hyperinsulinemia, hyperglycemia, dyslipidemia, hyperlipidemia, hyperketonemia, hyperglucagonemia, pancreatitis, pancreatic neoplasms, cardiovascular disease, hypertension, coronary artery disease, atherosclerosis, renal failure, neuropathy (e.g., autonomic neuropathy, parasympathetic neuropathy, and polyneuropathy), diabetic retinopathy, cataracts, endocrine disorders, and sleep apnea, polycystic ovarian syndrome, neoplasms of the breast, colon, prostate, rectum and ovarian, osteoarthritis steatosis of the liver.
  • neuropathy e.g., autonomic neuropathy, parasympathetic neuropathy, and polyneuropathy
  • diabetic retinopathy cataracts
  • endocrine disorders and sleep apnea
  • polycystic ovarian syndrome
  • the invention further encompasses methods of regulating insulin responsiveness in a patient, as well as methods of increasing glucose uptake by a cell, and methods of regulating insulin sensitivity of a cell, using the conjugates or fusions of the invention. Also provided are methods of stimulating insulin synthesis and release, enhancing adipose, muscle or liver tissue sensitivity towards insulin uptake, stimulating glucose uptake, slowing digestive process, slowing of gastric emptying, inhibition of gastric acid secretion, inhibition of pancreatic enzyme secretion, reducing appetite, inhibition of food intake, modifying energy expenditure, or blocking the secretion of glucagon in a patient, comprising administering to said patient a composition of the invention e.g. comprising administering at least one dose of a composition e.g. a pharmaceutical composition or pharmaceutical combination of the present invention.
  • a composition of the invention e.g. comprising administering at least one dose of a composition e.g. a pharmaceutical composition or pharmaceutical combination of the present invention.
  • the invention also provides for use of a composition of the invention in the manufacture of a medicament for treatment of a metabolic disease such as those described herein.
  • the invention also relates to use of any of the compositions described herein for use in therapy.
  • polypeptides of the present invention and their salts may be employed alone or in combination with other therapeutic agents (a “pharmaceutical combination”) for the treatment of the above-mentioned conditions.
  • the polypeptide of the present invention and the additional therapeutic agent or agents are administered together, while in other embodiments, the polypeptide of the invention and the additional therapeutic agent or agents are administered separately. When administered separately, administration may occur simultaneously or sequentially, in any order.
  • the amounts of the polypeptides(s) of the present invention and the other therapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the administration in combination of a compound of the present invention with other treatment agents may be in combination by administration concomitantly in: (1) a unitary pharmaceutical composition including both therapeutic agents; or (2) separate pharmaceutical compositions each including one of the therapeutic agents.
  • the combination may be administered separately in a sequential manner wherein one treatment agent is administered first and the other second or vice versa. Such sequential administration may be close in time or remote in time.
  • the pharmaceutical combinations of the invention include a polypeptide according to the invention and an exendin-4 peptide (see, for example, U.S. Pat. No. 5,424,286) or a fragment or analog thereof.
  • Exendin-4 (Ex-4) and analogs thereof that are useful for the present invention include Byetta® and Bydureon® (exenatide), Victoza® (liraglutide), lixisenatide, LY2189265 (dulaglutide), PF-4856883, ZYD-1, and HM11260C (LAPS exendin) as well as those described in PCT patent publications WO 99/25728 (Beeley et al.), WO 99/25727 (Beeley et al.), WO 98/05351 (Young at al.), WO 99/40788 (Young et al.), WO 99/07404 (Beeley at al.), and WO 99/43708 (Knudsen et al.).
  • the pharmaceutical combinations of the invention include a polypeptide according to the invention and GLP-1 (see, for example, Gutniak, M., et al. (1992) N. Engl. J. Bled. 326:1316-22), or a fragment or analog thereof, for example, GLP-1 (7-37), GLP-1(7-36), GLP-1 (7-35), GLP-1(7-38), GLP-1(7-39), GLP-1(7-40), GLP-1(7-41).
  • GLP-1 see, for example, Gutniak, M., et al. (1992) N. Engl. J. Bled. 326:1316-22
  • GLP-1 see, for example, Gutniak, M., et al. (1992) N. Engl. J. Bled. 326:1316-22
  • GLP-1 see, for example, Gutniak, M., et al. (1992) N. Engl. J. Bled. 326:1316-22
  • GLP-1
  • GLP-1 analogues are described in International Patent Application No. 90/11299, which relates to peptide fragments which comprise GLP-1 (7-36) and functional derivatives thereof and have an insulinotropic activity which exceeds the insulinotropic activity of GLP-1(1-36) or GLP-1(1-37) and to their use as insulinotropic agents (incorporated herein by reference, particularly by way of examples of drugs for use in the present invention).
  • the pharmaceutical combinations of the invention also include a polypeptide according to the invention and albiglutide.
  • the pharmaceutical combinations include a polypeptide according to the invention and an enhancer of GLP-1 action such as a DPP-IV inhibitor (e.g. sitagliptin and/or saxagliptin).
  • a DPP-IV inhibitor e.g. sitagliptin and/or saxagliptin.
  • the pharmaceutical combination comprises a PYY analog of the present invention and one or more therapeutic agents that are direct or indirect stimulators of GLP-1 secretion such as metformin, bile acid sequestrants (e.g. colestipol, cholestryramine, and/or colesevelam), ileal bile acid transport (iBAT) Inhibitors (e.g. ALBI-3309, AZD-7806, S-8921, SAR-58304, or those described in US20130029938), and SGLT-1 Inhibitors (e.g. DSP-3235 and/or LX-4211).
  • bile acid sequestrants e.g. colestipol, cholestryramine, and/or colesevelam
  • ileal bile acid transport (iBAT) Inhibitors e.g. ALBI-3309, AZD-7806, S-8921, SAR-58304, or those described in US20130029938
  • SGLT-1 Inhibitors
  • the invention provides for methods of treatment where a “therapeutically effective amount” of a polypeptide of the invention is administered to a subject in need of such treatment.
  • a “therapeutically effective amount” means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • an effective amount of therapeutic agent will vary with many factors including the age and weight of the patient, the patient's physical condition, the blood sugar level, the weight level to be obtained, and other factors
  • a therapeutically effective amount of a polypeptide of the present invention is the amount required to suppress appetite in the subject to a desired degree.
  • the effective daily appetite-suppressing dose of the compounds will typically be in the range of about 0.01 ⁇ g to about 500 ⁇ g/day, preferably about 0.05 ⁇ g to about 100 ⁇ g/day and more preferably about 1 ⁇ g to about 50 ⁇ g/day, most preferably about 5 ⁇ g to about 25 ⁇ g/day, for a 70 kg patient, administered in a single or divided doses.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide of the invention, and a pharmaceutically acceptable carrier, excipient or diluent.
  • compositions and pharmaceutical combinations of the invention can be administered by any route, including intravenously, intraperitoneal, subcutaneous, and intramuscular, orally, topically, transmucosally, or by pulmonary inhalation.
  • polypeptides of the invention can be provided in the form of formulations suitable for parenteral (including intravenous, intramuscular and subcutaneous), nasal or oral administration.
  • the invention encompasses a slow release formulation.
  • Such formulations allow for therapeutically effective amounts of the therapeutic polypeptide or polypeptides to be delivered into the bloodstream over many hours or days following injection or delivery to the subcutaneous space.
  • Slow release formulations of the invention may include one or more polymers useful in delaying the release of the therapeutic polypeptide.
  • Non-limiting examples of such polymers include poly(lactic-co-glycolic acid) PLGA, polycaprolactone, polydioxanone, polytrimethylene carbonate, polyanhydrides, PEG-PLGA, polyglutamic acid, polyethylene glycol terphthalate/polybutylene terphthalate/polybutylene terphthalate, poly(aminoacid)-Leu/Glu copolymer, polytyrosine carbonates, polyesteramides, poly(alpha aminoacid) based polymeric micelles, polyhydroxypropylmethacrylamide, polyalkylcyanoacrylate, collagen, hyaluronic acide, albumin, carboxymethylcellulose, fleximer, chitosan, maltodextrin, dextran, or dextran sulfate.
  • the polypeptides of the invention may be delivered via a miniature device such as an implantable infusion pump which is designed to provide long-term continuous or intermittent drug infusion.
  • a miniature device such as an implantable infusion pump which is designed to provide long-term continuous or intermittent drug infusion.
  • Such devices can be used to administer a therapeutic polypeptide of the invention via intravenous, intra-arterial, subcutaneous, intraperitoneal, intrathecal, epidural, or intraventricular routes.
  • Such devices may be erodible, non-erodible and/or durable.
  • Non-limiting examples of such devices include the DurasertTM device (pSivida), the DUROS® osmotic delivery system (Intarcia Therapeutics), MedLaunchTM Polymer Technology (Endo Health).
  • polypeptides of the invention may be administered in an in situ gel formulation.
  • Such formulations typically are administered as liquids which form a gel either by dissipation of the water miscible organic solvent or by aggregation of hydrophobic domains present in the matrix.
  • Non-limiting examples include the FLUID CRYSTAL technology (Camurus) and the SABER technology (Durect), and the formulations described in U.S. Pat. Nos. 5,612,051, 5,714,159, 6,413,539, 6,004,573, and 6,117,949.
  • the therapeutic polypeptides of the invention may also be encapsulated into a microsphere-based pharmaceutical formulation suitable for subcutaneous injection.
  • microsphere-based formulations for the delivery of peptides include ChronijectTM (Oakwood Labs), Medusa® (Flamel's), Q-Sphera (Q-CHIP), as well as those described in U.S. Pat. Nos. 4,675,189, 6,669,961, and Amin et al. (2001) J of Controlled Release 73: 49-57.
  • the formulation may contain antibacterial or antifungal agents such as meta-cresol, benzyl alcohol, parabens (methyl, propyl, butyl), chlorobutanol, phenol, phenylmercuric salts such as acetate, borate, or nitrate, or sorbic acid.
  • antibacterial or antifungal agents such as meta-cresol, benzyl alcohol, parabens (methyl, propyl, butyl), chlorobutanol, phenol, phenylmercuric salts such as acetate, borate, or nitrate, or sorbic acid.
  • compositions of this invention can be lyophilized for storage and reconstituted in a suitable carrier prior to use. Any suitable lyophilization method (e.g., spray drying, cake drying) and/or reconstitution techniques can be employed.
  • the invention provides a composition comprising a lyophilized (freeze dried) polypeptide as described herein.
  • the invention provides a nucleic acid encoding a polypeptide of the invention and recombinant expression vectors containing such nucleic acids.
  • Recombinant expression vectors of the invention include a nucleic acid encoding a polypeptide of the invention operably linked to one more expression control elements such as, e.g., a promoter.
  • Host cells containing a nucleic acid or recombinant expression vector encoding a polypeptide of the invention are also included.
  • Suitable host cells according to the invention include both prokaryotic host cells and eukaryotic hosts cells.
  • Possible host cells include, but are not limited to, mammalian host cells, bacterial host cells (e.g. E. coli ), yeast host cells, and plant host cells.
  • Nucleic acids and recombinant expression vectors encoding a polypeptide of the invention can be introduced into a suitable host cell to create a recombinant host cell using any method appropriate to the host cell selected, e.g., transformation, transfection, electroporation, or infection.
  • the nucleic acid or recombinant expression vector is integrated into the host cell genome.
  • the resulting recombinant host cell can be maintained under conditions suitable for expression (e.g., in the presence of an inducer, in a suitable animal, in suitable culture media supplemented with appropriate salts, growth factors, antibiotics, nutritional supplements, etc.), whereby the encoded polypeptide is produced. If desired, the encoded peptide or polypeptide can be isolated or recovered.
  • the invention further provides a method for producing a polypeptide of the present invention where the method comprises maintaining a host cell such as those described above that comprises a nucleic acid or recombinant expression vector that encodes a polypeptide of the invention under conditions suitable for expression of said nucleic acid or recombinant expression vector.
  • Methods for recombinant expression of polypeptides in host cells are well known in the art. See, for example, Rosalyn M. Bill, ed. Recombinant Protein Production in Yeast: Methods and Protocols ( Methods in Molecular Biology, Vol. 866), Humana Press 2012; James L. Hartley, ed.
  • nucleic acids of the invention are “isolated.” Nucleic acids referred to herein as “isolated” are nucleic acids which have been separated away from other material (e.g., other nucleic acids such as genomic DNA, cDNA and/or RNA) in its original environment (e.g., in cells or in a mixture of nucleic acids such as a library). An isolated nucleic acid can be isolated as part of a recombinant expression vector.
  • peptides shown in the following examples were synthesized by solid-phase methods using Fmoc strategy with 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate (HBTU) or 2-(6-chloro-1-H-benzotriazole-1-yl)-1,1,3,3-tetramethylaminium hexafluorophosphate (HCTU) activation (5 fold molar excess) in N,N-dimethylformamide (DMF), and N-methylmorpholine (NMM) as base, 20% piperidine/DMF for Fmoc deprotection, on an automated peptide synthesizer (model Prelude or Overture; Protein Technologies, Arlington, Ariz.).
  • HBTU 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyl uronium hexafluorophosphate
  • HCTU 2-(6-chloro-1-H
  • the resin was Rink Amide MBHA LL (Novabiochem) or Rink Amide AM LL (Novabiochem) with a loading of 0.29-0.38 mmol/g on a 20-400 ⁇ mol scale.
  • the side chain protection groups used were Trt for Asn, Gin, Cys and His; t-Bu for Ser, Thr, and Tyr; Boc for Lys and Trp; Ot-Bu for Asp and Glu; and Pbf for Arg.
  • Cleavage of peptide-resin was completed with a mixture of trifluoroacetic acid (TFA):anisole:water:triisopropylsilane (88:5:5:2).
  • the crude peptide was precipitated in cold diethyl ether, the diethyl ether was decanted and the solids triturated again with cold diethyl ether.
  • the crude solids were then purified by reverse phase HPLC on a Waters XBridgeTM BEH 130, C18, 10 ⁇ m, 130 ⁇ , 30 ⁇ 250 mm ID column, using a gradient within the ranges of 5-75% acetonitrile/water with 0.1% TFA over 30-45 minutes at a flow rate of 30 mL/min, ⁇ -215 nm.
  • Method A Performed using a Phenomenex UPLC AerisTM Peptide XB C18 column, 1.7 ⁇ m, 2.1 ⁇ 100 mm or ACQUITY BEH300 or BEH130 C18 column, 1.77 ⁇ m, 2.1 ⁇ 100 mm using 5-65% acetonitrile/water with 0.05% TFA over 30 minutes with a flow rate 0.5 mL/min, ⁇ -215 nm, 280 nm.
  • Method A Performed using a Waters XBridgeTM BEH130 C18 column, 5 ⁇ m, 4.6 ⁇ 250 mm, with 5-70% acetonitrile/water with 0.1% TFA over 15 minutes with a flow rate 1.5 mL/min, 40° C., ⁇ -215 nm, 280 nm.
  • Method B Performed using a Waters XBridgeTM BEH130 C18 column, 5 ⁇ m, 4.6 ⁇ 250 mm, 5-75% acetonitrile/water with 0.1% TFA over 20 minutes with a flow rate 1.5 mL/min, ⁇ -215 nm, 280 nm.
  • Method C Performed using a Waters XBridgeTM BEH130 C18 column, 5 ⁇ m, 4.6 ⁇ 250 mm, 20-37.5% acetonitrile/water with 0.1% TFA over 15 minutes with a flow rate 1.0 mL/min, 60° C., ⁇ -215 nm, 280 nm.
  • Method D Performed using a Waters XBridgeTM BEH300 C18 column, 5 ⁇ m, 4.6 ⁇ 250 mm, 5-70% acetonitrile/water with 0.1% TFA over 15 minutes with a flow rate 1.5 mL/min, ⁇ -215 nm, 280 nm.
  • Example 1 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1369 amu and (M+4)/4 ⁇ 1027 amu, which corresponds to a peptide with the parent molecular weight of 4105 amu (ESI-MS, LC/MS Method A).
  • Example 2 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1371 amu and (M+4)/4-1028 amu, which corresponds to a peptide with the parent molecular weight of 4111 amu (ESI-MS, LC/MS Method A).
  • Example 3 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1374 amu and (M+4)/4 ⁇ 1031 amu, which corresponds to a peptide with the parent molecular weight of 4120 amu (ESI-MS. LC/MS Method A).
  • Example 4 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1405 amu and (M+4)/4 ⁇ 1054 amu, which corresponds to a peptide with the parent molecular weight of 4212 amu (ESI-MS, LC/MS Method A).
  • Example 5 was prepared on a 6 ⁇ 50 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1407 amu and (M+4)/4 ⁇ 1056 amu, which corresponds to a peptide with the parent molecular weight of 4221 amu (ESI-MS, LC/MS Method A).
  • Example 5 was prepared via manual synthesis using a 250 mL jacketed reactor that was cooled to 15° C.
  • Rink Amide AM Resin LL (100-200 mesh, 13.8 g, 0.29 mmol/g loading) was swelled with DMF (50 mL) 3 times for 10 min each with nitrogen sparge.
  • the Fmoc group was removed with 20% piperidine in DMF (200 mL) over 5 min with nitrogen sparge, followed by 20% piperidine in DMF (200 mL) over 12 min with nitrogen sparge.
  • the resin was then washed with DMF (100 mL) and then twice with DMF (100 mL) for 1 min with nitrogen sparge.
  • the first amino acid (100 mL, 200 mM solution in DMF) was added, followed by DIPEA solution (50 mL, 800 mM solution in DMF).
  • DIPEA solution 50 mL, 800 mM solution in DMF.
  • a solution of HCTU in DMF 50 mL, 400 mM was added over a 12 min period via peristaltic pump. After a minimum of 15 min, a Kaiser test on an aliquot of resin was performed to ensure complete reaction. The resin was then washed with DMF (100 mL) and then twice with DMF (100 mL) for 1 min with nitrogen sparge.
  • the DMF was drained off and the final N-terminal Fmoc was removed with 20% piperidine in DMF (150 mL) over 5 min with nitrogen sparge, followed by 20% piperidine in DMF (150 mL) over 12 min with nitrogen sparge.
  • the resin was then washed with DMF (100 mL) and then twice with DMF (100 mL) for 1 min with nitrogen sparge, then washed three time with DCM (120 mL) for 5 min with nitrogen sparge, and then finally three times with methanol (120 mL) for 5 min with nitrogen sparge.
  • the resin was dried with nitrogen sparge for 30 min.
  • Cleavage of peptide from the resin was performed using 100-120 mL of cleavage cocktail: TFA:phenol:DODT:water:TIPS (90:2.5:2.5:2.5:2.5) for 2.5-3 h.
  • the filtrates were split into vessels and treated with cold diethyl ether.
  • the vessels were centrifuged for 10 min at 3000 RPM and the supernatant was poured off.
  • the material was treated with cold diethyl ether again, shaken and then centrifuged for another 10 min at 3000 RPM. The supernatant was poured off again.
  • the solids from the vessels were combined using 0.1% aqueous TFA and lyophilized to give batch 1.
  • the resin was subjected to second cleavage using the same procedure to give batch 2.
  • a salt exchange from TFA to HOAc using Example 5 prepared by peptide synthesizer and manual synthesis was performed using a 2 ⁇ 60 mL Agilent StratoSpheresTM PL-HCO 3 MP SPE column.
  • the column was equilibrated by first treating with 50 mL of MeOH, followed by 50 mL of DI water.
  • the column was then treated with 2 ⁇ 50 mL 1 N HOAc and then with 2 ⁇ 50 mL 0.1 N HOAc, and the filtrate was monitored to ensure pH ⁇ 3 (pH paper).
  • Example 6 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1395 amu and (M+4)/4 ⁇ 1047 amu, which corresponds to a peptide with the parent molecular weight of 4184 amu (ESI-MS, LC/MS Method A).
  • Example 7 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1398 amu and (M+4)/4 ⁇ 1049 amu, which corresponds to a peptide with the parent molecular weight of 4193 amu (ESI-MS, LC/MS Method A).
  • Example 8 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1400 amu and (M+4)/4 ⁇ 1050 amu, which corresponds to a peptide with the parent molecular weight of 4198 amu (ESI-MS, LC/MS Method A).
  • Example 9 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1403 amu and (M+4)/4 ⁇ 1052 amu, which corresponds to a peptide with the parent molecular weight of 4207 amu (ESI-MS, LC/MS Method A).
  • Example 10 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1391 amu and (M+4)/4 ⁇ 1043 amu, which corresponds to a peptide with the parent molecular weight of 4170 amu (ESI-MS, LC/MS Method A).
  • Example 11 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1393 amu and (M+4)/4 ⁇ 1045 amu, which corresponds to a peptide with the parent molecular weight of 4179 amu (ESI-MS, LC/MS Method A).
  • Example 12 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1369 amu and (M+4)/4 ⁇ 1027 amu, which corresponds to a peptide with the parent molecular weight of 4106 amu (ESI-MS, LC/MS Method A).
  • Example 13 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1407 amu and (M+4)/4 ⁇ 1056 amu, which corresponds to a peptide with the parent molecular weight of 4220 amu (ESI-MS, LC/MS Method A).
  • Example 14 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1377 amu and (M+4)/4 ⁇ 1033 amu, which corresponds to a peptide with the parent molecular weight of 4128 amu (ESI-MS, LC/MS Method A).
  • Example 15 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1391 amu and (M+4)/4 ⁇ 1044 amu, which corresponds to a peptide with the parent molecular weight of 4172 amu (ESI-MS, LC/MS Method A).
  • Example 16 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1377 amu and (M+4)/4 ⁇ 1033 amu, which corresponds to a peptide with the parent molecular weight of 4129 amu (ESI-MS, LC/MS Method A).
  • Example 17 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1416 amu and (M+4)/4 ⁇ 1062 amu, which corresponds to a peptide with the parent molecular weight of 4245 amu (ESI-MS, LC/MS Method A).
  • Example 18 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1425 amu and (M+4)/4 ⁇ 1069 amu, which corresponds to a peptide with the parent molecular weight of 4273 amu (ESI-MS, LC/MS Method A).
  • Example 19 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1425 amu and (M+4)/4 ⁇ 1069 amu, which corresponds to a peptide with the parent molecular weight of 4273 amu (ESI-MS, LC/MS Method A).
  • Example 20 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1408 amu and (M+4)/4 ⁇ 1057 amu, which corresponds to a peptide with the parent molecular weight of 4223 amu (ESI-MS, LC/MS Method A).
  • Example 21 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1408 amu and (M+4)/4 ⁇ 1057 amu, which corresponds to a peptide with the parent molecular weight of 4223 amu (ESI-MS, LC/MS Method A).
  • Example 22 was prepared on a 40 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1430 amu and (M+4)/4 ⁇ 1073 amu, which corresponds to a peptide with the parent molecular weight of 4287 amu (ESI-MS, LC/MS Method A).
  • Example 23 was prepared on a 40 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1386 amu and (M+4)/4 ⁇ 1040 amu, which corresponds to a peptide with the parent molecular weight of 4158 amu (ESI-MS, LC/MS Method A).
  • Example 24 was prepared on a 40 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1403 amu and (M+4)/4 ⁇ 1053 amu, which corresponds to a peptide with the parent molecular weight of 4207 amu (ESI-MS, LC/MS Method A).
  • Example 25 was prepared on a 40 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1412 amu and (M+4)/4 ⁇ 1060 amu, which corresponds to a peptide with the parent molecular weight of 4236 amu (ESI-MS, LC/MS Method A).
  • Example 26 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1378 amu and (M+4)/4 ⁇ 1034 amu, which corresponds to a peptide with the parent molecular weight of 4132 amu (ESI-MS, LC/MS Method A).
  • Example 27 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1385 amu and (M+4)/4 ⁇ 1039 amu, which corresponds to a peptide with the parent molecular weight of 4153 amu (ESI-MS, LC/MS Method A).
  • Example 28 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1368 amu and (M+4)/4 ⁇ 1027 amu, which corresponds to a peptide with the parent molecular weight of 4103 amu (ESI-MS, LC/MS Method A).
  • Example 29 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1382 amu and (M+4)/4 ⁇ 1037 amu, which corresponds to a peptide with the parent molecular weight of 4144 amu (ESI-MS, LC/MS Method A).
  • Example 30 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1371 amu and (M+4)/4 ⁇ 1029 amu, which corresponds to a peptide with the parent molecular weight of 4112 amu (ESI-MS, LC/MS Method A).
  • Example 31 was prepared on a 35 ⁇ mol scale as a white solid using the general method.
  • the isolated crude solid was stirred for several hours in 8 mL of 25% acetic acid to minimize the tryptophan CO 2 adduct formed during cleavage from the resin.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1366 amu and (M+4)/4 ⁇ 1025 amu, which corresponds to a peptide with the parent molecular weight of 4097 amu (ESI-MS, LC/MS Method A).
  • Example 32 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1387 amu and (M+4)/4 ⁇ 1041 amu, which corresponds to a peptide with the parent molecular weight of 4159 amu (ESI-MS, LC/MS Method A).
  • Example 33 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1385 amu and (M+4)/4 ⁇ 1039 amu, which corresponds to a peptide with the parent molecular weight of 4154 amu (ESI-MS, LC/MS Method A).
  • Example 34 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1381 amu and (M+4)/4 ⁇ 1036 amu, which corresponds to a peptide with the parent molecular weight of 4139 amu (ESI-MS, LC/MS Method A).
  • Example 35 was prepared on a 20 ⁇ mol scale as a white solid using the general method.
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1408 amu and (M+4)/4 ⁇ 1056 amu, which corresponds to a peptide with the parent molecular weight of 4222 amu (ESI-MS, LC/MS Method A).
  • the purified conjugate was desalted using size exclusion chromatography (GE HiPrep 26/10 Desalting column, 0.1 M acetic acid-, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 47379 amu (MALDI).
  • Example 36 (107 mg, as the 7 acetic acid salt) gave a retention time equal to 9.95 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 50% acetonitrile/water with 0.5% TFA over 20 min, flow rate 0.75 mL/min, ⁇ -220 nm).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine Desalting column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44568 amu (MALDI).
  • Example 37 (35 mg, as the 9 acetic acid salt) gave a retention time equal to 11.58 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm).
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1442 amu and (M+4)/4 ⁇ 1082 amu, which corresponds to a peptide with the parent molecular weight of 4323 amu (ESI-MS, LC/MS Method A).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine Desalting column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44346 amu (MALDI).
  • Example 38 (27 mg, as the 8 acetic acid salt) gave a retention time equal to 12.19 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm).
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1433 amu and (M+4)/4 ⁇ 1075 amu, which corresponds to a peptide with the parent molecular weight of 4298 amu (ESI-MS, LC/MS Method A).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine, 50 ⁇ 130 mm column, 0.1 Macetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44384 amu (MALDI).
  • Example 39 (26.7 mg, as the 8 acetic acid salt) gave a retention time equal to 12.30 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm), and a retention time equal to 12.31 min by C18 HPLC (C18 HPLC Method A).
  • size exclusion HPLC Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm
  • the molecular mass of the isolated peptide was confirmed by fragment ions (M+3)/3 ⁇ 1440 amu and (M+4)/4 ⁇ 1080 amu, which corresponds to a peptide with the parent molecular weight of 4318 amu (ESI-MS, LC/MS Method A).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine, 50 ⁇ 130 mm column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44514 amu (MALDI).
  • Example 40 (35 mg, as the 9 acetic acid salt) gave a retention time equal to 14.90 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm), and a retention time equal to 12.08 min by C18 HPLC (C18 HPLC Method A).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine Desalting column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44239 amu (MALDI).
  • Example 41 (41 mg, as the 8 acetic acid salt) gave a retention time equal to 9.20 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine, 50 ⁇ 130 mm column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44392 amu (MALDI).
  • Example 42 (32.2 mg, as the 8 acetic acid salt) gave a retention time equal to 13.83 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm), and a retention time equal to 12.06 min by C18 HPLC (C18 HPLC Method A).
  • the purified conjugate was desalted using size exclusion chromatography (Sephadex G 25 Fine, 50 ⁇ 130 mm column, 0.1 M acetic acid, ⁇ -254 nm) to afford a white solid after lyophilization.
  • the molecular mass of the isolated peptide was confirmed by positive fragment ion distribution with the apex at 44117 amu (MALDI).
  • Example 43 (32 mg, as the 8 acetic acid salt) gave a retention time equal to 10.65 min using size exclusion HPLC (Phenomenex BioSep-SEC-S3000 column, 7.8 ⁇ 300 mm, 5 ⁇ m, 0.15 mM NaCl in 30 mM PBS over 20 min, pH 6.8, flow rate 0.75 mL/min, ⁇ -215 nm), and a retention time equal to 12.10 min by C18 HPLC (C18 HPLC Method A).
  • the relative potency of PYY analogs at the human Neuropeptide Y receptor type 2 was determined using a melanophore assay essentially as described in Jayawickreme et al. (2005) Current Protocols in Pharmacology 12.9.1-12.
  • Model A Cumulative food intake after 6 h was determined for the PYY analogs in either lean (Model A) or diet-induced obese (DIO) (Model B) C57BL/6 mice in a BioDaQ system for continuous monitoring of food intake (Research Diets Inc., New Brunswick, N.J.).
  • Model A utilized 10 week old male C57BL/6 mice (Taconic, Germantown, N.Y.) fed a normal chow (Purina PMI 5001)
  • Model B utilized 25 week old male C57BL/6 mice fed a 45% high fat chow for 20 weeks (Research Diets D12451).
  • mice were placed singly into the BioDaQ cages and acclimatized for a minimum of 6 days and were allowed ad libitum access to food and water. Approximately 1 hour prior to lights-out, animals were dosed subcutaneously with either vehicle (20 mM Acetate buffer, pH 4.9 or 20% DMSO in water) or analogs dissolved in vehicle (1 mg/kg) (8 animals per group). Once all animals have been dosed, feeder gates were opened providing ad-libitum access to food. Continuous food intake was monitored and collected for 15 hours. Hourly food intake, as well as 6 and 15-hour cumulative food intake, was summarized as % inhibition relative to vehicle controls.
  • Table 1 shows potency at the human Neuropeptide Y receptor and food intake reduction for the PYY analogs shown in Examples 1-35.
  • Example 1 9.9 ⁇ 81 ⁇ 0.0001
  • Example 2 9.8 ⁇ 50 0.0038
  • Example 3 9.5 ⁇ 89 ⁇ 0.0001
  • Example 4 10.7 ⁇ 83 ⁇ 0.0001
  • Example 5 10.5 ⁇ 89 ⁇ 0.0001
  • Example 6 10.1 ⁇ 88 ⁇ 0.0001
  • Example 7 10.1 ⁇ 91 ⁇ 0.0001
  • Example 8 10.7 ⁇ 74 ⁇ 0.0001
  • Example 9 10.7 ⁇ 86 ⁇ 0.0001
  • Example 10 10.4 ⁇ 88 ⁇ 0.0001
  • Example 11 10.3 ⁇ 86 ⁇ 0.0001
  • Example 12 10.2 ⁇ 88 ⁇ 0.0001
  • Example 13 9.9 ⁇ 85 ⁇ 0.0001
  • Example 14 10.5 ⁇ 89 ⁇ 0.0001
  • Example 15 10 ⁇ 90 ⁇ 0.0001
  • Example 16 10.4 ⁇ 90 ⁇ 0.0001
  • Example 17 9.9 ⁇ 90 ⁇ 0.0001
  • Example 18 10.5 ⁇ 92 ⁇ 0.0001
  • Table 2 shows examples of PYY analogs which have potency at the human Neuropeptide Y receptor but do not show food intake reduction greater than human PYY(3-36)NH 2 at the 6 h time point.
  • Example 5 A chronic (41 days) in vivo efficacy study was conducted in a rodent model for obesity (diet-induced obese (DIO) Long Evans rat) to investigate the efficacy and durability of Example 5 singly and in combination with exendin-4 as anti-obesity agents.
  • DIO diet-induced obese
  • DIO Diet-Induced Obese
  • AIZET® mini-osmotic pumps (6 week; Model 2006, Durect Corporation, Cupertino, Calif.) were filled under sterile condition with either vehicle or peptide one day prior to the surgery. Each rat was implanted with two osmotic pumps subcutaneously in the scapula region containing vehicle or peptide according to their treatment group. Body weight and food intake were measured twice per week beginning three days before the 41-day treatment period. On day 41 of treatment, whole blood was collected by cardiac stick under isoflurane anesthesia. Plasma and serum were then prepared from the whole blood for serum chemistry analysis. All the data are presented as mean ⁇ SEM.
  • Example 5+exendin-4 combination based upon the food intake inhibition of each peptide administered alone ( ⁇ 11.5% and ⁇ 20.1%, respectively, with a projected additive food intake inhibition of ⁇ 31.6%).
  • Example 23 A chronic (26 days) in vivo efficacy study was conducted in a rodent model for diabetes (Zucker Diabetic Fatty (ZDF) rat) to investigate the efficacy and durability of Example 23 singly and in combination with exendin-4 as anti-diabetes agents.
  • ZDF Zinc Diabetic Fatty
  • ZDF rats Male ZDF rats were 12 weeks old at the start of the study (Charles River, Inc. Boston, Mass.). The ZDF rats were housed 1 per cage and given ad libitum access to diet (Purina PMI 5008) and water, maintained on a 12 hr light/dark cycle from 5:00 AM to 5:00 PM at 21° C. and 50% relative humidity and allowed to acclimate for at least 6 days prior to baseline measurements and 10 days prior to the surgeries. Baseline fat mass and non-fat mass measurements were taken 3 days before the start of peptide infusion and on day 26 of treatment using a QMR instrument (Echo Medical Systems, Houston, Tex.).
  • ALZET® mini-osmotic pumps (4-week; Model 2006, Durect Corporation, Cupertino, Calif.) were filled under sterile condition with either vehicle or peptide one day prior to the surgery. Similar surgical implantation of the mini-pumps was performed as described for the DIO rats above (except animals were injected ID with lidocaine (0.1 mL of 0.125% lidocaine). Body weight and food intake were measured twice per week beginning 3 days before the 26-day treatment period. On day 26 of treatment, whole blood was collected by cardiac stick under isoflurane anesthesia. The whole blood was used to determine the % HbA1c and the serum was used to measure glucose.
US14/888,085 2013-05-02 2014-04-30 Therapeutic peptides Abandoned US20160108098A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/888,085 US20160108098A1 (en) 2013-05-02 2014-04-30 Therapeutic peptides

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361818624P 2013-05-02 2013-05-02
US14/888,085 US20160108098A1 (en) 2013-05-02 2014-04-30 Therapeutic peptides
PCT/IB2014/061123 WO2014178018A1 (en) 2013-05-02 2014-04-30 Therapeutic peptides

Publications (1)

Publication Number Publication Date
US20160108098A1 true US20160108098A1 (en) 2016-04-21

Family

ID=50842285

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/888,085 Abandoned US20160108098A1 (en) 2013-05-02 2014-04-30 Therapeutic peptides
US14/265,831 Active US9441023B2 (en) 2013-05-02 2014-04-30 Peptide YY analogs

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/265,831 Active US9441023B2 (en) 2013-05-02 2014-04-30 Peptide YY analogs

Country Status (20)

Country Link
US (2) US20160108098A1 (es)
EP (1) EP2992008B1 (es)
JP (1) JP2016519130A (es)
KR (1) KR20160003848A (es)
CN (1) CN105263957A (es)
AR (1) AR096162A1 (es)
AU (1) AU2014261111B2 (es)
BR (1) BR112015027528B1 (es)
CA (1) CA2909045C (es)
CL (1) CL2015003199A1 (es)
EA (1) EA201591839A1 (es)
ES (1) ES2732291T3 (es)
HK (1) HK1214829A1 (es)
MX (1) MX2015015249A (es)
PE (1) PE20151808A1 (es)
RU (1) RU2015144632A (es)
SG (1) SG11201508469YA (es)
TW (1) TW201534616A (es)
UY (1) UY35548A (es)
WO (1) WO2014178018A1 (es)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018081370A1 (en) * 2016-10-27 2018-05-03 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide y receptors
US10875902B2 (en) 2018-04-25 2020-12-29 Janssen Pharmaceutica Nv Glucagon like peptide 1 (GLP-1) fusion peptide coupled cyclic peptide tyrosine tyrosine conjugates and uses thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101925620B1 (ko) 2010-12-16 2018-12-05 노보 노르디스크 에이/에스 Glp-1 아고니스트 및 n-(8-(2-히드록시벤조일)아미노)카프릴산의 염을 포함하는 고체 조성물
EP2827885B1 (en) 2012-03-22 2018-08-15 Novo Nordisk A/S Compositions of glp-1 peptides and preparation thereof
AU2014261111B2 (en) * 2013-05-02 2017-03-16 Glaxosmithkline Intellectual Property Development Limited Therapeutic peptides
EP3068421B1 (en) 2013-11-15 2019-04-17 Novo Nordisk A/S Selective pyy compounds and uses thereof
EP3068795B1 (en) 2013-11-15 2019-03-06 Novo Nordisk A/S Hpyy(1-36) having a beta-homoarginine substitution at position 35
WO2016124687A1 (en) * 2015-02-04 2016-08-11 Novo Nordisk A/S Selective pyy compounds and uses thereof
AR104984A1 (es) 2015-06-12 2017-08-30 Novo Nordisk As Compuestos selectivos para pyy y sus usos
EP3399033A4 (en) * 2015-12-28 2019-06-19 Idemitsu Kosan Co., Ltd. PEPTIDE MARKING AND MARKED PROTEIN THEREOF
WO2018165462A1 (en) 2017-03-08 2018-09-13 Intarcia Therapeutics, Inc Apparatus and methods for administration of a nauseogenic compound from a drug delivery device
PL3746111T3 (pl) 2018-02-02 2024-01-15 Novo Nordisk A/S Stałe kompozycje zawierające agonistę glp-1 i sól kwasu n-(8-(2- hydroksybenzoilo)amino kaprylowego i substancję poślizgową
TW202208410A (zh) * 2018-11-01 2022-03-01 美商美國禮來大藥廠 蛋白質酪胺酸-酪胺酸類似物及其使用方法
MX2022005661A (es) 2019-11-11 2022-09-07 Boehringer Ingelheim Int Agonistas del receptor npy2.
CN115052886A (zh) * 2019-12-04 2022-09-13 斯克利普斯研究所 Glp2受体激动剂和使用方法
BR112022020956A8 (pt) * 2020-04-17 2023-01-10 I2O Therapeutics Inc Análogos de peptídeo tirosina tirosina (pyy) de ação prolongada e métodos de uso
CA3185637A1 (en) 2020-08-07 2022-02-10 Boehringer Ingelheim International Gmbh Soluble npy2 receptor agonists
WO2024038067A1 (en) 2022-08-18 2024-02-22 Boehringer Ingelheim International Gmbh Combination therapy comprising long acting glp-1/glucagon and npy2 receptor agonists
CN115960258B (zh) * 2022-09-30 2024-01-12 广西医科大学附属肿瘤医院 一类GLP-1/glucagon/Y2受体三重激动剂及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9441023B2 (en) * 2013-05-02 2016-09-13 Glaxosmithkline Intellectual Property Development Limited Peptide YY analogs
US20160289283A1 (en) * 2013-11-15 2016-10-06 Novo Nordisk Selective pyy compounds and uses thereof

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4675189A (en) 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
DK134189D0 (da) 1989-03-20 1989-03-20 Nordisk Gentofte Insulinforbindelser
ATE164852T1 (de) 1990-01-24 1998-04-15 Douglas I Buckley Glp-1-analoga verwendbar in der diabetesbehandlung
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US5612052A (en) 1995-04-13 1997-03-18 Poly-Med, Inc. Hydrogel-forming, self-solvating absorbable polyester copolymers, and methods for use thereof
US6413539B1 (en) 1996-10-31 2002-07-02 Poly-Med, Inc. Hydrogel-forming, self-solvating absorbable polyester copolymers, and methods for use thereof
US5612051A (en) 1995-11-17 1997-03-18 Yue; Samuel K. Method of treating involuntary muscle dysfunction with relaxin hormone
ATE417622T1 (de) 1996-08-08 2009-01-15 Amylin Pharmaceuticals Inc Regulation gastrointestinaler beweglichkeit
ES2293688T5 (es) 1997-08-08 2011-05-04 Amylin Pharmaceuticals, Inc. Nuevos compuestos análogos de la exendina.
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6117949A (en) 1998-10-01 2000-09-12 Macromed, Inc. Biodegradable low molecular weight triblock poly (lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
WO1999025727A2 (en) 1997-11-14 1999-05-27 Amylin Pharmaceuticals, Inc. Novel exendin agonist compounds
ATE383867T1 (de) 1997-11-14 2008-02-15 Amylin Pharmaceuticals Inc Neuartige exendin agonisten
WO1999040788A1 (en) 1998-02-13 1999-08-19 Amylin Pharmaceuticals, Inc. Inotropic and diuretic effects of exendin and glp-1
EP1056775B1 (en) 1998-02-27 2010-04-28 Novo Nordisk A/S Glp-1 derivatives of glp-1 and exendin with protracted profile of action
ATE449596T1 (de) 2000-08-15 2009-12-15 Univ Illinois Verfahren zur herstellung von mikropartikeln
DE60230818D1 (de) * 2001-09-24 2009-02-26 Imp Innovations Ltd Pyy3-36 zur zur reduzierung oder vorbeugung von fettleibigkeit
JP5638177B2 (ja) * 2004-02-11 2014-12-10 アミリン・ファーマシューティカルズ, リミテッド・ライアビリティ・カンパニーAmylin Pharmaceuticals, Llc 膵臓ポリペプチドファミリーモチーフおよびそのモチーフを含むポリペプチド
CA2577821A1 (en) * 2004-09-24 2006-04-06 Merck & Co., Inc. Combination therapy for the treatment of obesity
SG158141A1 (en) * 2004-12-13 2010-01-29 Amylin Pharmaceuticals Inc Pancreatic polypeptide family motifs, polypeptides and methods comprising the same
PA8660701A1 (es) 2005-02-04 2006-09-22 Pfizer Prod Inc Agonistas de pyy y sus usos
GB0504857D0 (en) * 2005-03-09 2005-04-13 Imp College Innovations Ltd Novel compounds and their effects on feeding behaviour
MX2008012221A (es) * 2006-03-23 2009-03-06 Amylin Pharmaceuticals Inc Endotelina y agonistas del receptor de endotelina en el tratamiento de enfermedades metabolicas.
SG10201406063XA (en) 2009-09-30 2014-11-27 Glaxo Group Ltd Drug fusions and conjugates with extended half life
WO2011050008A2 (en) 2009-10-19 2011-04-28 Amylin Pharmaceuticals, Inc. Combination therapy comprising administration of an amylinomimetic and a pyy peptidomimetic for effecting weight loss and for treating obesity and related metabolic conditions and disorders
GB201001333D0 (en) * 2010-01-27 2010-03-17 Imp Innovations Ltd Novel compounds and their effects on feeding behaviour
JO3131B1 (ar) 2010-04-27 2017-09-20 Glaxosmithkline Llc مركبات كيميائية
WO2012006566A2 (en) 2010-07-09 2012-01-12 Amylin Pharmaceuticals, Inc. Microcrystalline y receptor agonists
GB201101459D0 (en) 2011-01-27 2011-03-16 Imp Innovations Ltd Novel compounds and thier effects on fedding behaviour

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9441023B2 (en) * 2013-05-02 2016-09-13 Glaxosmithkline Intellectual Property Development Limited Peptide YY analogs
US20160289283A1 (en) * 2013-11-15 2016-10-06 Novo Nordisk Selective pyy compounds and uses thereof

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10961293B2 (en) 2016-10-27 2021-03-30 Janssen Pharmaceutics Nv Cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
WO2018081370A1 (en) * 2016-10-27 2018-05-03 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide y receptors
US10640544B2 (en) 2016-10-27 2020-05-05 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US10787495B2 (en) 2016-10-27 2020-09-29 Janssen Pharmaceutica Nv Immunoglobulins and uses thereof
US10858413B2 (en) 2016-10-27 2020-12-08 Janssen Pharmaceutica Nv Immunoglobulins and uses thereof
US11767354B2 (en) 2016-10-27 2023-09-26 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US10428134B2 (en) 2016-10-27 2019-10-01 Janssen Pharmaceutica Nv Immunoglobulins and uses thereof
US10968265B2 (en) 2016-10-27 2021-04-06 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US11591379B2 (en) 2016-10-27 2023-02-28 Janssen Pharmaceutica Nv Antibody-coupled cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US11401315B2 (en) 2016-10-27 2022-08-02 Janssen Pharmaceutica Nv Immunoglobulins and uses thereof
US10968264B2 (en) 2016-10-27 2021-04-06 Janssen Pharmaceutica Nv Cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US11732019B2 (en) 2016-10-27 2023-08-22 Janssen Pharmaceutica Nv Cyclic peptide tyrosine tyrosine compounds as modulators of neuropeptide Y receptors
US10875902B2 (en) 2018-04-25 2020-12-29 Janssen Pharmaceutica Nv Glucagon like peptide 1 (GLP-1) fusion peptide coupled cyclic peptide tyrosine tyrosine conjugates and uses thereof
US11780900B2 (en) 2018-04-25 2023-10-10 Janssen Sciences Ireland Unlimited Company Glucagon like peptide 1 (GLP-1) fusion peptide coupled cyclic peptide tyrosine tyrosine conjugates and uses thereof

Also Published As

Publication number Publication date
AR096162A1 (es) 2015-12-09
UY35548A (es) 2014-11-28
BR112015027528A2 (pt) 2017-12-05
RU2015144632A (ru) 2017-06-07
SG11201508469YA (en) 2015-11-27
CL2015003199A1 (es) 2016-05-06
KR20160003848A (ko) 2016-01-11
MX2015015249A (es) 2016-02-09
BR112015027528B1 (pt) 2023-02-14
CN105263957A (zh) 2016-01-20
EA201591839A1 (ru) 2016-05-31
ES2732291T3 (es) 2019-11-21
PE20151808A1 (es) 2015-12-16
AU2014261111A1 (en) 2015-11-19
WO2014178018A1 (en) 2014-11-06
AU2014261111B2 (en) 2017-03-16
CA2909045C (en) 2022-12-06
US9441023B2 (en) 2016-09-13
HK1214829A1 (zh) 2016-08-05
EP2992008B1 (en) 2019-04-10
CA2909045A1 (en) 2014-11-06
TW201534616A (zh) 2015-09-16
EP2992008A1 (en) 2016-03-09
US20140329742A1 (en) 2014-11-06
JP2016519130A (ja) 2016-06-30

Similar Documents

Publication Publication Date Title
US9441023B2 (en) Peptide YY analogs
US10758592B2 (en) Exendin-4 derivatives as dual GLP1/glucagon agonists
US11001619B2 (en) GIP agonist compounds and methods
US10253079B2 (en) Functionalized Exendin-4 derivatives
US10093713B2 (en) GIP-GLP-1 dual agonist compounds and methods
US10246497B2 (en) Selective PYY compounds and uses thereof
US9181305B2 (en) Exendin-4 peptide analogues
AU2008257448B2 (en) Unacylated ghrelin as therapeutic agent in the treatment of metabolic disorders
TW201625672A (zh) 升糖素及glp-1受體之共促效劑
TW201202265A (en) Glucagon analogues
JP2007531711A5 (es)
US11713344B2 (en) Acylated oxyntomodulin peptide analog
CN116970062B (zh) 一种超长效glp-1多肽衍生物及其制备方法和用途
US20230063794A1 (en) Stapled triazole co-agonists of the glucagon and glp-1 receptors
US20240059753A1 (en) Compositions including multi-agonist peptides and methods of manufacture and use
CN114867742A (zh) 胰高血糖素和glp-1受体的钉合内酰胺共激动剂
US20230270825A1 (en) Stapled olefin co-agonists of the glucagon and glp-1 receptors

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLLNE INTELLECTUAL PROPERTY (NO.2) LIMIT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CARPENTER, ANDREW JAMES;DOCK, STEVEN THOMAS;HUNTER, III, ROBERT NEIL;AND OTHERS;SIGNING DATES FROM 20140513 TO 20140519;REEL/FRAME:037617/0806

Owner name: GLAXOSMITHKLINE INTELLECTUAL PROPERTY DEVEOLPMENT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED;REEL/FRAME:037617/0973

Effective date: 20140804

STCB Information on status: application discontinuation

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