WO2005058958A2 - Novel glp-1 analogues linked to albumin-like agents - Google Patents

Novel glp-1 analogues linked to albumin-like agents Download PDF

Info

Publication number
WO2005058958A2
WO2005058958A2 PCT/DK2004/000887 DK2004000887W WO2005058958A2 WO 2005058958 A2 WO2005058958 A2 WO 2005058958A2 DK 2004000887 W DK2004000887 W DK 2004000887W WO 2005058958 A2 WO2005058958 A2 WO 2005058958A2
Authority
WO
WIPO (PCT)
Prior art keywords
xaa
glp
lys
compound according
agonist
Prior art date
Application number
PCT/DK2004/000887
Other languages
French (fr)
Other versions
WO2005058958A3 (en
Inventor
Thomas Kruse Hansen
Magali Zundel
Kjeld Madsen
Anne Svendsen
Christine Bruun SCHIØDT
Jesper Lau
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2004298425A priority Critical patent/AU2004298425A1/en
Priority to BRPI0417684-7A priority patent/BRPI0417684A/en
Priority to CA002550050A priority patent/CA2550050A1/en
Priority to EP04803038A priority patent/EP1696962A2/en
Priority to JP2006544221A priority patent/JP2007537142A/en
Priority to MXPA06006746A priority patent/MXPA06006746A/en
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Publication of WO2005058958A2 publication Critical patent/WO2005058958A2/en
Publication of WO2005058958A3 publication Critical patent/WO2005058958A3/en
Priority to IL175938A priority patent/IL175938A0/en
Priority to US11/454,348 priority patent/US20070093417A1/en
Priority to NO20063242A priority patent/NO20063242L/en
Priority to US12/186,880 priority patent/US20090005312A1/en

Links

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
    • 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
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • 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
    • A61P3/06Antihyperlipidemics
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • 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
    • 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
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4705Regulators; Modulating activity stimulating, promoting or activating activity
    • 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
    • C07K14/57563Vasoactive intestinal peptide [VIP]; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to novel GLP-1 compounds, to pharmaceutical compositions comprising these compounds and to the use of the compounds for the treatment of diseases related to diabetes.
  • Diabetes mellitus is a metabolic disorder in which the ability to utilize glucose is partly or completely lost. About 5% of all people suffer from diabetes and the disorder approaches epidemic proportions. Since the introduction of insulin in the 1920 ' s, continuous efforts have been made to improve the treatment of diabetes mellitus.
  • GLP-1 glucagon-like peptide-1
  • Human GLP-1 is a 37 amino acid residue peptide originating from preproglucagon which is synthesized La. in the L-cells in the distal ileum, in the pancreas and in the brain. GLP-1 is an important gut hormone with regulatory function in glucose metabolism and gastrointestinal secretion and metabolism. GLP-1 stimulates insulin secretion in a glucose-dependant manner, stimulates insulin biosynthesis, promotes beta cell rescue, decreases glucagon secretion, gastric emptying and food intake. Human GLP-1 is hydrolysed to GLP-1 (7-37) and GLP-1 (7-36)-amide which are both insulinotropic peptides.
  • [Gly 8 ]GLP-1 (7-37) designates an analogue of GLP-1 (7-37) formally derived from GLP-1 (7-37) by substituting the naturally occurring amino acid residue in position 8 (Ala) by Gly.
  • (N ⁇ 3 -tetradecanoyl)[Lys 34 ]GLP-1(7-37) designates GLP-1 (7-37) wherein the ⁇ - amino group of the Lys residue in position 34 has been tetradecanoylated.
  • PCT publications WO 98/08871 and WO 99/43706 disclose stable derivatives of GLP-1 analogues, which have a lipophilic substituent. These stable derivatives of GLP-1 analogues have a protracted profile of action compared to the corresponding GLP-1 analogues.
  • Exendin-4 is a 39 amino acid residue peptide isolated from the venom of Heloderma suspectum, and this peptide shares 52% homology with GLP-1 (7-37) in the overlapping region.
  • Exendin-4 is a potent GLP-1 receptor agonist which has been shown to stimulate insulin release and ensuing lowering of the blood glucose level when injected into dogs.
  • the group of exendin-4(1-39), certain fragments thereof, analogs thereof and derivatives thereof, are potent insulinotropic agents.
  • the group of exendin-4(1-39), insulinotropic fragments thereof, insulinotropic analogs thereof and insulinotropic derivatives thereof are potent insulinotropic agents.
  • GLP-1 compounds including exendin compounds have been synthesized and studied in particular in relation the plasma half-life. Low plasma halflifes may be due to chemical stability towards peptidases (mainly dipeptidyl aminopeptidase IV) and to renal clearance. However, these variants of insulionotropic peptides have hitherto not showed protracted effects beyond what will suffice for at product to be administered to the patient once daily. A second generation GLP-1 compounds are needed which can be administered to the patients only once weekly or even less frequently.
  • US 6,329,336 discloses the injection of highly reactive GLP-1 peptides into plasma, wherein chemical reactions will take palce with blood components, such as serum albumin.
  • WO 02/46227 discloses fusion proteins between a GLP-1 compound and human serum albumin.
  • WO 2003/103572 discloses conjugates of GLP-1 analogs and a blood component. It is an object of the present invention to provide GLP-1 analogues including exendin peptides linked to protein having a long half-life in human plasma, threreby facilitating a once-weekly treatment of patients. It is also an object of the present invention to provide GLP-1 peptides which are less prone to aggregation, a well known problem associated with the glucagon-like peptides. Being less prone to aggregation facilitates economical manufacturing processes as well as enabling the compounds to be administered by medical infusion pumps.
  • polypeptide and peptide as used herein means a compound composed of at least five constituent amino acids connected by peptide bonds.
  • the constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids.
  • Natural amino acids which are not encoded by the genetic code are e.g. hydroxyproline, -carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine.
  • Synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e.
  • D- isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib ( ⁇ -aminoisobutyric acid), Abu ( ⁇ -aminobutyric acid), Tie (tert-butylglycine), ?-alanine, 3- aminomethyl benzoic acid, anthranilic acid.
  • analogue as used herein referring to a polypeptide means a modified peptide wherein one or more amino acid residues of the peptide have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the peptide and/or wherein one or more amino acid residues have been deleted from the peptide and or wherein one or more amino acid residues have been added to the peptide.
  • Such addition or deletion of amino acid residues can take place at the N-terminal of the peptide and/or at the C-terminal of the peptide.
  • GLP-1 agonist means a compound which stimulates the formation of cAMP in a suitable medium containing the human GLP-1 receptor. The potency of a GLP-1 agonist is determined by calculating the EC 50 value from the dose-response curve as described below.
  • Baby hamster kidney (BHK) cells expressing the cloned human GLP-1 receptor (BHK- 467-12A) were grown in DMEM media with the addition of 100 lU/mL penicillin, 100 ⁇ g/mL streptomycin, 5% fetal calf serum and 0.5 mg/mL Geneticin G-418 (Life Technologies). The cells were washed twice in phosphate buffered saline and harvested with Versene. Plasma membranes were prepared from the cells by homogenisation with an Ultraturrax in buffer 1 (20 mM HEPES-Na, 10 mM EDTA, pH 7.4). The homogenate was centrifuged at 48,000 x g for 15 min at 4°C.
  • the pellet was suspended by homogenization in buffer 2 (20 mM HEPES-Na, 0.1 mM EDTA, pH 7.4), then centrifuged at 48,000 x g for 15 min at 4°C. The washing procedure was repeated one more time. The final pellet was suspended in buffer 2 and used immediately for assays or stored at -80°C.
  • the functional receptor assay was carried out by measuring cyclic AMP (cAMP) as a response to stimulation by the insulinotropic agent.
  • cAMP formed was quantified by the AlphaScreenTM cAMP Kit (Perkin Elmer Life Sciences). Incubations were carried out in half- area 96-well microtiter plates in a total volume of 50 ⁇ L buffer 3 (50 mM Tris-HCI, 5 mM
  • GLP-1 peptide as used herein means GLP-1 (7-37) (SEQ ID No 2), a
  • GLP-1 (7-37) analogue a GLP-1 (7-37) derivative or a derivative of a GLP-1 (7-37) analogue.
  • the GLP-1 peptide is an insulinotropic agent.
  • exendin-4 peptide as used herein means exendin-4(1-39) (SEQ ID No 3), an exendin-4(1-39) analogue, an exendin-4(1-39) derivative or a derivative of an exendin- 4(1-39) analogue.
  • exendin-4 peptide is an insulinotropic agent.
  • DPP-IV protected as used herein referring to a polypeptide means a polypeptide which has been chemically modified in order to render said compound resistant to the plasma peptidase dipeptidyl aminopeptidase-4 (DPP-IV).
  • DPP-IV enzyme in plasma is known to be involved in the degradation of several peptide hormones, e.g. GLP-1, GLP-2, Exendin-4 etc.
  • GLP-1 peptide hormones
  • GLP-2 GLP-2
  • Exendin-4 peptide hormones
  • a considerable effort is being made to develop analogues and derivatives of the polypeptides susceptible to DPP-IV mediated hydrolysis in order to reduce the rate of degradation by DPP-IV.
  • a DPP-IV protected peptide is more resistant to
  • Resistance of a peptide to degradation by dipeptidyl aminopeptidase IV is determined by the following degradation assay : Aliquots of the peptide (5 nmol) are incubated at 37 °C with 1 ⁇ L of purified dipeptidyl aminopeptidase IV corresponding to an enzymatic activity of 5 mU for 10-180 minutes in 100 ⁇ L of 0.1 M triethylamine-HCI buffer, pH 7.4. Enzymatic reactions are terminated by the addition of 5 ⁇ L of 10% trifluoroacetic acid, and the peptide degradation products are separated and quantified using HPLC analysis.
  • Peptides and their degradation products may be monitored by their absorbance at 220 nm (peptide bonds) or 280 nm (aromatic amino acids), and are quantified by integration of their peak areas related to those of standards.
  • the rate of hydrolysis of a peptide by dipeptidyl aminopeptidase IV is estimated at incubation times which result in less than 10% of the peptide being hydrolysed.
  • C 1-6 -alkyl as used herein means a saturated, branched, straight or cyclic hydrocarbon group having from 1 to 6 carbon atoms.
  • Representative examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, te/t-butyl, n- pentyl, isopentyl, neopentyl, ferf-pentyl, n-hexyl, isohexyl, cyclohexane and the like.
  • pharmaceutically acceptable as used herein means suited for normal pharmaceutical applications, i.e. giving rise to no adverse events in patients etc.
  • excipient as used herein means the chemical compounds which are normally added to pharmaceutical compositions, e.g. buffers, tonicity agents, preservatives and the like.
  • effective amount as used herein means a dosage which is sufficient to be effective for the treatment of the patient compared with no treatment.
  • composition means a product comprising an active compound or a salt thereof together with pharmaceutical excipients such as buffer, preservative, and optionally a tonicity modifier and/or a stabilizer.
  • a pharmaceutical composition is also known in the art as a pharmaceutical formulation.
  • treatment of a disease means the management and care of a patient having developed the disease, condition or disorder. The purpose of treatment is to combat the disease, condition or disorder. Treatment includes the administration of the active compounds to eliminate or control the disease, condition or disorder as well as to alleviate the symptoms or complications associated with the disease, condition or disorder. DESCRIPTION OF THE INVENTION
  • the present invention relates to a compound having the structure of the formula (I) : GLP-1 agonist - L - RR - protraction protein (I) wherein
  • GLP-1 agonist is a polypeptide which is an agonist of the human GLP-1 receptor
  • L is a linker connecting an amino acid side chain of said GLP-1 agonist or the C-terminal amino acid residue of said GLP-1 agonist with RR,
  • RR is the remains of a reactive residue that has formed a covalent bond with an amino acid residue of the protraction protein
  • protraction protein is a protein having a molar weight of at least 5 kDa, having a plasma half- life of at least 24 hours in human plasma, and said protraction protein has been synthesised by a non-mammalian organism or synthetically.
  • the protraction protein is recombinant human serum albumin (SEQ ID NO 1). In another embodiment of the invention the protraction protein is a human serum albumin variant. In another embodiment of the invention the human serum albumin variant has reduced binding affinities towards copper and nickel as compared to the corresponding binding affinities of human serum albumin towards copper and nickel. In another embodiment of the invention the protraction protein is an N-terminal fragment of human serum albumin, or an analogue thereof. In another embodiment of the invention the protraction protein is a human serum albumin variant comprising a modification of the Asp-Ala-His-Lys N-terminal sequence.
  • the protraction protein comprises at least one deletion among the three N-terminal amino acid residues Asp-Ala-His.
  • the protraction protein comprises an N- terminal extension, such as Glu "3 ,Ala "2 Glu "1 ,Phe°-HSA(1-585) or an N-terminal fragment thereof.
  • the human serum albumin (HSA) variant is selected from the group consisting of HSA(2-585), HSA(3-585), HSA(4-585), Asp-Ala- HSA(4-585), Xaa 3 -HSA(1-585) where Xaa 3 is an amino acid residue which has substituted the His residue occupying position 3 in native HSA, and N-terminal fragments thereof.
  • a recombinant human serum albumin variant is commercially available from New Century Pharma under the name Albagen. Albagen is HSA(2-585) and is hypoallergenic due to the modified metal binding properties caused by the single N-terminal deletion.
  • the said protration protein comprises an amino acid sequence of from 60-200 such as from 100 to 150 amino acid residues, and said amino acid sequence being identical to a fragment of SEQ ID NO 1 or a fragment of SEQ ID NO 1 with one or two amino acid substitutions and/or deletions.
  • the protraction protein is the Fc portion of an immunoglobulin, an analogue or a fragment thereof.
  • the GLP-1 agonist has at least 50% amino acid homology with either GLP-1 (7-37) (SEQ ID NO 2) or Exendin-4(1-39) (SEQ ID NO 3).
  • the GLP-1 agonist has at least 80% amino acid homology with either GLP-1(7-37) (SEQ ID NO 2) or Exendin-4(1-39) (SEQ ID NO 3).
  • the GLP-1 agonist comprises the amino acid sequence of the formula (II):
  • Xaa 8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid;
  • Xaa 16 is Val or Leu
  • Xaa 18 is Ser, Lys or Arg
  • Xaaig is Tyr or Gin
  • Xaa 20 is Leu or Met
  • Xaa 22 is Gly, Glu or Aib;
  • Xaa 23 is Gin, Glu, Lys or Arg;
  • Xaa 25 is Ala or Val
  • Xaa 26 is Lys, Glu or Arg
  • Xaa 27 is Glu or Leu
  • Xaa 30 is Ala, Glu or Arg
  • Xaa 33 is Val or Lys
  • Xaa 34 is Lys, Glu, Asn or Arg;
  • Xaa 35 is Gly or Aib
  • Xaa 36 is Arg, Gly or Lys
  • Xaa 37 is Gly, Ala, Glu, Pro, Lys, amide or is absent;
  • Xaa 38 is Lys, Ser, amide or is absent.
  • Xaa 39 is Ser, Lys, amide or is absent;
  • Xaa 0 is Gly, amide or is absent;
  • Xaa 41 is Ala, amide or is absent;
  • Xaa 2 is Pro, amide or is absent;
  • Xaa 43 is Pro, amide or is absent;
  • Xaa ⁇ is Pro, amide or is absent
  • Xaa 45 is Ser, amide or is absent
  • Xaa 46 is amide or is absent ; provided that if Xaa 38 , Xaa 39 , Xaa 40 , Xaa 41 , Xaa 42 , Xaa 3 , Xaa 44 , Xaa 45 orXaa 46 is absent then each amino acid residue downstream is also absent.
  • the GLP-1 agonist comprises the amino acid sequence of formula (III): Xaa 7 -Xaa 8 -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Xaa 18 -Tyr-Leu-Glu-Xaa 22 -Xaa 23 -Ala-Ala- Xaa ⁇ -Glu-Phe-lle-Xaaso-Trp-Leu-Val-Xaa ⁇ -Xaas E s-Xaase-Xaas ⁇ Xaass Formula (III) (SEQ ID No: 5) wherein
  • Xaa 7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, -hydroxy-histidine, homohistidine, N ⁇ -acetyl-histidine, -fluoromethyl-histidine, ⁇ -methyl-histidine,* 3- pyridylalanine, 2-pyridylalanine or 4-pyridylalanine;
  • Xaa 8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, or (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid; Xaa 2 s Gly, Glu or Aib; Xaa 3 s Gin, Glu, Lys or Arg; Xaa 30 s Ala, Glu or Arg; Xaa 34 s Lys, Glu or Arg; Xaa 37 s Gly, Ala, Glu or Lys; Xaa 38 s Lys, amide or is absent.
  • the said GLP-1 agonist is dipeptidyl aminopeptidase IV protected.
  • the GLP-1 agonist is hydrolysed by DPP-IV at a rate lower than the rate of hydrolysis of GLP-1 (7-37) using the DPP-IV hydrolysis assay disclosed herein.
  • the GLP-1 agonist is a position 8 analogue, i.e. the alanine residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No: 2) has been substituted by another amino acid residue.
  • the GLP-1 agonist comprises an Aib residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No:2).
  • the amino acid residue in position 7 of the GLP-1 peptide is selected from the group consisting of D-histidine, desamino-histidine, 2-amino-histidine, Miydroxy-histidine, homohistidine, N°-acetyl-histidine , ⁇ -fluoromethyl-histidine, ⁇ -methyl-histidine, 3-pyridylalanine, 2-pyridylalanine and 4- pyridylalanine.
  • the GLP-1 agonist comprises no more than twelve amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist comprises no more than six amino acid residues which have been exchanged, added or deleted as compared to GLP- 1(7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
  • the GLP-1 agonist comprises no more than four amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist comprises no more than 4 amino acid residues which are not encoded by the genetic code. In another embodiment of the invention the GLP-1 agonist comprises no more than two amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
  • the GLP-1 agonist is selected from the group consisting of [Arg 34 ]GLP-1(7-37), [Arg 26 ' 34 ]GLP-1(7-37)Lys, [Lys 36 Arg 26 ' 34 ]GLP-1(7-36), [Aib 8 ' 22 ' 35 ]GLP-1 (7-37),
  • the GLP-1 agonist is ZP-10, i.e. [Ser 38 Lys 39 ]Exendin-4(1-39)LysLysLysLysLys-amide (SEQ ID No. 4).
  • the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the amino acid residue in position 23, 26, 34, 36 or 38 relative to the amino acid sequence SEQ ID No:2 (GLP-1 (7-37)), (corresponding to position 17, 20, 28, 30 or 32 relative to amino acid sequence SEQ ID No:3(Exendin-4(1-39)).
  • the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the C-terminal amino acid residue.
  • the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of an amino acid residue selected from arginine, lysine, cysteine, glutamic acid, aspartic acid, histidine, serine, threonine and tyrosine.
  • the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of a cysteine residue.
  • the linker L is selected from the group consisting of the bivalent connecting chemical groups amides: -C(O)-NR- where R is hydrogen or C 1-6 -alkyl, amine: -NR-, where R is hydrogen or C 1-6 -alkyI,
  • GLP-1 agonist - CH 2 (CH 2 ) n (OCH 2 CH 2 ) m - RR - protraction protein
  • the compound of general formula (I) is selected from the group consisting of
  • the compound of the general formula (I) is selected from the group consisting of S-gamma 3 -(1 - ⁇ 2-[2-(2-([D-Ala 8 , Lys 37 ]-GLP-1 -(7- 37)amide-N ⁇ 37 -yl)acetyloxyethoxy)ethylcarbamoyl]ethyl ⁇ -2,5-dioxo-pyrrolidin-3-yl)Albagen
  • the compounds of the present invention can be produced by classical peptide synthesis, e.g. solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques., see e.g. Green and Wuts, "Protecting Groups in Organic Synthesis", John Wiley & Sons, 1999. These methods are preferred when the insulinotropic agent is a peptide comprising non-natural amino acid residues.
  • the polypeptides can also be produced by a method which comprises cultuhng a host cell containing a DNA sequence encoding the polypeptide and capable of expressing the polypeptide in a suitable nutrient medium under conditions permitting the expression of the peptide, after which the resulting peptide is recovered from the culture and then derivatized to the compound of formula (I).
  • the medium used to culture the cells may be any conventional medium suitable for growing the host cells, such as minimal or complex media containing appropriate supplements. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection).
  • the peptide produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration.
  • the proteinaceous components of the supernatant are isolated by filtration, column chromatography or precipitation, e.g. microfiltation, ultrafiltration, isoelectric precipitation, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, hydrophobic interaction chromatography, gel filtration chromatography, affinity chromatography, or the like, dependent on the type of polypeptide in question.
  • chromatographic procedures e.g. ion exchange chromatography, hydrophobic interaction chromatography, gel filtration chromatography, affinity chromatography, or the like, dependent on the type of polypeptide in question.
  • the cells isolated from the culture medium are disintegrated or permeabilised and extracted to recover the product polypeptide or precursor thereof.
  • the DNA sequence encoding the therapeutic polypeptide may suitably be of genomic or cDNA origin, for instance obtained by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the peptide by hybridisation using synthetic oligonucleotide probes in accordance with standard techniques (see, for example, Sambrook, J, Fritsch, EF and Maniatis, T, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor
  • the DNA sequence encoding the polypeptide may also be prepared synthetically by established standard methods, e.g. the phosphoamidite method described by Beaucage and Caruthers, Tetrahedron Letters 22 (1981), 1859 - 1869, or the method described by Matthes et al., EMBO Journal 3 (1984), 801 - 805.
  • the DNA sequence may also be prepared by polymerase chain reaction using specific primers, for instance as described in US 4,683,202 or Saiki et al., Science 239 (1988), 487 - 491.
  • the DNA sequence may be inserted into any vector which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced.
  • the vector may be an autonomously replicating vector, i.e. a vector which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g. a plasmid.
  • the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated.
  • the vector is preferably an expression vector in which the DNA sequence encoding the polypeptide is operably linked to additional segments required for transcription of the DNA, such as a promoter.
  • the promoter may be any DNA sequence which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • Suitable promoters for directing the transcription of the DNA encoding the peptide of the invention in a variety of host cells are well known in the art, cf. for instance Sambrook et. al., supra.
  • the DNA sequence encoding the polypeptide may also, if necessary, be operably connected to a suitable terminator, polyadenylation signals, transcriptional enhancer sequences, and translational enhancer sequences.
  • the recombinant vector of the invention may further comprise a DNA sequence enabling the vector to replicate in the host cell in question.
  • the vector may also comprise a selectable marker, e.g. a gene the product of which complements a defect in the host cell or one which confers resistance to a drug, e.g.
  • a secretory signal sequence also known as a leader sequence, prepro sequence or pre sequence
  • a secretory signal sequence may be provided in the recombinant vector.
  • the secretory signal sequence is joined to the DNA sequence encoding the peptide in the correct reading frame.
  • Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide.
  • the secretory signal sequence may be that normally associated with the peptide or may be from a gene encoding another secreted protein.
  • the procedures used to ligate the DNA sequences coding for the present peptide, the promoter and optionally the terminator and/or secretory signal sequence, respectively, and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (cf., for instance, Sambrook et. al.., supra).
  • the host cell into which the DNA sequence or the recombinant vector is introduced may be any cell which is capable of producing the present peptide and includes bacteria, yeast, fungi and higher eukaryotic cells.
  • suitable host cells well known and used in the art are, without limitation, E. coli, Saccharomyces cerevisiae, or mammalian BHK or CHO cell lines.
  • Pharmaceutical compositions containing a compound according to the present invention may be prepared by conventional techniques, e.g. as described in Remington's Pharmaceutical Sciences, 1985 or in Remington: The Science and Practice of Pharmacy, 19 th edition, 1995.
  • One object of the present invention is to provide a pharmaceutical formulation comprising a compound according to the present invention which is present in a concentration from about 0.1 mg/ml to about 25 mg/ml, and wherein said formulation has a pH from 2.0 to 10.0.
  • the formulation may further comprise a buffer system, preservative(s), isotonicity agent(s), chelating agent(s), stabilizers and surfactants.
  • the pharmaceutical formulation is an aqueous formulation, i.e. formulation comprising water. Such formulation is typically a solution or a suspension.
  • the pharmaceutical formulation is an aqueous solution.
  • aqueous formulation is defined as a formulation comprising at least 50 %w/w water.
  • aqueous solution is defined as a solution comprising at least 50 %w/w water
  • aqueous suspension is defined as a suspension comprising at least 50 %w/w water.
  • pharmaceutical formulation is a freeze-dried formulation, whereto the physician or the patient adds solvents and/or diluents prior to use.
  • pharmaceutical formulation is a dried formulation (e.g. freeze-dried or spray-dried) ready for use without any prior dissolution.
  • the invention relates to a pharmaceutical formulation comprising an aqueous solution of a compound according to the present invention, and a buffer, wherein said compound is present in a concentration from 0.1 mg/ml or above, and wherein said formulation has a pH from about 2.0 to about 10.0.
  • the pH of the formulation is from about 7.0 to about 9.5.
  • the pH of the formulation is from about 3.0 to about 7.0.
  • the pH of the formulation is from about 5.0 to about 7.5.
  • the pH of the formulation is from about 7.5 to about 9.0.
  • the pH of the formulation is from about.7.5 to about 8.5.
  • the pH of the formulation is from about 6.0 to about 7.5. In another embodiment of the invention the pH of the formulation is from about 6.0 to about 7.0. In another embodiment of the invention the pH of the formulation is from about 3.0 to about 9.0, and said pH is at least 2.0 pH units from the isoelectric pH of compound of the present invention.
  • the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginin, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof.
  • Each one of these specific buffers constitutes an alternative embodiment of the invention.
  • the formulation further comprises a pharmaceutically acceptable preservative.
  • the preservative is selected from the group consisting of phenol, o-cresol, m-cresol, p-cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p-hydroxybenzoate, benzethonium chloride, chlorphenesine (3p-chlorphenoxypropane-1 ,2-diol) or mixtures thereof.
  • the preservative is present in a concentration from 0.1 mg/ml to 20 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 0.1 mg/ml to 5 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 5 mg/ml to 10 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 10 mg/ml to 20 mg/ml. Each one of these specific preservatives constitutes an alternative embodiment of the invention.
  • the use of a preservative in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19 th edition, 1995.
  • the formulation further comprises an isotonic agent.
  • the isotonic agent is selected from the group consisting of a salt (e.g. sodium chloride), a sugar or sugar alcohol, an amino acid (e.g. L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine), an alditol (e.g. glycerol (glycerine), 1 ,2-propanedioI (propyleneglycol), 1,3-propanediol, 1,3- butanediol) polyethyleneglycol (e.g.
  • Any sugar such as mono-, di-, or polysaccharides, or water-soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, soluble starch, hydroxyethyl starch and carboxymethylcellulose-Na may be used.
  • the sugar additive is sucrose.
  • Sugar alcohol is defined as a C4-C8 hydrocarbon having at least one -OH group and includes, for example, mannitol, sorbitol, inositol, galacititol, dulcitol, xylitol, and arabitol.
  • the sugar alcohol additive is mannitol.
  • the sugars or sugar alcohols mentioned above may be used individually or in combination. There is no fixed limit to the amount used, as long as the sugar or sugar alcohol is soluble in the liquid preparation and does not adversely effect the stabilizing effects achieved using the methods of the invention.
  • the sugar or sugar alcohol concentration is between about 1 mg/ml and about 150 mg/ml.
  • the isotonic agent is present in a concentration from 1 mg/ml to 50 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 1 mg/ml to 7 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 8 mg/ml to 24 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 25 mg/ml to 50 mg/ml. Each one of these specific isotonic agents constitutes an alternative embodiment of the invention.
  • the use of an isotonic agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19 th edition, 1995.
  • the formulation further comprises a chelating agent.
  • the chelating agent is selected from salts of ethylenediaminetetraacetic acid (EDTA), citric acid, and aspartic acid, and mixtures thereof.
  • EDTA ethylenediaminetetraacetic acid
  • the chelating agent is present in a concentration from 0.1 mg/ml to 5mg/ml.
  • the chelating agent is present in a concentration from 0.1 mg/ml to 2mg/ml.
  • the chelating agent is present in a concentration from 2mg/ml to 5mg/ml.
  • Each one of these specific chelating agents constitutes an alternative embodiment of the invention.
  • compositions of the invention are stabilized liquid pharmaceutical compositions whose therapeutically active components include a polypeptide that possibly exhibits aggregate formation during storage in liquid pharmaceutical formulations.
  • aggregate formation is intended a physical interaction between the polypeptide molecules that results in formation of oligomers, which may remain soluble, or large visible aggregates that precipitate from the solution.
  • liquid pharmaceutical composition or formulation once prepared, is not immediately administered to a subject. Rather, following preparation, it is packaged for storage, either in a liquid form, in a frozen state, or in a dried form for later reconstitution into a liquid form or other form suitable for administration to a subject.
  • dried form is intended the liquid pharmaceutical composition or formulation is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and Polli (1984) J. Parenteral Sci. Technol. 38:48-59), spray drying (see Masters (1991) in Spray-Drying Handbook (5th ed; Longman Scientific and Technical, Essez, U.K.), pp. 491- 676; Broadhead et al.
  • compositions of the invention may further comprise an amount of an amino acid base sufficient to decrease aggregate formation by the polypeptide during storage of the composition.
  • amino acid base is intended an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. Where a combination of amino acids is used, all of the amino acids may be present in their free base forms, all may be present in their salt forms, or some may be present in their free base forms while others are present in their salt forms.
  • amino acids to use in preparing the compositions of the invention are those carrying a charged side chain, such as arginine, lysine, aspartic acid, and glutamic acid.
  • Any stereoisomer i.e., L, D, or DL isomer
  • a particular amino acid e.g. glycine, methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof
  • a particular amino acid e.g. glycine, methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof
  • the L-stereoisomer is used.
  • Compositions of the invention may also be formulated with analogues of these amino acids.
  • amino acid analogue is intended a derivative of the naturally occurring amino acid that brings about the desired effect of decreasing aggregate formation by the polypeptide during storage of the liquid pharmaceutical compositions of the invention.
  • Suitable arginine analogues include, for example, aminoguanidine, ornithine and N-monoethyl L-arginine
  • suitable methionine analogues include S-ethyl homocysteine and S-butyl homocysteine
  • suitable cystein analogues include S-methyl-L cystein.
  • the amino acid analogues are incorporated into the compositions in either their free base form or their salt form:
  • the amino acids or amino acid analogues are used in a concentration, which is sufficient to prevent or delay aggregation of the protein.
  • methionine or other sulphur containing amino acids or amino acid analogous
  • methionine may be added to inhibit oxidation of methionine residues to methionine sulfoxide when the polypeptide acting as the therapeutic agent is a polypeptide comprising at least one methionine residue susceptible to such oxidation.
  • inhibit is intended minimal accumulation of methionine oxidized species over time.
  • Inhibiting methionine oxidation results in greater retention of the polypeptide in its proper molecular form.
  • Any stereoisomer of methionine (L, D, or DL isomer) or combinations thereof can be used.
  • the amount to be added should be an amount sufficient to inhibit oxidation of the methionine residues such that the amount of methionine sulfoxide is acceptable to regulatory agencies. Typically, this means that the composition contains no more than about 10% to about 30% methionine sulfoxide. Generally, this can be achieved by adding methionine such that the ratio of methionine added to methionine residues ranges from about 1:1 to about 1000:1, such as 10: 1 to about 100: 1.
  • the formulation further comprises a stabiliser selected from the group of high molecular weight polymers or low molecular compounds.
  • the stabilizer is selected from polyethylene glycol (e.g. PEG 3350), polyvinylalcohol (PVA), polyvinylpyrrolldone, carboxy- /hydroxycellulose or derivates thereof (e.g. HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, sulphur-containing substances as monothioglycerol, thioglycolic acid and 2- methylthioethanol, and different salts (e.g. sodium chloride).
  • PEG 3350 polyethylene glycol
  • PVA polyvinylalcohol
  • PVpyrrolldone polyvinylpyrrolldone
  • carboxy- /hydroxycellulose or derivates thereof e.g. HPC, HPC-SL, HPC-L and HPMC
  • cyclodextrins e.g. sulphur-containing substances as monothiogly
  • compositions may also comprise additional stabilizing agents, which further enhance stability of a therapeutically active polypeptide therein.
  • Stabilizing agents of particular interest to the present invention include, but are not limited to, methionine and EDTA, which protect the polypeptide against methionine oxidation, and a nonionic surfactant, which protects the polypeptide against aggregation associated with freeze-thawing or mechanical shearing.
  • the formulation further comprises a ; surfactant.
  • the surfactant is selected from a detergent, ethoxylated castor oil, polyglycolyzed glycehdes, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypropylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic ® F68, poloxamer 188 and 407, Triton X-100 ), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g.
  • Tween-20, Tween-40, Tween-80 and Brij-35 monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lecitins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmitoyl phosphatidic acid) and lysophospholipids (eg.
  • phospholipids eg. dipalmitoyl phosphatidic acid
  • lysophospholipids eg.
  • ceramides e.g. sodium tauro-dihydrofusidate etc.
  • C6-C12 e.g.
  • acylcamitines and derivatives N ⁇ -acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, N ⁇ -acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, N ⁇ -acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, DSS (docusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49- 4]), docusate potassium, CAS registry no [7491-09-0]), SDS (sodium dodecyl sulfate or sodium lauryl sulfate), sodium caprylate, cholic acid or derivatives thereof, bile acids and salts thereof and glycine or taurine
  • N-alkyl-N,N-dimethylammonio-1-propanesulfonates 3- cholamido-1-propyldimethylammonio-1-propanesulfonate
  • cationic surfactants quarternary ammonium bases
  • cetyl-trimethylammonium bromide cetylpyridinium chloride
  • non- ionic surfactants eg. dodecyl ⁇ -D-glucopyranoside
  • poloxamines eg.
  • Tetronic's which are tetrafunctional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention.
  • the use of a surfactant in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19 th edition, 1995.
  • Such additional ingredients may include wetting agents, emulsifiers, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous vehicles, proteins (e.g., human serum albumin, gelatin or proteins) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine).
  • additional ingredients should not adversely affect the overall stability of the pharmaceutical formulation of the present invention.
  • compositions containing a compound according to the present invention may be administered to a patient in need of such treatment at several sites, for example, at topical sites, for example, skin and mucosal sites, at sites which bypass absorption, for example, administration in an artery, in a vein, in the heart, and at sites which involve absorption, for example, administration in the skin, under the skin, in a muscle or in the abdomen.
  • topical sites for example, skin and mucosal sites
  • sites which bypass absorption for example, administration in an artery, in a vein, in the heart
  • sites which involve absorption for example, administration in the skin, under the skin, in a muscle or in the abdomen.
  • compositions according to the invention may be through several routes of administration, for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment.
  • the present invention relates to a pharmaceutical composition comprising a compound according to Formula (I), and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is suited for pulmonary administration.
  • compositions of the current invention may be administered in several dosage forms, for example, as solutions, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinses, capsules, for example, hard gelatine capsules and soft gelatine capsules, suppositories, rectal capsules, drops, gels, sprays, powder, aerosols, inhalants, eye drops, ophthalmic ointments, ophthalmic rinses, vaginal pessaries, vaginal rings, vaginal ointments, injection solution, in situ transforming solutions, for example in situ gelling, in situ setting, in situ precipitating, in situ crystallization, infusion solution, and implants.
  • compositions of the invention may further be compounded in, or attached to, for example through covalent, hydrophobic and electrostatic interactions, a drug carrier, drug delivery system and advanced drug delivery system in order to further enhance stability of the compound, increase bioavailability, increase solubility, decrease adverse effects, achieve chronotherapy well known to those skilled in the art, and increase patient compliance or any combination thereof.
  • carriers, drug delivery systems and advanced drug delivery systems include, but are not limited to, polymers, for example cellulose and derivatives, polysaccharides, for example dextran and derivatives, starch and derivatives, poly(vinyl alcohol), acrylate and methacrylate polymers, polylactic and polyglycolic acid and block co-polymers thereof, polyethylene glycols, carrier proteins, for example albumin, gels, for example, thermogelling systems, for example block co-polymeric systems well known to those skilled in the art, micelles, liposomes, microspheres, nanoparticulates, liquid crystals and dispersions thereof, L2 phase and dispersions there of well known to those skilled in the art of phase behaviour in lipid-water systems, polymeric micelles, multiple emulsions, self- emulsifying, self-microemulsifying, cyclodextrins and derivatives thereof, and dendrimers.
  • polymers for example cellulose and derivatives, polysaccharides, for example dextran and derivative
  • compositions of the current invention are useful in the formulation of solids, semisolids, powder and solutions for pulmonary administration of the compound, using, for example a metered dose inhaler, dry powder inhaler and a nebulizer, all being devices well known to those skilled in the art.
  • Compositions of the current invention are specifically useful in the formulation of controlled, sustained, protracting, retarded, and slow release drug delivery systems. More specifically, but not limited to, compositions are useful in formulation of parenteral controlled release and sustained release systems (both systems leading to a many-fold reduction in number of administrations), well known to those skilled in the art. Even more preferably, are controlled release and sustained release systems administered subcutaneous.
  • examples of useful controlled release system and compositions are hydrogels, oleaginous gels, liquid crystals, polymeric micelles, microspheres, nanoparticles,
  • Methods to produce controlled release systems useful for compositions of the current invention include, but are not limited to, crystallization, condensation, co- cystallization, precipitation, co-precipitation, emulsification, dispersion, high pressure homogenization, encapsulation, spray drying, microencapsulation, coacervation, phase separation, solvent evaporation to produce microspheres, extrusion and supercritical fluid processes.
  • General reference is made to Handbook of Pharmaceutical Controlled Release (Wise, D.L., ed.
  • Parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe. Alternatively, parenteral administration can be performed by means of an infusion pump.
  • a further option is a composition which may be a solution or suspension for the administration of the compound according to the present invention in the form of a nasal or pulmonal spray.
  • the pharmaceutical compositions containing the compound of the invention can also be adapted to transdermal administration, e.g.
  • stabilized formulation refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • physical stability of the protein formulation as used herein refers to the tendency of the protein to form biologically inactive and/or insoluble aggregates of the protein as a result of exposure of the protein to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces. Physical stability of the aqueous protein formulations is evaluated by means of visual inspection and/or turbidity measurements after exposing the formulation filled in suitable containers (e.g.
  • the turbidity of the formulation is characterized by a visual score ranking the degree of turbidity for instance on a scale from 0 to 3 (a formulation showing no turbidity corresponds to a visual score 0, and a formulation showing visual turbidity in daylight corresponds to visual score 3).
  • a formulation is classified physical unstable with respect to protein aggregation, when it shows visual turbidity in daylight.
  • the turbidity of the formulation can be evaluated by simple turbidity measurements well-known to the skilled person.
  • aqueous protein formulations can also be evaluated by using a spectroscopic agent or probe of the conformational status of the protein.
  • the probe is preferably a small molecule that preferentially binds to a non-native conformer of the protein.
  • One example of a small molecular spectroscopic probe of protein structure is Thioflavin T.
  • Thioflavin T is a fluorescent dye that has been widely used for the detection of amyloid fibrils. In the presence of fibrils, and perhaps other protein configurations as well, Thioflavin T gives rise to a new excitation maximum at about 450 nm and enhanced emission at about 482 nm when bound to a fibril protein form. Unbound Thioflavin T is essentially non-fluorescent at the wavelengths.
  • hydrophobic patch probes that bind preferentially to exposed hydrophobic patches of a protein.
  • the hydrophobic patches are generally buried within the tertiary structure of a protein in its native state, but become exposed as a protein begins to unfold or denature.
  • these small molecular, spectroscopic probes are aromatic, hydrophobic dyes, such as antrhacene, acridine, phenanthroline or the like.
  • spectroscopic probes are metal-amino acid complexes, such as cobalt metal complexes of hydrophobic amino acids, such as phenylalanine, leucine, isoleucine, methionine, and valine, or the like.
  • chemical stability refers to chemical covalent changes in the protein structure leading to formation of chemical degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native protein structure.
  • chemical degradation products can be formed depending on the type and nature of the native protein and the environment to which the protein is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the protein formulation as well- known by the person skilled in the art.
  • the chemical stability of the protein formulation can be evaluated by measuring the amount of the chemical degradation products at various time-points after exposure to different environmental conditions (the formation of degradation products can often be accelerated by for instance increasing temperature).
  • the amount of each individual degradation product is often determined by separation of the degradation products depending on molecule size and/or charge using various chromatography techniques (e.g. SEC-HPLC and/or RP-HPLC).
  • a "stabilized formulation” refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability. In general, a formulation must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 6 weeks of usage and for more than 3 years of storage. In another embodiment of the invention the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 4 weeks of usage and for more than 3 years of storage. In a further embodiment of the invention the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 4 weeks of usage and for more than two years of storage. In an even further embodiment of the invention the pharmaceutical formulation comprising the compound is stable for more than 2 weeks of usage and for more than two years of storage.
  • the present invention relates to the use of a compound according to the invention for the preparation of a medicament.
  • a compound according to the invention is used for the preparation of a medicament for the treatment or prevention of hyperglycemia, type 2 diabetes, impaired glucose tolerance, type 1 diabetes, obesity, hypertension, syndrome X, dyslipidemia, cognitive disorders, atheroschlerosis, myocardial infarction, coronary heart disease and other cardiovascular disorders, stroke, inflammatory bowel syndrome, dyspepsia and gastric ulcers.
  • a compound according to the invention is used for the preparation of a medicament for delaying or preventing disease progression in type 2 diabetes.
  • a compound according to the invention is used for the preparation of a medicament for decreasing food intake, decreasing ⁇ -ce ⁇ apoptosis, increasing ?-cell function and ⁇ -ce ⁇ mass, and/or for restoring glucose sensitivity to ?-cells.
  • the treatment with a compound according to the present invention may also be combined with combined with a second or more pharmacologically active substances, e.g. selected from antidiabetic agents, antiobesity agents, appetite regulating agents, antihypertensive agents, agents for the treatment and/or prevention of complications resulting from or associated with diabetes and agents for the treatment and/or prevention of complications and disorders resulting from or associated with obesity.
  • a second or more pharmacologically active substances e.g. selected from antidiabetic agents, antiobesity agents, appetite regulating agents, antihypertensive agents, agents for the treatment and/or prevention of complications resulting from or associated with diabetes and agents for the treatment and/or prevention of complications and disorders resulting from or associated with obesity.
  • Examples of these pharmacologically active substances are : Insulin, sulphonylureas, biguanides, meglitinides, glucosidase inhibitors, glucagon antagonists, DPP-IV (dipeptidyl peptidase-IV) inhibitors, inhibitors of hepatic enzymes involved in stimulation of gluconeogenesis and/or glycogenolysis, glucose uptake modulators, compounds modifying the lipid metabolism such as antihyperlipidemic agents as HMG CoA inhibitors (statins), compounds lowering food intake, RXR agonists and agents acting on the ATP-dependent potassium channel of the ⁇ - cells; Cholestyramine, colestipol, clofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, probucol, dextrothyroxine, neteglinide, repaglinide; ⁇ -blockers such as alprenolol, atenolol,
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to general formula (I), and a pharmaceutically acceptable preservative.
  • the pharmaceutical composition comprises a compound according to the general formula (I) and a pharmaceutically acceptable stabilizer.
  • the pharmaceutical composition is suited for parenteral administration.
  • the present invention relates to the use of a compound according to the general formula (I) for the preparation of a medicament.
  • the protected amino acid derivatives used were standard Fmoc- amino acids (Anaspec) supplied in preweighed cartridges suitable for the ABI433A synthesizer with the exception of unnatural aminoacids such as Fmoc-Aib-OH (Fmoc- aminoisobutyric acid).
  • the amino acid (4 molar equivalents relative to resin) was dissolved in N- methyl pyrrolidone/methylene chloride (1 :1 , 20 ml). Hydroxybenzotriazole (HOBt) (4 molar equivalents relative to resin) and diisopropylcarbodiimide (4 molar equivalents relative to resin) was added and the solution was stirred for 15 min. The solution was added to the resin and diisopropyethylamine (4 molar equivalents relative to resin) was added. The resin was shaken 24 hours at room temperature. The resin was was washed with N-methyl pyrrolidone (2x20 ml), N-methyl pyrrolidone/Methylene chloride (1 :1) (2x20ml) and methylene chloride (2x20 ml).
  • the cleavage mixture was filtered and the filtrate was concentrated to an oil by a stream of nitrogen.
  • the crude peptide was precipitated from this oil with 45 ml diethyl ether and washed 3 times with 45 ml diethyl ether.
  • the crude peptide was purified by semipreparative HPLC on a 20 mm x 250 mm column packed with 7 ⁇ C-18 silica. Depending on the peptide two one or two purification systems were used. TFA: After drying the crude peptide was dissolved in 5 ml 50% acetic acid H 2 O and diluted to 20 ml with H 2 O and injected on the column which then was eluted with a gradient of 40-60 % CH 3 CN in 0.1% TFA 10 ml/min during 50 min at 40 °C. The peptide containing fractions were collected. The purified peptide was lyophilized after dilution of the eluate with water.
  • Ammonium sulphate The column was equilibrated with 40% CH3CN in 0.05M (NH 4 ) 2 SO 4 , which was adjusted to pH 2.5 with concentrated H 2 SO . After drying the crude peptide was dissolved in 5 ml 50% acetic acid H 2 O and diluted to 20 ml with H 2 O and injected on the column which then was eluted with a gradient of 40% - 60% CH 3 CN in 0.05M (NH 4 ) 2 SO 4 , pH 2.5 at 10 ml/min during 50 min at 40 °C. The peptide containing fractions were collected and diluted with 3 volumes of H 2 O and passed through a Sep-Pak ® C18 cartridge (Waters part.
  • A1 Equilibration of the column with in a buffer consisting of 0.1 M (NH 4 ) 2 SO 4 , which was adjusted to pH 2.5 with concentrated H 2 SO 4 and elution by a gradient of 0% to 60% CH 3 CN in the same buffer during 50 min.
  • B1 Equilibration of the column with 0.1 % TFA / H 2 O and elution by a gradient of 0% CHsCN / 0.1 % TFA / H 2 O to 60% CH 3 CN / 0.1 % TFA / H 2 O during 50 min.
  • LCMS was performed on a setup consisting of Hewlett Packard series 1100 G1312A Bin Pump, Hewlett Packard series 1100 Column compartment, Hewlett Packard series 1100 G1315A DAD diode array detector, Hewlett Packard series 1100 MSD and Sedere 75 Evaporative Light Scattering detectorcontrolled by HP Chemstation software.
  • the HPLC pump is connected to two eluent reservoirs containing: A: 10mM NH 4 OH in water B: 10mM NH 4 OH in 90% acetonitrile
  • the analysis was performed at 23° C by injecting an appropriate volume of the sample (preferably 20 ⁇ l) onto the column which is eluted with a gradient of A and B.
  • HPLC conditions, detector settings and mass spectrometer settings used are giving in the following table.
  • MS was performed on a Voyager RP MALDI-TOF instrument (Perseptive Biosystems Inc., Framingham, MA) equipped with a nitrogen laser (337 nm). The instrument was operated in linear mode with delayed extraction, and the accelerating voltage in the ion source was 25kV. Sample preparation was done as follows: 1 ⁇ l sample-solution (0.5-1.0 mg/ml) was mixed with 10 ⁇ l matrix-solution (Sinapinic acid dissolved in a 5:4:1 mixture of acetonitrile:water:3% TFA) and 1 ⁇ l was deposited on the sample plate and allowed to dry. Only external calibration was performed as the normal peptide standards used are in the low molecular weight range and insufficient to assure proper determination of masses in the range of serum albumin (> 60 KDa). As a result the absolute mass values determined are only within 0.2 % accuracy.
  • a resin (Rink amide, 0.68 mmol/g Novabiochem 0.25 mmole) was used to produce the primary sequence on an ABI433A machine according to manufacturers guidelines. All protecting groups were acid labile with the exception of the residue used in position 37 (FmocLys(ivDde)-OH, Novabiochem) allowing specific deprotection of this lysine rather than any other lysine.
  • 3-maleimido propionic acid (4 molar equivalents relative to resin) was dissolved in N-methyl pyrrolidone/methylene chloride (1 :1 , 20 ml). Hydroxybenzotriazole hydrate (HOBt; H 2 O) (4 molar equivalents relative to resin) and diisopropylcarbodiimide (4 molar equivalents relative to resin) was added and the solution was stirred for 15 min. The solution was added to the resin and diisopropylethylamine (4 molar equivalents relative to resin) was added. The resin was shaken 24 hours at room temperature.
  • HOBt Hydroxybenzotriazole hydrate
  • diisopropylcarbodiimide 4 molar equivalents relative to resin
  • the resin was washed with N-methyl pyrrolidone (2x20 ml), N-methyl pyrrolidone/methylene chloride (1 :1 ) (2x20ml) and methylene chloride (2x20 ml).
  • the peptide was cleaved from the resin by stirring for 180 min at room temperature with a mixture of trifluoroacetic acid, water and triisopropylsilane (95:2.5:2.5).
  • the cleavage mixture was filtered and the filtrate was concentrated to an oil by a stream of nitrogen.
  • the crude peptide was precipitated from this oil with 45 ml diethyl ether and washed 3 times with 45 ml diethyl ether.
  • the crude peptide was purified by semipreparative HPLC on a 20 mm x 250 mm column packed with 7 ⁇ C-18 silica.
  • the crude peptide was dissolved in 5 ml 50% acetic acidin water and diluted to 20 ml with H 2 O and injected on the column which then was eluted with a gradient of 40-60 % (CH 3 CN in water with 0.1% TFA) 10 ml/min during 50 min at 40 °C.
  • the peptide containing fractions were collected.
  • the conjugate was purified on a ResourceTM HIC ISO with a total volume of 1 ml. Flow rate was kept at 1 ml/min. Buffer: 50mM NaPi pH 7.0 + 4 % HP-jff-CD. A gradient from 2 M to 0 M ammonium sulphate over 20 column volumes was used to separate Albagen from the conjugate. The chromatogram showed two eluting peaks. The first peak was due to Albagen, the second peak contained the conjugate. The conjugate was concentrated and separated from non- reacted analogue on a centriprepTM device with a MW cut -off at 30,000 Da. The overall yield • was 25-35%. The site of conjugation was determined to be Cys-34 by peptide mapping.
  • the mass found by MALDI is 70023 Da
  • Theoretical molecular weight of conjugate is 70046 Da.
  • the mass found by MALDI is 70102
  • Theoretical molecular weight of conjugate is 70116 Da.
  • the mass found by MALDI is 70208 Da.
  • Theoretical molecular weight of conjugate is 70304 Da.
  • Example 4 Other compounds which are synthesised according to the method described in example 1 are :
  • Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Psychiatry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Immunology (AREA)
  • Emergency Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a compound having the structure of the formula (I) : GLP-1 agonist-L-RR-protraction protein (I) wherein GLP-1 agonist is a polypeptide which is an agonist of the human GLP-1 receptor, L is a linker connecting an amino acid side chain of said GLP-1 agonist with RR, RR is the remains of a reactive residue that has formed a covalent bond with an amino acid residue of the protraction protein, and protraction protein is a protein having a molar weight of at least 5 kDa, having a plasma halflife of at least 24 hours in human plasma , and said protraction protein has been synthesised by a non-mammalian organism or synthetically.

Description

NOVEL GLP-1 ANALOGUES LINKED TO ALBUMIN-LIKE AGENTS
FIELD OF THE INVENTION
The present invention relates to novel GLP-1 compounds, to pharmaceutical compositions comprising these compounds and to the use of the compounds for the treatment of diseases related to diabetes.
BACKGROUND OF THE INVENTION
Diabetes mellitus is a metabolic disorder in which the ability to utilize glucose is partly or completely lost. About 5% of all people suffer from diabetes and the disorder approaches epidemic proportions. Since the introduction of insulin in the 1920's, continuous efforts have been made to improve the treatment of diabetes mellitus.
One peptide expected to become very important in the treatment of diabetes is glucagon-like peptide-1 (GLP-1). Human GLP-1 is a 37 amino acid residue peptide originating from preproglucagon which is synthesized La. in the L-cells in the distal ileum, in the pancreas and in the brain. GLP-1 is an important gut hormone with regulatory function in glucose metabolism and gastrointestinal secretion and metabolism. GLP-1 stimulates insulin secretion in a glucose-dependant manner, stimulates insulin biosynthesis, promotes beta cell rescue, decreases glucagon secretion, gastric emptying and food intake. Human GLP-1 is hydrolysed to GLP-1 (7-37) and GLP-1 (7-36)-amide which are both insulinotropic peptides. A simple system is used to describe fragments and analogues of this peptide. Thus, for example, [Gly8]GLP-1 (7-37) designates an analogue of GLP-1 (7-37) formally derived from GLP-1 (7-37) by substituting the naturally occurring amino acid residue in position 8 (Ala) by Gly. Similarly, (Nε3 -tetradecanoyl)[Lys34]GLP-1(7-37) designates GLP-1 (7-37) wherein the ε- amino group of the Lys residue in position 34 has been tetradecanoylated. PCT publications WO 98/08871 and WO 99/43706 disclose stable derivatives of GLP-1 analogues, which have a lipophilic substituent. These stable derivatives of GLP-1 analogues have a protracted profile of action compared to the corresponding GLP-1 analogues. In the last decade a number of peptides have been isolated from the venom of the Gila monster lizards (Heloderma suspectum and Heloderma horridum). Exendin-4 is a 39 amino acid residue peptide isolated from the venom of Heloderma suspectum, and this peptide shares 52% homology with GLP-1 (7-37) in the overlapping region. Exendin-4 is a potent GLP-1 receptor agonist which has been shown to stimulate insulin release and ensuing lowering of the blood glucose level when injected into dogs. The group of exendin-4(1-39), certain fragments thereof, analogs thereof and derivatives thereof, are potent insulinotropic agents. Most importantly the group of exendin-4(1-39), insulinotropic fragments thereof, insulinotropic analogs thereof and insulinotropic derivatives thereof.
A range of GLP-1 compounds including exendin compounds have been synthesized and studied in particular in relation the plasma half-life. Low plasma halflifes may be due to chemical stability towards peptidases (mainly dipeptidyl aminopeptidase IV) and to renal clearance. However, these variants of insulionotropic peptides have hitherto not showed protracted effects beyond what will suffice for at product to be administered to the patient once daily. A second generation GLP-1 compounds are needed which can be administered to the patients only once weekly or even less frequently.
US 6,329,336 discloses the injection of highly reactive GLP-1 peptides into plasma, wherein chemical reactions will take palce with blood components, such as serum albumin. WO 02/46227 discloses fusion proteins between a GLP-1 compound and human serum albumin. WO 2003/103572 discloses conjugates of GLP-1 analogs and a blood component. It is an object of the present invention to provide GLP-1 analogues including exendin peptides linked to protein having a long half-life in human plasma, threreby facilitating a once-weekly treatment of patients. It is also an object of the present invention to provide GLP-1 peptides which are less prone to aggregation, a well known problem associated with the glucagon-like peptides. Being less prone to aggregation facilitates economical manufacturing processes as well as enabling the compounds to be administered by medical infusion pumps.
DEFINITIONS In the present specification, the following terms have the indicated meaning The term "polypeptide" and "peptide" as used herein means a compound composed of at least five constituent amino acids connected by peptide bonds. The constituent amino acids may be from the group of the amino acids encoded by the genetic code and they may natural amino acids which are not encoded by the genetic code, as well as synthetic amino acids. Natural amino acids which are not encoded by the genetic code are e.g. hydroxyproline, -carboxyglutamate, ornithine, phosphoserine, D-alanine and D-glutamine. Synthetic amino acids comprise amino acids manufactured by chemical synthesis, i.e. D- isomers of the amino acids encoded by the genetic code such as D-alanine and D-leucine, Aib (σ-aminoisobutyric acid), Abu (σ-aminobutyric acid), Tie (tert-butylglycine), ?-alanine, 3- aminomethyl benzoic acid, anthranilic acid. The term "analogue" as used herein referring to a polypeptide means a modified peptide wherein one or more amino acid residues of the peptide have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the peptide and/or wherein one or more amino acid residues have been deleted from the peptide and or wherein one or more amino acid residues have been added to the peptide. Such addition or deletion of amino acid residues can take place at the N-terminal of the peptide and/or at the C-terminal of the peptide. A simple system is often used to describe analogues : For example [Arg34]GLP-1 (7-37)Lys designates a GLP-1 (7-37) analogue wherein the naturally occuring lysine at position 34 has been substituted with arginine and wherein a lysine has been added to the terminal amino acid residue, i.e. to the Gly37. All amino acids for which the optical isomer is not stated is to be understood to mean the L-isomer. The term "derivative" as used herein in relation to a peptide means a chemically modified peptide or an analogue thereof, wherein at least one substituent is not present in the unmodified peptide or an analogue thereof, i.e. a peptide which has been covalently modified. Typical modifications are amides, carbohydrates, alkyl groups, acyl groups, esters and the like. An example of a derivative of GLP-1 (7-37) is Nε26-((4S)-4-(hexadecanoylamino)- butanoyl)[Arg34, Lys26] GLP-1 -(7-37). The term "GLP-1 agonist" as used herein means a compound which stimulates the formation of cAMP in a suitable medium containing the human GLP-1 receptor. The potency of a GLP-1 agonist is determined by calculating the EC50 value from the dose-response curve as described below. Baby hamster kidney (BHK) cells expressing the cloned human GLP-1 receptor (BHK- 467-12A) were grown in DMEM media with the addition of 100 lU/mL penicillin, 100 μg/mL streptomycin, 5% fetal calf serum and 0.5 mg/mL Geneticin G-418 (Life Technologies). The cells were washed twice in phosphate buffered saline and harvested with Versene. Plasma membranes were prepared from the cells by homogenisation with an Ultraturrax in buffer 1 (20 mM HEPES-Na, 10 mM EDTA, pH 7.4). The homogenate was centrifuged at 48,000 x g for 15 min at 4°C. The pellet was suspended by homogenization in buffer 2 (20 mM HEPES-Na, 0.1 mM EDTA, pH 7.4), then centrifuged at 48,000 x g for 15 min at 4°C. The washing procedure was repeated one more time. The final pellet was suspended in buffer 2 and used immediately for assays or stored at -80°C. The functional receptor assay was carried out by measuring cyclic AMP (cAMP) as a response to stimulation by the insulinotropic agent. cAMP formed was quantified by the AlphaScreen™ cAMP Kit (Perkin Elmer Life Sciences). Incubations were carried out in half- area 96-well microtiter plates in a total volume of 50 μL buffer 3 (50 mM Tris-HCI, 5 mM
HEPES, 10 mM MgCI2, pH 7.4) and with the following addiditions: 1 mM ATP, 1 μM GTP, 0.5 mM 3-isobutyl-1-methylxanthine (IBMX), 0.01 % Tween-20, 0.1% BSA, 6 μg membrane preparation, 15 μg/mL acceptor beads, 20μg/mL donor beads preincubated with 6 nM biotinyl-cAMP. Compounds to be tested for agonist activity were dissolved and diluted in buffer 3. GTP was freshly prepared for each experiment. The plate was incubated in the dark with slow agitation for three hours at room temperature followed by counting in the Fusion™ instrument (Perkin Elmer Life Sciences). Concentration-response curves were plotted for the individual compounds and EC50 values estimated using a four-parameter logistic model with
Prism v. 4.0 (GraphPad, Carlsbad, CA). The term "GLP-1 peptide" as used herein means GLP-1 (7-37) (SEQ ID No 2), a
GLP-1 (7-37) analogue, a GLP-1 (7-37) derivative or a derivative of a GLP-1 (7-37) analogue.
In one embodiment the GLP-1 peptide is an insulinotropic agent. The term "exendin-4 peptide" as used herein means exendin-4(1-39) (SEQ ID No 3), an exendin-4(1-39) analogue, an exendin-4(1-39) derivative or a derivative of an exendin- 4(1-39) analogue. In one embodiment the exendin-4 peptide is an insulinotropic agent. The term "DPP-IV protected" as used herein referring to a polypeptide means a polypeptide which has been chemically modified in order to render said compound resistant to the plasma peptidase dipeptidyl aminopeptidase-4 (DPP-IV). The DPP-IV enzyme in plasma is known to be involved in the degradation of several peptide hormones, e.g. GLP-1, GLP-2, Exendin-4 etc. Thus, a considerable effort is being made to develop analogues and derivatives of the polypeptides susceptible to DPP-IV mediated hydrolysis in order to reduce the rate of degradation by DPP-IV. In one embodiment a DPP-IV protected peptide is more resistant to
DPP-IV than GLP-1 (7-37) or Exendin-4(1-39).
Resistance of a peptide to degradation by dipeptidyl aminopeptidase IV is determined by the following degradation assay : Aliquots of the peptide (5 nmol) are incubated at 37 °C with 1 μL of purified dipeptidyl aminopeptidase IV corresponding to an enzymatic activity of 5 mU for 10-180 minutes in 100 μL of 0.1 M triethylamine-HCI buffer, pH 7.4. Enzymatic reactions are terminated by the addition of 5 μL of 10% trifluoroacetic acid, and the peptide degradation products are separated and quantified using HPLC analysis. One method for performing this analysis is : The mixtures are applied onto a Vydac C18 widepore (30 nm pores, 5 μm particles) 250 x 4.6 mm column and eluted at a flow rate of 1 ml/min with linear stepwise gradients of acetonitrile in 0.1% trifluoroacetic acid (0% acetonitrile for 3 min, 0-24% acetonitrile for 17 min, 24-48% acetonitrile for 1 min) according to Siegel et al., Regul. Pept. 1999;79:93-102 and Mentlein et al. Eur. J. Biochem. 1993;214:829-35. Peptides and their degradation products may be monitored by their absorbance at 220 nm (peptide bonds) or 280 nm (aromatic amino acids), and are quantified by integration of their peak areas related to those of standards. The rate of hydrolysis of a peptide by dipeptidyl aminopeptidase IV is estimated at incubation times which result in less than 10% of the peptide being hydrolysed. The term "C1-6-alkyl" as used herein means a saturated, branched, straight or cyclic hydrocarbon group having from 1 to 6 carbon atoms. Representative examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, te/t-butyl, n- pentyl, isopentyl, neopentyl, ferf-pentyl, n-hexyl, isohexyl, cyclohexane and the like. The term "pharmaceutically acceptable" as used herein means suited for normal pharmaceutical applications, i.e. giving rise to no adverse events in patients etc. The term "excipient" as used herein means the chemical compounds which are normally added to pharmaceutical compositions, e.g. buffers, tonicity agents, preservatives and the like. The term "effective amount" as used herein means a dosage which is sufficient to be effective for the treatment of the patient compared with no treatment.
The term "pharmaceutical composition" as used herein means a product comprising an active compound or a salt thereof together with pharmaceutical excipients such as buffer, preservative, and optionally a tonicity modifier and/or a stabilizer. Thus a pharmaceutical composition is also known in the art as a pharmaceutical formulation. The term "treatment of a disease" as used herein means the management and care of a patient having developed the disease, condition or disorder. The purpose of treatment is to combat the disease, condition or disorder. Treatment includes the administration of the active compounds to eliminate or control the disease, condition or disorder as well as to alleviate the symptoms or complications associated with the disease, condition or disorder. DESCRIPTION OF THE INVENTION
In one aspect the present invention relates to a compound having the structure of the formula (I) : GLP-1 agonist - L - RR - protraction protein (I) wherein
GLP-1 agonist is a polypeptide which is an agonist of the human GLP-1 receptor,
L is a linker connecting an amino acid side chain of said GLP-1 agonist or the C-terminal amino acid residue of said GLP-1 agonist with RR,
RR is the remains of a reactive residue that has formed a covalent bond with an amino acid residue of the protraction protein, and protraction protein is a protein having a molar weight of at least 5 kDa, having a plasma half- life of at least 24 hours in human plasma, and said protraction protein has been synthesised by a non-mammalian organism or synthetically.
The compounds encompassed by formula (I) is illustrated by the following illustration :
maleimide
vinylsulfone
Figure imgf000008_0001
In one embodiment of the invention the protraction protein is recombinant human serum albumin (SEQ ID NO 1). In another embodiment of the invention the protraction protein is a human serum albumin variant. In another embodiment of the invention the human serum albumin variant has reduced binding affinities towards copper and nickel as compared to the corresponding binding affinities of human serum albumin towards copper and nickel. In another embodiment of the invention the protraction protein is an N-terminal fragment of human serum albumin, or an analogue thereof. In another embodiment of the invention the protraction protein is a human serum albumin variant comprising a modification of the Asp-Ala-His-Lys N-terminal sequence. In another embodiment of the invention the protraction protein comprises at least one deletion among the three N-terminal amino acid residues Asp-Ala-His. In another embodiment of the invention the protraction protein comprises an N- terminal extension, such as Glu"3,Ala"2Glu"1,Phe°-HSA(1-585) or an N-terminal fragment thereof. In another embodiment of the invention the human serum albumin (HSA) variant is selected from the group consisting of HSA(2-585), HSA(3-585), HSA(4-585), Asp-Ala- HSA(4-585), Xaa3-HSA(1-585) where Xaa3 is an amino acid residue which has substituted the His residue occupying position 3 in native HSA, and N-terminal fragments thereof. A recombinant human serum albumin variant is commercially available from New Century Pharma under the name Albagen. Albagen is HSA(2-585) and is hypoallergenic due to the modified metal binding properties caused by the single N-terminal deletion. In another embodiment of the invention the said protration protein comprises an amino acid sequence of from 60-200 such as from 100 to 150 amino acid residues, and said amino acid sequence being identical to a fragment of SEQ ID NO 1 or a fragment of SEQ ID NO 1 with one or two amino acid substitutions and/or deletions. In another embodiment of the invention the protraction protein is the Fc portion of an immunoglobulin, an analogue or a fragment thereof. In another embodiment of the invention the GLP-1 agonist has at least 50% amino acid homology with either GLP-1 (7-37) (SEQ ID NO 2) or Exendin-4(1-39) (SEQ ID NO 3). In another embodiment of the invention the GLP-1 agonist has at least 80% amino acid homology with either GLP-1(7-37) (SEQ ID NO 2) or Exendin-4(1-39) (SEQ ID NO 3). In another embodiment of the invention the GLP-1 agonist comprises the amino acid sequence of the formula (II):
Xaa7-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Xaa16-Ser-Xaa18-Xaa19-Xaa2o-Glu-Xaa22-Xaa23-Ala- Xaa25-Xaa26-Xaa27-Phe-lle-Xaa3o-Trp-Leu-Xaa33-Xaa34-Xaa35-Xaa36-Xaa37-Xaa38-Xaa39-Xaa4o- Xaa4-)-Xaa42-Xaa43-Xaa44-Xaa45-Xaa46 Formula (II) (SEQ ID No: 4) wherein Xaa7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, ?-hydroxy-histidine, homohistidine, N°-acetyl-histidine, σ-fluoromethyl-histidine, σ-methyl-histidine, 3- pyridylalanine, 2-pyridylalanine or 4-pyridylalanine;
Xaa8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid;
Xaa16 is Val or Leu;
Xaa18 is Ser, Lys or Arg;
Xaaig is Tyr or Gin;
Xaa20 is Leu or Met;
Xaa22 is Gly, Glu or Aib;
Xaa23 is Gin, Glu, Lys or Arg;
Xaa25 is Ala or Val;
Xaa26 is Lys, Glu or Arg;
Xaa27 is Glu or Leu;
Xaa30 is Ala, Glu or Arg;
Xaa33 is Val or Lys;
Xaa34 is Lys, Glu, Asn or Arg;
Xaa35 is Gly or Aib;
Xaa36 is Arg, Gly or Lys;
Xaa37 is Gly, Ala, Glu, Pro, Lys, amide or is absent;
Xaa38 is Lys, Ser, amide or is absent.
Xaa39 is Ser, Lys, amide or is absent;
Xaa 0 is Gly, amide or is absent;
Xaa41 is Ala, amide or is absent;
Xaa 2 is Pro, amide or is absent;
Xaa43 is Pro, amide or is absent;
Xaa^ is Pro, amide or is absent;
Xaa45 is Ser, amide or is absent;
Xaa46 is amide or is absent ; provided that if Xaa38, Xaa39, Xaa40, Xaa41, Xaa42, Xaa 3, Xaa44, Xaa45 orXaa46 is absent then each amino acid residue downstream is also absent.
In another embodiment of the invention the GLP-1 agonist comprises the amino acid sequence of formula (III): Xaa7-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Xaa18-Tyr-Leu-Glu-Xaa22-Xaa23-Ala-Ala- Xaa^-Glu-Phe-lle-Xaaso-Trp-Leu-Val-Xaa^-XaasEs-Xaase-Xaas^Xaass Formula (III) (SEQ ID No: 5) wherein
Xaa7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, -hydroxy-histidine, homohistidine, Nσ-acetyl-histidine, -fluoromethyl-histidine, σ-methyl-histidine,* 3- pyridylalanine, 2-pyridylalanine or 4-pyridylalanine;
Xaa8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, or (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid;
Figure imgf000011_0001
Xaa2 s Gly, Glu or Aib; Xaa 3 s Gin, Glu, Lys or Arg;
Figure imgf000011_0002
Xaa30 s Ala, Glu or Arg; Xaa34 s Lys, Glu or Arg;
Figure imgf000011_0003
Xaa37 s Gly, Ala, Glu or Lys; Xaa38 s Lys, amide or is absent.
In another embodiment of the invention the said GLP-1 agonist is dipeptidyl aminopeptidase IV protected. In another embodiment of the invention the GLP-1 agonist is hydrolysed by DPP-IV at a rate lower than the rate of hydrolysis of GLP-1 (7-37) using the DPP-IV hydrolysis assay disclosed herein. In another embodiment of the invention the GLP-1 agonist is a position 8 analogue, i.e. the alanine residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No: 2) has been substituted by another amino acid residue. In another embodiment of the invention the GLP-1 agonist comprises an Aib residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No:2). In another embodiment of the invention the amino acid residue in position 7 of the GLP-1 peptide (the N-terminal) is selected from the group consisting of D-histidine, desamino-histidine, 2-amino-histidine, Miydroxy-histidine, homohistidine, N°-acetyl-histidine , σ-fluoromethyl-histidine, σ-methyl-histidine, 3-pyridylalanine, 2-pyridylalanine and 4- pyridylalanine. In another embodiment of the invention the GLP-1 agonist comprises no more than twelve amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist comprises no more than six amino acid residues which have been exchanged, added or deleted as compared to GLP- 1(7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist comprises no more than four amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist comprises no more than 4 amino acid residues which are not encoded by the genetic code. In another embodiment of the invention the GLP-1 agonist comprises no more than two amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3). In another embodiment of the invention the GLP-1 agonist is selected from the group consisting of [Arg34]GLP-1(7-37), [Arg26'34]GLP-1(7-37)Lys, [Lys36Arg26'34]GLP-1(7-36), [Aib8'22'35]GLP-1 (7-37),
[Aib8'35]GLP-1 (7-37), [Aib8'22]GLP-1 (7-37), [Aib8'22'35 Arg26'34]GLP-1 (7-37)Lys, [Aib8'35 Arg26'34]GLP-1 (7-37)Lys, [Aib8'22Arg 6'34]GLP-1 (7-37)Lys, [Aib8'22'35 Arg26'34]GLP-1 (7-37)Lys, [Aib8'35 Arg26'34]GLP-1 (7-37)Lys,
[Aib8'2235 Arg26]GLP-1 (7-37)Lys, [Aib8'35 Arg26]GLP-1 (7-37)Lys, [Aib8'22 Arg26] GLP-1 (7-37)Lys, [Aib8'22'35 Arg34]GLP-1 (7-37)Lys, [Aib8'35Arg34]GLP-1 (7-37)Lys, [Aib8'22Arg34]GLP-1 (7-37)Lys, [Aib8'22'35Ala37]GLP-1 (7-37)Lys, [Aib8'35Ala37]GLP-1 (7-37)Lys, [Aib8'22Ala37]GLP-1 (7-37)Lys, [Aib8'22'35 Lys37]GLP-1 (7-37), [Aib8'35Lys37]GLP-1 (7-37) and [Aib8'22Lys37]GLP-1(7-37). In another embodiment of the invention the GLP-1 agonist is Exendin-4(1-39) (SEQ
ID No. 3). In another embodiment of the invention the GLP-1 agonist is ZP-10, i.e. [Ser38Lys39]Exendin-4(1-39)LysLysLysLysLys-amide (SEQ ID No. 4). In another embodiment of the invention the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the amino acid residue in position 23, 26, 34, 36 or 38 relative to the amino acid sequence SEQ ID No:2 (GLP-1 (7-37)), (corresponding to position 17, 20, 28, 30 or 32 relative to amino acid sequence SEQ ID No:3(Exendin-4(1-39)). In another embodiment of the invention the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the C-terminal amino acid residue. In another embodiment of the invention the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of an amino acid residue selected from arginine, lysine, cysteine, glutamic acid, aspartic acid, histidine, serine, threonine and tyrosine. In another embodiment of the invention the GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of a cysteine residue. In another embodiment of the invention the linker L is selected from the group consisting of the bivalent connecting chemical groups amides: -C(O)-NR- where R is hydrogen or C1-6-alkyl, amine: -NR-, where R is hydrogen or C1-6-alkyI,
thioethers: -S-, -S-(CH2)2-SO2-
Figure imgf000013_0001
ethers: -O-, urethanes: -N(R )-CO-N(R2)-, where R1 and R2 independently is hydrogen or C1-6-alkyl, carbamates: -O-CO-N(R)-, where R is hydrogen or C1-6-alkyl, — N— = ?C — hydrazines: H where R is hydrogen or C1-6-alkyl, oximes: -O-N=C(-R)-, where R is hydrogen or C1-6-alkyl, oxazolidines or thiazolidines:
Figure imgf000013_0002
In another embodiment of the invention the compound of general formula (I) is selected from the group consisting of GLP-1 agonist - C(=O)CH2O(CH2)2O(CH2)2- RR - protraction protein,
GLP-1 agonist - C(=O)(CH2)n(OCH2CH2)m- RR - protraction protein,
GLP-1 agonist - S(=O)2(CH2)n(OCH2CH2)m- RR - protraction protein,
GLP-1 agonist - CH2(CH2)n(OCH2CH2)m- RR - protraction protein,
GLP-1 agonist - C(=O)O(CH2)n(OCH2CH2)m- RR - protraction protein, wherein n is an interger in the range from 0 to 10, and m is an integer in the range from 0 to
100.
In another embodiment of the invention the compound of general formula (I) is selected from the group consisting of
GLP-1 agonist - L - NC(=O)CH2- sulphur in cysteine residue in protraction protein, GLP-1 agonist- L- S(=O)2(CH2)2- sulphur in cysteine residue in protraction protein,
GLP-1 agonist - L - NC(=O)CH2- sulphur in cysteine residue in protraction protein, and in cysteine in protraction protein
Figure imgf000014_0001
In another embodiment of the invention the compound of the general formula (I) is selected from the group consisting of S-gamma3 -(1 -{2-[2-(2-([D-Ala8, Lys37]-GLP-1 -(7- 37)amide-Nε37-yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
— H—
Figure imgf000014_0002
S-gamma34-(1-{2-[2-(2-([Aib8'22'25, Lys37]-GLP-1-(7-37)amide-Nε37- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
Figure imgf000014_0003
S-gamma34-((1 -{2-[2-(2-([ Aib8,Arg26,34,Glu22,23,30]-GLP-1 -(7-37))Lys amide-Nε- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
Figure imgf000014_0004
The compounds of the present invention can be produced by classical peptide synthesis, e.g. solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques., see e.g. Green and Wuts, "Protecting Groups in Organic Synthesis", John Wiley & Sons, 1999. These methods are preferred when the insulinotropic agent is a peptide comprising non-natural amino acid residues. When the insulinotropic agent is a polypeptide comprising only amino acid residues encoded by the genetic code, the polypeptides can also be produced by a method which comprises cultuhng a host cell containing a DNA sequence encoding the polypeptide and capable of expressing the polypeptide in a suitable nutrient medium under conditions permitting the expression of the peptide, after which the resulting peptide is recovered from the culture and then derivatized to the compound of formula (I). The medium used to culture the cells may be any conventional medium suitable for growing the host cells, such as minimal or complex media containing appropriate supplements. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection). The peptide produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration. For extracellular products the proteinaceous components of the supernatant are isolated by filtration, column chromatography or precipitation, e.g. microfiltation, ultrafiltration, isoelectric precipitation, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, hydrophobic interaction chromatography, gel filtration chromatography, affinity chromatography, or the like, dependent on the type of polypeptide in question. For intracellular or periplasmic products the cells isolated from the culture medium are disintegrated or permeabilised and extracted to recover the product polypeptide or precursor thereof. The DNA sequence encoding the therapeutic polypeptide may suitably be of genomic or cDNA origin, for instance obtained by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the peptide by hybridisation using synthetic oligonucleotide probes in accordance with standard techniques (see, for example, Sambrook, J, Fritsch, EF and Maniatis, T, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor
Laboratory Press, New York, 1989). The DNA sequence encoding the polypeptide may also be prepared synthetically by established standard methods, e.g. the phosphoamidite method described by Beaucage and Caruthers, Tetrahedron Letters 22 (1981), 1859 - 1869, or the method described by Matthes et al., EMBO Journal 3 (1984), 801 - 805. The DNA sequence may also be prepared by polymerase chain reaction using specific primers, for instance as described in US 4,683,202 or Saiki et al., Science 239 (1988), 487 - 491. The DNA sequence may be inserted into any vector which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced. Thus, the vector may be an autonomously replicating vector, i.e. a vector which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g. a plasmid. Alternatively, the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated. The vector is preferably an expression vector in which the DNA sequence encoding the polypeptide is operably linked to additional segments required for transcription of the DNA, such as a promoter. The promoter may be any DNA sequence which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell. Examples of suitable promoters for directing the transcription of the DNA encoding the peptide of the invention in a variety of host cells are well known in the art, cf. for instance Sambrook et. al., supra. The DNA sequence encoding the polypeptide may also, if necessary, be operably connected to a suitable terminator, polyadenylation signals, transcriptional enhancer sequences, and translational enhancer sequences. The recombinant vector of the invention may further comprise a DNA sequence enabling the vector to replicate in the host cell in question. The vector may also comprise a selectable marker, e.g. a gene the product of which complements a defect in the host cell or one which confers resistance to a drug, e.g. ampicillin, kanamycin, tetracyclin, chloramphenicol, neomycin, hygromycin or methotrexate. For large scale manufacture the selectable marker preferably is not antibiotic resistance, e.g. antibiotic resistance genes in the vector are preferably excised when the vector is used for large scale manufacture. Methods for eliminating antibiotic resistance genes from vectors are known in the art, see e.g. US 6,358,705 which is incorporated herein by reference. To direct a parent peptide of the present invention into the secretory pathway of the host cells, a secretory signal sequence (also known as a leader sequence, prepro sequence or pre sequence) may be provided in the recombinant vector. The secretory signal sequence is joined to the DNA sequence encoding the peptide in the correct reading frame. Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide. The secretory signal sequence may be that normally associated with the peptide or may be from a gene encoding another secreted protein. The procedures used to ligate the DNA sequences coding for the present peptide, the promoter and optionally the terminator and/or secretory signal sequence, respectively, and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (cf., for instance, Sambrook et. al.., supra). The host cell into which the DNA sequence or the recombinant vector is introduced may be any cell which is capable of producing the present peptide and includes bacteria, yeast, fungi and higher eukaryotic cells. Examples of suitable host cells well known and used in the art are, without limitation, E. coli, Saccharomyces cerevisiae, or mammalian BHK or CHO cell lines. Pharmaceutical compositions containing a compound according to the present invention may be prepared by conventional techniques, e.g. as described in Remington's Pharmaceutical Sciences, 1985 or in Remington: The Science and Practice of Pharmacy, 19th edition, 1995. One object of the present invention is to provide a pharmaceutical formulation comprising a compound according to the present invention which is present in a concentration from about 0.1 mg/ml to about 25 mg/ml, and wherein said formulation has a pH from 2.0 to 10.0. The formulation may further comprise a buffer system, preservative(s), isotonicity agent(s), chelating agent(s), stabilizers and surfactants. In one embodiment of the ; invention the pharmaceutical formulation is an aqueous formulation, i.e. formulation comprising water. Such formulation is typically a solution or a suspension. In a further embodiment of the invention the pharmaceutical formulation is an aqueous solution. The term "aqueous formulation" is defined as a formulation comprising at least 50 %w/w water. Likewise, the term "aqueous solution" is defined as a solution comprising at least 50 %w/w water, and the term "aqueous suspension" is defined as a suspension comprising at least 50 %w/w water. In another embodiment the pharmaceutical formulation is a freeze-dried formulation, whereto the physician or the patient adds solvents and/or diluents prior to use. In another embodiment the pharmaceutical formulation is a dried formulation (e.g. freeze-dried or spray-dried) ready for use without any prior dissolution. In a further aspect the invention relates to a pharmaceutical formulation comprising an aqueous solution of a compound according to the present invention, and a buffer, wherein said compound is present in a concentration from 0.1 mg/ml or above, and wherein said formulation has a pH from about 2.0 to about 10.0. In another embodiment of the invention the pH of the formulation is from about 7.0 to about 9.5. In another embodiment of the invention the pH of the formulation is from about 3.0 to about 7.0. In another embodiment of the invention the pH of the formulation is from about 5.0 to about 7.5. In another embodiment of the invention the pH of the formulation is from about 7.5 to about 9.0. In another embodiment of the invention the pH of the formulation is from about.7.5 to about 8.5. In another embodiment of the invention the pH of the formulation is from about 6.0 to about 7.5. In another embodiment of the invention the pH of the formulation is from about 6.0 to about 7.0. In another embodiment of the invention the pH of the formulation is from about 3.0 to about 9.0, and said pH is at least 2.0 pH units from the isoelectric pH of compound of the present invention.
In a further embodiment of the invention the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginin, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof. Each one of these specific buffers constitutes an alternative embodiment of the invention.
In a further embodiment of the invention the formulation further comprises a pharmaceutically acceptable preservative. In a further embodiment of the invention the preservative is selected from the group consisting of phenol, o-cresol, m-cresol, p-cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p-hydroxybenzoate, benzethonium chloride, chlorphenesine (3p-chlorphenoxypropane-1 ,2-diol) or mixtures thereof. In a further embodiment of the invention the preservative is present in a concentration from 0.1 mg/ml to 20 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 0.1 mg/ml to 5 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 5 mg/ml to 10 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 10 mg/ml to 20 mg/ml. Each one of these specific preservatives constitutes an alternative embodiment of the invention. The use of a preservative in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995. In a further embodiment of the invention the formulation further comprises an isotonic agent. In a further embodiment of the invention the isotonic agent is selected from the group consisting of a salt (e.g. sodium chloride), a sugar or sugar alcohol, an amino acid (e.g. L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine), an alditol (e.g. glycerol (glycerine), 1 ,2-propanedioI (propyleneglycol), 1,3-propanediol, 1,3- butanediol) polyethyleneglycol (e.g. PEG400), or mixtures thereof. Any sugar such as mono-, di-, or polysaccharides, or water-soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, soluble starch, hydroxyethyl starch and carboxymethylcellulose-Na may be used. In one embodiment the sugar additive is sucrose. Sugar alcohol is defined as a C4-C8 hydrocarbon having at least one -OH group and includes, for example, mannitol, sorbitol, inositol, galacititol, dulcitol, xylitol, and arabitol. In one embodiment the sugar alcohol additive is mannitol. The sugars or sugar alcohols mentioned above may be used individually or in combination. There is no fixed limit to the amount used, as long as the sugar or sugar alcohol is soluble in the liquid preparation and does not adversely effect the stabilizing effects achieved using the methods of the invention. In one embodiment, the sugar or sugar alcohol concentration is between about 1 mg/ml and about 150 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 1 mg/ml to 50 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 1 mg/ml to 7 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 8 mg/ml to 24 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 25 mg/ml to 50 mg/ml. Each one of these specific isotonic agents constitutes an alternative embodiment of the invention. The use of an isotonic agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995.
In a further embodiment of the invention the formulation further comprises a chelating agent. In a further embodiment of the invention the chelating agent is selected from salts of ethylenediaminetetraacetic acid (EDTA), citric acid, and aspartic acid, and mixtures thereof. In a further embodiment of the invention the chelating agent is present in a concentration from 0.1 mg/ml to 5mg/ml. In a further embodiment of the invention the chelating agent is present in a concentration from 0.1 mg/ml to 2mg/ml. In a further embodiment of the invention the chelating agent is present in a concentration from 2mg/ml to 5mg/ml. Each one of these specific chelating agents constitutes an alternative embodiment of the invention. The use of a chelating agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995. In a further embodiment of the invention the formulation further comprises a stabiliser. The use of a stabilizer in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995. More particularly, compositions of the invention are stabilized liquid pharmaceutical compositions whose therapeutically active components include a polypeptide that possibly exhibits aggregate formation during storage in liquid pharmaceutical formulations. By "aggregate formation" is intended a physical interaction between the polypeptide molecules that results in formation of oligomers, which may remain soluble, or large visible aggregates that precipitate from the solution. By "during storage" is intended a liquid pharmaceutical composition or formulation once prepared, is not immediately administered to a subject. Rather, following preparation, it is packaged for storage, either in a liquid form, in a frozen state, or in a dried form for later reconstitution into a liquid form or other form suitable for administration to a subject. By "dried form" is intended the liquid pharmaceutical composition or formulation is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and Polli (1984) J. Parenteral Sci. Technol. 38:48-59), spray drying (see Masters (1991) in Spray-Drying Handbook (5th ed; Longman Scientific and Technical, Essez, U.K.), pp. 491- 676; Broadhead et al. (1992) Drug Devel. Ind. Pharm. 18:1169-1206; and Mumenthaler et al. (1994) Pharm. Res. 11:12-20), or air drying (Carpenter and Crowe (1988) Cryobiology 25:459-470; and Roser (1991) Biopharm. 4:47-53). Aggregate formation by a polypeptide during storage of a liquid pharmaceutical composition can adversely affect biological activity of that polypeptide, resulting in loss of therapeutic efficacy of the pharmaceutical composition. Furthermore, aggregate formation may cause other problems such as blockage of tubing, membranes, or pumps when the polypeptide-containing pharmaceutical composition is administered using an infusion system. The pharmaceutical compositions of the invention may further comprise an amount of an amino acid base sufficient to decrease aggregate formation by the polypeptide during storage of the composition. By "amino acid base" is intended an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. Where a combination of amino acids is used, all of the amino acids may be present in their free base forms, all may be present in their salt forms, or some may be present in their free base forms while others are present in their salt forms. In one embodiment, amino acids to use in preparing the compositions of the invention are those carrying a charged side chain, such as arginine, lysine, aspartic acid, and glutamic acid. Any stereoisomer (i.e., L, D, or DL isomer) of a particular amino acid (e.g. glycine, methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof) or combinations of these stereoisomers, may be present in the pharmaceutical compositions of the invention so long as the particular amino acid is present either in its free base form or its salt form. In one embodiment the L-stereoisomer is used. Compositions of the invention may also be formulated with analogues of these amino acids. By "amino acid analogue" is intended a derivative of the naturally occurring amino acid that brings about the desired effect of decreasing aggregate formation by the polypeptide during storage of the liquid pharmaceutical compositions of the invention. Suitable arginine analogues include, for example, aminoguanidine, ornithine and N-monoethyl L-arginine, suitable methionine analogues include S-ethyl homocysteine and S-butyl homocysteine and suitable cystein analogues include S-methyl-L cystein. As with the other amino acids, the amino acid analogues are incorporated into the compositions in either their free base form or their salt form: In a further embodiment of the invention the amino acids or amino acid analogues are used in a concentration, which is sufficient to prevent or delay aggregation of the protein. In a further embodiment of the invention methionine (or other sulphur containing amino acids or amino acid analogous) may be added to inhibit oxidation of methionine residues to methionine sulfoxide when the polypeptide acting as the therapeutic agent is a polypeptide comprising at least one methionine residue susceptible to such oxidation. By "inhibit" is intended minimal accumulation of methionine oxidized species over time. Inhibiting methionine oxidation results in greater retention of the polypeptide in its proper molecular form. Any stereoisomer of methionine (L, D, or DL isomer) or combinations thereof can be used. The amount to be added should be an amount sufficient to inhibit oxidation of the methionine residues such that the amount of methionine sulfoxide is acceptable to regulatory agencies. Typically, this means that the composition contains no more than about 10% to about 30% methionine sulfoxide. Generally, this can be achieved by adding methionine such that the ratio of methionine added to methionine residues ranges from about 1:1 to about 1000:1, such as 10: 1 to about 100: 1. In a further embodiment of the invention the formulation further comprises a stabiliser selected from the group of high molecular weight polymers or low molecular compounds. In a further embodiment of the invention the stabilizer is selected from polyethylene glycol (e.g. PEG 3350), polyvinylalcohol (PVA), polyvinylpyrrolldone, carboxy- /hydroxycellulose or derivates thereof (e.g. HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, sulphur-containing substances as monothioglycerol, thioglycolic acid and 2- methylthioethanol, and different salts (e.g. sodium chloride). Each one of these specific stabilizers constitutes an alternative embodiment of the invention. The pharmaceutical compositions may also comprise additional stabilizing agents, which further enhance stability of a therapeutically active polypeptide therein. Stabilizing agents of particular interest to the present invention include, but are not limited to, methionine and EDTA, which protect the polypeptide against methionine oxidation, and a nonionic surfactant, which protects the polypeptide against aggregation associated with freeze-thawing or mechanical shearing.
In a further embodiment of the invention the formulation further comprises a ; surfactant. In a further embodiment of the invention the surfactant is selected from a detergent, ethoxylated castor oil, polyglycolyzed glycehdes, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypropylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic® F68, poloxamer 188 and 407, Triton X-100 ), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g. Tween-20, Tween-40, Tween-80 and Brij-35), monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lecitins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmitoyl phosphatidic acid) and lysophospholipids (eg. palmitoyl lysophosphatidyl-L-serine and 1-acyl-sn-glycero-3- phosphate esters of ethanolamine, choline, serine or threonine) and alkyl, alkoxyl (alkyl ester), alkoxy (alkyl ether)- derivatives of lysophosphatidyl and phosphatidylcholines, e.g. lauroyl and myristoyl derivatives of lysophosphatidylcholine, dipalmitoylphosphatidylcholine, and modifications of the polar head group, that is cholines, ethanolamines, phosphatidic acid, serines, threonines, glycerol, inositol, and the positively charged DODAC, DOTMA, DCP, BISHOP, lysophosphatidylserine and lysophosphatidylthreonine, and glycerophospholipids (eg. cephalins), glyceroglycolipids (eg. galactopyransoide), sphingoglycolipids (eg. ceramides, gangliosides), dodecylphosphocholine, hen egg lysolecithin, fusidic acid derivatives- (e.g. sodium tauro-dihydrofusidate etc.), long-chain fatty acids and salts thereof C6-C12 (eg. oleic acid and caprylic acid), acylcamitines and derivatives, Nα-acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, Nα-acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, Nα-acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, DSS (docusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49- 4]), docusate potassium, CAS registry no [7491-09-0]), SDS (sodium dodecyl sulfate or sodium lauryl sulfate), sodium caprylate, cholic acid or derivatives thereof, bile acids and salts thereof and glycine or taurine conjugates, ursodeoxycholic acid, sodium cholate, sodium deoxycholate, sodium taurocholate, sodium glycocholate, N-hexadecyl-N,N-dimethyl- 3-ammonio-1-propanesulfonate, anionic (alkyl-aryl-sulphonates) monovalent surfactants, zwitterionic surfactants (e.g. N-alkyl-N,N-dimethylammonio-1-propanesulfonates, 3- cholamido-1-propyldimethylammonio-1-propanesulfonate, cationic surfactants (quarternary ammonium bases) (e.g. cetyl-trimethylammonium bromide, cetylpyridinium chloride), non- ionic surfactants (eg. dodecyl β-D-glucopyranoside), poloxamines (eg. Tetronic's), which are tetrafunctional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention. The use of a surfactant in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995.
It is possible that other ingredients may be present in the peptide pharmaceutical formulation of the present invention. Such additional ingredients may include wetting agents, emulsifiers, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous vehicles, proteins (e.g., human serum albumin, gelatin or proteins) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine). Such additional ingredients, of course, should not adversely affect the overall stability of the pharmaceutical formulation of the present invention.
Pharmaceutical compositions containing a compound according to the present invention may be administered to a patient in need of such treatment at several sites, for example, at topical sites, for example, skin and mucosal sites, at sites which bypass absorption, for example, administration in an artery, in a vein, in the heart, and at sites which involve absorption, for example, administration in the skin, under the skin, in a muscle or in the abdomen. Administration of pharmaceutical compositions according to the invention may be through several routes of administration, for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment. In one aspect the present invention relates to a pharmaceutical composition comprising a compound according to Formula (I), and a pharmaceutically acceptable excipient. In one embodiment the pharmaceutical composition is suited for pulmonary administration. In another aspect the present invention relates to the use of a compound of formula (I) for the preparation of a pulmonary medicament. Compositions of the current invention may be administered in several dosage forms, for example, as solutions, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinses, capsules, for example, hard gelatine capsules and soft gelatine capsules, suppositories, rectal capsules, drops, gels, sprays, powder, aerosols, inhalants, eye drops, ophthalmic ointments, ophthalmic rinses, vaginal pessaries, vaginal rings, vaginal ointments, injection solution, in situ transforming solutions, for example in situ gelling, in situ setting, in situ precipitating, in situ crystallization, infusion solution, and implants. Compositions of the invention may further be compounded in, or attached to, for example through covalent, hydrophobic and electrostatic interactions, a drug carrier, drug delivery system and advanced drug delivery system in order to further enhance stability of the compound, increase bioavailability, increase solubility, decrease adverse effects, achieve chronotherapy well known to those skilled in the art, and increase patient compliance or any combination thereof. Examples of carriers, drug delivery systems and advanced drug delivery systems include, but are not limited to, polymers, for example cellulose and derivatives, polysaccharides, for example dextran and derivatives, starch and derivatives, poly(vinyl alcohol), acrylate and methacrylate polymers, polylactic and polyglycolic acid and block co-polymers thereof, polyethylene glycols, carrier proteins, for example albumin, gels, for example, thermogelling systems, for example block co-polymeric systems well known to those skilled in the art, micelles, liposomes, microspheres, nanoparticulates, liquid crystals and dispersions thereof, L2 phase and dispersions there of well known to those skilled in the art of phase behaviour in lipid-water systems, polymeric micelles, multiple emulsions, self- emulsifying, self-microemulsifying, cyclodextrins and derivatives thereof, and dendrimers. Compositions of the current invention are useful in the formulation of solids, semisolids, powder and solutions for pulmonary administration of the compound, using, for example a metered dose inhaler, dry powder inhaler and a nebulizer, all being devices well known to those skilled in the art. Compositions of the current invention are specifically useful in the formulation of controlled, sustained, protracting, retarded, and slow release drug delivery systems. More specifically, but not limited to, compositions are useful in formulation of parenteral controlled release and sustained release systems (both systems leading to a many-fold reduction in number of administrations), well known to those skilled in the art. Even more preferably, are controlled release and sustained release systems administered subcutaneous. Without limiting the scope of the invention, examples of useful controlled release system and compositions are hydrogels, oleaginous gels, liquid crystals, polymeric micelles, microspheres, nanoparticles, Methods to produce controlled release systems useful for compositions of the current invention include, but are not limited to, crystallization, condensation, co- cystallization, precipitation, co-precipitation, emulsification, dispersion, high pressure homogenization, encapsulation, spray drying, microencapsulation, coacervation, phase separation, solvent evaporation to produce microspheres, extrusion and supercritical fluid processes. General reference is made to Handbook of Pharmaceutical Controlled Release (Wise, D.L., ed. Marcel Dekker, New York, 2000) and Drug and the Pharmaceutical Sciences vol. 99: Protein Formulation and Delivery (MacNally, E.J., ed. Marcel Dekker, New York, 2000). Parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe. Alternatively, parenteral administration can be performed by means of an infusion pump. A further option is a composition which may be a solution or suspension for the administration of the compound according to the present invention in the form of a nasal or pulmonal spray. As a still further option, the pharmaceutical compositions containing the compound of the invention can also be adapted to transdermal administration, e.g. by needle-free injection or from a patch, optionally an iontophoretic patch, or transmucosal, e.g. buccal, administration. The term "stabilized formulation" refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability. The term "physical stability" of the protein formulation as used herein refers to the tendency of the protein to form biologically inactive and/or insoluble aggregates of the protein as a result of exposure of the protein to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces. Physical stability of the aqueous protein formulations is evaluated by means of visual inspection and/or turbidity measurements after exposing the formulation filled in suitable containers (e.g. cartridges or vials) to mechanical/physical stress (e.g. agitation) at different temperatures for various time periods. Visual inspection of the formulations is performed in a sharp focused light with a dark background. The turbidity of the formulation is characterized by a visual score ranking the degree of turbidity for instance on a scale from 0 to 3 (a formulation showing no turbidity corresponds to a visual score 0, and a formulation showing visual turbidity in daylight corresponds to visual score 3). A formulation is classified physical unstable with respect to protein aggregation, when it shows visual turbidity in daylight. Alternatively, the turbidity of the formulation can be evaluated by simple turbidity measurements well-known to the skilled person. Physical stability of the aqueous protein formulations can also be evaluated by using a spectroscopic agent or probe of the conformational status of the protein. The probe is preferably a small molecule that preferentially binds to a non-native conformer of the protein. One example of a small molecular spectroscopic probe of protein structure is Thioflavin T. Thioflavin T is a fluorescent dye that has been widely used for the detection of amyloid fibrils. In the presence of fibrils, and perhaps other protein configurations as well, Thioflavin T gives rise to a new excitation maximum at about 450 nm and enhanced emission at about 482 nm when bound to a fibril protein form. Unbound Thioflavin T is essentially non-fluorescent at the wavelengths. Other small molecules can be used as probes of the changes in protein structure from native to non-native states. For instance the "hydrophobic patch" probes that bind preferentially to exposed hydrophobic patches of a protein. The hydrophobic patches are generally buried within the tertiary structure of a protein in its native state, but become exposed as a protein begins to unfold or denature. Examples of these small molecular, spectroscopic probes are aromatic, hydrophobic dyes, such as antrhacene, acridine, phenanthroline or the like. Other spectroscopic probes are metal-amino acid complexes, such as cobalt metal complexes of hydrophobic amino acids, such as phenylalanine, leucine, isoleucine, methionine, and valine, or the like. The term "chemical stability" of the protein formulation as used herein refers to chemical covalent changes in the protein structure leading to formation of chemical degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native protein structure. Various chemical degradation products can be formed depending on the type and nature of the native protein and the environment to which the protein is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the protein formulation as well- known by the person skilled in the art. Most proteins are prone to deamidation, a process in which the side chain amide group in glutaminyl or asparaginyl residues is hydrolysed to form a free carboxylic acid. Other degradations pathways involves formation of high molecular weight transformation products where two or more protein molecules are covalently bound to each other through transamidation and/or disulfide interactions leading to formation of covalently bound dimer, oligomer and polymer degradation products (Stability of Protein Pharmaceuticals, Ahern. TJ. & Manning M.C., Plenum Press, New York 1992). Oxidation (of for instance methionine residues) can be mentioned as another variant of chemical degradation. The chemical stability of the protein formulation can be evaluated by measuring the amount of the chemical degradation products at various time-points after exposure to different environmental conditions (the formation of degradation products can often be accelerated by for instance increasing temperature). The amount of each individual degradation product is often determined by separation of the degradation products depending on molecule size and/or charge using various chromatography techniques (e.g. SEC-HPLC and/or RP-HPLC). Hence, as outlined above, a "stabilized formulation" refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability. In general, a formulation must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached. In one embodiment of the invention the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 6 weeks of usage and for more than 3 years of storage. In another embodiment of the invention the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 4 weeks of usage and for more than 3 years of storage. In a further embodiment of the invention the pharmaceutical formulation comprising the compound according to the present invention is stable for more than 4 weeks of usage and for more than two years of storage. In an even further embodiment of the invention the pharmaceutical formulation comprising the compound is stable for more than 2 weeks of usage and for more than two years of storage.
In another aspect the present invention relates to the use of a compound according to the invention for the preparation of a medicament. In one embodiment a compound according to the invention is used for the preparation of a medicament for the treatment or prevention of hyperglycemia, type 2 diabetes, impaired glucose tolerance, type 1 diabetes, obesity, hypertension, syndrome X, dyslipidemia, cognitive disorders, atheroschlerosis, myocardial infarction, coronary heart disease and other cardiovascular disorders, stroke, inflammatory bowel syndrome, dyspepsia and gastric ulcers. In another embodiment a compound according to the invention is used for the preparation of a medicament for delaying or preventing disease progression in type 2 diabetes. In another embodiment a compound according to the invention is used for the preparation of a medicament for decreasing food intake, decreasing β-ce\\ apoptosis, increasing ?-cell function and β-ce\\ mass, and/or for restoring glucose sensitivity to ?-cells.
The treatment with a compound according to the present invention may also be combined with combined with a second or more pharmacologically active substances, e.g. selected from antidiabetic agents, antiobesity agents, appetite regulating agents, antihypertensive agents, agents for the treatment and/or prevention of complications resulting from or associated with diabetes and agents for the treatment and/or prevention of complications and disorders resulting from or associated with obesity. Examples of these pharmacologically active substances are : Insulin, sulphonylureas, biguanides, meglitinides, glucosidase inhibitors, glucagon antagonists, DPP-IV (dipeptidyl peptidase-IV) inhibitors, inhibitors of hepatic enzymes involved in stimulation of gluconeogenesis and/or glycogenolysis, glucose uptake modulators, compounds modifying the lipid metabolism such as antihyperlipidemic agents as HMG CoA inhibitors (statins), compounds lowering food intake, RXR agonists and agents acting on the ATP-dependent potassium channel of the β- cells; Cholestyramine, colestipol, clofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, probucol, dextrothyroxine, neteglinide, repaglinide; β-blockers such as alprenolol, atenolol, timolol, pindolol, propranolol and metoprolol, ACE (angiotensin converting enzyme) inhibitors such as benazepril, captopril, enalapril, fosinopril, lisinopril, alatriopril, quinapril and ramipril, calcium channel blockers such as nifedipine, felodipine, nicardipine, isradipine, nimodipine, diltiazem and verapamil, and α-blockers such as doxazosin, urapidil, prazosin and terazosin; CART (cocaine amphetamine regulated transcript) agonists, NPY (neuropeptide Y) antagonists, MC4 (melanocortin 4) agonists, orexin antagonists, TNF (tumor necrosis factor) agonists, CRF (corticotropin releasing factor) agonists, CRF BP (corticotropin releasing factor binding protein) antagonists, urocortin agonists, β3 agonists, MSH (melanocyte- stimulating hormone) agonists, MCH (melanocyte-concentrating hormone) antagonists, CCK (cholecystokinin) agonists, serotonin re-uptake inhibitors, serotonin and noradrenaline re- uptake inhibitors, mixed serotonin and noradrenergic compounds, 5HT (serotonin) agonists, bombesin agonists, galanin antagonists, growth hormone, growth hormone releasing compounds, TRH (thyreotropin releasing hormone) agonists, UCP 2 or 3 (uncoupling protein 2 or 3) modulators, leptin agonists, DA agonists (bromocriptin, doprexin), lipase/amylase inhibitors, RXR (retinoid X receptor) modulators, TR β agonists; histamine H3 antagonists.
It should be understood that any suitable combination of the compounds according to the invention with one or more of the above-mentioned compounds and optionally one or more further pharmacologically active substances are considered to be within the scope of the present invention.
The present invention is further illustrated by the following examples which, however, are not to be construed as limiting the scope of protection. The features disclosed in the foregoing description and in the following examples may, both separately and in any combination thereof be material for realising the invention in diverse forms thereof.
In another aspect the present invention relates to a pharmaceutical composition comprising a compound according to general formula (I), and a pharmaceutically acceptable preservative. In one embodiment of the invention the pharmaceutical composition comprises a compound according to the general formula (I) and a pharmaceutically acceptable stabilizer. In another embodiment of the invention the pharmaceutical composition is suited for parenteral administration. In another aspect the present invention relates to the use of a compound according to the general formula (I) for the preparation of a medicament.
EXAMPLES
General procedure (A) General synthetic methods The peptide was synthesized on Fmoc protected Rink amide resin
(Novabiochem) or chlorotrityl resin using Fmoc strategy on an Applied Biosystems 433A peptide synthesizer in 0.25 mmol scale using the manufacturer supplied FastMoc UV protocols which employ HBTU (2-(1H-Benzotriazol-1-yl-)-1 , 1 ,3,3 tetramethyluronium hexafluorophosphate) mediated couplings in N-methyl pyrrolidone (N-methyl pyrrolidone) and UV monitoring of the deprotection of the Fmoc protection group. The protected amino acid derivatives used were standard Fmoc- amino acids (Anaspec) supplied in preweighed cartridges suitable for the ABI433A synthesizer with the exception of unnatural aminoacids such as Fmoc-Aib-OH (Fmoc- aminoisobutyric acid).
The attachment of sidechains and linkers to specific lysine residues on the crude resin bound protected peptide was carried out in a specific position by incorporation of Fmoc-Lys(Dde)-OH during automated synthesis. Procedure for removal of Dde-protection. The resin (0.25 mmol) was placed in a manual shaker/filtration apparatus and treated with 2% hydrazine in N-methyl pyrrolidone (20 ml, 2x12 min) to remove the DDE group and wash with N-methyl pyrrolidone (4x20 ml). Procedure for attachment of sidechains to Lysine residues. The amino acid (4 molar equivalents relative to resin) was dissolved in N- methyl pyrrolidone/methylene chloride (1 :1 , 20 ml). Hydroxybenzotriazole (HOBt) (4 molar equivalents relative to resin) and diisopropylcarbodiimide (4 molar equivalents relative to resin) was added and the solution was stirred for 15 min. The solution was added to the resin and diisopropyethylamine (4 molar equivalents relative to resin) was added. The resin was shaken 24 hours at room temperature. The resin was washed with N-methyl pyrrolidone (2x20 ml), N-methyl pyrrolidone/Methylene chloride (1 :1) (2x20ml) and methylene chloride (2x20 ml).
Procedure for removal of Dde-protection: The resin (0.25 mmol) was placed in a filter flask in a manual shaking apparatus and treated with N-methyl pyrrolidone/methylene chloride (1 :1) (2x20 ml) and with N-methyl pyrrolidone (1x20 ml), a solution of 20% piperidine in N-methyl pyrrolidone (3x20 ml, 10 min each). The resin was washed with N-methyl pyrrolidone (2x20 ml), N-methyl pyrrolidone/Methylene chloride (1 :1) (2x20ml) and methylene chloride (2x20 ml).
Procedure for cleaving the peptide off the resin: The peptide was cleaved from the resin by stirring for 180 min at room temperature with a mixture of trifluoroacetic acid, water and triisopropylsilane
(95:2.5:2.5). The cleavage mixture was filtered and the filtrate was concentrated to an oil by a stream of nitrogen. The crude peptide was precipitated from this oil with 45 ml diethyl ether and washed 3 times with 45 ml diethyl ether.
Purification: The crude peptide was purified by semipreparative HPLC on a 20 mm x 250 mm column packed with 7μ C-18 silica. Depending on the peptide two one or two purification systems were used. TFA: After drying the crude peptide was dissolved in 5 ml 50% acetic acid H2O and diluted to 20 ml with H2O and injected on the column which then was eluted with a gradient of 40-60 % CH3CN in 0.1% TFA 10 ml/min during 50 min at 40 °C. The peptide containing fractions were collected. The purified peptide was lyophilized after dilution of the eluate with water. Ammonium sulphate: The column was equilibrated with 40% CH3CN in 0.05M (NH4)2SO4, which was adjusted to pH 2.5 with concentrated H2SO . After drying the crude peptide was dissolved in 5 ml 50% acetic acid H2O and diluted to 20 ml with H2O and injected on the column which then was eluted with a gradient of 40% - 60% CH3CN in 0.05M (NH4)2SO4, pH 2.5 at 10 ml/min during 50 min at 40 °C. The peptide containing fractions were collected and diluted with 3 volumes of H2O and passed through a Sep-Pak® C18 cartridge (Waters part. #:51910 ) which has been equilibrated with 0.1 % TFA. It was then eluted with 70% CH3CN containing 0.1 % TFA and the purified peptide was isolated by lyophilisation after dilution of the eluate with water. The final product obtained was characterised by analytical RP-HPLC (retention time) and by LCMS The RP-HPLC analysis was performed using UV detection at 214 nm and a Vydac 218TP54 4.6mm x 250mm 5μ C-18 silica column (The Separations Group, Hesperia, USA) which was eluted at 1 ml/min at 42 °C. Two different elution conditions were used: A1 : Equilibration of the column with in a buffer consisting of 0.1 M (NH4)2SO4, which was adjusted to pH 2.5 with concentrated H2SO4 and elution by a gradient of 0% to 60% CH3CN in the same buffer during 50 min. B1 : Equilibration of the column with 0.1 % TFA / H2O and elution by a gradient of 0% CHsCN / 0.1 % TFA / H2O to 60% CH3CN / 0.1 % TFA / H2O during 50 min. B6: Equilibration of the column with 0.1 % TFA / H2O and elution by a gradient of 0% CH3CN / 0.1 % TFA / H2O to 90% CH3CN / 0.1 % TFA / H2O during 50 min.
LCMS was performed on a setup consisting of Hewlett Packard series 1100 G1312A Bin Pump, Hewlett Packard series 1100 Column compartment, Hewlett Packard series 1100 G1315A DAD diode array detector, Hewlett Packard series 1100 MSD and Sedere 75 Evaporative Light Scattering detectorcontrolled by HP Chemstation software. The HPLC pump is connected to two eluent reservoirs containing: A: 10mM NH4OH in water B: 10mM NH4OH in 90% acetonitrile The analysis was performed at 23° C by injecting an appropriate volume of the sample (preferably 20 μl) onto the column which is eluted with a gradient of A and B. The HPLC conditions, detector settings and mass spectrometer settings used are giving in the following table. Column Waters Xterra MS C-18 X 3 mm id 5 μm Gradient 5% - 100% acetonitrile linear during 6.5 min at 1.5ml/min Detection 210 nm (analogue output from DAD) ELS (analogue output from ELS) MS ionisation mode API-ES. Scan 100-1000 amu step 0.1 amu Matrix Assisted Laser Desorption lonization Mass spectrometric analysis (MALDI-
MS) was performed on a Voyager RP MALDI-TOF instrument (Perseptive Biosystems Inc., Framingham, MA) equipped with a nitrogen laser (337 nm). The instrument was operated in linear mode with delayed extraction, and the accelerating voltage in the ion source was 25kV. Sample preparation was done as follows: 1 μl sample-solution (0.5-1.0 mg/ml) was mixed with 10 μl matrix-solution (Sinapinic acid dissolved in a 5:4:1 mixture of acetonitrile:water:3% TFA) and 1 μl was deposited on the sample plate and allowed to dry. Only external calibration was performed as the normal peptide standards used are in the low molecular weight range and insufficient to assure proper determination of masses in the range of serum albumin (> 60 KDa). As a result the absolute mass values determined are only within 0.2 % accuracy.
Example 1
S-gamma34-(1 -{2-[2-(2-([D-Ala8, Lys37]-GLP-1 -(7-37)amide-Nε37- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
IH2— H—
Figure imgf000034_0001
A resin (Rink amide, 0.68 mmol/g Novabiochem 0.25 mmole) was used to produce the primary sequence on an ABI433A machine according to manufacturers guidelines. All protecting groups were acid labile with the exception of the residue used in position 37 (FmocLys(ivDde)-OH, Novabiochem) allowing specific deprotection of this lysine rather than any other lysine.
Procedure The resin (0.25 mmole) was placed in a manual shaker/filtration apparatus and treated with 2% hydrazine in N-methyl pyrrolidone in (2x12 min. 2x20 ml) to remove the Dde group. The resin was washed with N-methyl pyrrolidone (4x20 ml). Fmoc-8-amino-3,6-dioxaoctanoic acid (Neosystem FA03202) (4 molar equivalents relative to resin) was dissolved in N-methyl pyrrolidone/methylene chloride (1 :1 , 20 ml). Hydroxybenzotriazole (HOBt) (4 molar equivalents relative to resin) and diisopropylcarbodiimide (4 molar equivalents relative to resin) was added and the solution was stirred for 15 min. The solution was added to the resin and diisopropylethylamine (4 molar equivalents relative to resin) was added. The resin was shaken 24 hours at room temperature. The resin was washed with N-methyl pyrrolidone (4x20 ml). A solution of 20% piperidine in N-methyl pyrrolidone (3x20 ml, 10 min each) was added to the resin while shaking. The resin was washed with N- methyl pyrrolidone (4x20 ml). 3-maleimido propionic acid (4 molar equivalents relative to resin) was dissolved in N-methyl pyrrolidone/methylene chloride (1 :1 , 20 ml). Hydroxybenzotriazole hydrate (HOBt; H2O) (4 molar equivalents relative to resin) and diisopropylcarbodiimide (4 molar equivalents relative to resin) was added and the solution was stirred for 15 min. The solution was added to the resin and diisopropylethylamine (4 molar equivalents relative to resin) was added. The resin was shaken 24 hours at room temperature. The resin was washed with N-methyl pyrrolidone (2x20 ml), N-methyl pyrrolidone/methylene chloride (1 :1 ) (2x20ml) and methylene chloride (2x20 ml). The peptide was cleaved from the resin by stirring for 180 min at room temperature with a mixture of trifluoroacetic acid, water and triisopropylsilane (95:2.5:2.5). The cleavage mixture was filtered and the filtrate was concentrated to an oil by a stream of nitrogen. The crude peptide was precipitated from this oil with 45 ml diethyl ether and washed 3 times with 45 ml diethyl ether. The crude peptide was purified by semipreparative HPLC on a 20 mm x 250 mm column packed with 7μ C-18 silica. The crude peptide was dissolved in 5 ml 50% acetic acidin water and diluted to 20 ml with H2O and injected on the column which then was eluted with a gradient of 40-60 % (CH3CN in water with 0.1% TFA) 10 ml/min during 50 min at 40 °C. The peptide containing fractions were collected. The purified peptide was lyophilized after dilution of the eluate with water affording Nε37-(2-(2-(3- (maleimido)propionylamino)ethoxy)ethoxy)acetyl)[D-Ala8,Lys37] GLP-1 (7-37)amide. HPLC: (method B6): RT= 36.8 min HPLC: (method A1): RT= 35.1 min LCMS: m/z = 931.4 (M+H)4+, 1241.5 (M+H)3+. Calculated (M+H)+ = 3722.1 Freeze dried Nε37-(2-(2-(3-(maleimido)propionylamino)ethoxy)ethoxy)acetyl)[D- Ala8,Lys37] GLP-1 (7-37)amide was dissolved in 10 μl 10% acetic acid and 800 μl 40mg/ml Albagen (New Century Pharma) in 50mM NaPi pH 7.0 + 4 % hydroxypropyl- ?-cyclodextrin (HP-/?-CD ) was added and stirred for 2 hours at ambient temperature. Subsequently,, solid ammonium sulphate was added slowly to a final concentration of 2M. The conjugate was purified on a Resource™ HIC ISO with a total volume of 1 ml. Flow rate was kept at 1 ml/min. Buffer: 50mM NaPi pH 7.0 + 4 % HP-jff-CD. A gradient from 2 M to 0 M ammonium sulphate over 20 column volumes was used to separate Albagen from the conjugate. The chromatogram showed two eluting peaks. The first peak was due to Albagen, the second peak contained the conjugate. The conjugate was concentrated and separated from non- reacted analogue on a centriprep™ device with a MW cut -off at 30,000 Da. The overall yield • was 25-35%. The site of conjugation was determined to be Cys-34 by peptide mapping.
The mass found by MALDI is 70023 Da
Theoretical molecular weight of conjugate is 70046 Da.
Example 2 S-gamma34-(1 -{2-[2-(2-([Aib8'22'25, Lys37]-GLP-1 -(7-37)amide-Nε37- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen This compound was prepared as in example 1. Data for GLP1 precursor HPLC: (method B1 ): RT= 38.5 min HPLC: (method A1 ): RT= 36.9 min LCMS: m/z = 949.0 (M+H)4+, 1264.9 (M+H)3+. Calculated (M+H)+ = 3792.2 G T F T S D V S S Y L
Figure imgf000037_0001
The mass found by MALDI is 70102
Figure imgf000037_0002
Theoretical molecular weight of conjugate is 70116 Da.
Example 3
S-gamma34-((1 -{2-[2-(2-([ Aib8,Arg26,34,Glu22,23,30]-GLP-1 -(7-37))Lys amide-Nε- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
This compound was prepared as in example 1. Data for GLP1 -precursor HPLC: (method B1): RT= 36.5 min HPLC: (method A1 ): RT= 34.9 min LCMS: m/z = (M+H)3+ = 1327.8 Calculated (M+H)+ = 3980.3
NH 2-H-
Figure imgf000037_0003
The mass found by MALDI is 70208 Da. Theoretical molecular weight of conjugate is 70304 Da.
Example 4 Other compounds which are synthesised according to the method described in example 1 are :
S-γ3 -(1-{2-[2-(2-([Lys32]-exendin-(1-39)amide-N-ε32-yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}- 2,5-dioxo-pyrrolidin-3-yl) Albumin, (wherein Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).
S-γ34-(1 -{2-[2-(2-([Lys20]-exendin-(1 -39)amide-N- ε20- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin, (wherein
Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).
S-γ34-(1-{2-[2-(2-([Arg12, Lys27]-exendin-(1-39)amide-N- ε27- yl)acetyloxyethoxy)ethy!carbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin. (Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).
S-γ34-(1-{2-[2-(2-([Arg12' 27, Lys32]-exendin-(1-39)amide-N- ε32- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin. (Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).

Claims

Claims
1. A compound of the general formula (I): GLP-1 agonist - L - RR - protraction protein (I) wherein
GLP-1 agonist is a polypeptide which is an agonist of the human GLP-1 receptor,
L is a linker connecting an amino acid side chain of said GLP-1 agonist or the C-terminal amino acid residue of said GLP-1 agonist with RR,
RR is the remains of a reactive residue that has formed a covalent bond with an amino acid residue of the protraction protein, and
protraction protein is a protein having a molar weight of at least 5 kDa, having a plasma half- life of at least 24 hours in human plasma, and said protraction protein has been synthesised by a non-mammalian organism or synthetically.
2. The compound according to claim 1 , wherein said protraction protein is recombinant human serum albumin (SEQ ID NO 1).
3. The compound according to claim 1 , wherein said protraction protein is a human serum albumin variant.
4. The compound according to claim 3, wherein said human serum albumin variant has reduced binding affinities towards copper and nickel as compared to the corresponding binding affinities of human serum albumin towards copper and nickel.
5. The compound according to any one of claims 3-4, wherein said protraction protein is an N-terminal fragment of human serum albumin, or an analogue thereof.
6. The compound according to any one of claims 3-5, wherein said protraction protein is a human serum albumin variant comprising a modification of the Asp-Ala-His-Lys N-terminal sequence.
7. The compound according to claim 6, wherein said protraction protein comprises at least one deletion among the three N-terminal amino acid residues Asp-Ala-His.
8. The compound according to claim 6, wherein said protraction protein comprises an N- terminal extension, such as Glu"3,AIa"2Glu"1,Phe°-HSA(1-585) or an N-terminal fragment thereof.
9. The compound according to any one of claims 6-7, wherein said human serum albumin (HSA) variant is selected from the group consisting of HSA(2-585), HSA(3-585), HSA(4-585), Asp-Ala-HSA(4-585), Xaa3-HSA(1-585) where Xaa3 is an amino acid residue which has substituted the His residue occupying position 3 in native HSA, and N-terminal fragments thereof.
10. The compound according to any one of the previous claims, wherein said protration protein comprises an amino acid sequence of from 60-200 such as from 100 to 150 amino acid residues, and said amino acid sequence being identical to a fragment of SEQ ID NO 1 or a fragment of SEQ ID NO 1 with one or two amino acid substitutions and/or deletions.
11. The compound according to claim 1 , wherein said protraction protein is the Fc portion of an immunoglobulin, an analogue or a fragment thereof.
12. The compound according to any one of the previous claims, wherein said GLP-1 agonist has at least 50% amino acid homology with either GLP-1 (7-37) (SEQ ID NO 2) or Exendin- 4(1-39) (SEQ ID NO 3).
13. The compound according to claim 12, wherein said GLP-1 agonist has at least 80% amino acid homology with either GLP-1 (7-37) (SEQ ID NO 2) or Exendin-4(1-39) (SEQ ID NO 3).
14. The compound according to any one of the previous claims, wherein said GLP-1 agonist comprises the amino acid sequence of the formula (II):
Xaa7-Xaa8-Glu-GIy-Thr-Phe-Thr-Ser-Asp-Xaa16-Ser-Xaa18-Xaai9-Xaa20-Glu-Xaa22-Xaa23-Ala- Xaa25-Xaa26-Xaa27-Phe-lle-Xaa3o-Trp-Leu-Xaa33-Xaa34-Xaa35-Xaa36-Xaa37-Xaa38-Xaa39-Xaa4o- Xaa4ι -Xaa -Xaa43-Xaa44-Xaa45-Xaa4β Formula (II) (SEQ ID No: 4) wherein
Xaa7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, ?-hydroxy-histidine, homohistidine, Nσ-acetyl-histidine, σ-fluoromethyl-histidine, σ-methyl-histidine, 3- pyridylalanine, 2-pyridylalanine or 4-pyridylalanine;
Xaa8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid; Xaa16 is Val or Leu; Xaa18 is Ser, Lys or Arg; Xaa19 is Tyr or Gin; Xaa20 is Leu or Met; Xaa22 is Gly, Glu or Aib; Xaa23 is Gin, Glu, Lys or Arg; Xaa25 is Ala or Val; Xaa26 is Lys, Glu or Arg; Xaa27 is Glu or Leu; Xaa30 is Ala, Glu or Arg; Xaa33 is Val or Lys; Xaa3 is Lys, Glu, Asn or Arg; Xaa35 is Gly or Aib; Xaa36 is Arg, Gly or Lys;
Xaa37 is Gly, Ala, Glu, Pro, Lys, amide or is absent; Xaa38 is Lys, Ser, amide or is absent. Xaa39 is Ser, Lys, amide or is absent; Xaa40 is Gly, amide or is absent; Xaa41 is Ala, amide or is absent; Xaa42 is Pro, amide or is absent; Xaa43 is Pro, amide or is absent; Xaa4 is Pro, amide or is absent; Xaa45 is Ser, amide or is absent; Xaa46 is amide or is absent ; provided that if Xaa38, Xaa39, Xaa40, Xaa41, Xaa42, Xaa43, Xaaψj, Xaa45 or Xaa46 is absent then each amino acid residue downstream is also absent.
15. The compound according to claim 14, wherein said GLP-1 agonist comprises the amino acid sequence of formula (III):
Xaa7-Xaa8-Glu-GIy-Thr-Phe-Thr-Ser-Asp-Val-Ser-Xaa18-Tyr-Leu-Glu-Xaa22-Xaa23-Ala-Ala- Xaa26-Glu-Phe-lle-Xaa3o-Trp-Leu-Val-Xaa34-Xaa35-Xaa36-Xaa37-Xaa3s Formula (III) (SEQ ID No: 5) wherein
Xaa7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, ?-hydroxy-histidine, homohistidine, Nσ-acetyl-histidine, σ-fluoromethyl-histidine, σ-methyl-histidine, 3- pyridylalanine, 2-pyridylalanine or 4-pyridylalanine; Xaa8 is Ala, D-Ala, Gly, Val, Leu, lie, Lys, Aib, (1-aminocyclopropyl) carboxylic acid, (1- aminocyclobutyl) carboxylic acid, 1-aminocyclopentyl) carboxylic acid, or (1-aminocyclohexyl) carboxylic acid, (1-aminocycloheptyl) carboxylic acid, or (1-aminocyclooctyl) carboxylic acid;
Xaa18 is Ser, Lys or Arg;
Xaa22 is Gly, Glu or Aib; Xaa23 is Gin, Glu, Lys or Arg;
Xaa26 is Lys; Glu or Arg;
Xaa30 is Ala, Glu or Arg;
Xaa3 is Lys, Glu or Arg;
Xaa35 is Gly or Aib; Xaa36 is Arg or Lys;
Xaa37 is Gly, Ala, Glu or Lys;
Xaa38 is Lys, amide or is absent.
16. The compound according to any one of claims 1-15, wherein said GLP-1 agonist is dipeptidyl aminopeptidase IV protected.
17. The compound according to claim 16, wherein said GLP-1 agonist is a position 8 analogue, i.e. the alanine residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No: 2) has been substituted by another amino acid residue.
18. The compound according to claim 17, wherein said GLP-1 agonist comprises an Aib residue in position 8 relative to the GLP-1 (7-37) sequence (SEQ ID No:2).
19. The compound according to any of the previous claims, wherein the amino acid residue in position 7 of said GLP-1 peptide (the N-terminal) is selected from the group consisting of D-histidine, desamino-histidine, 2-amino-histidine, /?-hydroxy-histidine, homohistidine, Nσ- acetyl-histidine , σ-fluoromethyl-histidine, σ-methyl-histidine, 3-pyridylalanine, 2- pyridylalanine and 4-pyridylalanine.
20. The compound according to any of the previous claims, wherein said GLP-1 agonist comprises no more than twelve amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
21. The compound according to any one of the previous claims, wherein said GLP-1 agonist comprises no more than six amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
22. The compound according to any one of the previous claims, wherein said GLP-1 agonist comprises no more than four amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
23. The compound according to any one of the previous claims, wherein said GLP-1 agonist comprises no more than 4 amino acid residues which are not encoded by the genetic code.
24. The compound according to any one of claims 1-22, wherein said GLP-1 agonist comprises no more than two amino acid residues which have been exchanged, added or deleted as compared to GLP-1 (7-37) (SEQ ID No:2) or Exendin-4(1-39) (SEQ ID No:3).
25. The compound according to any one of the previous claims, wherein said GLP-1 agonist is selected from the group consisting of [Arg34]GLP-1 (7-37), [Arg2634]GLP-1 (7-37)Lys,
[Lys36Arg 6'34]GLP-1 (7-36), [Aib8'22'35]GLP-1 (7-37),
[Aib8'35]GLP-1 (7-37), [Aib8'22]GLP-1 (7-37), [Aib8'22'35 Arg26'34] GLP-1 (7-37)Lys,
[Aib8'35 Arg26'34]GLP-1 (7-37)Lys, [Aib8'22 Arg26'34]GLP-1 (7-37)Lys,
[Aib8'22'35 Arg26'3 ]GLP-1 (7-37)Lys, [Aib8'35 Arg26'34]GLP-1 (7-37)Lys, [Aib8'22'35 Arg26]GLP-1 (7-37)Lys, [Aib8'35 Arg26]GLP-1 (7-37)Lys, [Aib8'22 Arg26]GLP-1 (7-37)Lys,
[Aib8'22'35 Arg34]GLP-1 (7-37)Lys, [Aib8'35Arg34]GLP-1 (7-37)Lys, [Aib8'22Arg34]GLP-1 (7-37)Lys,
[Aib8,2 '35Ala37]GLP-1 (7-37)Lys, [Aib8'35Ala37]GLP-1 (7-37)Lys, [Aib8'22Ala37]GLP-1 (7-37)Lys,
[Aib8'22'35 Lys37]GLP-1 (7-37), [Aib8'35Lys37]GLP-1(7-37) and [Aib8'22Lys37]GLP-1(7-37).
26. The compound according to any one of claims 1-13, wherein said GLP-1 agonist is Exendin-4(1-39) (SEQ ID No. 3).
27. The compound according to any one of claims 1-13, wherein said GLP-1 agonist is ZP- 10, i.e. [Ser38Lys39]Exendin-4(1 -39)LysLysLysLysLys-amide (SEQ ID No. 4).
28. The compound according to any one of the previous claims, wherein said GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the amino acid residue in position 23, 26, 34, 36 or 38 relative to the amino acid sequence SEQ ID No:2 (GLP-1 (7-37)), (corresponding to position 17, 20, 28, 30 or 32 relative to amino acid sequence SEQ ID No:3(Exendin-4(1-39)).
29. The compound according to any one of the previous claims, wherein said GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of the C-terminal amino acid residue.
30. The compound according to any one of the previous claims, wherein said GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of an amino acid residue selected from arginine, lysine, cysteine, glutamic acid, aspartic acid, histidine, serine, threonine and tyrosine.
31. The compound according to any one of the previous claims, wherein said GLP-1 agonist is attached to the moiety : -L-RR-protraction protein via the side chain of a cysteine residue.
32. The compound according to any one of the previous claims, wherein said linker L is selected from the group consisting of the bivalent connecting chemical groups amides: -C(O)-NR- where R is hydrogen or C1-6-alkyl, amine: -NR-, where R is hydrogen or C1-6-alkyl, thioethers: -S-,
Figure imgf000045_0001
, ethers: -O-, urethanes: -N(R1)-CO-N(R2)-, where R1 and R2 independently is hydrogen or C1-6-alkyl, carbamates: -O-CO-N(R)-, where R is hydrogen or Cι-6-alkyl, ? — N-N=C — hydrazines: H where R is hydrogen or C1-6-alkyl, oximes: -O-N=C(-R)-, where R is hydrogen or C1-6-alkyl, oxazolidines or thiazolidines:
Figure imgf000045_0002
33. The compound according to any one of the previous claims, which is selected from the group consisting of
GLP-1 agonist - C(=O)CH2O(CH2)2O(CH2)2- RR - protraction protein, GLP-1 agonist - C(=O)(CH2)n(OCH2CH2)m- RR - protraction protein, GLP-1 agonist - S(=O)2(CH2)n(OCH2CH2)m - RR - protraction protein, GLP-1 agonist - CH2(CH2)n(OCH2CH2)m- RR - protraction protein, GLP-1 agonist - C(=O)O(CH2)n(OCH2CH2)m- RR - protraction protein, wherein n is an interger in the range from 0 to 10, and m is an integer in the range from 0 to 100.
34. The compound according to any of the previous claims, which is selected from the group consisting of
GLP-1 agonist - L - NC(=O)CH2- sulphur in cysteine residue in protraction protein, GLP-1 agonist - L - S(=O)2(CH2)2- sulphur in cysteine residue in protraction protein, GLP-1 agonist - L - NC(=O)CH2- sulphur in cysteine residue in protraction protein, and in cysteine in protraction protein
GLP-., agonist -
Figure imgf000045_0003
35. The compound according to any one of the previous claims which is selected from the group consisting of S-gamma3 -(1-{2-[2-(2-([D-Ala8, Lys37]-GLP-1-(7-37)amide-Nε37- yl)acetyloxyethoxy)ethylcarbamoyI]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
<2~ H—
Figure imgf000046_0001
S-gamma34-(1 -{2-[2-(2-([Aib8'22'25, Lys37]-GLP-1 -(7-37)amide-Nε37- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
Figure imgf000046_0002
S-gamma34-((1-{2-[2-(2-([ Aib8,Arg26,34,Glu22,23,30]-GLP-1-(7-37))Lys amide-Nε- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl)Albagen
Figure imgf000046_0003
36. A compound according to any one of claims 1-34, which is selected from the group consisting of S-γ34-(1-{2-[2-(2-([Lys32]-exendin-(1-39)amide-N-ε32-yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}- 2,5-dioxo-pyrrolidin-3-yl) Albumin (wherein Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)), S-γ3 -(1 -{2-[2-(2-([Lys20]-exendin-(1 -39)amide-N- ε20- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin (wherein Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)), S-γ34-(1-{2-[2-(2-([Arg12, Lys27]-exendin-(1-39)amide-N- ε27- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin (wherein Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)), and S-γ34-(1 -{2-[2-(2-([Arg12' 27, Lys32]-exendin-(1 -39)amide-N- ε32- yl)acetyloxyethoxy)ethylcarbamoyl]ethyl}-2,5-dioxo-pyrrolidin-3-yl) Albumin (wherein Albumin is recombinant Albagen from New Century Pharma, i.e. recombinant HSA(2-585)).
37. A pharmaceutical composition comprising a compound according to any one of the previous claims, and a pharmaceutically acceptable preservative.
38. A pharmaceutical composition comprising a compound according to any one of claims 1-36, and a pharmaceutically acceptable stabilizer.
39. The pharmaceutical composition according to any one of claims 37-38 which is suited for parenteral administration.
40. Use of a compound according to any one of the claims 1-36 for the preparation of a medicament.
41. Use of a compound according to any one of the claims 1-36 for the preparation of a medicament for the treatment or prevention of hyperglycemia, type 2 diabetes, impaired glucose tolerance, type 1 diabetes, obesity, hypertension, syndrome X, dyslipidemia, cognitive disorders, atheroschlerosis, myocardial infarction, coronary heart disease and other cardiovascular disorders, stroke, inflammatory bowel syndrome, dyspepsia and gastric ulcers.
42. Use of a compound according to any one of the claims 1 -36 for the preparation of a medicament for delaying or preventing disease progression in type 2 diabetes.
43. Use of a compound according to any one of the claims 1-36 for the preparation of a medicament for decreasing food intake, decreasing ?-cell apoptosis, increasing β-ce\\ function and β-ce\\ mass, and/or for restoring glucose sensitivity to β-cells.
PCT/DK2004/000887 2003-12-18 2004-12-17 Novel glp-1 analogues linked to albumin-like agents WO2005058958A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
BRPI0417684-7A BRPI0417684A (en) 2003-12-18 2004-12-17 compound, pharmaceutical composition, and use of a compound
CA002550050A CA2550050A1 (en) 2003-12-18 2004-12-17 Novel glp-1 analogues linked to albumin-like agents
EP04803038A EP1696962A2 (en) 2003-12-18 2004-12-17 Novel glp-1 analogues linked to albumin-like agents
JP2006544221A JP2007537142A (en) 2003-12-18 2004-12-17 Novel GLP-1 analogues bound to albumin-like substances
MXPA06006746A MXPA06006746A (en) 2003-12-18 2004-12-17 Novel glp-1 analogues linked to albumin-like agents.
AU2004298425A AU2004298425A1 (en) 2003-12-18 2004-12-17 Novel GLP-1 analogues linked to albumin-like agents
IL175938A IL175938A0 (en) 2003-12-18 2006-05-25 Novel glp-1 analogues linked to albumin-like agents
US11/454,348 US20070093417A1 (en) 2003-12-18 2006-06-16 Novel GLP-1 analogues linked to albumin-like agents
NO20063242A NO20063242L (en) 2003-12-18 2006-07-12 New GLP-1 analogs linked to albumin-like agents
US12/186,880 US20090005312A1 (en) 2003-12-18 2008-08-06 Novel glp-1 analogues linked to albumin-like agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200301883 2003-12-18
DKPA200301883 2003-12-18

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/454,348 Continuation US20070093417A1 (en) 2003-12-18 2006-06-16 Novel GLP-1 analogues linked to albumin-like agents

Publications (2)

Publication Number Publication Date
WO2005058958A2 true WO2005058958A2 (en) 2005-06-30
WO2005058958A3 WO2005058958A3 (en) 2005-11-24

Family

ID=34684451

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2004/000887 WO2005058958A2 (en) 2003-12-18 2004-12-17 Novel glp-1 analogues linked to albumin-like agents

Country Status (14)

Country Link
US (2) US20070093417A1 (en)
EP (1) EP1696962A2 (en)
JP (1) JP2007537142A (en)
KR (1) KR20060109940A (en)
CN (2) CN101665538A (en)
AU (1) AU2004298425A1 (en)
BR (1) BRPI0417684A (en)
CA (1) CA2550050A1 (en)
IL (1) IL175938A0 (en)
MX (1) MXPA06006746A (en)
NO (1) NO20063242L (en)
RU (1) RU2006120077A (en)
WO (1) WO2005058958A2 (en)
ZA (1) ZA200604912B (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006005667A2 (en) * 2004-07-08 2006-01-19 Novo Nordisk A/S Polypeptide protracting tags comprising a tetrazole moiety
WO2007039140A1 (en) 2005-09-22 2007-04-12 Biocompatibles Uk Ltd. Glp-1 ( glucagon-like peptide-1 ) fusion polypeptides with increased peptidase resistance
WO2007071068A1 (en) * 2005-12-22 2007-06-28 Conjuchem Biotechnologies Inc. Process for the production of preformed conjugates of albumin and a therapeutic agent
JP2007535507A (en) * 2004-04-23 2007-12-06 コンジュシェム バイオテクノロジーズ インコーポレイティド Purification method of albumin conjugate
WO2008023050A1 (en) * 2006-08-25 2008-02-28 Novo Nordisk A/S Acylated exendin-4 compounds
EP1948676A1 (en) * 2005-10-27 2008-07-30 Peptron Co., Ltd. Bioactive substance-blood protein conjugate and stabilization of a bioactive substance using the same
EP1965823A2 (en) 2005-11-04 2008-09-10 SmithKline Beecham Corporation Methods for administering hypoglycemic agents
WO2008132477A1 (en) * 2007-05-01 2008-11-06 Activotec Spp Limited Compounds and uses thereof
WO2009075859A2 (en) * 2007-12-11 2009-06-18 Conjuchem Biotechnologies Inc. Formulation of insulinotropic peptide conjugates
WO2009121884A1 (en) 2008-04-01 2009-10-08 Novo Nordisk A/S Insulin albumin conjugates
JP2010514835A (en) * 2007-01-05 2010-05-06 コヴェックス・テクノロジーズ・アイルランド・リミテッド Glucagon-like protein 1 receptor (GLP-1R) agonist compound
EP2258398A1 (en) 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
WO2010084169A3 (en) * 2009-01-23 2011-02-17 Novo Nordisk A/S Fgf21 derivatives with albumin binder a-b-c-d-e- and their use
WO2011033068A1 (en) * 2009-09-18 2011-03-24 Novo Nordisk A/S Long-acting y2 receptor agonists
WO2011073328A1 (en) 2009-12-16 2011-06-23 Novo Nordisk A/S Glp-1 receptor agonist compounds with a modified n-terminus
WO2011109784A1 (en) * 2010-03-05 2011-09-09 Conjuchem, Llc Formulation of insulinotropic peptide conjugates
WO2012080471A1 (en) 2010-12-16 2012-06-21 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid
US8513192B2 (en) 2009-01-22 2013-08-20 Novo Nordisk A/S Stable growth hormone compounds resistant to proteolytic degradation
US8536122B2 (en) 2005-03-18 2013-09-17 Novo Nordisk A/S Acylated GLP-1 compounds
US8603972B2 (en) 2005-03-18 2013-12-10 Novo Nordisk A/S Extended GLP-1 compounds
WO2014060472A1 (en) 2012-10-17 2014-04-24 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
US8779109B2 (en) 2010-01-22 2014-07-15 Novo Nordisk Health Care Ag Growth hormones with prolonged in-vivo efficacy
US8790705B2 (en) 2006-05-10 2014-07-29 Biocompatibles Uk Ltd. Spherical microcapsules comprising GLP-1 peptides, their production and use
WO2014096440A3 (en) * 2012-12-21 2014-08-14 Novozymes Biopharma Dk A/S Albumin composition for preventing and/or reducing formation of peptide fibrils
US8841249B2 (en) 2009-08-06 2014-09-23 Novo Nordisk A/S Growth hormones with prolonged in-vivo efficacy
US8865868B2 (en) 2008-08-06 2014-10-21 Novo Nordisk Healthcare Ag Conjugated proteins with prolonged in vivo efficacy
US9085637B2 (en) 2013-11-15 2015-07-21 Novo Nordisk A/S Selective PYY compounds and uses thereof
US9211342B2 (en) 2010-01-22 2015-12-15 Novo Nordisk Healthcare Ag Stable growth hormone compounds resistant to proteolytic degradation
US9266940B2 (en) 2011-04-12 2016-02-23 Novo Nordisk A/S Double-acylated GLP-1 derivatives
WO2016055550A1 (en) 2014-10-07 2016-04-14 Cyprumed Gmbh Pharmaceutical formulations for the oral delivery of peptide or protein drugs
WO2017060500A1 (en) 2015-10-07 2017-04-13 Cyprumed Gmbh Pharmaceutical formulations for the oral delivery of peptide drugs
US9732137B2 (en) 2007-12-28 2017-08-15 Novo Nordisk A/S Semi-recombinant preparation of GLP-1 analogues
US9744213B2 (en) 2014-12-23 2017-08-29 Novo Nordisk A/S FGF21 derivatives and uses thereof
WO2018065634A1 (en) 2016-10-07 2018-04-12 Cyprumed Gmbh Pharmaceutical compositions for the nasal delivery of peptide or protein drugs
US10005824B2 (en) 2015-06-12 2018-06-26 Novo Nordisk A/S Selective PYY compounds and uses thereof
EP3488857A1 (en) 2012-03-22 2019-05-29 Novo Nordisk A/S Compositions of glp-1 peptides and preparation thereof
WO2019149880A1 (en) 2018-02-02 2019-08-08 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist, a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid and a lubricant
EP3542790A1 (en) 2012-03-22 2019-09-25 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
WO2019193204A1 (en) 2018-04-06 2019-10-10 Cyprumed Gmbh Pharmaceutical compositions for the transmucosal delivery of therapeutic peptides and proteins
WO2019215063A1 (en) 2018-05-07 2019-11-14 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid
US10583172B2 (en) 2013-11-15 2020-03-10 Novo Nordisk A/S HPYY(1-36) having a beta-homoarginine substitution at position 35
WO2020125744A1 (en) 2018-12-21 2020-06-25 江苏恒瑞医药股份有限公司 Bispecific protein
US10954267B2 (en) 2011-12-29 2021-03-23 Novo Nordisk A/S Dipeptide comprising a non-proteogenic amino acid
US11033499B2 (en) 2012-06-20 2021-06-15 Novo Nordisk A/S Tablet formulation comprising a GLP-1 peptide and a delivery agent
US11045523B2 (en) 2013-04-05 2021-06-29 Novo Nordisk Healthcare Ag Formulation of growth hormone albumin-binder conjugate
US11123296B2 (en) 2012-03-22 2021-09-21 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
WO2021219710A1 (en) 2020-04-29 2021-11-04 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and histidine
WO2022049310A1 (en) 2020-09-07 2022-03-10 Cyprumed Gmbh Improved pharmaceutical formulations of glp-1 receptor agonists
WO2023012263A1 (en) 2021-08-04 2023-02-09 Novo Nordisk A/S Solid oral peptide formulations

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8039432B2 (en) 2005-11-09 2011-10-18 Conjuchem, Llc Method of treatment of diabetes and/or obesity with reduced nausea side effect
WO2008019147A2 (en) * 2006-08-04 2008-02-14 Amylin Pharmaceuticals, Inc. Use of exendins, exendin agonists and glp-1 receptor agonists for altering the concentration of fibrinogen
JP2008169195A (en) 2007-01-05 2008-07-24 Hanmi Pharmaceutical Co Ltd Insulinotopic peptide drug combo using carrier material
CN101041693B (en) * 2007-02-06 2011-08-17 珠海联邦制药股份有限公司 Novel blood sugar lowing polypeptide and uses thereof
EP2109457B1 (en) * 2007-02-12 2016-01-06 CSL Behring GmbH Therapeutic application of kazal-type serine protease inhibitors
US9238878B2 (en) * 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
CA2797431C (en) * 2010-04-27 2016-06-21 Zhejiang Beta Pharma Inc. Glucagon-like peptide-1 analogue and use thereof
CN103415621A (en) 2011-01-14 2013-11-27 雷德伍德生物科技股份有限公司 Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
GB2493540A (en) 2011-08-10 2013-02-13 Follicum Ab Agents for stimulating hair growth in mammals
ES2626013T3 (en) * 2011-09-06 2017-07-21 Novo Nordisk A/S GLP-1 derivatives
CN104411322B (en) * 2012-05-08 2017-05-24 诺和诺德股份有限公司 double-acylated GLP-1 derivatives
CN103408669B (en) * 2013-08-01 2016-01-20 江苏泰康生物医药有限公司 GLP-1 analog fusion, and its production and use
CN105399834A (en) * 2015-10-29 2016-03-16 岳阳新华达制药有限公司 Compound of human GLP-1 (glucagon-like peptide) analogue and preparation method thereof
JP2019515677A (en) 2016-04-26 2019-06-13 アール.ピー.シェーラー テクノロジーズ エルエルシー Antibody conjugates and methods of making and using the same
CN113597434B (en) 2019-12-31 2022-07-01 北京质肽生物医药科技有限公司 Fusion proteins of GLP-1 and GDF15 and conjugates thereof
CN115925995A (en) 2020-09-30 2023-04-07 北京质肽生物医药科技有限公司 Polypeptide Conjugates and Methods of Use

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000069911A1 (en) * 1999-05-17 2000-11-23 Conjuchem, Inc. Long lasting insulinotropic peptides
IL155812A0 (en) * 2000-12-07 2003-12-23 Lilly Co Eli Glp-1 fusion proteins
WO2003059934A2 (en) * 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
ES2298785T3 (en) * 2003-06-12 2008-05-16 Eli Lilly And Company FUSION PROTEINS.

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1696962A2 *

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007535507A (en) * 2004-04-23 2007-12-06 コンジュシェム バイオテクノロジーズ インコーポレイティド Purification method of albumin conjugate
WO2006005667A3 (en) * 2004-07-08 2006-10-12 Novo Nordisk As Polypeptide protracting tags comprising a tetrazole moiety
US9061067B2 (en) 2004-07-08 2015-06-23 Novo Nordisk A/S Polypeptide protracting tags
WO2006005667A2 (en) * 2004-07-08 2006-01-19 Novo Nordisk A/S Polypeptide protracting tags comprising a tetrazole moiety
US8536122B2 (en) 2005-03-18 2013-09-17 Novo Nordisk A/S Acylated GLP-1 compounds
US8603972B2 (en) 2005-03-18 2013-12-10 Novo Nordisk A/S Extended GLP-1 compounds
US8853159B2 (en) 2005-09-22 2014-10-07 Biocompatibles Uk Ltd GLP-1 Fusion Peptides
AU2006299134B2 (en) * 2005-09-22 2012-02-23 Biocompatibles Uk Ltd. GLP-1 ( glucagon-like peptide-1 ) fusion polypeptides with increased peptidase resistance
US8431533B2 (en) 2005-09-22 2013-04-30 Biocompatibles Uk Ltd. GLP-1 fusion peptides, their production and use
WO2007039140A1 (en) 2005-09-22 2007-04-12 Biocompatibles Uk Ltd. Glp-1 ( glucagon-like peptide-1 ) fusion polypeptides with increased peptidase resistance
EP2261245A1 (en) * 2005-09-22 2010-12-15 Biocompatibles Uk Ltd. GLP-1 (glucagon-like peptide-1) fusion polypeptides with increased peptidase resistance
EA013796B1 (en) * 2005-09-22 2010-06-30 Байокомпатиблз Юк Лтд. Glp-1 (glucagon-like peptide-1) fusion polypeptides with increased peptidase resistance
EP2174952A3 (en) * 2005-09-22 2010-11-17 Biocompatibles Uk Ltd. glp-1 (glucagon-like peptide-1) fusion polypeptides with increased peptidase resistance
EP1948676A1 (en) * 2005-10-27 2008-07-30 Peptron Co., Ltd. Bioactive substance-blood protein conjugate and stabilization of a bioactive substance using the same
KR101367867B1 (en) * 2005-10-27 2014-05-07 주식회사 펩트론 Bioactive substance carrier for in vivo stable delivery thereof conjugate containing the same and method of in vivo stable delivery of the bioactive substance
EP1948676A4 (en) * 2005-10-27 2011-05-25 Peptron Co Ltd Bioactive substance-blood protein conjugate and stabilization of a bioactive substance using the same
EP1965823A2 (en) 2005-11-04 2008-09-10 SmithKline Beecham Corporation Methods for administering hypoglycemic agents
EP1965823B1 (en) * 2005-11-04 2016-05-18 Glaxosmithkline LLC Methods for administering hypoglycemic agents
EP3095456A1 (en) * 2005-11-04 2016-11-23 Glaxosmithkline LLC Methods for administering hypoglycemic agents
WO2007071068A1 (en) * 2005-12-22 2007-06-28 Conjuchem Biotechnologies Inc. Process for the production of preformed conjugates of albumin and a therapeutic agent
CN101384623B (en) * 2005-12-22 2013-07-24 常山凯捷健生物药物研发(河北)有限公司 Process for the production of preformed conjugates of albumin and a therapeutic agent
JP2009520469A (en) * 2005-12-22 2009-05-28 コンジュクヘム ビオテクフノロギエス インコーポレイテッド Method for the production of preformed conjugates of albumin and therapeutic agents
US8790705B2 (en) 2006-05-10 2014-07-29 Biocompatibles Uk Ltd. Spherical microcapsules comprising GLP-1 peptides, their production and use
WO2008023050A1 (en) * 2006-08-25 2008-02-28 Novo Nordisk A/S Acylated exendin-4 compounds
US9012398B2 (en) 2006-08-25 2015-04-21 Novo Nordisk A/S Acylated exendin-4 compounds
JP2010514835A (en) * 2007-01-05 2010-05-06 コヴェックス・テクノロジーズ・アイルランド・リミテッド Glucagon-like protein 1 receptor (GLP-1R) agonist compound
JP2012184232A (en) * 2007-01-05 2012-09-27 Covx Technologies Ireland Ltd Glucagon-like protein-1 receptor (glp-1r) agonist compound
WO2008132477A1 (en) * 2007-05-01 2008-11-06 Activotec Spp Limited Compounds and uses thereof
WO2009075859A2 (en) * 2007-12-11 2009-06-18 Conjuchem Biotechnologies Inc. Formulation of insulinotropic peptide conjugates
WO2009075859A3 (en) * 2007-12-11 2010-01-28 Conjuchem Biotechnologies Inc. Formulation of insulinotropic peptide conjugates
US9732137B2 (en) 2007-12-28 2017-08-15 Novo Nordisk A/S Semi-recombinant preparation of GLP-1 analogues
WO2009121884A1 (en) 2008-04-01 2009-10-08 Novo Nordisk A/S Insulin albumin conjugates
US9242011B2 (en) 2008-04-01 2016-01-26 Novo Nordisk A/S Insulin albumin conjugates
CN102065903B (en) * 2008-04-01 2015-02-18 诺沃-诺迪斯克有限公司 Insulin albumin conjugates
US8865868B2 (en) 2008-08-06 2014-10-21 Novo Nordisk Healthcare Ag Conjugated proteins with prolonged in vivo efficacy
US8513192B2 (en) 2009-01-22 2013-08-20 Novo Nordisk A/S Stable growth hormone compounds resistant to proteolytic degradation
US9480753B2 (en) 2009-01-23 2016-11-01 Novo Nordisk A/S FGF21 derivatives with albumin binder A-B-C-D-E- and their use
WO2010084169A3 (en) * 2009-01-23 2011-02-17 Novo Nordisk A/S Fgf21 derivatives with albumin binder a-b-c-d-e- and their use
RU2525393C2 (en) * 2009-01-23 2014-08-10 Ново Нордиск А/С Fgf21 derivatives with albumin binding agent a-b-c-d-e- and their application
EP2258398A1 (en) 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
US8841249B2 (en) 2009-08-06 2014-09-23 Novo Nordisk A/S Growth hormones with prolonged in-vivo efficacy
WO2011033068A1 (en) * 2009-09-18 2011-03-24 Novo Nordisk A/S Long-acting y2 receptor agonists
CN102573888A (en) * 2009-09-18 2012-07-11 诺和诺德公司 Long-acting Y2 receptor agonists
US8815802B2 (en) 2009-12-16 2014-08-26 Novo Nordisk A/S GLP-1 analogues and derivatives
WO2011073328A1 (en) 2009-12-16 2011-06-23 Novo Nordisk A/S Glp-1 receptor agonist compounds with a modified n-terminus
WO2011080102A2 (en) 2009-12-16 2011-07-07 Novo Nordisk A/S Glp-1 analogues and derivatives
US9556250B2 (en) 2009-12-16 2017-01-31 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US9211342B2 (en) 2010-01-22 2015-12-15 Novo Nordisk Healthcare Ag Stable growth hormone compounds resistant to proteolytic degradation
US8779109B2 (en) 2010-01-22 2014-07-15 Novo Nordisk Health Care Ag Growth hormones with prolonged in-vivo efficacy
US9695226B2 (en) 2010-01-22 2017-07-04 Novo Nordisk Healthcare Ag Growth hormones with prolonged in-vivo efficacy
WO2011109784A1 (en) * 2010-03-05 2011-09-09 Conjuchem, Llc Formulation of insulinotropic peptide conjugates
EP3326620A1 (en) 2010-12-16 2018-05-30 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2- hydroxybenzoyl)amino)caprylic acid
WO2012080471A1 (en) 2010-12-16 2012-06-21 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid
US10960052B2 (en) 2010-12-16 2021-03-30 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl) amino) caprylic acid
EP3730127A1 (en) 2010-12-16 2020-10-28 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid
US11382957B2 (en) 2010-12-16 2022-07-12 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
US10005827B2 (en) 2011-04-12 2018-06-26 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US11034746B2 (en) 2011-04-12 2021-06-15 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US9527900B2 (en) 2011-04-12 2016-12-27 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US9266940B2 (en) 2011-04-12 2016-02-23 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US11117947B2 (en) 2011-04-12 2021-09-14 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US10954267B2 (en) 2011-12-29 2021-03-23 Novo Nordisk A/S Dipeptide comprising a non-proteogenic amino acid
US11759502B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
EP4324475A1 (en) 2012-03-22 2024-02-21 Novo Nordisk A/S Compositions of glp-1 peptides and preparation thereof
EP4331667A2 (en) 2012-03-22 2024-03-06 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US11759501B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US10933120B2 (en) 2012-03-22 2021-03-02 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
EP3488857A1 (en) 2012-03-22 2019-05-29 Novo Nordisk A/S Compositions of glp-1 peptides and preparation thereof
US11759503B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
EP3542790A1 (en) 2012-03-22 2019-09-25 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US11123296B2 (en) 2012-03-22 2021-09-21 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
US11033499B2 (en) 2012-06-20 2021-06-15 Novo Nordisk A/S Tablet formulation comprising a GLP-1 peptide and a delivery agent
WO2014060472A1 (en) 2012-10-17 2014-04-24 Novo Nordisk A/S Fatty acid acylated amino acids for oral peptide delivery
WO2014096440A3 (en) * 2012-12-21 2014-08-14 Novozymes Biopharma Dk A/S Albumin composition for preventing and/or reducing formation of peptide fibrils
US11045523B2 (en) 2013-04-05 2021-06-29 Novo Nordisk Healthcare Ag Formulation of growth hormone albumin-binder conjugate
US10583172B2 (en) 2013-11-15 2020-03-10 Novo Nordisk A/S HPYY(1-36) having a beta-homoarginine substitution at position 35
US9085637B2 (en) 2013-11-15 2015-07-21 Novo Nordisk A/S Selective PYY compounds and uses thereof
US10246497B2 (en) 2013-11-15 2019-04-02 Novo Nordisk A/S Selective PYY compounds and uses thereof
WO2016055550A1 (en) 2014-10-07 2016-04-14 Cyprumed Gmbh Pharmaceutical formulations for the oral delivery of peptide or protein drugs
US9744213B2 (en) 2014-12-23 2017-08-29 Novo Nordisk A/S FGF21 derivatives and uses thereof
US9895417B2 (en) 2014-12-23 2018-02-20 Novo Nordisk A/S FGF21 derivatives and uses thereof
US10124039B2 (en) 2014-12-23 2018-11-13 Novo Nordisk A/S FGF21 derivatives and uses thereof
US10005824B2 (en) 2015-06-12 2018-06-26 Novo Nordisk A/S Selective PYY compounds and uses thereof
WO2017060500A1 (en) 2015-10-07 2017-04-13 Cyprumed Gmbh Pharmaceutical formulations for the oral delivery of peptide drugs
WO2018065634A1 (en) 2016-10-07 2018-04-12 Cyprumed Gmbh Pharmaceutical compositions for the nasal delivery of peptide or protein drugs
EP4299118A2 (en) 2018-02-02 2024-01-03 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist, a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid and a lubricant
WO2019149880A1 (en) 2018-02-02 2019-08-08 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist, a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid and a lubricant
US11833248B2 (en) 2018-02-02 2023-12-05 Novo Nordisk A/S Solid compositions comprising a GLP-1 agonist and a salt of N-(8-(2-hydroxybenzoyl)amino)caprylic acid
WO2019193204A1 (en) 2018-04-06 2019-10-10 Cyprumed Gmbh Pharmaceutical compositions for the transmucosal delivery of therapeutic peptides and proteins
WO2019215063A1 (en) 2018-05-07 2019-11-14 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and a salt of n-(8-(2-hydroxybenzoyl)amino)caprylic acid
WO2020125744A1 (en) 2018-12-21 2020-06-25 江苏恒瑞医药股份有限公司 Bispecific protein
WO2021219710A1 (en) 2020-04-29 2021-11-04 Novo Nordisk A/S Solid compositions comprising a glp-1 agonist and histidine
WO2022049310A1 (en) 2020-09-07 2022-03-10 Cyprumed Gmbh Improved pharmaceutical formulations of glp-1 receptor agonists
WO2023012263A1 (en) 2021-08-04 2023-02-09 Novo Nordisk A/S Solid oral peptide formulations

Also Published As

Publication number Publication date
IL175938A0 (en) 2006-10-05
CA2550050A1 (en) 2005-06-30
US20070093417A1 (en) 2007-04-26
CN101665538A (en) 2010-03-10
EP1696962A2 (en) 2006-09-06
JP2007537142A (en) 2007-12-20
CN1893980A (en) 2007-01-10
US20090005312A1 (en) 2009-01-01
AU2004298425A1 (en) 2005-06-30
WO2005058958A3 (en) 2005-11-24
RU2006120077A (en) 2008-01-27
ZA200604912B (en) 2007-09-26
BRPI0417684A (en) 2007-03-20
NO20063242L (en) 2006-07-12
MXPA06006746A (en) 2006-08-18
KR20060109940A (en) 2006-10-23

Similar Documents

Publication Publication Date Title
US20090005312A1 (en) Novel glp-1 analogues linked to albumin-like agents
EP1704165B1 (en) Glp-1 compounds
US8030273B2 (en) Protracted exendin-4 compounds
US7893017B2 (en) Protracted GLP-1 compounds
EP2932981B1 (en) Albumin-binding derivatives of GLP-1
US20090062192A1 (en) Dimeric Peptide Agonists of the Glp-1 Receptor
WO2006097535A2 (en) Peptide agonists of the glucagon family with secretin like activity
WO2009030774A1 (en) Truncated glp-1 derivatives and their therapeutical use
EP2190872A1 (en) Glucagon-like peptide-1 derivatives and their pharmaceutical use
EP2004213A1 (en) Glp-1 peptide agonists

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480037741.1

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 175938

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2004298425

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3283/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/006746

Country of ref document: MX

Ref document number: 200604912

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 1020067011910

Country of ref document: KR

Ref document number: 11454348

Country of ref document: US

Ref document number: 2550050

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006544221

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 2004803038

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2004298425

Country of ref document: AU

Date of ref document: 20041217

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004298425

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006120077

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2004803038

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067011910

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0417684

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 11454348

Country of ref document: US