US20100292133A1 - Truncated glp-1 derivaties and their therapeutical use - Google Patents

Truncated glp-1 derivaties and their therapeutical use Download PDF

Info

Publication number
US20100292133A1
US20100292133A1 US12/676,453 US67645308A US2010292133A1 US 20100292133 A1 US20100292133 A1 US 20100292133A1 US 67645308 A US67645308 A US 67645308A US 2010292133 A1 US2010292133 A1 US 2010292133A1
Authority
US
United States
Prior art keywords
glp
xaa
ethoxy
analogue
derivative
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/676,453
Other languages
English (en)
Inventor
Jane Spetzler
Lauge Schäffer
Jesper Lau
János Tibor Kodra
Kjeld Madsen
Patrick William Garibay
Jacob Kofoed
Steffen Reedtz-Runge
Henning Thøgersen
Igrid Pettersson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novo Nordisk AS
Original Assignee
Novo Nordisk AS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novo Nordisk AS filed Critical Novo Nordisk AS
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LAU, JESPER, KODRA, JANOS TIBOR, PETTERSSON, INGRID, REEDTZ-RUNGE, STEFFEN, KOFOED, JACOB, GARIBAY, PATRICK WILLIAM, MADSEN, KJELD, SCHAFFER, LAUGE, SPETZLER, JANE, THOGERSEN, HENNING
Publication of US20100292133A1 publication Critical patent/US20100292133A1/en
Abandoned legal-status Critical Current

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
    • 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
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to the field of therapeutic peptides, i.e. to new truncated Glucagon-Like Peptide-1 (GLP-1) analogues and derivatives thereof.
  • GLP-1 Glucagon-Like Peptide-1
  • GLP-1 glucagon-like peptide 1
  • WO 98/08871 and US 2001/0011071 describes various GLP-1 analogues and derivatives thereof.
  • Runge et al Journal of Biological Chemistry, vol. 283, no. 17, pp. 11340-11347, which was published after the priority dates of the present invention, discloses the crystal structure of the extracellular domain of the ligand-bound GLP-1 receptor.
  • One object of the present invention is to provide a chemically, physically and enzymatically stable GLP-1 analogue or derivative thereof.
  • a further object of the invention is to provide a long acting, i.e. having an administration regimen as described above, GLP-1 analogue or derivative thereof.
  • Another object of this invention is to provide a GLP-1 analogue or derivative thereof with high potency (receptor affinity) in order to reduce the therapeutic dose used for example for once weekly s.c. dosing or alternatively for non-invasive delivery.
  • Another object of this invention is to provide a GLP-1 compound with a high binding affinity to the GLP-1 receptor (GLP-1R).
  • a still further object of this invention is to provide a GLP-1 compound with a high binding affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R).
  • Another object of this invention is to provide a GLP-1 analogue or derivative thereof with high albumin binding affinity which protects the peptide for proteolytic degradation and reduce renal clearance of the peptide.
  • Potency, binding affinity to the GLP-1 receptor, and possibly also to the extracellular domain of the GLP-1 receptor are properties of potential relevance for an overall object of achieving long-acting, stable and of course therapeutically active GLP-1 derivatives with a potential for once weekly administration.
  • the invention relates to truncated analogues of GLP-1 (7-37), and derivatives thereof.
  • the invention relates to a GLP-1 analogue which is a modified GLP-1(7-35) (SEQ ID No 1) having: i) a total of 2, 3, 4, 5 6, 7, 8, or 9 amino acid substitutions as compared to GLP-1(7-35), including a) a Glu residue at a position equivalent to position 22 of GLP-1(7-35), and b) an Arg residue at a position equivalent to position 26 of GLP-1(7-35); or a derivative thereof.
  • SEQ ID No 1 modified GLP-1(7-35) having: i) a total of 2, 3, 4, 5 6, 7, 8, or 9 amino acid substitutions as compared to GLP-1(7-35), including a) a Glu residue at a position equivalent to position 22 of GLP-1(7-35), and b) an Arg residue at a position equivalent to position 26 of GLP-1(7-35); or a derivative thereof.
  • the invention also relates to compositions and uses of these derivatives and analogues.
  • amino acid(s) at a position equivalent to position 30, 31, 32, 33, 34, or 35 of GLP-1(7-35) can be absent provided that if the amino acid at position 30, 31, 32, 33 or 34 is absent then each amino acid residue downstream is also absent
  • the GLP-1 analogue of the invention comprises a C-terminal amide group, or a C-terminal carboxylic acid group.
  • compositions and methods and uses of the analogues and derivatives according to the invention is provided.
  • 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 be 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., ⁇ -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), Tle (tert-butylglycine), ⁇ -alanine, 3-aminomethyl benzoic acid, anthranilic acid.
  • the 22 proteogenic amino acids are: Alanine, Arginine, Asparagine, Aspartic acid, Cysteine, Cystine, Glutamine, Glutamic acid, Glycine, Histidine, Hydroxyproline, Isoleucine, Leucine, Lysine, Methionine, Phenylalanine, Proline, Serine, Threonine, Tryptophan, Tyrosine, Valine.
  • non-proteogenic amino acid is a moiety which can be incorporated into a peptide via peptide bonds but is not a proteogenic amino acid.
  • examples are ⁇ -carboxyglutamate, ornithine, phosphoserine, the D-amino acids such as D-alanine and D-glutamine,
  • Synthetic non-proteogenic 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), Tle (tert-butylglycine), 3-aminomethyl benzoic acid, anthranilic acid, des-amino-Histidine, the beta analogs of amino acids such as ⁇ -alanine etc., D-histidine, desamino-histidine, 2-amino-histidine, ⁇ -hydroxy-histidine, homohistidine, N ⁇ -acetyl-histidine, ⁇ -fluoromethyl-histidine, ⁇ -methyl-histidine, 3-pyridylalanine, 2-pyridylalanine or 4-pyridylalanine, (1-aminocyclopropyl)carboxylic acid, (1-aminocyclobutyl)carboxylic acid, (1-aminocycl
  • 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 in the C-terminal of the peptide.
  • modified peptide refers to a modified peptide as defined above. For the present purposes this term is used interchangeably with the term “modified peptide sequence”. Consistently herewith, the term “modification” when used herein in connection with peptide sequences refers to amino acid substitutions, additions, and/or deletions.
  • any amino acid substitution, deletion, and/or addition refers to the sequence of human GLP-1(7-35) which is included herein as SEQ ID No: 1.
  • the numbering of the amino acid residues in the sequence listing always starts with no. 1, whereas for the present purpose we want, following the established practice in the art, to start with amino acid residue no. 7 and assign number 7 to it. Therefore, generally, any reference herein to a position number of the GLP-1(7-35) sequence is to the sequence starting with His at position 7 and ending with Gly at position 35.
  • GLP-1(7-37)Lys designates a GLP-1(7-37) analogue wherein the naturally occurring 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 Gly 37 .
  • a position equivalent to when used herein to characterize a modified GLP-1(7-35) sequence refers to the corresponding position in the natural GLP-1(7-35) sequence (having the sequence of SEQ ID No: 1). Corresponding positions are easily deduced, e.g. by simple handwriting and eyeballing.
  • a standard protein or peptide alignment program may be used, such as “align” which is a Needleman-Wunsch alignment. The algorithm is described in Needleman, S. B. and Wunsch, C. D., (1970), Journal of Molecular Biology, 48: 443-453, and the align program by Myers and W.
  • the default scoring matrix BLOSUM50 and the default identity matrix may be used, and the penalty for the first residue in a gap may be set at ⁇ 12 and the penalties for additional residues in a gap at ⁇ 2.
  • each amino acid residue downstream refers to each amino acid positioned towards the C-terminal relative to a specific amino acid.
  • Lys34 and Gly35 are each amino acid residues downstream of Val33 in GLP-1 (7-35).
  • a maximum of 8 amino acids have been modified. In embodiments of the invention a maximum of 7 amino acids have been modified. In embodiments of the invention a maximum of 6 amino acids have been modified. In embodiments of the invention a maximum of 5 amino acids have been modified. In embodiments of the invention a maximum of 4 amino acids have been modified. In embodiments of the invention a maximum of 3 amino acids have been modified. In embodiments of the invention a maximum of 2 amino acids have been modified.
  • one or more amino acid(s) have been deleted in the C-terminal end.
  • the C-terminal of the analogue or derivative according to the invention may be terminated as either an acid or amide.
  • the C-terminal of the analogue or derivative of the invention is an amide.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 35, 36 and 37 are absent, and wherein the total length of the
  • GLP-1 analogue is 28 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 34, 35, 36 and 37 are absent, and wherein the total length of the GLP-1 analogue is 27 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 33, 34, 35, 36 and 37 are absent, and wherein the total length of the GLP-1 analogue is 26 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 32, 33, 34, 35, 36 and 37 are absent, and wherein the total length of the GLP-1 analogue is 25 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 31, 32, 33, 34, 35, 36 and 37 are absent, and wherein the total length of the GLP-1 analogue is 24 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein the amino acids at position 30, 31, 32, 33, 34, 35, 36 and 37 are absent, and wherein the total length of the GLP-1 analogue is 23 amino acids.
  • the invention relates to a GLP-1 analogue or derivative thereof having a C-terminal amide group.
  • the invention relates to a GLP-1 analogue or derivative thereof having 3 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has a substitution selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has a substitution selected from the group consisting of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has a substitution selected from the group consisting of desaminoHis7, Aib8, Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has a substitution selected from the group consisting of desaminoHis7 and Aib8.
  • the invention relates to a GLP-1 analogue or derivative thereof having 4 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has two substitutions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has two substitutions selected from the group consisting of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and an amino acid substitution selected from the group consisting Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting desaminoHis7 and Aib8, and an amino acid substitution selected from the group consisting Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having 5 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has three amino acid substitutions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has three amino acid substitutions selected from the group of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8, and two amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8, and two amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having 6, 7 or 8 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has four, five or six amino acid substitutions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has four, five or six amino acid substitutions selected from the group consisting of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and three, four or five amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and three, four or five amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the invention relates to a GLP-1 analogue or derivative thereof having the sequence of formula (I)
  • Xaa 7 is L-histidine, D-histidine, desamino-histidine, 2-amino-histidine, ⁇ -hydroxy-homohistidine, N ⁇ -acetyl-histidine, ⁇ -fluoromethyl-histidine, ⁇ -methyl-histidine, 3-pyridylalanine, 2-pyridylalanine or 4-pyridylalanine;
  • Xaa 8 is Ala, Gly, Val, Leu, Ile, 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 9 is Glu or a Glu derivative such as alpha, alpha dimethyl-Glu;
  • Xaa 16 is Val or Leu
  • Xaa 18 is Ser, Lys, Cys or Arg
  • Xaa 20 is Leu, Lys or Cys
  • Xaa 23 is Gln, Glu, Lys, Cys or Arg;
  • Xaa 24 is Ala or Asn
  • Xaa 25 is Ala or Val
  • Xaa 27 is Glu, Ala or Leu;
  • Xaa 30 is Ala, Glu, Lys, Arg or absent;
  • Xaa 31 is Trp, Lys, Cys or absent
  • Xaa 33 is Val, Lys, Cys or absent
  • Xaa 34 is Lys, Glu, Asn, Arg, Cys or absent;
  • Xaa 35 is Gly, Aib or absent
  • R is amide or is absent
  • each amino acid residue downstream is also absent.
  • the invention relates to a GLP-1 analogue or derivative thereof having the sequence of formula (II)
  • 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, Gly, Val, Leu, Ile, 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 18 is Ser, Lys or Arg
  • Xaa 30 is Ala, Glu, Lys, Arg or is absent;
  • Xaa 33 is Val, Lys or absent
  • Xaa 34 is Lys, Glu, Arg or is absent;
  • Xaa 35 is Gly, Aib or is absent;
  • R is amide or is absent.
  • R is absent.
  • Xaa 35 and R are absent.
  • Xaa 34 , Xaa 35 and R are absent.
  • Xaa 33 , Xaa 34 , Xaa 35 and R are absent.
  • Xaa 30 , Xaa 33 , Xaa 34 , Xaa 35 and R are absent.
  • 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 according to the invention is N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Val25,Arg26,Leu27,Glu30, Lys33)GLP-1(7-33)amide (structure 1) wherein the naturally occurring Tyr at position 20 has been substituted with lysine which has been derivatised at N-epsilon20 with epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy
  • alanine at position 8 has been substituted with Aib and glycine in pos 22 with glutamate and alanine at position 25 with valine and lysine at position 26 with arginine and glutamate at position 27 with leucine and alanine at position 30 with glutamate and valine at position 33 with lysineamide.
  • a GLP-1 analogue or derivative thereof which is derivatised with an albumin binding residue or is pegylated, is provided.
  • the invention relates to a GLP-1 analogue or derivative, wherein the amino acid which is pegylated or derivatised with an albumin binding residue is a Lys-residue or a Cys-residue.
  • the amino acid which is pegylated or derivatised with an albumin binding residue is a Lys-residue.
  • the amino acid which is pegylated or derivatised with an albumin binding residue is a Cys-residue.
  • the C-terminal amino acid is pegylated or derivatised with an albumin binding residue.
  • the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 18, 20, 23, 31, 33, 34 or at the C-terminal amino acid. In a further aspect, the invention relates to a GLP-1 analogue or derivative pegylated or derivatised with an albumin binding residue at position 18. In further aspect, the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 20. In a further aspect, the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 23.
  • the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 31. In a further aspect, the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 33. In a further aspect, the invention relates to a GLP-1 analogue or derivative thereof pegylated or derivatised with an albumin binding residue at position 34.
  • the invention relates to a GLP-1 analogue or derivative thereof, which has been derivatised with an albumin binding residue.
  • lysine residue or cysteine residue is linked to an albumin binding residue via a chemical bond.
  • a chemical bond can as an example be obtained by derivatisation of an epsilon amino group of lysine by acylation with an active ester of an albumin binding residue such as a long fatty acid.
  • connecting two chemical moieties as used in the present invention includes but is not limited to alkylation, ester formation, amide formation or maleimide coupling.
  • linker means a spacer (the two terms spacer and linker is used interchangeably in the present specification) that separates a peptide and an albumin binding residue or a polyethylene glycol polymer.
  • the linker comprises one or more alkylene glycol units, such as 1 to 5 alkylene glycol units.
  • the alkylene glycol units are in a further aspect ethylene glycol, propylene glycol or butylene glycol but can also be higher alkylene glycols.
  • the linker is a hydrophilic linker selected from
  • I, m and n independently are 1-20 and p is 0-10,
  • q is an integer in the range from 0 to 5
  • each D, E, and G are independently selected from —O—, —NR 3 —, —N(COR 4 )—, —PR 5 (O)—, and —P(OR 6 )(O)—, wherein R 3 , R 4 , R 5 , and R 6 independently represent hydrogen or C 1-6 -alkyl,
  • Z is selected from —C(O)NH—, —C(O)NHCH 2 —, —OC(O)NH—, —C(O)NHCH 2 CH 2 —, —C(O)CH 2 —, —C(O)CH ⁇ CH—, —(CH 2 ) s —, —C(O)—, —C(O)O— or —NHC(O)—, wherein s is 0 or 1.
  • the linker is a hydrophilic linker as defined above wherein I is 1 or 2, n and m are independently 1-10 and p is 0-10.
  • the linker is a hydrophilic linker as defined above wherein D is —O—.
  • the linker is a hydrophilic linker as defined above wherein E is —O—.
  • the hydrophilic linker is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoe hydrophilic linker
  • the linker is a hydrophilic linker as defined above wherein q is 1.
  • the linker is a hydrophilic linker as defined above wherein G is —O—.
  • the linker is a hydrophilic linker as defined above wherein Z is selected from the group consisting of —C(O)NH—, —C(O)NHCH 2 —, and —OC(O)NH—.
  • the linker is a hydrophilic linker as defined above wherein q is 0.
  • the linker is a hydrophilic linker as defined above wherein l is 2.
  • the linker is a hydrophilic linker as defined above wherein n is 2.
  • a “hydrophilic linker” is used that separates a peptide and an albumin binding residue with a chemical moiety.
  • the hydrophilic linker is —C(O)—(CH 2 ) l —O—[(CH 2 CH 2 —O] m —(CH 2 ) p —[NHC(O)—(CH 2 ) l —O—[(CH 2 ) n —O] m —(CH 2 ) p ] q —NH—, wherein l, m, n, and p independently are 1-5, and q is 0-5.
  • the hydrophilic linker is —C(O)—CH 2 —O—CH 2 CH 2 —O—CH 2 CH 2 [NHC(O)—CH 2 —O—CH 2 CH 2 O—CH 2 CH 2 ] q —NH—, wherein q is 0-5.
  • the hydrophilic linker is —C(O)—CH 2 —O—CH 2 CH 2 —O—CH 2 CH 2 —NHC(O)—CH 2 —O—CH 2 CH 2 O—CH 2 CH 2 —NH—.
  • the hydrophilic linker is —[CH 2 CH 2 O] m+1 (CH 2 ) p Q q — wherein m and p independently is 0-10, and Q is —Z—(CH 2 ) l D[(CH 2 ) n G] m (CH 2 ) p — as defined above.
  • the hydrophilic linker is —(CH 2 ) l —O—[(CH 2 ) n —O] m —(CH 2 ) p —[C(O)NH—(CH 2 ) l —O—[(CH 2 ) n —O] m —(CH 2 ) p ] q —, wherein l, m, n, and p independently are 1-5, and q is 0-5.
  • the linker comprises an amino acid residue except Cys, or a dipeptide such as Gly-Lys.
  • a dipeptide such as Gly-Lys is used to designate a dipeptide wherein the C-terminal amino acid residue is Lys, His or Trp, preferably Lys, and wherein the N-terminal amino acid residue is selected from the group comprising Ala, Arg, Asp, Asn, Gly, Glu, Gln, Ile, Leu, Val, Phe and Pro.
  • Suitable PEG polymers are typically commercially available or may be made by techniques well-known to those skilled in the art.
  • the PEG polymer has a molecular weight of greater than 700D, in a further aspect a molecular weight greater than 5 kD, in yet a further aspect greater than 10 kD, and in a even further aspect greater that 20kD.
  • the PEG polymer may be linear or branched. In cases where the PEG polymer is greater than 20KDa, the PEG polymer is preferable having a branched structure, such as for example, a 43 kD branched PEG-peptide (Shearwater 2001 catalog #2D3XOT01, mPEG2-MAL).
  • the attachment of a PEG on an intact peptide can be accomplished by attaching the PEG on the opposite side of the peptide surface that interacts with the receptor.
  • cysteine PEGylation is one method for site-specific PEGylation, and can be accomplished by introducing a unique cysteine mutation at one of the specific positions on human amylin or the amylin analog and then reacting the resulting peptide with a cysteine-specific PEGylation reagent, such as PEG-maleimide. It may be necessary to mutate the peptide in order to allow for site-specific PEGylation. For example, if the peptide contains cysteine residues, these will need to be substituted with conservative amino acids in order to ensure site-specific PEGylation.
  • rigid linkers including but not limited to “GGS”, “GGSGGS”, and “PPPS” may be added to the C-terminus, but before the site of PEG attachment (i.e. a unique cysteine residue).
  • the albumin binding residue is a lipophilic residue.
  • the lipophilic residue is attached to a lysine residue optionally via a linker by conjugation chemistry such as by alkylation, acylation, ester formation, or amide formation or to a cysteine residue by maleimide coupling.
  • the albumin binding residue is negatively charged at physiological pH.
  • the albumin binding residue comprises a group which can be negatively charged.
  • One preferred group which can be negatively charged is a carboxylic acid group.
  • the albumin binding residue is selected from the group consisting of a straight chain alkyl group, a branched alkyl group, a group which has an w-carboxylic acid group, and a partially or completely hydrogenated cyclopentanophenanthrene skeleton.
  • the albumin binding residue is a cibacronyl residue.
  • the albumin binding residue has from 6 to 40 carbon atoms, from 8 to 26 carbon atoms or from 8 to 20 carbon atoms.
  • the albumin binding residue is an acyl group selected from the group comprising CH 3 (CH 2 ) r CO—, wherein r is an integer from 4 to 38, preferably an integer from 4 to 24, more preferred selected from the group comprising CH 3 (CH 2 ) 6 CO—, CH 3 (CH 2 ) 8 CO—, CH 3 (CH 2 ) 10 CO—, CH 3 (CH 2 ) 12 CO—, CH 3 (CH 2 ) 14 CO—, CH 3 (CH 2 ) 16 CO—, CH 3 (CH 2 ) 18 CO—, CH 3 (CH 2 ) 20 CO— and CH 3 (CH 2 ) 22 CO—.
  • r is an integer from 4 to 38, preferably an integer from 4 to 24, more preferred selected from the group comprising CH 3 (CH 2 ) 6 CO—, CH 3 (CH 2 ) 8 CO—, CH 3 (CH 2 ) 10 CO—, CH 3 (CH 2 ) 12 CO—, CH 3 (CH 2 ) 14 CO—, CH 3 (CH 2 ) 16 CO—, CH 3 (CH
  • the albumin binding residue is an acyl group of a straight-chain or branched alkane ⁇ , ⁇ -dicarboxylic acid.
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein at least one amino acid residue is derivatised with A-B-C-D-
  • A- is selected from the group consisting of
  • n is selected from the group consisting of 14, 15, 16 17, 18 and 19
  • p is selected from the group consisting of 10, 11, 12, 13 and 14
  • d is selected from the group consisting of 0, 1, 2, 3, 4 and 5,
  • -B- is selected from the group consisting of
  • x is selected from the group consisting of 0, 1, 2, 3 and 4, and ⁇ is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12,
  • -C- is selected from the group consisting of
  • b and e are each independently selected from the group consisting of 0, 1 and 2
  • c and f are each independently selected from the group consisting of 0, 1 and 2 with the proviso that b is 1 or 2 when c is 0, or b is 0 when c is 1 or 2, and e is 1 or 2 when f is 0, or e is 0 when f is 1 or 2, and
  • -D- is attached to said amino acid residue and is a linker.
  • one amino acid residue of the analogue according to the invention is derivatised with A-B-C-D-.
  • the derivatised amino acid residue comprises an amino group. In a further aspect, the derivatised amino acid residue comprises a primary amino group in a side chain. In yet a further aspect, the derivatised amino acid residue is lysine.
  • A- is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • n is selected from the group consisting of 15 and 17, and more is preferred 17.
  • A- is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • p is selected from the group consisting of 12, 13, and 14 and more preferred is 13.
  • d is selected from the group consisting of 0, 1, 2, 3 and 4, more preferred 0, 1 and 2 and most preferred 1.
  • d is selected from the group consisting of 0, 1 and 2 and p is selected from the group consisting of 12, 13 or 14, more preferred d is selected from the group consisting of 1 and 2 and p is selected from the group consisting of 13 and 14, and most preferred d is 1 and p is 13.
  • -B- is
  • -B- is
  • -B- is
  • -B- is
  • x is selected from the group consisting of 0, 1 and 2, more preferred x is selected from the group consisting of 0 and 1 and most preferred x is 1.
  • -B- is
  • is selected from the group consisting of 2, 3, 4, 5, 6, 7, 8, 9 and 10 and more preferred ⁇ is selected from the group consisting of 2, 3, 4, 5, 6, 7, and 8.
  • -C- is
  • c is selected from the group consisting of 0 and 1 and b is selected from the group consisting of 1 and 2, more preferred b is 1 and c is 0.
  • -C- is
  • f is selected from the group consisting of 0 and 1 and e is selected from the group consisting of 1 and 2, more preferred e is 1 and f is 0.
  • -C- is
  • D is selected from the group consisting of
  • k is selected from the group consisting of 0, 1, 2, 3, 4, 5, 11 and 27, and m is selected from the group consisting of 0, 1, 2, 3, 4, 5 and 6.
  • -D- is
  • k is selected from the group consisting of 1, 2, 3, 11 and 27 and more preferred k is 1.
  • m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • -D- is
  • -D- is
  • -D- is
  • -D- is
  • -D- is
  • m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • A-B-C-D- is selected and combined from
  • A-B-C-D- is selected and combined from
  • A-B-C-D- is selected from the group consisting of
  • the invention relates to a GLP-1 analogue or derivative thereof, wherein at least one amino acid residue is derivatised with A-B-C-D-, and where the derivative binds to albumin.
  • A-B-C-D is composed of an albumin binding fragment A-B-C- and a hydrophilic linker D.
  • GLP-1 peptide as used herein means GLP-1(7-35) (SEQ ID No 1) or a GLP-1(7-35) analogue thereof.
  • the GLP-1 analogue or derivative thereof according to the invention is an insulinotropic agent.
  • any amino acid position in the GLP-1 analogue may be derivatised.
  • the amino acid residue which is derivatised comprises an amino group.
  • amino acid residues comprising an amino group is lysine, ornithine, Epsilon-N-alkylated lysine such as Epsilon-N methylLysine, O-aminoethylserine, O-aminopropylserine or longer O alkylated serines containing a primary or secondary amino group in the side chain.
  • the derivatised amino acid residue comprises a primary amino group in a side chain.
  • Examples of amino acid residues comprising a primary amino group is lysine ornithine, O-aminoethylserine, O-aminopropylserine or longer O alkylated serines containing a primary amino group in the side chain.
  • the derivatised amino acid residue is lysine.
  • the derivative according to the invention is only derivatised in one position, e.g. only one amino acid residue is derivatised.
  • the amino acid residue which is derivatised is cysteine.
  • GLP-1 compounds of the invention have been synthesized and tested as described in the experimental part.
  • the GLP-1 compounds of the invention have several advantageous and beneficial properties as explained in the following, by reference to the Examples.
  • insulinotropic agent means a GLP-1 analogue or derivative thereof which is an agonist of the human GLP-1 receptor, i.e. a GLP-1 analogue or derivative thereof which stimulates the formation of cAMP in a suitable medium containing the human GLP-1 receptor (one such medium disclosed below).
  • the GLP-1 analogue or derivative of the invention has an acceptable, preferably high potency (at the receptor).
  • the potency of an insulinotropic agent such as the GLP-1 compounds of the invention may be determined by calculating the EC 50 value from the dose-response curve, e.g. as described in Example 21.
  • baby hamster kidney (BHK) cells expressing the cloned human GLP-1 receptor are used, preferably BHK-467-12A, more preferably BHK-467-12A (tk-ts13);
  • the cells are grown in DMEM media with the addition of 100 IU/mL penicillin, 100 ⁇ g/mL streptomycin, 5% fetal calf serum and 0.5 mg/mL Geneticin G-418 (Life Technologies), preferably at 5% CO2;
  • the cells preferably at approximately 80% confluence, are washed twice in phosphate buffered saline;
  • the cells are harvested with an aqueous solution of tetrasodium salt of ethylenediaminetetraacetic acid, such as Versene;
  • plasma membranes are prepared from the cells by homogenisation, preferably in buffer 1 ; the homogenate is centrifuged, e.g.
  • Steps (vi) and (vii) are preferably repeated, e.g. one or two times more.
  • the functional receptor assay may be carried out as described in Example 21 by measuring cyclic AMP (cAMP) as a response to stimulation by the insulinotropic agent.
  • cAMP formed is preferably quantified by the AlphaScreenTM cAMP Kit (Perkin Elmer Life Sciences).
  • Incubations may be carried out in half-area 96-well microtiter plates in a total volume of 50 ⁇ L buffer 3 (50 mM Tris-HCl, 5 mM HEPES, 10 mM MgCl 2 , pH 7.4) and with the following addiditions: 1 mM ATP, 1 ⁇ M GTP, 0.5 mM 3-isobutyl-1-methylxanthine (IBMX), 0.01% Tween-20, 0.1% BSA, 6 ⁇ g membrane preparation, 15 ⁇ g/mL acceptor beads, 20 ⁇ g/mL donor beads preincubated with 6 nM biotinyl-cAMP.
  • buffer 3 50 mM Tris-HCl, 5 mM HEPES, 10 mM MgCl 2 , pH 7.4
  • 1 mM ATP 1 ⁇ M GTP
  • IBMX 3-isobutyl-1-methylxanthine
  • BSA 0.1% BSA
  • 6 ⁇ g membrane preparation 15
  • Analogues or derivatives to be tested for agonist activity are preferably dissolved and diluted in buffer 3.
  • GTP is freshly prepared for each experiment.
  • the plate is incubated in the dark with slow agitation for three hours at room temperature followed by counting in the FusionTM instrument (Perkin Elmer Life Sciences).
  • Concentration-response curves are plotted for the individual analogues or derivatives and EC 50 values estimated using a four-parameter logistic model with Prism v. 4.0, or 5.0 (GraphPad, Carlsbad, Calif.).
  • the GLP-1 derivative of the invention has a potency (EC 50 in nM), as determined using the cAMP assay, below 10.00, preferably below 9.00, more preferably below 8.00, even more preferably below 7.00, and most preferably below 6.00 (nM).
  • the GLP-1 derivative of the invention has a potency (EC 50 in nM), as determined using the cAMP assay, below 5.00, preferably below 4.00, more preferably below 3.00, even more preferably below 2.00, and most preferably below 1.00 (nM).
  • the GLP-1 derivative of the invention has a potency (EC 50 in nM), as determined using the cAMP assay, below 0.80, preferably below 0.60, more preferably below 0.40, even more preferably below 0.20, and most preferably below 0.10 (nM).
  • the GLP-1 derivative of the invention has a potency (EC 50 in nM), as determined using the cAMP assay, below 0.090, preferably below 0.080, more preferably below 0.070, even more preferably below 0.060, and most preferably below 0.050 (nM).
  • the GLP-1 derivative of the invention has a potency (EC 50 in nM), as determined using the cAMP assay, below 0.040, preferably below 0.030, more preferably below 0.020, and most preferably below 0.010 (nM).
  • exemplary ranges of potency (EC 50 in nM, as determined using the cAMP assay) of GLP-1 derivatives of the invention are 0.010-10.0, 0.010-8.0, 0.010-6.0, 0.010-4.0, 0.010-2.0, 0.010-1.00, 0.010-0.80, 0.010-0.60, 0.010-0.40, 0.010-0.30, 0.010-0.20, 0.010-0.10, and 0.010-0.90 (nM), preferably 0.010-0.40, 0.010-0.30, 0.010-0.20, 0.010-0.10, and 0.010-0.90 (nM).
  • the GLP-1 analogue or derivative thereof (“GLP-1 compound”) has a high affinity to the GLP-1 receptor.
  • the affinity to the GLP-1 receptor may be determined as described in Example 23, i.e. by way of displacement of 125 I-GLP-1 from the receptor.
  • BHK cells may be used for membrane preparation, preferably strain tk-ts13.
  • the membranes may be purified, preferably as described in Example 23,
  • a preferred binding assay method is the SPA assay of Example 23.
  • the IC 50 value may be read from the resulting (binding) curve as the concentration which displaces 50% of 125 I-GLP-1 from the receptor.
  • the IC 50 value is below 500 nM, preferably below 400 nM, more preferably below 300 nM, even more preferably below 200 nM, and most preferably below 100 nM.
  • the IC 50 value is below 80 nM, preferably below 60 nM, more preferably below 50 nM, even more preferably below 40 nM, and most preferably below 30 nM.
  • the IC 50 value is below 20 nM, preferably below 15 nM, more preferably below 10 nM, even more preferably below 5.0 nM, and most preferably below 4.0 nM. In a fourth particular embodiment, the IC 50 value is below 3.0 nM, preferably below 2.0 nM, more preferably below 1.0 nM, even more preferably below 0.80 nM, and most preferably below 0.60 nM.
  • the IC 50 value is below 0.50 nM, preferably below 0.40 nM, more preferably below 0.30 nM, even more preferably below 0.20 nM, and most preferably below 0.10 nM.
  • exemplary ranges of IC 50 are: 0.1-400, 0.2-300, 0.3-200, 0.4-100, 0.5-50, and 1-10 nM.
  • the present invention relates to a GLP-1 analogue or derivative thereof with high affinity binding to the isolated N-terminal extracellular domain of the GLP-1 receptor (nGLP-1R).
  • the affinity may be measured as the ability to displace 125 I-Exendin-4(9-39) from binding to nGLP-1R, e.g. as described in Example 22.
  • Exendin-4 binds nGLP-1R with an IC 50 value of 5 nM
  • GLP-1(7-37) binds nGLP-1R with an IC 50 value of 1120 nM
  • liraglutide binds nGLP-1R with an IC 50 value of 1500 nM.
  • the GLP-1 analogues or derivatives thereof of this invention binds nGLP-1R with an IC 50 value lower than that of liraglutide. More preferable the GLP-1 analogues or derivatives thereof of this invention binds nGLP-1R with an IC 50 value lower than 100 nM and even more preferable below 10 nM or even below 5 nM.
  • the protein nGLP-1R may be prepared as described by Runge et al 2007 (In Biochemistry, vol. 46, pp. 5830-5840). The protein is then biotinylated and immobilized, preferably on streptavidin-coated SPA beads.
  • the nGLP1R in a suitable buffer such as 0.1M NaHCO 3 may be biotinylated using 75 ⁇ g BNHS (Sigma H1759) to 1 mg protein.
  • the biotinylated nGLP1R is subsequently preferably dialyzed against PBS. All reagents and analogues or derivatives are preferably diluted in PBS with 0.05% v/v Tween 20.
  • the binding assay may e.g.
  • OptiPlates PerkinElmer 6005290
  • a final volume of 200 ⁇ l Each well may contain 2 mg streptavidin coated SPA beads (PerkinElmer RPNQ007), 0.1 pmol biotinylated nGLP1R, 50 pCi 125 I-Exendin (9-39) and test peptide in suitable final concentrations, e.g. ranging from 1000 nM to 0.064 nM.
  • the plates are incubated on a shaker, preferably at RT for 3 hours.
  • the SPA particles may be spun down by centrifugation, e.g. for 10 min at 1500 rpm, and the plates are counted, e.g. in a TopCount-NXT (PerkinElmer).
  • the affinity may be expressed by way of an IC 50 value, which is read from the curve as the concentration of the GLP-1 derivative which displaces 50% of 125 I-Exendin-4(9-39) from binding to nGLP-1R.
  • the GLP-1 derivative of the invention has an affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R), measured as IC 50 /nM in the assay of Example 22, of below 1500, preferably below 1000, even more preferably below 900, and most preferably below 800 (nM).
  • nGLP-1R GLP-1 receptor
  • the GLP-1 derivative of the invention has an affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R), measured as IC 50 /nM in the assay of Example 22, of below 700, preferably below 600, even more preferably below 500, and most preferably below 400 (nM).
  • nGLP-1R GLP-1 receptor
  • the GLP-1 derivative of the invention has an affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R), measured as IC 50 /nM in the assay of Example 22, of below 300, preferably below 200, even more preferably below 100, and most preferably below 80 (nM).
  • nGLP-1R GLP-1 receptor
  • the GLP-1 derivative of the invention has an affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R), measured as IC 50 /nM in the assay of Example 22, of below 60, preferably below 50, even more preferably below 40, and most preferably below 30 (nM).
  • nGLP-1R GLP-1 receptor
  • the GLP-1 derivative of the invention has an affinity to the extracellular domain of the GLP-1 receptor (nGLP-1R), measured as IC 50 /nM in the assay of Example 22, of below 20, preferably below 15, even more preferably below 10.0, and most preferably below 5.0 (nM).
  • nGLP-1R GLP-1 receptor
  • exemplary ranges of affinity to nGLP-1R (IC 50 in nM) of the GLP-1 derivative of the invention are: 5-1500, 5-1000, 10-500, 20-300, 50-500, 10-500, and 5-50 (nM).
  • DPP-IV protected as used herein referring to a polypeptide means a polypeptide which has been chemically modified in order to render said derivative 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, GLP-2, Exendin-4 etc e.g. GLP-1, GLP-2, Exendin-4 etc.
  • a GLP-1 analogue or derivative thereof according to the invention is a DPP-IV protected GLP-1 analogue or derivative thereof.
  • a GLP-1 analogue or derivative thereof according to the invention is a DPP-IV protected GLP-1 analogue or derivative thereof which is more resistant to DPP-IV than liraglutide.
  • 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 resistance of a peptide to degradation by dipeptidyl aminopeptidase IV is determined by the following degradation assay:
  • One method for performing this analysis is: The mixtures are applied onto an Agilent Zorbax 300SB-C18 (5 ⁇ m particles) 150 ⁇ 2.1 mm column and eluted at a flow rate of 0.5 ml/min with a linear gradient from 0.1% trifluoroacetic acid to 100% acetonitrile with 0.07% TFA in 30 minutes. Peptides and their degradation products are monitored by their absorbance at 214 nm, and are quantified by integration of their peak areas. The stability of a peptide against dipeptidyl aminopeptidase IV is determined as the peak area of the intact peptide relative to the sum of the peak areas of the intact peptide and the degradation product lacking the two aminoterminal amino acids after cleavage.
  • pharmaceutically acceptable means suited for normal pharmaceutical applications, i.e. giving rise to no serious adverse events in patients etc.
  • excipient means the chemical compounds which are normally added to pharmaceutical compositions, e.g. buffers, tonicity agents, preservatives and the like.
  • an effective amount means a dosage which is sufficient to be effective for the treatment of the patient compared with no treatment.
  • pharmaceutical composition means a product comprising an active GLP-1 analogue or derivative thereof according to the invention together with pharmaceutical excipients such as buffer, preservative, and optionally a tonicity modifier and/or a stabilizer.
  • 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 analogue or derivative according to the invention 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.
  • the present invention relates to a GLP-1 analogue or derivative thereof that can bind to albumin and the GLP-1 receptor simultaneously.
  • the present invention relates to a GLP-1 analogue or derivative thereof that bind to the GLP-1 receptor with an affinity below 100nM, preferable below 30 nM in the presence of 2% albumin.
  • the GLP-1 analogue or derivative thereof (“GLP-1 compound”) has an affinity to the GLP-1 receptor which is only partly decreased when comparing the affinity in the presence of very low concentration (e.g. 0.005% to 0.2%) of human albumin to the affinity in the presence of 2% human albumin.
  • the shift in binding affinity under these conditions is less than 50 fold, preferable below 30 fold and more preferable below 10 fold.
  • the present invention relates to a GLP-1 analogue or derivative thereof which is stable against the chemical degradation normally seen with exendin-4-especially oxidation and deamidation.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has a high potency at the receptor.
  • the GLP-1 potency is better than 3 micro molar and preferable the potency is better than 1 micromolar in the cAMP assay.
  • the GLP-1 potency is better than 1 micro molar and preferable the potency is better than 0.2 micromolar in the cAMP assay.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has high albumin binding affinity.
  • the analogues or derivatives of this invention have an albumin binding affinity that is below 1 micromolar. More preferable the analogues or derivatives of this invention has an albumin binding affinity that is below 500 nM and even more preferable below 200 nM or even below 100 nM.
  • the albumin binding affinity can be measured using the following assay:
  • affinities of the GLP-1 analogue or derivative thereof for human serum albumin are measured by a competition scintillation proximity assay (SPA).
  • Streptavidin-SPA beads (GE Healthcare RPNQ0009) are incubated with biotinylated HSA for 5 hours. The beads are washed with buffer to remove unbound HSA.
  • the beads are mixed with an 125 I-labeled acylated GLP-1 analogue such as N-epsilon26-[2-(2- ⁇ 2-[2-(2- ⁇ 2-[(S)-4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl][A ib8, 125 I-Tyr19,Arg34]GLP-1(7-37) or N-epsilon37-[2-(2-[2-((S)-4-((S)-4-(12-[4-(16-(1H-tetrazol-5-yl)hexadecanoylsulfamoyl)butyrylamino]dodecanoylamino)-4-carboxybutyrylamino)-4-carboxybutyrylamino)ethoxy]ethoxy)acety
  • the mixture is pipetted into the wells of a Perkin Elmer Optiplate-96 6005290 (100 ⁇ l per well) and 100 ⁇ l of a dilution series of the GLP-1 analogue or derivative to be measured is added in the same buffer. After 20 hours of gentle rocking at room temperature the plates are centrifuged and counted on a TopCounter. Bound cpm are plotted as a function of GLP-1 analogue or derivative concentration end the EC50 value of the competition curve is used as a measure of the affinity of the analogue or derivative for HSA.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has substantially improved terminal half-life in rodent and in a non-rodent model relative to liraglutide.
  • the terminal half-life in rodent or in a non-rodent model is improved at least 3 fold relative to liraglutide.
  • the terminal half-life in a non-rodent model is improved at least 6 fold relative to liraglutide.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has an in vivo half-life of at least 10 hrs after i.v. administration to rats.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has an in vivo half-life of at least 50 hrs after s.c. administration to mini pigs, and preferable an in vivo half-life of at least 80 hrs after s.c. administration to mini pigs.
  • the present invention relates to a GLP-1 analogue or derivative thereof which can be formulated into particles suitable for pulmonary administration.
  • the present invention relates to a GLP-1 analogue or derivative thereof which is chemically and physically stable at neutral pH, most preferably in the range 6-8.
  • the present invention relates to a GLP-1 analogue or derivative thereof which has little or no tendency to aggregate.
  • the aggregation tendency is significantly improved relatively to the aggregation tendency of liraglutide when tested in a thioflavin assay.
  • the present invention relates to a GLP-1 analogue or derivative thereof which is suitable for pulmonal delivery. This may be with regard to physical or chemical aspects which are useful for a pulmonal formulation. Alternatively, the analogues or derivatives are stable against degradation by enzymes in the airways and lungs.
  • albumin binding moiety means a residue which binds non-covalently to human serum albumin.
  • the albumin binding residue attached to the therapeutic polypeptide typically has an albumin binding affinity that is below 1 micromolar, preferable below 500 nM and even more preferable below 200 nM or even below 100 nM.
  • a range of albumin binding residues are known among linear and branched lipohophillic moieties containing 4-40 carbon atoms having a distal acidic group.
  • hydrophilic linker as used herein means a spacer that separates a peptide and an albumin binding residue with a chemical moiety which comprises at least 5 non-hydrogen atoms where 30-50% of these are either N or O.
  • the invention relates to a GLP-1 analogue or derivative thereof, which comprises a hydrophilic linker between the modified GLP-1 sequence and one or more albumin binding residue(s).
  • the hydrophilic linker is an unbranched oligo ethylene glycol moiety with appropriate functional groups at both terminals that forms a bridge between an amino group of the modified GLP-1 sequence and a functional group of the albumin binding residue.
  • terminal bonds from the attached groups are to be regarded as attachment bonds and not ending in methylene groups unless stated.
  • the GLP-1 analogue or derivative is selected form the group consisting of
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Va125,Arg26,Leu27,Glu30, Lys33)GLP-1(7-33)amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26,Glu30) GLP-1(7-33) amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26,Glu30) GLP-1(7-33) amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -[Aib8,Lys20,Glu22,Va125,Arg26,Glu30] GLP-1 (7-33) amide, and
  • Another object of the present invention is to provide a pharmaceutical formulation comprising an analogue or derivative according to the present invention which is present in a concentration from 0.1 mg/ml to 25 mg/ml, and wherein said formulation has a pH from 3.0 to 9.0.
  • the formulation may further comprise a buffer system, preservative(s), tonicity agent(s), chelating agent(s), stabilizers and surfactants.
  • the pharmaceutical formulation is an aqueous formulation, i.e. formulation comprising water.
  • 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, and the term “aqueous suspension” is defined as a suspension comprising at least 50% w/w water.
  • the pharmaceutical formulation is a freeze-dried formulation, whereto the physician or the patient adds solvents and/or diluents prior to use.
  • the 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
  • a pharmaceutical formulation comprising an aqueous solution of an analogue or derivative according to the present invention, and a buffer, wherein said analogue or derivative is present in a concentration from 0.1 mg/ml or above, and wherein said formulation has a pH from about 3.0 to about 9.0.
  • 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, the pharmaceutical formulation is from 8.0 to 8.5.
  • each administered dose contains from 0.01 mg-10 mg of active analogue or derivative according to the invention. In an embodiment, the dose administered contains more than 0.05 mg active analogue or derivative. In an embodiment, the dose administered contains more than 0.1 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 10 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 9 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 8 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 7 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 6 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains up to 5 mg active analogue or derivative according to the invention. In an embodiment, the dose administered contains from 0.2 mg to 5 mg active analogue or derivative according to the invention.
  • the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, 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 phenol or m-cresol. 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, 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-propanediol (propyleneglycol), 1,3-propanediol, 1,3-butanediol) polyethyleneglycol (e.g.
  • the isotoncity agent is propyleneglycol.
  • 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, alfa and beta HPCD, 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, galactitol, 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 an embodiment of the invention, the isotonic agent is present in a concentration from 5 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.
  • the chelating agent is present in a concentration from 0.1 mg/ml to 5 mg/ml.
  • the chelating agent is present in a concentration from 0.1 mg/ml to 2 mg/ml.
  • the chelating agent is present in a concentration from 2 mg/ml to 5 mg/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, 19 th edition, 1995.
  • the formulation further comprises a stabilizer.
  • 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, 19 th edition, 1995.
  • 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.
  • 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.
  • 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.
  • 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 a mixture thereof
  • a particular amino acid e.g. methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof
  • a particular amino acid e.g. methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof
  • 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.
  • 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 ethionine and buthionine and suitable cysteine analogues include S-methyl-L cysteine.
  • 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 sulphuric 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.
  • inhibitor 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 or D) 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 stabilizer selected from the group of high molecular weight polymers or low molecular compounds.
  • the stabilizer is selected from polyethylene glycol (e.g. PEG 3350), polyvinyl alcohol (PVA), polyvinylpyrrolidone, 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 polyvinyl alcohol
  • PVC polyvinylpyrrolidone
  • carboxy-/hydroxycellulose or derivates thereof e.g. HPC, HPC-SL, HPC-L and HPMC
  • cyclodextrins e.g. sulphur-containing substances as monothioglycerol, thioglycolic acid and 2-methylthioethanol, and different salts (e.g. sodium chloride).
  • 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 pharmaceutical composition comprises two different surfactants.
  • the term “Surfactant” as used herein refers to any molecules or ions that are comprised of a water-soluble (hydrophilic) part, the head, and a fat-soluble (lipophilic) segment. Surfactants accumulate preferably at interfaces, which the hydrophilic part is orientated towards the water (hydrophilic phase) and the lipophilic part towards the oil- or hydrophobic phase (i.e. glass, air, oil etc.). The concentration at which surfactants begin to form micelles is known as the critical micelle concentration or CMC. Furthermore, surfactants lower the surface tension of a liquid. Surfactants are also known as amphipathic compounds.
  • the term “Detergent” is a synonym used for surfactants in general.
  • Anionic surfactants may be selected from the group of: Chenodeoxycholic acid, Chenodeoxycholic acid sodium salt, Cholic acid, Dehydrocholic acid, Deoxycholic acid, Deoxycholic acid methyl ester, Digitonin, Digitoxigenin, N,N-Dimethyldodecylamine N-oxide, Docusate sodium, Glycochenodeoxycholic acid sodium, Glycocholic acid hydrate, Glycodeoxycholic acid monohydrate, Glycodeoxycholic acid sodium salt, Glycodeoxycholic acid sodium salt, Glycolithocholic acid 3-sulfate disodium salt, Glycolithocholic acid ethyl ester, N-Lauroylsarcosine sodium salt, N-Lauroylsarcosine sodium salt, N-Lauroylsarcosine, N-Lauroylsarcosine, Lithium dodecyl sulfate, Lugol, 1-Octanesulf
  • Cationic surfactants may be selected from the group of: Alkyltrimethylammonium bromide
  • Benzalkonium chloride Benzalkonium chloride, Benzalkonium chloride, Benzyldimethylhexadecylammonium chloride, Benzyldimethyltetradecylammonium chloride, Benzyltrimethylammonium tetrachloroiodate, Dimethyldioctadecylammonium bromide, Dodecylethyldimethylammonium bromide, Dodecyltrimethylammonium bromide, Dodecyltrimethylammonium bromide, Ethylhexadecyldimethylammonium bromide, Hexadecyltrimethylammonium bromide, Hexadecyltrimethylammonium bromide, Polyoxyethylene(10)-N-tallow-1,3-diaminopropane, Thonzonium bromide, and/or Trimethyl(tetradecyl)ammonium bromide.
  • Nonionic surfactants may be selected from the group of: BigCHAP, Bis(polyethylene glycol bis[imidazoyl carbonyl]), block copolymers as polyethyleneoxide/polypropyleneoxide block copolymers such as poloxamers, poloxamer 188 and poloxamer 407, Brij® 35, Brij® 56, Brij® 72, Brij® 76, Brij® 92V, Brij® 97, Brij® 58P, Cremophor® EL, Decaethylene glycol monododecyl ether, N-Decanoyl-N-methylglucamine, n-Dodecanoyl-N-methylglucamide, alkyl-polyglucosides, ethoxylated castor oil, Heptaethylene glycol monodecyl ether, Heptaethylene glycol monododecyl ether, Heptaethylene glycol monotetradecyl ether, Hexaethylene glycol monodode
  • Zwitterionic surfactants may be selected from the group of: CHAPS, CHAPSO, 3-(Decyldimethylammonio)propanesulfonate inner salt, 3-(Dodecyldimethylammonio)-propanesulfonate inner salt, 3-(Dodecyldimethylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylmyristylammonio)propanesulfonate, 3-(N,N-Dimethyloctadecyl-ammonio)propanesulfonate, 3-(N,N-Dimethyloctylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylpalmitylammonio)propanesulfonate, N-alkyl-N,N-dimethylammonio-1-propanesulfonates, 3-cholamido-1-propyldimethylammonio-1-propanesulfonate
  • oleic acid and caprylic acid N-Hexadecyl-N,N-dimethyl-3-ammonio-1-propanesulfonate, anionic (alkyl-aryl-sulphonates) monovalent surfactants, palmitoyl lysophosphatidyl-L-serine, lysophospholipids (e.g. 1-acyl-sn-glycero-3-phosphate esters of ethanolamine, choline, serine or threonine), or mixtures thereof.
  • anionic (alkyl-aryl-sulphonates) monovalent surfactants palmitoyl lysophosphatidyl-L-serine
  • lysophospholipids e.g. 1-acyl-sn-glycero-3-phosphate esters of ethanolamine, choline, serine or threonine
  • alkyl-polyglucosides as used herein in relates to an straight or branched C 5-20 -alkyl, -alkenyl or -alkynyl chain which is substituted by one or more glucoside moieties such as maltoside, saccharide etc.
  • Embodiments of these alkyl-polyglucosides include C 6-18 -alkyl-polyglucosides.
  • Specific embodiments of these alkyl-polyglucosides includes the even numbered carbon-chains such as C 6 , C 8 , C 10 , C 12 , C 14 , C 16 , C 18 and C 20 alkyl chain.
  • glucoside moieties include pyranoside, glucopyranoside, maltoside, maltotrioside and sucrose. In embodiments of the invention, less than 6 glucosid moieties are attached to the alkyl group. In embodiments of the invention, less than 5 glucosid moieties are attached to the alkyl group. In embodiments of the invention, less than 4 glucosid moieties are attached to the alkyl group. In embodiments of the invention, less than 3 glucosid moieties are attached to the alkyl group. In embodiments of the invention, less than 2 glucosid moieties are attached to the alkyl group.
  • alkyl-polyglucosides are alkyl glucosides such n-decyl ⁇ -D-glucopyranoside, decyl ⁇ -D-maltopyranoside, dodecyl ⁇ -D-glucopyranoside, n-dodecyl ⁇ -D-maltoside, n-dodecyl ⁇ -D-maltoside, n-dodecyl ⁇ -D-maltoside, tetradecyl ⁇ -D-glucopyranoside, decyl ⁇ -D-maltoside, hexadecyl ⁇ -D-maltoside, decyl ⁇ -D-maltotrioside, dodecyl ⁇ -D-maltotrioside, tetradecyl ⁇ -D-maltotrioside, hexadecyl ⁇ -D-maltotrioside, n-dodecyl-s
  • the formulation further comprises protease inhibitors such as EDTA (ethylenediamine tetraacetic acid) and benzamidineHCl, but other commercially available protease inhibitors may also be used.
  • protease inhibitors such as EDTA (ethylenediamine tetraacetic acid) and benzamidineHCl, but other commercially available protease inhibitors may also be used.
  • EDTA ethylenediamine tetraacetic acid
  • benzamidineHCl benzamidineHCl
  • the use of a protease inhibitor is particular useful in pharmaceutical compositions comprising zymogens of proteases in order to inhibit autocatalysis.
  • 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, gelatine 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 an analogue or derivative according to the 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.
  • 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.
  • 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.
  • 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, chewing gum, 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.
  • solutions for example, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets
  • 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 analogue or derivative of the present invention, 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 an analogue or derivative according to the invention, 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-crystallization, precipitation, co-precipitation, emulsification, dispersion, high pressure homogenisation, encapsulation, spray drying, microencapsulating, 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.
  • parenteral administration can be performed by means of an infusion pump.
  • a further option is a composition which may be a solution or suspension or a powder for the administration of the analogue or derivative of the present invention in the form of a nasal or pulmonal liquid or powder spray.
  • the pharmaceutical compositions containing the analogue or derivative according to 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 analogues or derivatives according to the present invention can be administered via the pulmonary route in a vehicle, as a solution, suspension or dry powder using any of known types of devices suitable for pulmonary drug delivery.
  • Examples of these comprise, but are not limited to, the three general types of aerosol-generating for pulmonary drug delivery, and may include jet or ultrasonic nebulizers, metered-dose inhalers, or dry powder inhalers (Cf. Yu J, Chien Y W. Pulmonary drug delivery: Physiologic and mechanistic aspects. Crit Rev Ther Drug Carr Sys 14(4) (1997) 395-453).
  • the aerodynamic diameter (d a ) of a particle is defined as the geometric equivalent diameter of a reference standard spherical particle of unit density (1 g/cm 3 ).
  • d a is related to a reference diameter (d) as a function of the square root of the density ratio as described by:
  • Mass median aerodynamic diameter (MMAD) and mass median effective aerodynamic diameter (MMEAD) are used inter-changeably, are statistical parameters, and empirically describe the size of aerosol particles in relation to their potential to deposit in the lungs, independent of actual shape, size, or density (cf. Edwards D A, Ben-Jebria A, Langer R. Recent advances in pulmonary drug delivery using large, porous inhaled particles. J Appl Physiol 84(2) (1998) 379-385).
  • MMAD is normally calculated from the measurement made with impactors, an instrument that measures the particle inertial behaviour in air.
  • the formulation could be aerosolized by any known aerosolisation technology, such as nebulisation, to achieve a MMAD of aerosol particles less than 10 ⁇ m, more preferably between 1-5 ⁇ m, and most preferably between 1-3 ⁇ m.
  • the preferred particle size is based on the most effective size for delivery of drug to the deep lung, where protein is optimally absorbed (cf. Edwards D A, Ben-Jebria A, Langer A, Recent advances in pulmonary drug delivery using large, porous inhaled particles. J Appl Physiol 84(2) (1998) 379-385).
  • Deep lung deposition of the pulmonal formulations comprising the analogue or derivative according to the invention may optional be further optimized by using modifications of the inhalation techniques, for example, but not limited to: slow inhalation flow (eg. 30 L/min), breath holding and timing of actuation.
  • 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. 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.
  • 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.
  • 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 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.
  • 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.
  • a “stabilized formulation” refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • 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 analogue or derivative according to the present invention is stable for more than 6 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical formulation comprising the analogue or derivative according to the present invention is stable for more than 4 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical formulation comprising the analogue or derivative according to the present invention is stable for more than 4 weeks of usage and for more than two years of storage.
  • the pharmaceutical formulation comprising the analogue or derivative according to the present invention 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 an analogue or derivative according to the present for the preparation of a medicament.
  • an analogue or derivative according to the present 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, stroke, coronary heart disease and other cardiovascular disorders, inflammatory bowel syndrome, dyspepsia and gastric ulcers.
  • an analogue or derivative according to the present is used for the preparation of a medicament for delaying or preventing disease progression in type 2 diabetes.
  • an analogue or derivative according to the present is used for the preparation of a medicament for decreasing food intake, decreasing ⁇ -cell apoptosis, increasing ⁇ -cell function and ⁇ -cell mass, and/or for restoring glucose sensitivity to ⁇ -cells.
  • the treatment with an analogue or derivative according to the present invention may also be 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), Gastric Inhibitory Polypeptides (GIP analogs), 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
  • the treatment with an analogue or derivative according to the present may also be combined with surgery—a surgery that influence the glucose levels and/or lipid homeostasis such as gastric banding or gastric bypass.
  • the analogues of this invention can be produced by a method which comprises culturing 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.
  • 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, precipitating the proteinaceous components of the supernatant or filtrate by means of a salt, e.g. ammonium sulphate, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, gel filtration chromatography, affinity chromatography, or the like, dependent on the type of peptide in question.
  • a salt e.g. ammonium sulphate
  • 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 polypeptide by hybridisation using synthetic oligonucleotide probes in accordance with standard techniques (see, for example, Sambrook, J, Fritsch, E F and Maniatis, T, Molecular Cloning: A Laboratory
  • 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 U.S. Pat. No. 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 peptide 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 peptide 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.
  • 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.
  • 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 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.
  • Xaa 34 is Lys, Glu, Asn, Arg, Cys or absent;
  • n is selected from the group consisting of 14, 15, 16 17, 18 and 19
  • p is selected from the group consisting of 10, 11, 12, 13 and 14
  • d is selected from the group consisting of 0, 1, 2, 3, 4 and 5,
  • x is selected from the group consisting of 0, 1, 2, 3 and 4, and y is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12,
  • b and e are each independently selected from the group consisting of 0, 1 and 2
  • c and f are each independently selected from the group consisting of 0, 1 and 2 with the proviso that b is 1 or 2 when c is 0, or b is 0 when c is 1 or 2, and e is 1 or 2 when f is 0, or e is 0 when f is 1 or 2, and
  • k is selected from the group consisting of 0, 1, 2, 3, 4, 5, 11 and 27, and m is selected from the group consisting of 0, 1, 2, 3, 4, 5 and 6.
  • amino acid sequence of human GLP-1(7-35) is included in the Sequence Listing as SEQ ID No 1, and SEQ ID Nos 2 and 3 are derivatives thereof according to the invention.
  • position 1 of SEQ ID No 1 is equivalent to position 7 of GLP-1(7-35) (His)
  • position 16 of SEQ ID No 1 is equivalent to position 22 of GLP-1(7-35) (Gly)
  • position 20 of SEQ ID No 1 is equivalent to position 26 of GLP-1(7-35) (Lys)—and vice versa for the other positions and the other sequences.
  • the invention also provides, in above embodiment 1, a GLP-1 analogue or derivative which comprises a modified GLP-1(7-35) sequence having:
  • amino acid(s) at a position equivalent to position 30, 31, 32, 33, 34, or 35 of GLP-1(7-35) can be absent provided that if the amino acid at position 30, 31, 32, 33 or 34 of GLP1(7-35) (position 24, 25, 26, 27, or 28 of SEQ ID No 1) is absent then each amino acid residue downstream is also absent, and
  • the invention furthermore provides GLP-1 analogues or derivatives, methods and uses thereof, and pharmaceutical compositions with a content thereof corresponding to any of the above embodiments and particular embodiments according to the invention, in which corresponding position numbering amendments have been made as explained above, and shown above for the GLP-1 analogue or derivative of above embodiment 1.
  • a GLP-1 analogue or derivative thereof which comprises a modified GLP-1 sequence 7-35 (SEQ ID No 1) having:
  • amino acid(s) at a position equivalent to position 30, 31, 32, 33, 34, or 35 of SEQ ID No 1 can be absent provided that if the amino acid at position 30, 31, 32, 33 or 34 is absent then each amino acid residue downstream is also absent, and
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 26 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 25 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 31, 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 24 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 30, 31, 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 23 amino acids.
  • 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, Gly, Val, Leu, Ile, 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 9 is Glu or a Glu derivative such as alpha, alpha dimethyl-Glu;
  • Xaa 16 is Val or Leu
  • Xaa 18 is Ser, Lys, Cys or Arg
  • Xaa 20 is Leu, Lys or Cys
  • Xaa 23 is Gln, Glu, Lys, Cys or Arg;
  • Xaa 24 is Ala or Asn
  • Xaa 25 is Ala or Val
  • Xaa 27 is Glu, Ala or Leu;
  • Xaa 30 is Ala, Glu, Lys, Arg or absent;
  • Xaa 31 is Trp, Lys, Cys or absent
  • Xaa 33 is Val, Lys, Cys or absent
  • Xaa 34 is Lys, Glu, Asn, Arg, Cys or absent;
  • Xaa 35 is Gly, Aib or absent
  • R is amide or is absent
  • each amino acid residue downstream is also absent.
  • 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, Gly, Val, Leu, Ile, 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 18 is Ser, Lys or Arg
  • Xaa 30 is Ala, Glu, Lys, Arg or is absent;
  • Xaa 33 is Val, Lys or absent
  • Xaa 34 is Lys, Glu, Arg or is absent;
  • Xaa 35 is Gly, Aib or is absent;
  • R is amide or is absent.
  • A- is selected from the group consisting of
  • n is selected from the group consisting of 14, 15, 16 17, 18 and 19
  • p is selected from the group consisting of 10, 11, 12, 13 and 14
  • d is selected from the group consisting of 0, 1, 2, 3, 4 and 5,
  • -B- is selected from the group consisting of
  • x is selected from the group consisting of 0, 1, 2, 3 and 4, and y is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12,
  • -C- is selected from the group consisting of
  • b and e are each independently selected from the group consisting of 0, 1 and 2
  • c and f are each independently selected from the group consisting of 0, 1 and 2 with the proviso that b is 1 or 2 when c is 0, or b is 0 when c is 1 or 2, and e is 1 or 2 when f is 0, or e is 0 when f is 1 or 2, and
  • -D- is attached to said amino acid residue and is a linker.
  • k is selected from the group consisting of 0, 1, 2, 3, 4, 5, 11 and 27, and m is selected from the group consisting of 0, 1, 2, 3, 4, 5 and 6.
  • a pharmaceutical composition comprising a GLP-1 analogue or derivative thereof according to any one of embodiments 1-13, and a pharmaceutically acceptable excipient.
  • a GLP-1 analogue which is a modified GLP-1(7-35) (SEQ ID No 1) having:
  • GLP-1 analogue or derivative thereof wherein the amino acid(s) at a position equivalent to position 30, 31, 32, 33, 34, or 35 of GLP-1(7-35) are absent, provided that if the amino acid at position 30, 31, 32, 33, or 34 is absent then each amino acid residue downstream is also absent.
  • GLP-1 analogue or derivative thereof according to any one of embodiments 1-2, wherein the GLP-1 analogue comprises i) a C-terminal carboxylic acid group; or iii) a C-terminal amide group.
  • GLP-1 analogue or derivative thereof according to any one of embodiments 1-3, which is derivatised with an albumin binding residue or is pegylated.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-4, wherein the amino acid at position 35 is absent, and wherein the total length of the GLP-1 analogue is 28 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-6, wherein the amino acids at position 33, 34, and 35 are absent, and wherein the total length of the GLP-1 analogue is 26 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-7, wherein the amino acids at position 32, 33, 34, and 35 are absent, and wherein the total length of the GLP-1 analogue is 25 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-8, wherein the amino acids at position 31, 32, 33, 34, and 35 are absent, and wherein the total length of the GLP-1 analogue is 24 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9, wherein the amino acids at position 30, 31, 32, 33, 34, and 35 are absent, and wherein the total length of the GLP-1 analogue is 23 amino acids.
  • a GLP-1 analogue or derivative thereof having the sequence of formula (I) having the sequence of formula (I)
  • 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, Gly, Val, Leu, Ile, 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 9 is Glu or a Glu derivative such as alpha, alpha dimethyl-Glu;
  • Xaa 16 is Val or Leu
  • Xaa 18 is Ser, Lys, Cys or Arg
  • Xaa 20 is Leu, Lys or Cys
  • Xaa 23 is Gln, Glu, Lys, Cys or Arg;
  • Xaa 24 is Ala or Asn
  • Xaa 25 is Ala or Val
  • Xaa 27 is Glu, Ala or Leu;
  • Xaa 30 is Ala, Glu, Lys, Arg or absent;
  • Xaa 31 is Trp, Lys, Cys or absent
  • Xaa 33 is Val, Lys, Cys or absent
  • Xaa 34 is Lys, Glu, Asn, Arg, Cys or absent;
  • Xaa 35 is Gly, Aib or absent
  • R is amide or is absent
  • each amino acid residue downstream is also absent.
  • a GLP-1 analogue or derivative thereof having the sequence of formula (II) having the sequence of formula (II)
  • 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, Gly, Val, Leu, Ile, 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 18 is Ser, Lys or Arg
  • Xaa 30 is Ala, Glu, Lys, Arg or is absent;
  • Xaa 33 is Val, Lys or absent
  • Xaa 34 is Lys, Glu, Arg or is absent;
  • Xaa 35 is Gly, Aib or is absent;
  • R is amide or is absent.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-12, wherein at least one amino acid residue is derivatised with A-B-C-D-
  • A- is selected from the group consisting of
  • n is selected from the group consisting of 14, 15, 16 17, 18 and 19
  • p is selected from the group consisting of 10, 11, 12, 13 and 14
  • d is selected from the group consisting of 0, 1, 2, 3, 4 and 5,
  • -B- is selected from the group consisting of
  • x is selected from the group consisting of 0, 1, 2, 3 and 4, and y is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12,
  • -C- is selected from the group consisting of
  • b and e are each independently selected from the group consisting of 0, 1 and 2
  • c and f are each independently selected from the group consisting of 0, 1 and 2 with the proviso that b is 1 or 2 when c is 0, or b is 0 when c is 1 or 2, and e is 1 or 2 when f is 0, or e is 0 when f is 1 or 2, and
  • -D- is attached to said amino acid residue and is a linker.
  • k is selected from the group consisting of 0, 1, 2, 3, 4, 5, 11 and 27, and m is selected from the group consisting of 0, 1, 2, 3, 4, 5 and 6.
  • a GLP-1 analogue or derivative which is selected from the following:
  • a pharmaceutical composition comprising a GLP-1 analogue or derivative thereof according to any one of embodiments 1-15 or a pharmaceutically acceptable salt, amide, alkyl, or ester thereof, and a pharmaceutically acceptable excipient.
  • GLP-1 analogue or derivative thereof in the manufacture of a medicament for use in 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 method of treating or preventing 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 by administering a pharmaceutically active amount of a GLP-1 analogue or derivative thereof according to any one of embodiments 1-15 or a pharmaceutical composition according to embodiment 16.
  • a GLP-1 analogue or derivative thereof which comprises a modified GLP-1 sequence 7-35 (SEQ ID No 1) having:
  • amino acid(s) at a position equivalent to position 30, 31, 32, 33, 34, or 35 of SEQ ID No 1 can be absent provided that if the amino acid at position 30, 31, 32, 33 or 34 is absent then each amino acid residue downstream is also absent, and optionally a C-terminal amide group.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 26 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 25 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 31, 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 24 amino acids.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-2, wherein the amino acids at position 30, 31, 32, 33, 34 and 35 are absent, and wherein the total length of the GLP-1 analogue is 23 amino acids.
  • GLP-1 analogue or derivative thereof having 3 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 1-10, which has a substitution at a position selected from the group of position 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 11-12, which has a substitution selected from the group consisting of desaminoHis7, Aib8, Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 11-13, which has a substitution selected from the group consisting of desaminoHis7 and Aib8.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 having 4 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 and 15, which has two substitutions at positions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 15-16, which has two substitutions selected from the group consisting of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • GLP-1 analogue or derivative thereof having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and an amino acid substitution selected from the group consisting Lys18, Lys20, Lys23, Glu30, Lys31,Lys33 and Lys34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 15-18 having an amino acid substitution selected from the group consisting desaminoHis7 and Aib8, and an amino acid substitution selected from the group consisting Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 having 5 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 and 20, which has three amino acid substitutions at positions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34 compared to the sequence 7-35 of SEQ ID NO 1.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 20-21, which has three amino acid substitutions selected from the group of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 20-22 having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8, and two amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 20-23 having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8, and two amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 having 6 amino acid substitutions compared to the sequence 7-35 of SEQ ID NO 1 including the substitutions at position 22 and 26.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-9 and 25, which has four amino acid substitutions at positions selected from the group of positions 7, 8, 18, 20, 23, 24, 25, 27, 30, 31, 33 and 34.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 25-26, which has four amino acid substitutions selected from the group consisting of desaminoHis7, Aib8, Lys18, Cys18, Lys20, Cys20, Lys23, Cys23, Asn24, Val25, Ala27, Leu27, Glu30, Lys31, Cys31, Lys33, Cys33, Lys34, Cys34 and Asn34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 25-27 having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and three amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 25-28 having an amino acid substitution selected from the group consisting of desaminoHis7 and Aib8 and three amino acid substitutions selected from the group consisting of Lys18, Lys20, Lys23, Glu30, Lys31, Lys33 and Lys34.
  • 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, Gly, Val, Leu, Ile, 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 9 is Glu or a Glu derivative such as alpha, alpha dimethyl-Glu;
  • Xaa 16 is Val or Leu
  • Xaa 18 is Ser, Lys, Cys or Arg
  • Xaa 20 is Leu, Lys or Cys
  • Xaa 23 is Gln, Glu, Lys, Cys or Arg;
  • Xaa 24 is Ala or Asn
  • Xaa 25 is Ala or Val
  • Xaa 27 is Glu, Ala or Leu;
  • Xaa 30 is Ala, Glu, Lys, Arg or absent;
  • Xaa 31 is Trp, Lys, Cys or absent
  • Xaa 33 is Val, Lys, Cys or absent
  • Xaa 34 is Lys, Glu, Asn, Arg, Cys or absent;
  • Xaa 35 is Gly, Aib or absent
  • R is amide or is absent
  • each amino acid residue downstream is also absent.
  • 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, Gly, Val, Leu, Ile, 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 18 is Ser, Lys or Arg
  • Xaa 30 is Ala, Glu, Lys, Arg or is absent;
  • Xaa 33 is Val, Lys or absent
  • Xaa 34 is Lys, Glu, Arg or is absent;
  • Xaa 35 is Gly, Aib or is absent;
  • R is amide or is absent.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 30-32, wherein Xaa 35 and R are absent.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 30-33, wherein Xaa 34 , Xaa 35 and R are absent.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 30-34, wherein Xaa 33 , Xaa 34 , Xaa 35 and and R are absent.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 30 and 32-35 having a total of 2 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2 and an Arg residue at a position equivalent to position 26 of SEQ ID No 2.
  • the GLP-1 analogue or derivative thereof having a total of 3 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and one amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof having a total of 4 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and 2 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof having a total of 5 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and 3 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 30 and 32-35 having a total of 6 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and 4 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof having a total of 7 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and 5 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 30 and 32-35 having a total of 8 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 2, an Arg residue at a position equivalent to position 26 of SEQ ID No 2 and 3 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 9 , Xaa 16 , Xaa 18 , Xaa 20 , Xaa 23 , Xaa 24 , Xaa 25 , Xaa 27 , Xaa 30 , Xaa 31 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 2 compared to the sequence 7-35 of SEQ ID No 1.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 2 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3 and an Arg residue at a position equivalent to position 26 of SEQ ID No 3.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 3 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and one amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 4 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and 2 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 5 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and 3 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 6 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and 4 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 7 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and 5 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 31-35 having a total of 8 amino acids substitutions compared to the sequence 7-35 of SEQ ID NO 1, which are a Glu residue at a position equivalent to position 22 of SEQ ID No 3, an Arg residue at a position equivalent to position 26 of SEQ ID No 3 and 3 amino acid substitution selected from the group consisting of Xaa 7 , Xaa 8 , Xaa 18 , Xaa 30 , Xaa 33 , Xaa 34 and Xaa 35 in SEQ ID No 3 compared to the sequence 7-35 of SEQ ID No 1.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 30-49 wherein Xaa 7 is desamino-histidine.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 30-49 wherein Xaa 8 is Aib.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-51, wherein the amino acid which is pegylated or derivatised with an albumin binding residue is a Lys-residue or a Cys-residue.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-51, wherein the amino acid which is pegylated or derivatised with an albumin binding residue is a Lys-residue.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-51, wherein the C-terminal amino acid is pegylated or derivatised with an albumin binding residue.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-54, which has been pegylated or derivatised with an albumin binding residue at position 18, 20, 23, 31, 33, 34 or at the C-terminal amino acid.
  • GLP-1 analogue or derivative thereof according to any one of the embodiments 1-55, which has been pegylated or derivatised with an albumin binding residue at position 18.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 1-55, which has been pegylated or derivatised with an albumin binding residue at position 31.
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 1-55, which has been pegylated or derivatised with an albumin binding residue at position 34.
  • A- is selected from the group consisting of
  • n is selected from the group consisting of 14, 15, 16 17, 18 and 19
  • p is selected from the group consisting of 10, 11, 12, 13 and 14
  • d is selected from the group consisting of 0, 1, 2, 3, 4 and 5,
  • -B- is selected from the group consisting of
  • x is selected from the group consisting of 0, 1, 2, 3 and 4, and y is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12,
  • -C- is selected from the group consisting of
  • b and e are each independently selected from the group consisting of 0, 1 and 2
  • c and f are each independently selected from the group consisting of 0, 1 and 2 with the proviso that b is 1 or 2 when c is 0, or b is 0 when c is 1 or 2, and e is 1 or 2 when f is 0, or e is 0 when f is 1 or 2, and
  • -D- is attached to said amino acid residue and is a linker.
  • GLP-1 analogue or derivative thereof according to embodiment 61, wherein one amino acid residue is derivatised with A-B-C-D-.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 63-69, wherein n is selected from the group consisting of 15 and 17, and more is preferred 17.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 63-68 and 69, wherein p is selected from the group consisting of 12, 13, and 14 and more preferred is 13.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 61-68 and 69-70, wherein d is selected from the group consisting of 0, 1, 2, 3 and 4, more preferred 0, 1 and 2 and most preferred 1.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 63-68 and 69-71, wherein d is selected from the group consisting of 0, 1 and 2 and p is selected from the group consisting of 12, 13 or 14, more preferred d is selected from the group consisting of 1 and 2 and p is selected from the group consisting of 13 and 14, and most preferred d is 1 and p is 13.
  • GLP-1 analogue or derivative thereof according to embodiment 80, wherein y is selected from the group consisting of 2, 3, 4, 5, 6, 7, 8, 9 and 10 and more preferred y is selected from the group consisting of 2, 3, 4, 5, 6, 7, and 8.
  • GLP-1 analogue or derivative thereof according to embodiment 82, wherein c is selected from the group consisting of 0 and 1 and b is selected from the group consisting of 1 and 2, more preferred b is 1 and c is 0.
  • GLP-1 analogue or derivative thereof according to embodiment 84, wherein f is selected from the group consisting of 0 and 1 and e is selected from the group consisting of 1 and 2, more preferred e is 1 and f is 0.
  • k is selected from the group consisting of 0, 1, 2, 3, 4, 5, 11 and 27, and m is selected from the group consisting of 0, 1, 2, 3, 4, 5 and 6.
  • GLP-1 analogue or derivative thereof according to embodiment 88, wherein k is selected from the group consisting of 1, 2, 3, 11 and 27 and more preferred k is 1.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 88-89, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to embodiment 91, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to embodiment 93, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to embodiment 95, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to embodiment 97, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to embodiment 99, wherein m is selected from the group consisting of 0, 1, 2, 3, and 4 and more preferred m is selected from the group consisting of 0, 1 and 2.
  • GLP-1 analogue or derivative thereof according to any of the embodiments 1-100, wherein A-B-C-D- is selected and combined from
  • GLP-1 analogue or derivative thereof according to any of the embodiments 1-100, wherein A-B-C-D- is selected from the group consisting of
  • the GLP-1 analogue or derivative thereof according to any one of the embodiments 1-103, which comprises a hydrophilic linker between the modified GLP-1 sequence and one or more albumin binding residue(s).
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Val25,Arg26,Leu27,Glu30,Lys33)GLP-1(7-33)amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26,Glu30)GLP-1(7-33)amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26,Glu30)GLP-1(7-33)amide,
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -[Aib8,Lys20,Glu22,Val25,Arg26,Glu30]GLP-1(7-33)amide, and
  • a pharmaceutical composition comprising a GLP-1 analogue or derivative thereof according to any one of embodiments 1-107, and a pharmaceutically acceptable excipient.
  • composition according to embodiment 109 which is suited for parenteral administration.
  • GLP-1 analogue or derivative thereof 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.
  • GLP-1 analogue or derivative thereof for the preparation of a medicament for delaying or preventing disease progression in type 2 diabetes.
  • GLP-1 analogue or derivative thereof for the preparation of a medicament for decreasing food intake, decreasing ⁇ -cell apoptosis, increasing ⁇ -cell function and ⁇ -cell mass, and/or for restoring glucose sensitivity to ⁇ -cells.
  • a GLP-1 analogue or derivative thereof for use in 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.
  • the protected peptidyl resin was synthesized according to the Fmoc strategy on an Applied Biosystems 433 peptide synthesizer in 0.25 mmol or 1.0 mmol scale using the manufacturer supplied FastMoc UV protocols which employ HBTU (2-(1H-Benzotriazol-1-yl-)-1,1,3,3 tetramethyluronium hexafluorophosphate) or HATU (O-(7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) mediated couplings in NMP (N-methyl pyrrolidone), and UV monitoring of the deprotection of the Fmoc protection group.
  • the starting resin used for the synthesis of the peptide amides was Rink-Amide resin and either Wang or chlorotrityl resin was used for peptides with a carboxy C-terminal.
  • the protected amino acid derivatives used were standard Fmoc-amino acids (supplied from e.g. Anaspec, or Novabiochem) supplied in preweighed cartridges suitable for the ABI433A synthesizer with the exception of unnatural aminoacids such as Fmoc-Aib-OH (Fmoc-aminoisobutyric acid).
  • the N terminal amino acid was Boc protected at the alpha amino group (e.g. Boc-His(Boc)OH was used for peptides with His at the N-terminal).
  • the epsilon amino group of lysine at position 26 was either protected with Mtt, Mmt, Dde, ivDde, or Boc, depending on the route for attachment of the albumin binding moiety and spacer.
  • the synthesis of the peptides may in some cases be improved by the use of dipeptides protected on the dipeptide amide bond with a group that can be cleaved under acidic conditions such but not limited to 2-Fmoc-oxy-4-methoxybenzyl or 2,4,6-trimethoxybenzyl.
  • the use of pseudoproline dipeptides may be used (see e.g. catalogue from Novobiochem 2002/2003 or newer version, or W. R. Sampson (1999), J. Pep. Sci. 5, 403.
  • Method B One alternative method (method B) of peptide synthesis was by Fmoc chemistry on a microwave-based Liberty peptide synthesizer (CEM Corp., North Carolina).
  • the resin was Tentagel S RAM with a loading of 0.24 mmol/g.
  • the coupling chemistry was DIC/HOAt in NMP using amino acid solutions of 0.3 M in NMP and a molar excess of 8-10 fold. Coupling conditions was 5 minutes at up to 70° C. Deprotection was with 5% piperidine in NMP at up to 70° C. When a chemical modification of a lysine side chain was desired, the lysine was incorporated as Lys(Mtt).
  • the Mtt group was removed by suspending the resin in neat hexafluoroisopropanol for 20 minutes followed by washing with DCM and NMP.
  • the chemical modification of the lysine was performed either by manual synthesis or by one or more automated steps on the Liberty followed by a manual coupling.
  • Another method of peptide synthesis was by Fmoc chemistry on an ABI 433 with HBTU coupling. After synthesis the resin was washed with DCM and dried, and the peptide was cleaved from the resin by a 2 hour treatment with TFA/TIS/water (92.5/5/2.5) followed by precipitation with diethylether. the peptide was redissolved in 30% acetic acid or similar solvent and purified by standard RP-HPLC on a C18 column using acetonitrile/TFA. The identity of the peptide was confirmed by MALDI-MS.
  • the protected peptidyl resin was synthesized according to the Fmoc strategy on an Advanced ChemTech Synthesiser (APEX 348) 0.25 mmol scale using the manufacturer supplied protocols which employ DIC (dicyclohexylcarbodiimide) and HOBt (1-Hydroxybenzotriazole) mediated couplings in NMP (N-methyl pyrrolidone.
  • the starting resin used for the synthesis of the peptide amides was Rink-Amide resin and either Wang or chlorotrityl resin was used for peptides with a carboxy C-terminal.
  • the protected amino acid derivatives used were standard Fmoc-amino acids (supplied from e.g. Anaspec, or Novabiochem.
  • the N terminal amino acid was Boc protected at the alpha amino group (e.g. Boc-His(Boc)OH was used for peptides with His at the N-terminal).
  • the epsilon amino group of lysine at position 26 was either protected with Mtt, Mmt, Dde, ivDde, or Boc, depending on the route for attachment of the albumin binding moiety and spacer.
  • the synthesis of the peptides may in some cases be improved by the use of dipeptides, e.g., pseudoprolines from Novabiochem, Fmoc-Ser(tbu)- ⁇ Ser(Me,Me)-OH, see e.g. catalogue from Novobiochem 2002/2003 or newer version, or W. R. Sampson (1999), J. Pep. Sci. 5, 403
  • the resin (0.25 mmol) was placed in a manual shaker/filtration apparatus and treated with 2% hydrazine in N-methyl pyrrolidone (20 ml, 2 ⁇ 12 min) to remove the Dde or ivDde group and wash with N-methyl pyrrolidone (4 ⁇ 20 ml).
  • the resin (0.25 mmol) was placed in a manual shaker/filtration apparatus and treated with 2% TFA and 2-3% TIS in DCM (20 ml, 5-10 min repeated 6-12 times) to remove the Mtt or Mmt group and wash with DCM (2 ⁇ 20 ml), 10% MeOH and 5% DIPEA in DCM (2 ⁇ 20 ml) and N-methyl pyrrolidone (4 ⁇ 20 ml).
  • the resin was placed in a syringe and treated with hexafluroisopropanol for 2 ⁇ 10 min to remove the Mtt group. The resin was then washed with DCM and NMP as described above.
  • the albumin binding residue (B—U-sidechain of formula I) can be attached to the peptide either by acylation to resin bound peptide or acylation in solution to the unprotected peptide using standard acylating reagent such as but not limited to DIC, HOBt/DIC, HOAt/DIC, or HBTU.
  • Activated (active ester or symmetric anhydride) albumin binding residue (A-B)-sidechain of formula I) such as octadecanedioic acid mono-(2,5-dioxo-pyrrolidin-1-yl)ester (Ebashi et al. EP511600, 4 molar equivalents relative to resin bound peptide) was dissolved in NMP (25 mL), added to the resin and shaken overnight at room temperature. The reaction mixture was filtered and the resin was washed extensively with NMP, dichloromethane, 2-propanol, methanol and diethyl ether.
  • the albumin binding residue (A-(B)-sidechain of formula I) was dissolved in N-methyl pyrrolidone/methylene chloride (1:1, 10 ml).
  • the activating reagent such as 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 2 to 24 hours at room temperature.
  • the resin was washed with N-methyl pyrrolidone (2 ⁇ 20 ml), N-methyl pyrrolidone/Methylene chloride (1:1) (2 ⁇ 20 ml) and methylene chloride (2 ⁇ 20 ml).
  • Activated (active ester or symmetric anhydride) albumin binding residue such as octadecanedioic acid mono-(2,5-dioxo-pyrrolidin-1-yl)ester (Ebashi et al. EP511600, 1-1.5 molar equivalents relative to the peptide was dissolved in an organic solvent such as acetonitrile, THF, DMF, DMSO or in a mixture of water/organic solvent (1-2 ml) and added to a solution of the peptide in water (10-20 ml) together with 10 molar equivalents of DIPEA.
  • organic solvent such as acetonitrile, THF, DMF, DMSO or in a mixture of water/organic solvent (1-2 ml)
  • the reaction mixture was lyophilized O/N and the isolated crude peptide deprotected afterwards—in case of a tert-butyl group the peptide was dissolved in a mixture of trifluoroacetic acid, water and triisopropylsilane (90:5:5). After for 30 min the mixture was, evaporated in vacuo and the finale peptide purified by preparative HPLC.
  • 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 to 92:4:4).
  • 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 1 to 3 times with 45 ml diethyl ether.
  • the crude peptide was purified by semipreparative HPLC on a 20 mm ⁇ 250 mm column packed with either 5 ⁇ or 7 ⁇ C-18 silica. Depending on the peptide 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% CH 3 CN in 0.05M (NH 4 ) 2 SO 4 , which was adjusted to pH 2.5 with concentrated H 2 SO 4 . 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.
  • the RP-HPLC analysis may be performed using UV detection at 214 nm and e.g. a Vydac 218TP54 4.6 mm ⁇ 250 mm 5 ⁇ C-18 silica column (The Separations Group, Hesperia, USA) and eluted at e.g. 1 ml/min at 42° C. Most often one of following specific conditions were used:
  • HPLC Method 03_A1 — 1: The RP-analysis was performed using a Waters 2690 systems fitted with a Waters 996 diode array detector. UV detections were collected at 214, 254, 276, and 301 nm on a 218TP54 4.6 mm ⁇ 250 mm 5 ⁇ C-18 silica column (The Separations Group, Hesperia), which was eluted at 1 ml/min at 42° C. The column was equilibrated with 10% of a 0.5 M ammonium sulfate, which was adjusted to pH 2.5 with 4M sulfuric acid. After injection, the sample was eluted by a gradient of 0% to 60% acetonitrile in the same aqueous buffer during 50 min.
  • HPLC Method 03_B1 — 2: The RP-analysis was performed using a Waters 2690 systems fitted with a Waters 996 diode array detector. UV detections were collected at 214, 254, 276, and 301 nm on a Zorbax 300SB C-18 (4.5 ⁇ 150 mm, 5 ⁇ ), which was eluted at 0.5 ml/min at 42° C. The column was equilibrated with an aqueous solution of TFA in water (0.1%). After injection, the sample was eluted by a gradient of 0% to 60% acetonitrile (+0.1% TFA) in an aqueous solution of TFA in water (0.1%) during 50 min.
  • HPLC Method 02_B1 — 1: The RP-analyses was performed using a Alliance Waters 2695 system fitted with a Waters 2487 dualband detector. UV detections at 214 nm and 254 nm were collected using a Vydac 218TP53, C18, 300 ⁇ , 5 um, 3.2 mm ⁇ 250 mm column, 42° C. Eluted with a linear gradient of 0-60% acetonitrile, 95-35% water and 5% trifluoroacetic acid (1.0%) in water over 50 minutes at a flow-rate of 0.50 ml/min.
  • HPLC Method 01_B4 — 2: RP-analyses was performed using a Waters 600S system fitted with a Waters 996 diode array detector. UV detections at 214 nm and 254 nm were collected using a Symmetry300 C18, 5 um, 3.9 mm ⁇ 150 mm column, 42° C. Eluted with a linear gradient of 5-95% acetonitrile, 90-0% water, and 5% trifluoroacetic acid (1.0%) in water over 15 minutes at a flow-rate of 1.0 min/min.
  • HPLC Method 02_B4 — 4
  • the RP-analyses was performed using a Alliance Waters 2695 system fitted with a Waters 2487 dualband detector. UV detections at 214 nm and 254 nm were collected using a Symmetry300 C18, 5 um, 3.9 mm ⁇ 150 mm column, 42° C. Eluted with a linear gradient of 5-95% acetonitrile, 90-0% water, and 5% trifluoroacetic acid (1.0%) in water over 15 minutes at a flow-rate of 1.0 min/min.
  • HPLC Method 02_B6 — 1: The RP-analyses was performed using a Alliance Waters 2695 system fitted with a Waters 2487 dualband detector. UV detections at 214 nm and 254 nm were collected using a Vydac 218TP53, C18, 300 ⁇ , 5 um, 3.2 mm ⁇ 250 mm column, 42° C. Eluted with a linear gradient of 0-90% acetonitrile, 95-5% water, and 5% trifluoroacetic acid (1.0%) in water over 50 minutes at a flow-rate of 0.50 ml/min.
  • HPLC Method 03_B1 — 1: The RP-analysis was performed using a Waters 2690 systems fitted with a Waters 996 diode array detector. UV detections were collected at 214, 254, 276, and 301 nm on a 218TP54 4.6 mm ⁇ 250 mm 5 ⁇ C-18 silica column (The Separations Group, Hesperia), which was eluted at 1 ml/min at 42° C. The column was equilibrated with 5% acetonitrile (+0.1% TFA) in an aqueous solution of TFA in water (0.1%). After injection, the sample was eluted by a gradient of 0% to 90% acetonitrile (+0.1% TFA) in an aqueous solution of TFA in water (0.1%) during 50 min.
  • Buffer A 10 mM tris, 15 mM (NH4)2SO4, pH adjusted to 7.3 with 4N H2SO4, 20% v/v acetonitrile
  • Buffer B 80% v/v acetonitrile
  • LCMS was performed on a setup consisting of Sciex API 100 Single quadropole mass spectrometer, Perkin Elmer Series 200 Quard pump, Perkin Elmer Series 200 autosampler, Applied Biosystems 785A UV detector, Sedex 75 evaporative light scattering detector
  • the instrument control and data acquisition were done by the Sciex Sample control software running on a Windows 2000 computer.
  • the HPLC pump is connected to two eluent reservoirs containing:
  • the analysis is performed at room temperature by injecting an appropriate volume of the sample (preferably 20 ⁇ l) onto the column which is eluted with a gradient of acetonitrile.
  • HPLC conditions, detector settings and mass spectrometer settings used are giving in the following table.
  • ELS analogue output from ELS
  • 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 detector controlled by HP Chemstation software.
  • the HPLC pump is connected to two eluent reservoirs containing:
  • 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.
  • ELS analogue output from ELS
  • Molecular weights of the peptides were determined using matrix-assisted laser desorption time of flight mass spectroscopy (MALDI-MS), recorded on a Microflex (Bruker). A matrix of ⁇ -cyano-4-hydroxy cinnamic acid was used.
  • H is L-histidine
  • A is L-alanine ect.
  • Three letter abbreviations for amino acids may also be uses, e.g. His is L-histidine, Ala is L-alanine etc.
  • three letter abbreviations are used, such as Aib for aminoisobutyric acid.
  • the position of the amino acids may either be indicated with a number in superscript after the amino acid symbols such as Lys 33 , or as Lys33.
  • the N-terminal amino group may be symbolised either as NH 2 or as H.
  • the C-terminal carboxylic group may be symbolised either as —OH or as —COOH.
  • the C-terminal amide group is symbolised as —NH 2
  • the epsilon amino group of Lysine may be described either as the greek symbol ⁇ or spelled “epsilon”.
  • the peptide was eluted at 64% acetonitrile.
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Val25,Arg26,Leu27,Glu30,Lys33)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and MALDI-MS.
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26,Glu30)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and MALDI-MS.
  • the peptide was eluted at 60% acetonitrile.
  • N epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -[Aib8,Lys20,Glu22,Val25,Arg26,Glu30]GLP-1(7-33)amide
  • the peptide was eluted at 70% acetonitrile.
  • N-epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Arg26)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and LC-MS.
  • N-epsilon31 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Glu22,Val25,Arg26,Lys31)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and LC-MS.
  • N-epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(DesaminoHis7,Lys20,Glu22,Arg26)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and LC-MS.
  • N-epsilon31 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(DesaminoHis7,Glu22,Arg26,Lys31)GLP-1(7-33)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and LC-MS.
  • N-epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Val25,Arg26,Leu27, Glu30,Lys31)GLP-1(7-32)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and MALDI-MS.
  • N-epsilon20 ⁇ 2-(2- ⁇ 2-[2-(2- ⁇ 2-[4-Carboxy-4-(17-carboxy-heptadecanoylamino)butyrylamino]ethoxy ⁇ ethoxy)acetylamino]ethoxy ⁇ ethoxy)acetyl ⁇ -(Aib8,Lys20,Glu22,Val25,Arg26,Leu27,Nle30,Lys31)GLP-1(7-32)amide
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and MALDI-MS.
  • Preparation method Method C except that the peptide was prepared on an Apex396 from Advanced Chemtech using a molar excess of 8-10 fold amino acid, DIC and HOAt/HOBt (1:1) and the Mtt group was deprotected with hexaflouroisopropanol.
  • the final product was characterized by analytical HPLC and MALDI-MS.
  • MALDI-MS alpha-cyano-4-hydroxycinnamic acid; m/z: 3824.543 (NEGATIVE MODE)
  • the protraction of a number GLP-1 compounds of the invention is determined by monitoring the concentration thereof in plasma after sc administration to healthy pigs, using the methods described below. For comparison also the concentration in plasma of GLP-1(7-37) after sc. administration is followed.
  • the protraction of other GLP-1 compounds of the invention can be determined in the same way.
  • test substances are dissolved in a vehicle suitable for subcutaneous or intravenous administration.
  • concentration is adjusted so the dosing volume is approximately 1 ml.
  • the study is performed in 12 male Göttingen minipigs from Ellegaard Göttingen Minipigs ApS. An acclimatisation period of approximately 10 days is allowed before the animals enter the study. At start of the acclimatisation period the minipigs are about 5 months old and in the weight range of 8-10 kg.
  • the study is conducted in a suitable animal room with a room temperature set at 21-23° C. and the relative humidity to ⁇ 50%.
  • the room is illuminated to give a cycle of 12 hours light and 12 hours darkness.
  • Light is from 06.00 to 18.00 h.
  • the animals are housed in pens with straw as bedding, six together in each pen.
  • the animals have free access to domestic quality drinking water during the study, but are fasted from approximately 4 pm the day before dosing until approximately 12 hours after dosing.
  • the animals are weighed on arrival and on the days of dosing.
  • the animals receive a single intravenous or subcutaneous injection.
  • the subcutaneous injection is given on the right side of the neck, approximately 5-7 cm from the ear and 7-9 cm from the middle of the neck.
  • the injections are given with a stopper on the needle, allowing 0.5 cm of the needle to be introduced.
  • Each test substance is given to three animals. Each animal receives a dose of 2 nmol/kg body weight.
  • a full plasma concentration-time profile is obtained from each animal. Blood samples are collected according to the following schedule:
  • Predose (0) 0.17 (10 minutes), 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72, 96, and 120 hours after injection.
  • Predose (0) 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72, 96, and 120 hours after injection.
  • 2 ml of blood is drawn from each animal.
  • the blood samples are taken from a jugular vein.
  • the blood samples are collected into test tubes containing a buffer for stabilisation in order to prevent enzymatic degradation of the GLP-1 compounds.
  • Plasma is immediately transferred to Micronic-tubes. Approximately 200 ⁇ l plasma is transferred to each Micronic-tube. The plasma is stored at ⁇ 20° C. until assayed. The plasma samples are assayed for the content of GLP-1 compounds using an immunoassay.
  • the plasma concentration-time profiles are analysed by a non-compartmental pharmacokinetic analysis.
  • the following pharmacokinetic parameters are calculated at each occasion: AUC, AUC/Dose, AUC % Extrap , C max , t max , ⁇ z , t 1/2 , CL, CL/f, V z , V z /f and MRT.
  • Selected compounds of the invention are tested in Danish Landrace pigs.
  • Pigs (50% Duroc, 25% England, 25% Danish Landrace, app 40 kg) are fasted from the beginning of the experiment.
  • To each pig 0.5 nmol of test compound per kg body weight is administered in a 50 ⁇ M isotonic solution (5 mM phosphate, pH 7.4, 0.02% Tween® ⁇ 20 (Merck), 45 mg/ml mannitol (pyrogen free, Novo Nordisk). Blood samples are drawn from a catheter in vena jugularis.
  • the plasma concentrations of the peptides are determined in a sandwich ELISA or by RIA using different mono- or polyclonal antibodies. Choice of antibodies depends of the GLP-1 compounds. The time at which the peak concentration in plasma is achieved varies within wide limits, depending on the particular GLP-1 compound selected.
  • the assay is carried out as follows (volume/well):
  • the concentration in the samples is calculated from standard curves.
  • the assay is carried out in minisorp tubes 12 ⁇ 75 mm (volumen/tube) as follows:
  • the concentration in the samples is calculated from individual standard curves.
  • the method is a radiometric-ligand binding assay using LEADseeker imaging particles.
  • the assay is composed of membrane fragments containing the GLP-1 receptor, unlabeled GLP-1 analogues, human GLP-1 labelled with 125 I and PS LEADseeker particles coated with wheat germ agglutinin (WGA). Cold and 125 I-labelled GLP-1 will compete for the binding to the receptor.
  • WGA wheat germ agglutinin
  • the LEADseeker particles When the LEADseeker particles are added they will bind to carbohydrates residues on the membrane fragments via the WGA-residues.
  • the proximity between the 125 I-molecules and the LEADseeker particles causes light emission from the particles.
  • the LEADseeker will image the emitted light and it will be reversibly correlated to the amount of GLP-1 analogue present in the sample.
  • Pre treatment of animal plasma Animal plasma is heat treated for 4 hrs at 56° C. and centrifuged at 10.000 rpm for 10 minutes. Afterwards, Val-Pyr (10 ⁇ M) and aprotenin (500 KIE/mL) is added and stored at ⁇ 18° C. until use.
  • GLP-1 analogues calibrators GLP-1 analogues are spiked into heat-treated plasma to produce dilution lines ranging from approximately 1 ⁇ M to 1 ⁇ M.
  • GLP-1 receptor suspension GLP-1 receptor membrane fragments are purified from baby hamster kidney (BHK) cells stably expressing the human pancreatic GLP-1 receptor. Stored ⁇ 80° C. until use.
  • WGA-coupled polystyrene LEADseeker imaging beads (RPNQ0260, Amersham): The beads are reconstituted with GLP-1 RRA assay buffer to a concentration of 13.3 mg/mL. The GLP-1 receptor membrane suspension is then added and incubated cold (2-8° C.) at end-over-end for at least 1 hr prior to use.
  • MultiScreen® Solvinert 0.45 ⁇ m hydrophobic PTFE plates (MSRPN0450, Millipore Corp.)
US12/676,453 2007-09-05 2008-09-05 Truncated glp-1 derivaties and their therapeutical use Abandoned US20100292133A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP07115743.2 2007-09-05
EP07115743 2007-09-05
EP08101010.0 2008-01-28
EP08101010 2008-01-28
PCT/EP2008/061833 WO2009030774A1 (fr) 2007-09-05 2008-09-05 Dérivés glp-1 tronqués et utilisations à des fins thérapeutiques de ceux-ci

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/061833 A-371-Of-International WO2009030774A1 (fr) 2007-09-05 2008-09-05 Dérivés glp-1 tronqués et utilisations à des fins thérapeutiques de ceux-ci

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/299,638 Continuation US9657079B2 (en) 2007-09-05 2014-06-09 Truncated GLP-1 derivatives and their therapeutical use

Publications (1)

Publication Number Publication Date
US20100292133A1 true US20100292133A1 (en) 2010-11-18

Family

ID=40043001

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/676,453 Abandoned US20100292133A1 (en) 2007-09-05 2008-09-05 Truncated glp-1 derivaties and their therapeutical use
US14/299,638 Expired - Fee Related US9657079B2 (en) 2007-09-05 2014-06-09 Truncated GLP-1 derivatives and their therapeutical use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/299,638 Expired - Fee Related US9657079B2 (en) 2007-09-05 2014-06-09 Truncated GLP-1 derivatives and their therapeutical use

Country Status (6)

Country Link
US (2) US20100292133A1 (fr)
EP (1) EP2190873B1 (fr)
JP (1) JP2010538049A (fr)
CN (1) CN101842386A (fr)
ES (1) ES2550363T3 (fr)
WO (1) WO2009030774A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013037690A1 (fr) 2011-09-06 2013-03-21 Novo Nordisk A/S Dérivés de glp-1
US9764003B2 (en) 2012-07-01 2017-09-19 Novo Nordisk A/S Use of long-acting GLP-1 peptides
US10933120B2 (en) 2012-03-22 2021-03-02 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
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
US11034746B2 (en) 2011-04-12 2021-06-15 Novo Nordisk A/S Double-acylated GLP-1 derivatives
US11033499B2 (en) 2012-06-20 2021-06-15 Novo Nordisk A/S Tablet formulation comprising a GLP-1 peptide and a delivery agent
US11123296B2 (en) 2012-03-22 2021-09-21 Novo Nordisk A/S Compositions comprising a delivery agent and preparation thereof
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

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2507098T3 (es) 2005-11-07 2014-10-14 Indiana University Research And Technology Corporation Análogos de glucagón que muestran solubilidad y estabilidad fisiológicas
EP2124974B1 (fr) 2007-01-05 2017-03-15 Indiana University Research and Technology Corporation Analogues de glucagon présentant une solubilité améliorée dans des tampons à ph physiologiques
US8454971B2 (en) 2007-02-15 2013-06-04 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
WO2009058662A2 (fr) 2007-10-30 2009-05-07 Indiana University Research And Technology Corporation Antagonistes du glucagon
JP5771005B2 (ja) 2007-10-30 2015-08-26 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation グルカゴンアンタゴニスト及びglp−1アゴニスト活性を示す化合物
JP5753779B2 (ja) 2008-06-17 2015-07-22 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation 生理学的pHの緩衝液中で向上した溶解性及び安定性を示すグルカゴン類縁体
PL2300035T3 (pl) 2008-06-17 2016-04-29 Univ Indiana Res & Tech Corp Mieszani agoniści na bazie GIP do leczenia zaburzeń metabolicznych i otyłości
US8546327B2 (en) 2008-06-17 2013-10-01 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
KR20110110174A (ko) 2008-12-19 2011-10-06 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 아미드 기반 글루카곤 슈퍼패밀리 펩티드 프로드럭
MX2011013625A (es) 2009-06-16 2012-01-20 Univ Indiana Res & Tech Corp Compuestos glucagon activo de receptor de gip.
WO2011075393A2 (fr) 2009-12-18 2011-06-23 Indiana University Research And Technology Corporation Co-agonistes du récepteur du glucagon/glp-i
RU2012136450A (ru) 2010-01-27 2014-03-10 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн Конъюгаты антагонист глюкагона - агонист gip и композиции для лечения метаболических расстройств и ожирения
WO2011107494A1 (fr) 2010-03-03 2011-09-09 Sanofi Nouveaux dérivés aromatiques de glycoside, médicaments contenants ces composés, et leur utilisation
DE102010015123A1 (de) 2010-04-16 2011-10-20 Sanofi-Aventis Deutschland Gmbh Benzylamidische Diphenylazetidinone, diese Verbindungen enthaltende Arzneimittel und deren Verwendung
CA2797095A1 (fr) 2010-05-13 2011-11-17 Indiana University Research And Technology Corporation Peptides de la superfamille des glucagons presentant une activite de recepteur nucleaire d'hormone
KR20130111923A (ko) 2010-05-13 2013-10-11 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 G-단백결합 수용체 활성을 나타내는 글루카곤 슈퍼패밀리 펩티드
US8530413B2 (en) 2010-06-21 2013-09-10 Sanofi Heterocyclically substituted methoxyphenyl derivatives with an oxo group, processes for preparation thereof and use thereof as medicaments
CA2796894A1 (fr) 2010-06-24 2011-12-29 Indiana University Research And Technology Corporation Promedicaments peptidiques a base d'amides de la superfamille du glucagon
TW201215387A (en) 2010-07-05 2012-04-16 Sanofi Aventis Spirocyclically substituted 1,3-propane dioxide derivatives, processes for preparation thereof and use thereof as a medicament
TW201215388A (en) 2010-07-05 2012-04-16 Sanofi Sa (2-aryloxyacetylamino)phenylpropionic acid derivatives, processes for preparation thereof and use thereof as medicaments
TW201221505A (en) 2010-07-05 2012-06-01 Sanofi Sa Aryloxyalkylene-substituted hydroxyphenylhexynoic acids, process for preparation thereof and use thereof as a medicament
MA34885B1 (fr) 2010-12-22 2014-02-01 Indiana Unversity Res And Technology Corp Analogues du glucagon presentant una ctivite de recepteur de gip
CN103748109A (zh) 2011-06-22 2014-04-23 印第安纳大学研究及科技有限公司 胰高血糖素/glp-1受体共同激动剂
LT2723367T (lt) 2011-06-22 2017-08-25 Indiana University Research And Technology Corporation Bendri gliukagono/glp-1 receptoriaus agonistai
EP2567959B1 (fr) 2011-09-12 2014-04-16 Sanofi Dérivés d'amide d'acide 6-(4-hydroxy-phényl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylique en tant qu'inhibiteurs de kinase
WO2013037390A1 (fr) 2011-09-12 2013-03-21 Sanofi Dérivés amides d'acide 6-(4-hydroxyphényl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylique en tant qu'inhibiteurs de kinase
EP2760862B1 (fr) 2011-09-27 2015-10-21 Sanofi Dérivés d'amide d'acide 6-(4-hydroxyphényl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylique utilisés comme inhibiteurs de kinase
WO2013074910A1 (fr) 2011-11-17 2013-05-23 Indiana University Research And Technology Corporation Peptides de la superfamille du glucagon présentant une action sur les récepteurs aux glucocorticoïdes
HRP20231613T1 (hr) 2012-03-22 2024-03-15 Novo Nordisk A/S Pripravci koji sadrže sredstvo za unošenje i njihova priprava
RU2015101697A (ru) 2012-06-21 2016-08-10 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн Аналоги глюкагона, обладающие активностью рецептора gip
EP2908844A1 (fr) 2012-10-17 2015-08-26 Novo Nordisk A/S Acides aminés acylés par un acide gras pour l'administration de peptides par voie orale
IN2015DN03795A (fr) 2012-10-24 2015-10-02 Inserm Inst Nat De La Santé Et De La Rech Médicale
EP3006045B3 (fr) 2014-10-07 2021-03-17 Cyprumed GmbH Formulations pharmaceutiques pour l'administration orale de médicaments peptidiques ou protéines
US10426818B2 (en) 2015-03-24 2019-10-01 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method and pharmaceutical composition for use in the treatment of diabetes
CA2997343A1 (fr) 2015-10-07 2017-04-13 Cyprumed Gmbh Formulations pharmaceutiques pour l'administration par voie orale de medicaments peptidiques
WO2018065634A1 (fr) 2016-10-07 2018-04-12 Cyprumed Gmbh Compositions pharmaceutiques pour l'administration par voie nasale de médicaments de type peptide ou protéine
US20210087250A1 (en) 2018-04-06 2021-03-25 Cyprumed Gmbh Pharmaceutical compositions for the transmucosal delivery of therapeutic peptides and proteins
TWI829687B (zh) 2018-05-07 2024-01-21 丹麥商諾佛 儂迪克股份有限公司 包含glp-1促效劑與n-(8-(2-羥基苯甲醯基)胺基)辛酸之鹽的固體組成物
CN110845601B (zh) * 2019-10-12 2021-01-19 广东药科大学 不同构型的glp-1类似肽修饰二聚体及其制备方法在治疗ii型糖尿病中的应用
CA3166496A1 (fr) * 2019-12-30 2021-07-08 Gan & Lee Pharmaceuticals Co., Ltd. Compose glp-1 a action prolongee
MX2022008130A (es) * 2019-12-30 2022-10-03 Gan & Lee Pharmaceuticals Co Ltd Derivado de insulina.
CN111333714A (zh) * 2020-03-05 2020-06-26 成都奥达生物科技有限公司 一种长效glp-1化合物
CN115461044A (zh) 2020-04-29 2022-12-09 诺和诺德股份有限公司 包含glp-1激动剂和组氨酸的固体组合物
WO2022049310A1 (fr) 2020-09-07 2022-03-10 Cyprumed Gmbh Formulations pharmaceutiques améliorées d'agonistes du récepteur glp-1
EP4222176A4 (fr) 2020-09-30 2024-02-28 Beijing Ql Biopharmaceutical Co Ltd Conjugués polypeptidiques et leurs procédés d'utilisation
WO2023012263A1 (fr) 2021-08-04 2023-02-09 Novo Nordisk A/S Formulations solides de peptides oraux
CN115960202B (zh) * 2021-10-09 2023-07-14 合肥天汇生物科技有限公司 一种glp-1类似物及其制备方法和应用
CN114874314B (zh) * 2021-12-28 2022-11-11 北京惠之衡生物科技有限公司 一种高表达glp-1类似物的重组工程菌及其构建方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5968899A (en) * 1994-06-03 1999-10-19 Tsumura & Co. Medicinal compositions of peptides with EACA or tranexamic acid for enhanced mucosal absorption
US6268343B1 (en) * 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6458924B2 (en) * 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US7235627B2 (en) * 1996-08-30 2007-06-26 Novo Nordisk A/S Derivatives of GLP-1 analogs
US20070203058A1 (en) * 2003-09-19 2007-08-30 Novo Nordisk A/S Novel Glp-1 Derivatives
US7271149B2 (en) * 2000-12-07 2007-09-18 Eli Lilly And Company GLP-1 fusion proteins

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69129226T2 (de) * 1990-01-24 1998-07-30 Douglas I Buckley Glp-1-analoga verwendbar in der diabetesbehandlung
US5705483A (en) 1993-12-09 1998-01-06 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
US5869602A (en) 1995-03-17 1999-02-09 Novo Nordisk A/S Peptide derivatives
BRPI9711437B8 (pt) * 1996-08-30 2021-05-25 Novo Nordisk As derivados de glp-1
ES2237790T3 (es) * 1996-11-12 2005-08-01 Novo Nordisk A/S Uso de peptidos glp-1.
EP1056775B1 (fr) 1998-02-27 2010-04-28 Novo Nordisk A/S Derives de gpl-1 et de l'extendine au profil d'action etendu
NZ527241A (en) 1998-12-07 2004-12-24 Sod Conseils Rech Applic Analogues of GLP-1
US6329336B1 (en) 1999-05-17 2001-12-11 Conjuchem, Inc. Long lasting insulinotropic peptides
EP1076066A1 (fr) 1999-07-12 2001-02-14 Zealand Pharmaceuticals A/S Peptides abaissant le taux de glucose sanguin
EP2275117B1 (fr) * 2001-07-31 2016-10-26 The Government of the United States of America, as represented by the Secretary of the Department of Health and Human Services GLP-1, exendine-4, analogues de peptides et utilisations associées
PL1605897T3 (pl) 2003-03-19 2012-12-31 Lilly Co Eli Związki będące połączeniem GLP-1 z poli(glikolem etylenowym)
RU2006120079A (ru) 2003-12-18 2008-01-27 Ново Нордиск А/С (DK) Производные глюкагоноподобного пептида-1 (glp-1)
US20090111730A1 (en) * 2004-07-08 2009-04-30 Novo Nordisk A/S Polypeptide protracting tags
JP2008515856A (ja) * 2004-10-07 2008-05-15 ノボ ノルディスク アクティーゼルスカブ 遅延性glp−1化合物
WO2006097538A1 (fr) * 2005-03-18 2006-09-21 Novo Nordisk A/S Composes glp-1 avances
TWI372629B (en) * 2005-03-18 2012-09-21 Novo Nordisk As Acylated glp-1 compounds
WO2006097535A2 (fr) * 2005-03-18 2006-09-21 Novo Nordisk A/S Agonistes de peptides de la famille du glucagone presentant une activite similaire a la secretine
US20090062192A1 (en) * 2005-03-18 2009-03-05 Novo Nordisk A/S Dimeric Peptide Agonists of the Glp-1 Receptor
EP2679597A1 (fr) * 2007-09-05 2014-01-01 Novo Nordisk A/S Dérivés de peptide 1 de type glucagon et leur utilisation pharmaceutique

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5968899A (en) * 1994-06-03 1999-10-19 Tsumura & Co. Medicinal compositions of peptides with EACA or tranexamic acid for enhanced mucosal absorption
US6268343B1 (en) * 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6458924B2 (en) * 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US7235627B2 (en) * 1996-08-30 2007-06-26 Novo Nordisk A/S Derivatives of GLP-1 analogs
US8097698B2 (en) * 1996-08-30 2012-01-17 Novo Nordisk A/S Derivatives of GLP-1 analogs
US7271149B2 (en) * 2000-12-07 2007-09-18 Eli Lilly And Company GLP-1 fusion proteins
US20070203058A1 (en) * 2003-09-19 2007-08-30 Novo Nordisk A/S Novel Glp-1 Derivatives

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
[Retrieved from]http://cancerweb.ncl.ac.uklomd/about.html, 5 pages, 7-7-2005 [Retrieved on 7/7/05] *
Irwin, 1995, Molecular Endocrinology, 9, 267-277. *
Leger, 2004, Biooranic & Medicinal Chemistry Letters, 14, 4395-4398 *
Pan, 2006, JBC, 281, 12506-12515 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
US11034746B2 (en) 2011-04-12 2021-06-15 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
US9758560B2 (en) 2011-09-06 2017-09-12 Novo Nordisk A/S GLP-1 derivatives
US10308700B2 (en) 2011-09-06 2019-06-04 Novo Nordisk A/S GLP-1 derivatives
WO2013037690A1 (fr) 2011-09-06 2013-03-21 Novo Nordisk A/S Dérivés de glp-1
US11759503B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides and preparation thereof
US11759502B2 (en) 2012-03-22 2023-09-19 Novo Nordisk A/S Compositions of GLP-1 peptides 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
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
US10335462B2 (en) 2012-07-01 2019-07-02 Novo Nordisk A/S Use of long-acting GLP-1 peptides
US9764003B2 (en) 2012-07-01 2017-09-19 Novo Nordisk A/S Use of long-acting GLP-1 peptides
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

Also Published As

Publication number Publication date
WO2009030774A1 (fr) 2009-03-12
US9657079B2 (en) 2017-05-23
ES2550363T3 (es) 2015-11-06
US20140296131A1 (en) 2014-10-02
EP2190873A1 (fr) 2010-06-02
EP2190873B1 (fr) 2015-07-22
JP2010538049A (ja) 2010-12-09
CN101842386A (zh) 2010-09-22

Similar Documents

Publication Publication Date Title
US9657079B2 (en) Truncated GLP-1 derivatives and their therapeutical use
US9409966B2 (en) Glucagon-like peptide-1 derivatives and their pharmaceutical use
US9067977B2 (en) Peptides derivatized with A-B-C-D- and their therapeutical use
US9012398B2 (en) Acylated exendin-4 compounds
US8536122B2 (en) Acylated GLP-1 compounds
US8603972B2 (en) Extended GLP-1 compounds

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVO NORDISK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SPETZLER, JANE;SCHAFFER, LAUGE;LAU, JESPER;AND OTHERS;SIGNING DATES FROM 20100511 TO 20100602;REEL/FRAME:024500/0025

STCB Information on status: application discontinuation

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