WO2023139106A2 - Long-acting gipr antagonists - Google Patents

Long-acting gipr antagonists Download PDF

Info

Publication number
WO2023139106A2
WO2023139106A2 PCT/EP2023/051098 EP2023051098W WO2023139106A2 WO 2023139106 A2 WO2023139106 A2 WO 2023139106A2 EP 2023051098 W EP2023051098 W EP 2023051098W WO 2023139106 A2 WO2023139106 A2 WO 2023139106A2
Authority
WO
WIPO (PCT)
Prior art keywords
xxx
gip
lys
compound
derivative according
Prior art date
Application number
PCT/EP2023/051098
Other languages
French (fr)
Other versions
WO2023139106A3 (en
Inventor
Brian Patrick Finan
Bin Shubin YANG
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Publication of WO2023139106A2 publication Critical patent/WO2023139106A2/en
Publication of WO2023139106A3 publication Critical patent/WO2023139106A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • 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
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor

Definitions

  • the present application relates to novel antagonists that are derivatives of glucosedependent insulinotropic peptide (GIP) analogues with improved GIP antagonistic activity, and to the pharmaceutical use of the GIP derivatives.
  • GIP glucosedependent insulinotropic peptide
  • GIP Glucose-dependent insulinotropic polypeptide
  • GLP-1 glucagon-like Peptide-1
  • GIP receptor Due to the incretin effect, the GIP receptor has become an attractive drug target in the treatment of metabolic diseases such as obesity and diabetes, with GIP receptor agonists either as a standalone, in combination with GLP-1 receptor agonists, or in combination with GLP-1/glucagon receptor co-agonists.
  • GIP itself has a short plasma half-life due to dipeptidyl peptidase-4 (DPP-IV) mediated inactivation, and poor physical stability due to high tendency to form fibrils in solution. If GIP(1-42) or GIP(1-30) are secreted into the circulation in humans, the cleavages catalyzed by DPP-4 would result in GIP(3-42) or GIP(3- 30) which both are inactive GIP hormones.
  • DPP-IV dipeptidyl peptidase-4
  • GIP analogues that act as hGIP receptor antagonists. These GIP analogues are modified by introducing a modified lysine at one or more of positions 3 to 29, wherein the modification is primarily a fatty acid in the form of a diacid attached to the epsilon-amino group of the modified lysine and the primary site for attachment of said modification is in position 18.
  • the invention concerns a glucose-dependent insulinotropic peptide (GIP) derivative consisting of a modifying group and a GIP analogue wherein the GIP analogue is defined by Formula la, Ila, or Illa:
  • GIP glucose-dependent insulinotropic peptide
  • the invention concerns a dosage form comprising a combination of a GIP derivative according to the invention and a GLP-1 receptor agonist.
  • the invention concerns a kit-of-parts comprising a first dosage form comprising a GIP derivative according to the invention and a second dosage form comprising a GLP-1 receptor agonist.
  • Fig. 1 shows percent change in body weight over 26 days of treatment of DIO mice when administering vehicle (open circles), semaglutide 2 nmol/kg (closed squares), compound 7 500 nmol/kg (open triangles-up), compound 7 1500 nmol/kg (open triangles-down), semaglutide 2 nmol/kg + compound 7 500 nmol/kg (closed triangles-up), and semaglutide 2 nmol/kg + compound 7 1500 nmol/kg (closed triangles-down).
  • Fig. 2 shows cumulative food intake over 26 days of treatment of DIO mice when administering vehicle (open circles), semaglutide 2 nmol/kg (closed squares), compound 7 500 nmol/kg (open triangles-up), compound 7 1500 nmol/kg (open triangles-down), semaglutide 2 nmol/kg + compound 7 500 nmol/kg (closed triangles-up), and semaglutide 2 nmol/kg + compound 7 1500 nmol/kg (closed triangles-down).
  • GIP analogues also described herein are derivatives of GIP analogues, pharmaceutical compositions and uses thereof in which open ended terms like “comprises” and ’’comprising” are replaced with closed terms such as “consists of”, “consisting of”, and the like.
  • GIP receptor antagonists are also described herein.
  • a receptor antagonist may be defined as a compound that binds to a receptor and is capable of inhibiting or reducing a response typical of the natural ligand.
  • a “GIP antagonist” may be described as a compound capable of binding to the GIP receptor without activating the GIP receptor or reducing the activation compared to the native ligand.
  • hGIP(1-42) refers to the human glucose-dependent insulinotropic polypeptide, the sequence of which is included in the sequence listing as SEQ ID NO: 5.
  • the peptide having the sequence of SEQ ID NO: 5 may also be designated native hGIP or hGIP.
  • hGIP(1-31) refers to a truncated version of hGIP(1-42), comprising amino acids 1-31 of hGIP(1-42), the sequence of hGIP(1-31) is included in the sequence listings as SEQ ID NO: 4.
  • hGIP(3-31) refers to a truncated version of hGIP(1-42) comprising amino acids 3-31 of hGIP(1-42), and is a GIP antagonist.
  • the sequence of hGIP(3-31) is included in the sequence listings a SEQ ID NO: 6.
  • hGIP(3-42) refers to truncated version of hGIP(1-42) comprising amino acids 3-42 of hGIP(1-42), and is a GIP antagonist.
  • the sequence of hGIP(3-42) is included in the sequence listings as SEQ ID NO: 7.
  • GIP analogue refers to a peptide, or a compound, which is a variant of hGIP(1-31) or hGIP(1-42).
  • variant is used for peptides comprising at least one amino acid substitution as compared to hGIP(1-31) or hGIP(1-42) and is capable of binding to the GIP receptor.
  • amino acid modification refers to the substitution of an amino acid for another, the deletion of an amino acid, or the addition of an amino acid.
  • substitution refers to one amino acid being replaced by another in the backbone of the peptide.
  • the GIP derivatives of the invention may comprise substitutions of one or more unnatural and/or non-amino acids, e.g., amino acid mimetics, into the sequence of the GIP derivative.
  • amino acid mimetics e.g., amino acid mimetics
  • deletion refers to one amino acid in the backbone of the peptide being removed without replacing it with another moiety.
  • deleting the first four amino acids of hGIP(1-31) to prepare hGIP(5-31) (SEQ ID NO: 8) is considered a “deletion” of each of the four amino acids.
  • addition refers to adding one amino acid, or another moiety that may form part of the peptide backbone, into the backbone of the peptide.
  • adding an amino acid at the C-terminal end of hGIP(1-31) to prepare (a modified) hGIP(1-32) is considered an “addition”.
  • GIP analogues of the derivatives of the invention may be described by reference to i) the number of the amino acid residue in hGIP(1-31) or hGIP(1-42) which corresponds to the amino acid residue which is changed (i.e. the corresponding position in hGIP(1-31) or hGIP(1-42), and to ii) the actual change.
  • [M14L]-hGIP(1-31) refers to a GIP analogue in which the methionine of position 14 of hGIP(1-31) has been replaced by a leucine.
  • the numbering of positions in the GIP analogues of the present invention is made with reference to hGIP(1-31) or hGIP(1-42), even if some of the amino acids in the N- terminal end have been deleted.
  • the first position of hGIP(5-31) is referred to as position 5, even though it is the first amino acid of the sequence.
  • the GIP analogues of the derivatives of the invention comprises Formula la, Ila, or Illa (SEQ ID NOs: 46, 47, 48):
  • the GIP analogues of the derivatives of the invention comprises Formula I, II, or III (SEQ ID NOs: 1, 2, 3):
  • the position of the modified lysine of the GIP analogue of the derivative of the invention depends on whether the GIP derivative of the invention comprises a GIP analogue of Formula I, II, III, la, Ila, or Illa. If the GIP derivative of the invention comprises a GIP analogue of Formula I, II, la or Ila, the modified lysine is in one of positions 11 or 10. If the GIP derivative of the invention comprises a GIP analogue of Formula III or Illa, the modified lysine is in position 43.
  • the GIP derivative of the invention comprises a GIP analogue of Formula la or Ila. In a further embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula la. In another embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula Illa.
  • the GIP derivative of the invention comprises a GIP analogue of Formula I or II. In a further embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula I. In another embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula III.
  • the GIP derivative of the invention comprises a GIP analogue of Formula I, II, la or Ila, wherein the modified lysine is in one of positions 11 and 10. In yet a further embodiment, the modified lysine is in position 11 .
  • the GIP analogues of the derivatives of the present invention do not have any amino acids in positions 1 and 2 corresponding to positions in hGIP(1-31). In one embodiment, the derivatives do not have any amino acids in position 1 , 2, 3 and 4 corresponding to hGIP(1-31). In one embodiment, Xxx 5 is Thr and Xxx 6 is Phe. In a further embodiment, an additional deletion may be present in position 5. Accordingly, in one embodiment, Xxxs is absent. In another embodiment, Xxxs is absent and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention may have a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31) (i.e. compared to hGIP(3-31)), some of which may be substitutions. Accordingly, in one embodiment, Met in position 14 is substituted by Leu. In a further embodiment, His in position 18 is substituted by Arg. In still a further embodiment, Asp in position 21 is substituted by Glu. In yet a further embodiment, the GIP analogue of the derivative of the invention has a maximum of 8 amino acid modifications as compared to hGIP(3-31).
  • the GIP analogue of the derivative of the invention has a maximum of 7 amino acid modifications as compared to hGIP(3-31). In still another embodiment, the GIP analogue of the derivative of the invention has a maximum of 6 amino acid modifications as compared to hGIP(3-31). In one embodiment, Xxxi4 is Leu, Xxxis is Arg and XXX21 is Glu.
  • the GIP analogue of the derivatives of the present invention may have a combination of the features defined above. Accordingly, in one embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxx 5 is Thr, Xxx 6 is Phe, Xxx 3 o is Lys, Xxx 3i is Gly, and the N-terminal end is substituted with an a-acetylate.
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula la or Ila, wherein the modified lysine is in position 10 (Xxx is Lys), Xxxs is Thr, Xxxe is Phe, Xxx 3 o is Lys, Xxx 3i is Gly, and the N-terminal end is substituted with an a-acetylate.
  • the GIP analogue of the derivatives of the present invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx 3 and Xxx4 are absent, Xxx 3 o is Lys, Xxx 3i is Gly, and the N-terminal end is substituted with an a-acetylate.
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula I or II, wherein the modified lysine is in position 10, Xxx 3 and Xxx4 are absent, Xxx 3 o is Lys, Xxx 3i is Gly, and the N-terminal end is substituted with an a- acetylate.
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula Illa, wherein the modified lysine is in position 43 and the N-terminal end is substituted with an a-acetylate.
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxxs is absent, and Xxx 3 is L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxxs is absent, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent, and Xxx 3 is L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula la or Ila, wherein the modified lysine is in position 10 (Xxx is Lys), Xxxs is absent, Xxx 3 o is Lys, Xxx 3i is Gly, and Xxx 3 is L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula III, wherein the modified lysine is in position 43, Xxx 3 and Xxx4 are absent, and the N-terminal end is substituted with an a-acetylate.
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx 3 , Xxx4 and Xxxs are absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx 3 , Xxx4, and Xxxs are absent, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the GIP analogue of the derivative of the invention is a GIP analogue of Formula I or II, wherein the modified lysine is in position 10, Xxx 3 , Xxx4, and Xxxs are absent, Xxx 3 o is Lys, Xxx 3i is Gly, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the GIP analogues of the derivatives of the invention may comprise a substitution selected from the group consisting of: M14L, H18R, D21 E as compared to hGIP(1-31). In some embodiment, the GIP analogue of the derivative of the invention comprises all of substitutions M14L, H18R and D21 E as compared to hGIP(1-31). In one embodiment, the GIP analogue of the derivatives of the invention comprises the backbone of [S11 K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 9).
  • the GIP analogue of the derivatives of the invention comprises the backbone of [F6Pla, S11 K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 10). Also or alternatively, in one embodiment, the GIP analogue of the derivatives of the invention comprises the backbone of [Y10K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 11).
  • the amino acid in position 13 is selected from Ala or Lys.
  • position 16 is selected from Lys or Aib.
  • the amino acid in position 17 is selected from lie or Lys.
  • position 20 is selected from Gin or Aib.
  • the amino acid in position 24 is selected from Asn, Gin or Lys.
  • the amino acid in position 30 is selected from Lys, Arg or is absent.
  • the amino acid in position 31 is selected from Gly, Pro or is absent.
  • the GIP analogue of the derivative of the invention is selected from SEQ ID NO: 12-45, such as 17-45.
  • the GIP analogue of the derivative of the invention is be selected from any one of SEQ ID NO: 17-31 and 43-45, such as SEQ ID NO: 17-26, 28-31 and 43-44.
  • the GIP analogue of the derivative of the invention is selected from the group consisting of SEQ ID NO: 17-23, 31, 43- 44 such as 17-18, 21-23 or such as 19-20.
  • the GIP analogue of the derivative of the invention is selected from the group consisting of SEQ ID NO: 24-25, 28-30, such as 24-25, 30 or such as 28-30. In some embodiment the GIP analogue of the derivative of the invention is SEQ ID NO: 26.
  • Analogues “comprising” certain specified changes may comprise further changes, when compared to the respective formula.
  • the analogue "has" the specified changes.
  • amino acid residues may be identified by their full name, their one-letter code, and/or their three-letter code. These three ways are fully equivalent.
  • GIP derivative refers to a chemically modified GIP analogue, in which one or more substituents have been covalently attached to the peptide backbone.
  • the substituent may be an N-terminal substituent.
  • the substituent may be a modifying group or, alternatively, referred to as a protracting moiety or albumin binding moiety.
  • N-terminal substituent or “modifying group” as used herein, means a chemical moiety or group replacing a hydrogen atom.
  • the derivative of a GIP analogue comprises a substituent covalently attached to the alpha-amino group of the amino acid residue in the N-terminus of the analogue.
  • the N-terminal substituent is an alkanoyl or acyl group.
  • the N-terminal substituent is an acetyl group or glutaric acid.
  • an N-terminal substituted amino acid is Ac-Thr at position 5.
  • N-acetylation would not count as a substitution in the peptide backbone compared with hGI P(1-31 ), because the amino acid in the GIP analogue is the native Thr, e.g. N“-Ac- [Lys11 ,Met14,Arg18,Glu21]-hGIP(5-31) comprises 4 substitutions as compared to hGIP(5- 31).
  • the GIP analogue comprises a modifying group covalently attached to a lysine residue at one of positions 11, 10, 21 or 43.
  • the modifying group is capable of forming non-covalent conjugates with proteins, e.g. albumin, thereby promoting the circulation of the derivative with the blood stream, and also having the effect of protracting the time of action of the derivative, due to the fact that the conjugate of the GIP derivative and albumin is only slowly disintegrated to release the active pharmaceutical ingredient.
  • the modifying group may be covalently attached to a lysine residue of the GIP analogue by acylation, i.e. via an amide bond formed between a carboxylic acid group of the modifying group and the epsilon amino group of said lysine group.
  • the modifying group is covalently attached to a lysine residue at one of positions 11 , 10, 21 or 43 by acylation, i.e. via an amide bond formed between a carboxylic acid group of the modifying group and the epsilon amino group of the lysine residue.
  • the modifying group is defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present.
  • A- provides increased hGIP receptor antagonistic activity. It has in particular been found that antagonistic activity of comparative compounds, wherein an additional carboxylic acid group, has been added to A- to prepare a diacid derivative instead of a mono fatty acid derivative, is lower.
  • A- is a mono fatty acid.
  • the mono fatty acid is a straight-chain mono fatty acid.
  • the straight-chain mono fatty acid is a C14 to C18 fatty acid, such as a C16 fatty acid.
  • the fatty acid is a saturated fatty acid or an unsaturated fatty acid, such as a mono-unsaturated acid.
  • the fatty acid is a saturated fatty acid.
  • the fatty acid is palmitic acid (C16).
  • the term mono fatty acid refers to a monoacid having one carboxylic acid group attached to the lysine in the GIP analogue.
  • a mono fatty acid may be straight or branched and it may be saturated or unsaturated.
  • a mono fatty acid typically has an even number of carbon atoms from 4 to 28, such as from 14 to 18 carbon atoms.
  • the monoacids could use abbreviation such as C14, C16 and C18, herein C16 refer to palmitic acid.
  • B-C- The type of linker represented by B-C- in the GIP derivatives of the present invention is known in the art and may vary.
  • B is an amino acid.
  • B is an amino acid linker.
  • B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp, wherein y-Glu is gamma-glutamic acid represented by Chem. 1 :
  • B is selected from y-Glu and y-Glu-y-Glu. In yet a further embodiment, B is y-Glu. In another embodiment, B is absent.
  • C is absent or selected from AEEA2 and AEEA, wherein AEEA is 8-amino-3,6-dioxaoctanoic acid represented by Chem. 2:
  • C is absent or AEEA2. In still another embodiment, C is absent. In yet another embodiment, B-C is y-Glu. In a further embodiment, B-C is y-Glu and A is a C16 fatty acid, such as palmitic acid and as represented by Chem. 3:
  • the GIP derivative of the invention is selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 19, compound No. 17, compound No. 21, compound No. 22, compound No. 49, compound No. 50, compound No. 52 and compound No. 53 of example 1 herein.
  • Inhibitors of the human GIP receptor are known as useful candidates for treating or preventing obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome. Accordingly, one aspect of the invention concerns the GIP derivative according to the invention for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
  • GLP-1 receptor agonists are GLP-1 receptor agonists.
  • the GIP derivative according to the invention in combination with a GLP-1 receptor agonist for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • the GLP-1 receptor agonist is semaglutide.
  • compositions comprising a derivative of the invention or a pharmaceutically acceptable salt, or amide thereof, and a pharmaceutically acceptable excipient may be prepared as is known in the art.
  • excipient broadly refers to any component other than the active therapeutic ingredient(s).
  • the excipient may be an inert substance, an inactive substance, and/or a not medicinally active substance.
  • the pharmaceutical composition comprising the derivative of the invention may be of several dosage forms, e.g. a solution, a suspension, a tablet, and a capsule.
  • the pharmaceutical composition comprising the derivative of the invention may be administered to a patient in need thereof at several sites, e.g. at topical sites such as skin or mucosal sites; at sites which bypass absorption such as in an artery, in a vein, or in the heart; and at sites which involve absorption, such as in the skin, under the skin, in a muscle, orally, or in the abdomen.
  • the composition comprising the derivative of the invention is an injectable composition comprising GIP derivatives of the present invention
  • a GIP derivative of this invention is dissolved in a suitable buffer at a suitable pH so precipitation is minimised or avoided.
  • the injectable composition is made sterile, for example, by sterile filtration.
  • the composition comprising the derivative of the invention is a solid formulation, e.g. a freeze-dried or spray-dried composition, which may be used as is, or whereto the physician or the patient adds solvents, and/or diluents prior to use.
  • the pharmaceutical composition is in the form of a tablet.
  • a composition may be a stabilised formulation.
  • stabilized formulation refers to a formulation with increased physical and/or chemical stability, preferably both. In general, a formulation must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • a pharmaceutical composition may also be a dosage form combining a GIP derivative of the invention and a GLP-1 receptor agonist.
  • the present invention concerns a dosage form comprising a combination of a GIP derivative according to the invention and a GLP-1 receptor agonist, in terms of loose-dose combination and fixed-dose combination therapy of the GIP derivatives and the GLP-1 receptor agonists.
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • the GLP-1 receptor agonist is semaglutide.
  • the invention concerns a kit-of-parts comprising a first dosage form comprising a GIP derivative according to the invention and a second dosage form comprising a GLP-1 receptor agonist.
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • the GLP-1 receptor agonist is semaglutide.
  • the derivatives as described herein are in the form of a pharmaceutically acceptable salt.
  • Salts are e.g. formed by a chemical reaction between a base and an acid, e.g.: 2NH3 + H2SO4 — > (NH ⁇ SC .
  • the salt may be a basic salt, an acid salt, or it may be neither (i.e. a neutral salt).
  • Basic salts produce hydroxide ions and acid salts hydronium ions in water.
  • the salts of the derivatives may be formed with added cations or anions between anionic or cationic groups, respectively. These groups may be situated in the peptide and/or in the substituent of the derivatives.
  • Non-limiting examples of anionic groups include any free carboxylic acid groups in the substituent, if any, as well as in the peptide.
  • the peptide may include a free carboxylic acid group at the C-terminus, if present, as well as any free carboxylic acid group of internal acidic amino acid residues such as aspartic acid and glutamic acid.
  • Non-limiting examples of cationic groups include any free amino groups in the substituent, if any, as well as in the peptide.
  • the peptide may include a free amino group at the N-terminus, if present, as well as any free imidazole or amino group of internal basic amino acid residues such as histidine, arginine, and lysine.
  • the GIP derivatives of the invention may for instance be produced by classical peptide synthesis, e.g., solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques, see, e.g., Greene and Wuts, “Protective Groups in Organic Synthesis”, John Wiley & Sons, 1999, Florencio Zaragoza Dorwald, “Organic Synthesis on solid Phase”, Wiley-VCH Verlag GmbH, 2000, and moc Solid Phase Peptide Synthesis”, Edited by W.C. Chan and P.D. White, Oxford University Press, 2000.
  • telomeres may be produced by recombinant methods, viz. by culturing a host cell containing a DNA sequence encoding the analogue and capable of expressing the peptide in a suitable nutrient medium under conditions permitting the expression of the peptide.
  • host cells suitable for expression of these peptides are: Escherichia coli, Saccharomyces cerevisiae, as well as mammalian BHK or CHO cell lines.
  • Those derivatives of the invention which include non-natural amino acids and/or a covalently attached N-terminal mono- or dipeptide mimetic may e.g. be produced as described in the experimental part. Or see e.g., Hodgson et al: "The synthesis of peptides and proteins containing non-natural amino acids", Chemical Society Reviews, vol. 33, no. 7 (2004), p. 422-430.
  • Embodiment 1 is a diagrammatic representation of Embodiment 1:
  • GIP glucose-dependent insulinotropic peptide
  • Embodiment 2 A glucose-dependent insulinotropic peptide (GIP) derivative comprising a GIP analogue comprising Formula I, II, or III:
  • Embodiment 3 is a diagrammatic representation of Embodiment 3
  • Embodiment 4 is a diagrammatic representation of Embodiment 4:
  • Embodiment 5 is a diagrammatic representation of Embodiment 5:
  • Embodiment 7 is a diagrammatic representation of Embodiment 7:
  • Embodiment 8 is a diagrammatic representation of Embodiment 8
  • Embodiment 9 is a diagrammatic representation of Embodiment 9:
  • Embodiment 10 is a diagrammatic representation of Embodiment 10:
  • Embodiment 11 is a diagrammatic representation of Embodiment 11 :
  • Embodiment 12 is a diagrammatic representation of Embodiment 12
  • Embodiment 13 is a diagrammatic representation of Embodiment 13:
  • Embodiment 14 is a diagrammatic representation of Embodiment 14:
  • Embodiment 16 is a diagrammatic representation of Embodiment 16:
  • Embodiment 17 is a diagrammatic representation of Embodiment 17:
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of formula I, wherein the modified lysine is in position 11 , Xxx 3 and Xxx4 are absent, Xxx 3 o is Lys, Xxxsi is Gly, and the N-terminal end is substituted with an a-acetylate.
  • Embodiment 18 is a diagrammatic representation of Embodiment 18:
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I or II, wherein the modified lysine is in position 10, Xxx 3 and Xxx4 are absent, Xxx 3 o is Lys, Xxx 3i is Gly, and the N-terminal end is substituted with an a-acetylate.
  • Embodiment 19 is a diagrammatic representation of Embodiment 19:
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula III, wherein the modified lysine is in position 43, Xxx 3 and Xxx4 are absent, and the N-terminal end is substituted with an a-acetylate.
  • Embodiment 20 is a diagrammatic representation of Embodiment 20.
  • Embodiment 21 is a diagrammatic representation of Embodiment 21 :
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I, wherein the modified lysine is in position 11 , Xxx 3 , Xxx4, and Xxxs are absent, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the modified lysine is in position 11 , Xxx 3 , Xxx4, and Xxxs are absent, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • Embodiment 22 The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I or II, wherein the modified lysine is in position 10, Xxxs, Xxx4, and Xxxs are absent, Xxxso is Lys or absent, Xxxsi is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • the GIP analogue is of Formula I or II, wherein the modified lysine is in position 10, Xxxs, Xxx4, and Xxxs are absent, Xxxso is Lys or absent, Xxxsi is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
  • Embodiment 23 is a diagrammatic representation of Embodiment 23.
  • Embodiment 24 is a diagrammatic representation of Embodiment 24.
  • Embodiment 25 is a diagrammatic representation of Embodiment 25.
  • Embodiment 26 is a diagrammatic representation of Embodiment 26.
  • Embodiment 27 is a diagrammatic representation of Embodiment 27.
  • A is a C14 to C18 fatty acid, such as a C16 fatty acid.
  • Embodiment 28 is a diagrammatic representation of Embodiment 28:
  • Embodiment 29 is a diagrammatic representation of Embodiment 29.
  • Embodiment 30 The GIP derivative according to any one of the preceding Embodiments, wherein A is an unsaturated fatty acid.
  • Embodiment 31
  • Embodiment 32 is a diagrammatic representation of Embodiment 32.
  • GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp.
  • Embodiment 33 is a diagrammatic representation of Embodiment 33.
  • GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu and y-Glu-y-Glu.
  • Embodiment 34 is a diagrammatic representation of Embodiment 34.
  • Embodiment 35 is a diagrammatic representation of Embodiment 35.
  • Embodiment 36 is a diagrammatic representation of Embodiment 36.
  • Embodiment 37 is a diagrammatic representation of Embodiment 37.
  • Embodiment 38 is a diagrammatic representation of Embodiment 38.
  • Embodiment 40 is a diagrammatic representation of Embodiment 40.
  • Embodiment 41
  • Embodiment 42 is a diagrammatic representation of Embodiment 42.
  • Embodiment 43 is a diagrammatic representation of Embodiment 43.
  • Embodiment 44 is a diagrammatic representation of Embodiment 44.
  • Embodiment 45 is a diagrammatic representation of Embodiment 45.
  • Embodiment 46 The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, and compound No. 19 as shown in example 1 herein.
  • Embodiment 47 is a diagrammatic representation of Embodiment 47.
  • Embodiment 48 is a diagrammatic representation of Embodiment 48.
  • Embodiment 49 is a diagrammatic representation of Embodiment 49.
  • Embodiment 50 is a diagrammatic representation of Embodiment 50.
  • the GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor.
  • Embodiment 51
  • Embodiment 52 is a diagrammatic representation of Embodiment 52.
  • a pharmaceutical composition comprising a derivative according to any one of the preceding embodiments, and at least one pharmaceutically acceptable excipient.
  • Embodiment 53 is a diagrammatic representation of Embodiment 53.
  • a pharmaceutical composition comprising a derivative according to any one of embodiments 1-51 , a GLP-1 receptor agonist, and at least one pharmaceutically acceptable excipient.
  • Embodiment 54 The pharmaceutical composition according to embodiment 53, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 55 is a diagrammatic representation of Embodiment 55:
  • composition according to embodiments 53-54 wherein the GLP-1 receptor agonist is semaglutide.
  • Embodiment 56 is a diagrammatic representation of Embodiment 56.
  • composition according to any one of embodiments 52-55 for use as a medicament.
  • Embodiment 57
  • Embodiment 58
  • Embodiment 59 is a diagrammatic representation of Embodiment 59.
  • the GIP derivative for use according to Embodiment 58 in combination with a GLP-1 receptor agonist for use according to Embodiment 58 in combination with a GLP-1 receptor agonist.
  • Embodiment 60 is a diagrammatic representation of Embodiment 60.
  • Embodiment 61 is a diagrammatic representation of Embodiment 61 :
  • Embodiment 62 A dosage form comprising a combination of a GIP derivative according to any one of Embodiments 1 to 51 and a GLP-1 receptor agonist.
  • Embodiment 63
  • Embodiment 64 is a diagrammatic representation of Embodiment 64.
  • Embodiment 65 is a diagrammatic representation of Embodiment 65.
  • kits-of-parts comprising a first dosage form comprising a GIP derivative according to any one of Embodiments 1 to 51 and a second dosage form comprising a GLP-1 receptor agonist.
  • Embodiment 66
  • kits-of-parts according to Embodiment 67 wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 67 is a diagrammatic representation of Embodiment 67.
  • kits-of-parts according to Embodiment 66 wherein the GLP-1 receptor agonist is semaglutide.
  • Embodiment 68
  • GIP derivative for the manufacture of a medicament for the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
  • Embodiment 69
  • a method of prevention and/or treatment of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome comprising administering a pharmaceutically active amount of the derivative according to any one of embodiments 1-51.
  • Embodiment 70 is a diagrammatic representation of Embodiment 70.
  • the method according to embodiment 69 administering the derivative according to any one of embodiments in combination with a pharmaceutically active amount of a GLP-1 receptor agonist.
  • Embodiment 71
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 72 is a diagrammatic representation of Embodiment 72.
  • Embodiment 1 is a diagrammatic representation of Embodiment 1:
  • a glucose-dependent insulinotropic peptide (GIP) derivative comprising of a GIP analogue comprising Formula la, Ila, or Illa and a modifying group, wherein Formula la, Ila, and Illa are defined by:
  • Embodiment 2 is a diagrammatic representation of Embodiment 1:
  • a glucose-dependent insulinotropic peptide (GIP) derivative consisting of a GIP analogue of Formula la, Ila, or Illa and a modifying group, wherein Formula la, Ila, and Illa are defined by:
  • Embodiment 3 is a diagrammatic representation of Embodiment 3
  • GIP glucose-dependent insulinotropic peptide
  • Embodiment 4 is a diagrammatic representation of Embodiment 4:
  • Embodiment 5 is a diagrammatic representation of Embodiment 5:
  • Embodiment 6 is a diagrammatic representation of Embodiment 6
  • Embodiment 7 is a diagrammatic representation of Embodiment 7:
  • Embodiment 8 is a diagrammatic representation of Embodiment 8
  • Embodiment 9 is a diagrammatic representation of Embodiment 9:
  • Embodiment 10 The GIP derivative according to any one of the preceding embodiments, wherein the GIP analogue is of Formula la or Ila.
  • Embodiment 11 is a diagrammatic representation of Embodiment 11 :
  • Embodiment 12 is a diagrammatic representation of Embodiment 12
  • GIP derivative according to any one of the preceding embodiments, wherein the GIP analogue is of Formula la or Ila, wherein Xxx is Tyr and Xxxn is Lys.
  • Embodiment 13 is a diagrammatic representation of Embodiment 13:
  • Embodiment 14 is a diagrammatic representation of Embodiment 14:
  • GIP derivative according to any one of embodiments 1-10 or 13, wherein the GIP analogue is of Formula la or Ila, wherein Xxx is Lys and Xxxn is Ser.
  • Embodiment 15 is a diagrammatic representation of Embodiment 15:
  • Embodiment 16 is a diagrammatic representation of Embodiment 16:
  • Embodiment 17 is a diagrammatic representation of Embodiment 17:
  • Embodiment 18 is a diagrammatic representation of Embodiment 18:
  • Embodiment 20 is a diagrammatic representation of Embodiment 20.
  • Embodiment 21 is a diagrammatic representation of Embodiment 21 :
  • Embodiment 22 is a diagrammatic representation of Embodiment 22.
  • Embodiment 23 is a diagrammatic representation of Embodiment 23.
  • Embodiment 24 is a diagrammatic representation of Embodiment 24.
  • Embodiment 25 is a diagrammatic representation of Embodiment 25.
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of formula la, wherein the modified lysine is in position 11 , Xxx is Tyr, Xxxn is Lys, Xxxso is Lys, Xxxsi is Gly, and the N-terminal end is substituted with an a-acetylate.
  • Embodiment 26 is a diagrammatic representation of Embodiment 26.
  • Embodiment 27 is a diagrammatic representation of Embodiment 27.
  • Embodiment 28 is a diagrammatic representation of Embodiment 28:
  • Embodiment 29 is a diagrammatic representation of Embodiment 29.
  • GIP derivative according to any one of Embodiments 1-24, wherein the GIP analogue is of Formula Illa, wherein the modified lysine is in position 43, and the N-terminal end is substituted with an a-acetylate.
  • Embodiment 30 is a diagrammatic representation of Embodiment 30.
  • Embodiment 31
  • Embodiment 32 is a diagrammatic representation of Embodiment 32.
  • GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula la, wherein the modified lysine is in position 11 , Xxxs is absent, Xxx 8 is L-3-phenyllactic acid (Pla), Xxx is Tyr, Xxxn is Lys, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent.
  • the GIP analogue is of Formula la, wherein the modified lysine is in position 11 , Xxxs is absent, Xxx 8 is L-3-phenyllactic acid (Pla), Xxx is Tyr, Xxxn is Lys, Xxx 3 o is Lys or absent, Xxx 3i is Gly or absent.
  • Embodiment 33 is a diagrammatic representation of Embodiment 33.
  • Embodiment 34 is a diagrammatic representation of Embodiment 34.
  • Embodiment 36 is a diagrammatic representation of Embodiment 36.
  • Embodiment 37 is a diagrammatic representation of Embodiment 37.
  • Embodiment 38 is a diagrammatic representation of Embodiment 38.
  • A is a C14 to C18 mono fatty acid, such as a C16 mono fatty acid.
  • Embodiment 39 is a diagrammatic representation of Embodiment 39.
  • Embodiment 40 is a diagrammatic representation of Embodiment 40.
  • Embodiment 41
  • GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp.
  • Embodiment 42 is a diagrammatic representation of Embodiment 42.
  • GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu and y-Glu-y-Glu.
  • Embodiment 43 is a diagrammatic representation of Embodiment 43.
  • Embodiment 45 is a diagrammatic representation of Embodiment 45.
  • Embodiment 46 is a diagrammatic representation of Embodiment 46.
  • Embodiment 47 is a diagrammatic representation of Embodiment 47.
  • Embodiment 48 is a diagrammatic representation of Embodiment 48.
  • Embodiment 49 is a diagrammatic representation of Embodiment 49.
  • Embodiment 50 is a diagrammatic representation of Embodiment 50.
  • Embodiment 51
  • Embodiment 53 is a diagrammatic representation of Embodiment 53.
  • Embodiment 54 is a diagrammatic representation of Embodiment 54:
  • Embodiment 55 is a diagrammatic representation of Embodiment 55:
  • Embodiment 56 is a diagrammatic representation of Embodiment 56.
  • the GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor.
  • Embodiment 57
  • the GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor and an antagonist at the mouse GIP receptor.
  • Embodiment 58
  • a pharmaceutical composition comprising a derivative according to any one of the preceding embodiments, and at least one pharmaceutically acceptable excipient.
  • Embodiment 59 is a diagrammatic representation of Embodiment 59.
  • a pharmaceutical composition comprising a derivative according to any one of embodiments 1-57, a GLP-1 receptor agonist, and at least one pharmaceutically acceptable excipient.
  • Embodiment 60 :
  • composition according to embodiment 59 wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 61 is a diagrammatic representation of Embodiment 61 :
  • composition according to embodiments 59-60, wherein the GLP-1 receptor agonist is semaglutide.
  • Embodiment 62
  • composition according to any one of embodiments 58-61 for use as a medicament.
  • Embodiment 63
  • the GIP derivative according to any one of Embodiments 1-57 for use as a medicament for use as a medicament.
  • Embodiment 64 is a diagrammatic representation of Embodiment 64.
  • Embodiment 65 is a diagrammatic representation of Embodiment 65.
  • the GIP derivative for use according to Embodiment 64 in combination with a GLP-1 receptor agonist for use according to Embodiment 64 in combination with a GLP-1 receptor agonist.
  • Embodiment 66
  • the GIP derivative for use according to Embodiment 65 wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 67 is a diagrammatic representation of Embodiment 67.
  • Embodiment 65-66 wherein the GLP-1 receptor agonist is semaglutide.
  • GIP derivative for the manufacture of a medicament for the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
  • Embodiment 69
  • a method of prevention and/or treatment of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome comprising administering a pharmaceutically active amount of the derivative according to any one of embodiments 1-57.
  • Embodiment 70 is a diagrammatic representation of Embodiment 70.
  • the method according to embodiment 69 administering the derivative according to any one of embodiments in combination with a pharmaceutically active amount of a GLP-1 receptor agonist.
  • Embodiment 71
  • the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
  • Embodiment 72 is a diagrammatic representation of Embodiment 72.
  • Aib a-aminoisobutyric acid
  • 6-CI-HOBt 6-chloro-1 -hydroxybenzotriazole
  • AEEA 8-amino-3,6-dioxaoctanoic acid
  • DIEA/DIPEA N,N-Diisopropylethylamine
  • GIP Glucose-dependent insulinotropic peptide
  • GIPR Glucose-dependent insulinotropic peptide receptor
  • GLP-1 Glucagon-like peptide 1
  • HFIP Hexafluoroisopropanol
  • hGIP Human glucose-dependent insulinotropic peptide
  • hGIPR Human glucose-dependent insulinotropic peptide receptor
  • IPGTT Intra-peritoneal glucose tolerance test
  • mGIP Mouse glucose-dependent insulinotropic peptide
  • mGIPR Mouse glucose-dependent insulinotropic peptide receptor
  • peptides were synthesized by automated Fmoc/tBu solid-phase methodology employing a Symphony peptide synthesizer (Gyros Protein Technologies, Arlington, AZ) and Applied Biosystems ABI 433A peptide synthesizer, starting with preloaded Wang resin (AAPPtec, Louisville, KY; Novabiochem, San Diego, CA) for acid C-terminal peptides, and H-Rink Amide ChemMatrix® (PCAS BioMatrix Inc, Saint-Jean-sur-Richelieu, Quebec, Canada J3B 8J8) for C-terminal amide peptides.
  • Fmoc-amino acids were coupling with 6-CI-HOBt/DIC or OxymaPure/DIC activations in DMF. All common Fmoc- amino acids, 6-CI-HOBt, OxymaPure and DIC were purchased from Midwest Biotech (Fisher, IN), AAPPtec and Gyros Protein Technologies. The Fmoc were removed by 20% piperidine in DMF. The N-terminal acetylation was performed on resin in the presence of tenfold excess of acetic anhydride/DIEA in DCM for 1 h. The N-terminal L-3-Phenyllactic acid (Pla) was coupled manually in 5-fold excess by DEPBT in DMF/DIEA.
  • the residues were pre-incorporated with Fmoc-L-Lys(Mtt) - OH, after peptide assembling done and the N-terminal acetylation, the Mtt was deprotected with 1 - 2%TFA/5%Tis in DCM or with 30% HFIP/5% Tis in DCM.
  • the linkage amino acid of y-glutamic acid was coupled by Fmoc-Glu-OtBu; AEEA was coupled by Fmoc-8-amino-3,6-dioxaoctanoic acid/Fmoc-AEEA-OH (AAPPTec, Louisville, KY). Palmitic acid was coupled by 5-fold excess with DEPBT/DIEA in DMF for about 4h; the octadecanedioic acid acylation was done by coupling octadecanedioic acid mono-tert-butyl ester (Novo Nordisk, Malov, DK) with 5-fold excess DEPBT/DIEA in DMF for about 4h.
  • Completed peptidyl resins were cleaved by standard TFA cleavage containing 5% Tis and 5% H 2 O, added 2.5% 2-Mercaptoethanol or the odourless thiol scavenger DODT (Sigma-Aldrich, St. Louis, MS) for the cysteine and methionine residue containing peptides for 2 h with agitation.
  • DODT odourless thiol scavenger
  • the cleaved peptides in TFA were precipitated with ether, centrifuged and washed twice with ether, the crude peptides were dissolved in 20% ACN containing 2% acetic acid and injected to a Luna 19 x 250nm/10 pm C8 column (Phenomenex, Torrance, CA) to purify with 0.1% TFA/ACN eluent solvents on the Waters 2545 preparative HPLC system.
  • Peptide molecular weights characterization were measured by liquid chromatography-mass spectrometry on an Agilent 1260 lnfinity/6120 Quadrupole instrument with a Kinetex C8 column.
  • Eluent A is water with 0.05% TFA and eluent B is 10% water, 90% ACN and 0.05% TFA.
  • Eluent gradient methods are Method A: a gradient of 10%-80% eluent B in 10 min with 1 ml/min flow rate; Method B: a gradient of 20%-100% eluent B in 10 min with 1 ml/min flow rate; Method C: a gradient of 30%-80% eluent B in 3.5 min with 4 ml/min flow rate; Method D: a gradient of 40%-100% eluent B in 3.5 min with 4 ml/min flow rate.
  • the utility of the derivatives of the present invention as pharmacologically active agents e.g. in the reduction of weight gain and treatment of obesity in mammals, such as humans may be demonstrated by the activity of the antagonists in conventional assays and in the in vitro and in vivo assays described below.
  • Such assays also provide a means whereby the activities of the antagonists of the invention can be compared with activities of known compounds
  • the receptor binding assay may be performed at the desired receptors, herein the human GIP receptors (“hGIPR”) and mouse GIP receptors (“mGIPR”) are tested.
  • hGIPR human GIP receptors
  • mGIPR mouse GIP receptors
  • GIP receptors were inserted in pcDNA3 vectors under the control of a CMV promotor with Geneticin as the selectable marker.
  • the pGL4.29[luc2P/CRE/Hygro] vector from Promega (USA, E8471) contains a cAMP response element (CRE), that drives the transcription of the luciferase (luc) reporter gene luc2P.
  • Luc2P is a synthetically-derived luciferase sequence optimized for expression in mammalian cells.
  • BHK Baby Hamster Kidney
  • stable clones expressing receptor as well as reporter gene were selected in culture media containing hygromycin and geneticin (G418).
  • the clones used were: Human (hGIPR/BHK CRELuc2p clone#5), mouse (mGIPR/BHK CRELuc2p clone#3).
  • Binding of peptide antagonists to the full length GIP receptor was assessed in whole cells with competitive binding using radiolabelled human GIP ( 125 l-NNC0090-0554, produced at Novo Nordisk A/S).
  • BHK cells stably expressing the GIP receptor were seeded in poly-D- lysine coated white 96-well plates with clear bottom (Corning, cat. no. 354651).
  • 5000-10000 cells are seeded pr. well in DMEM medium (cat. no 61965-026) supplemented with Geneticin (Gibco, cat. no 10131-027), Hygromycin B (Gibco cat no 10687-010) and 10% fetal bovine serum (FBS) (Gibco, cat. no 16140-071).
  • the assay buffer consists of HBSS (Gibco, cat no 14025), 10mM HEPES (Gibco, cat. no 15630), 0.1 % pluronic F-68 (Gibco, cat. no 2404,) and 0.1 % ovalbumin (SigmaAldrich, cat. no A5503), and pH is adjusted to 7.4.
  • GIP receptor peptide antagonists were diluted in assay buffer and tested in 10-fold dilutions from 10 pM downwards.
  • Nonlinear regression analysis on the output files was performed in the Windows program GraphPad Prism 7 (GraphPad software, USA) using the equation “log(inhibitor) vs response (three parameters)”. Data were reported as geometric mean of IC50 values with 95% Cl.
  • BHK-21 cells (ATCC CCL-10) were transfected with human or mouse GLP-1 or GIP receptor and firefly luciferase under control of cAMP-response element.
  • the cells were grown in Dulbecco’s modified Eagle medium (DMEM, Thermo Fisher 10566-016) supplemented with 10% Fetal Bovine Serum (Thermo Fisher 10082147), 100 lU/ml penicillin, 100 pg/mL streptomycin, and 10 mM HEPES at 37°C, 5% CO2 and 90% humidity for 2-3 days. Sixteen to twenty hours prior to the experiment the cells were plated at 5x104 cells per well in 96 well Isoplate (Perkin-Elmer 6005040, Waltham, MA).
  • the plate was gently washed with warm Hank’s Balanced Salt Solution pH 7.4 (14025092, Invitrogen, Carlsbad, CA) and filled with 0.1 ml/well of serial dilutions of peptides prepared in sterile DMEM containing 1% Ovalbumin (A5503, Sigma-Aldrich).
  • the peptides were premixed with EC90 concentration- (75 - 95% maximal signal) of native ligand.
  • mice After 3h incubation at 37°C and 5% CO2 in humidified atmosphere, the plates were washed with warm Hank’s Balanced Salt Solution pH 7.4 and 0.1 ml/well of Steady Lite HTS luminescence substrate reagent (Perkin-Elmer, Waltham, MA) was added to each well. The plate was sealed and shaken 10 min at 800 rpm. Luminescence signal was measured on Perkin-Elmer Envision plate reader. The luminescence data were plotted against peptide concentrations and EC50 or IC50 values were calculated by using logistic nonlinear 3 parameter regression analysis in GraphPad Prism 7 (GraphPad Software, La Jolla, CA). General methods for pharmacokinetic study in mice
  • Plasma was collected at time points of 5min, 30min, 1 h, 2h, 6h, 8h, 24h, 48h, 72h, 96h, and 120h. The last time point of compound 9 is 48h.
  • standards of each peptide were prepared in mouse plasma ranging from 0.5 to 4000 nM.
  • a protein precipitation was performed by organic solvent extraction with a 14 - fold dilution with methanol. The samples were centrifuged for 20 min at 4°C at 13000g- force. Supernatants of samples were collected and diluted 3-fold with 0.1% formic acid in water. The diluted samples were then subjected to LC - MS analysis.
  • LC-MS analyses were carried out on a Thermo Q Exactive HF mass spectrometer interfaced with a Vanquish LIPLC. LC separations were performed on an Acquity LIPLC BEH C18 1.7 pm, 1.0 x 50 mm column. Mobile phase A was composed of 0.1% formic acid in water and mobile phase B was composed of 0.1% formic acid in ACN. The LC flow rate was set to 0.4 pL/min using a gradient elution from 10 to 95% B over the course of 4.0 min.
  • mice studies were performed in accordance with Institutional Animal Care and Use Committee guidelines at University of Cincinnati.
  • Male C57BL/6J mice (Jackson Laboratories) were housed 4 per cage under 12 h/12 h light-dark cycle at 22°C with ad libitum access to water and 58% fat, high-sugar diet (D12331 , Research Diets) for ⁇ 12 weeks.
  • Treatment groups received either vehicle, semaglutide (2nmol/kg/d), GIPR antagonist (500 or 1500nmol/kg/d), semaglutide + GIPR antagonist (2 + 500 or 1500nmol/kg/d; co-formulated single injection), or semaglutide + GIPR antibody (2nmol/kg/d + 30mg/kg/w; separate injections).
  • Test compounds 200pM were dissolved in a vehicle (pH 7.4) containing 0.05% polysorbate-80, 50 mM sodium phosphate, and 70 mM sodium chloride; test compounds were administered once daily (semaglutide and GIPR antagonist) for 27d, subcutaneously during the light cycle at a volume of 3.9
  • Tail blood glucose levels were measured 0, 15, 30, 60, 90, and 120 minutes following the glucose load.
  • An additional measure of 6h fasting insulin (Crystal Chem 90080), resistin (R&D Systems MRSN00), CTX (Novus Biologicals NBP2-69074) and total GIP (Crystal Chem 81527) levels were taken at day 27.
  • peptide analogues and derivative of the present invention were synthesized according to the General Methods of Peptide Synthesis as describe above. Name, structure, and properties are shown below for each compound. SEQ ID NO’s are included for the peptide backbone.
  • the purpose of this example is to test the activity, or potency, of the derivatives in vitro at the human or mouse GIP receptor.
  • the potencies and binding of the analogues and derivatives of Example 1 were determined as described under General methods of measuring in vitro receptor binding and General methods of measuring in vitro functional potency above at the human and mouse GIP receptor. All IC50 data are average ⁇ SD of at least two independent experiments or average from one duplicate experiment. Percentage indicating the maximal inhibition, the inhibition is 95 ⁇ 100% for those without notice. Results are shown in Table 1.
  • Table 1 Potency and binding of test compounds at hGIPR and mGIPR inhibitions are above 95%. **: compound has di-fatty acid in side chain; nd: not determined or not available; Partial weak antagonism: with below 60% inhibition. All IC50 data are average ⁇ SD of at least two independent experiments or average from one duplicate experiment.
  • acylation with mono fatty acid acylation at one of positions 10, 11 , or 43, in particular 11 or 10, compared to fatty acid acylation at positions 13, 16, 17, 18, 21 , 24, or 30, result in very potent antagonists on both hGIPR and mGIPR, with mono fatty acid acylation at position 11 being the best antagonists on mGIPR. 210081WO01 62
  • mono fatty acid acylated derivatives show better antagonism than comparative derivatives with diacid acylation (see e.g. compound 7 and 16).
  • Example 3 In vivo pharmacokinetic study 5
  • the purpose of this study is to determine the half-life in vivo of the derivatives of the present invention after a single s.c. administration to mice, i.e. the prolongation of their time in the body and thereby their time of action. This is done in a pharmacokinetic (PK) study, as described under General methods for pharmacokinetic study in mice where the terminal half- life of the derivative in question is determined.
  • PK pharmacokinetic
  • terminal half-life is generally meant the 10 period of time it takes to halve a certain plasma concentration, measured after the initial distribution phase.
  • Example 4 In vivo pharmacodynamic study The purpose of this example is to assess the in vivo effect of the derivatives of the 20 present invention alone and in combination with a GLP-1 receptor agonist on food intake, body weight, and glucose tolerance in diet-induced obese (DIO) mice.
  • the GLP-1 receptor agonist used for this example was semaglutide.
  • dosing compound No. 7 at 500 nmol/kg did not affect glucose level compared to vehicle.
  • dosing compound No. 7 at 1500 nmol/kg increased glucose levels on day 0. This effect was overcome when combining both the low dose and high dose of compound No. 7 with GLP-1R agonist semaglutide 2 nmol/kg.
  • results from day 22 OGTT and day 27 IPGTT shows that after chronical administration of compound No. 7 both alone and in combination with GLP-1R agonist semaglutide, the GIPR antagonist improves glucose control. Both high and low dose of compound No. 7 on day 22 (OGTT) and day 27 (IPGTT) improves glucose level compared to vehicle, and in combination with semaglutide showed significantly improved glucose levels compared to semaglutide alone on day 27 IPGTT.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Endocrinology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Otolaryngology (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present application relates to novel peptides that are derivatives of glucose-dependent insulinotropic peptide (GIP) analogues with improved GIP antagonistic activity, and to the pharmaceutical use of the GIP derivatives. The GIP derivatives of the invention are modified by including a lipophilic moiety.

Description

LONG-ACTING GIPR ANTAGONISTS
TECHNICAL FIELD
The present application relates to novel antagonists that are derivatives of glucosedependent insulinotropic peptide (GIP) analogues with improved GIP antagonistic activity, and to the pharmaceutical use of the GIP derivatives.
INCORPORATION-BY-REFERENCE OF THE SEQUENCE LISTING
SEQUENCE LISTING
The present application is filed with a Sequence Listing in electronic form. The entire contents of the sequence listing are hereby incorporated by reference.
BACKGROUND
Glucose-dependent insulinotropic polypeptide (GIP, also known as gastric inhibitory peptide) is one of two endogenous incretins and is a 42 amino acids peptide hormone released from intestinal K-cells following food intake. GIP and another incretin, glucagon-like Peptide-1 (GLP-1), are gut enteroendocrine cell-derived hormones accounting for the incretin effect, which estimated to account for over 70% of the total insulin response to an oral glucose challenge.
Due to the incretin effect, the GIP receptor has become an attractive drug target in the treatment of metabolic diseases such as obesity and diabetes, with GIP receptor agonists either as a standalone, in combination with GLP-1 receptor agonists, or in combination with GLP-1/glucagon receptor co-agonists. GIP itself has a short plasma half-life due to dipeptidyl peptidase-4 (DPP-IV) mediated inactivation, and poor physical stability due to high tendency to form fibrils in solution. If GIP(1-42) or GIP(1-30) are secreted into the circulation in humans, the cleavages catalyzed by DPP-4 would result in GIP(3-42) or GIP(3- 30) which both are inactive GIP hormones.
WO 2020/115048 and US2020/087373 discloses GIP analogues that act as hGIP receptor antagonists. These GIP analogues are modified by introducing a modified lysine at one or more of positions 3 to 29, wherein the modification is primarily a fatty acid in the form of a diacid attached to the epsilon-amino group of the modified lysine and the primary site for attachment of said modification is in position 18.
It is, however, desirable achieving hGIP receptor antagonists with increased activity. For the purpose of developing a GIP based drug, it is also important to have good correlation of the activity from in vitro assays to animal models. Accordingly, it is also desirable to obtain GIP receptor antagonists having antagonist activity at the human GIP receptor and at the same time on a model GIP receptor e.g. the mGIP receptor. These objectives have been achieved with the GIP derivatives according to the present invention.
SUMMARY OF THE INVENTION
Accordingly, in one aspect, the invention concerns a glucose-dependent insulinotropic peptide (GIP) derivative consisting of a modifying group and a GIP analogue wherein the GIP analogue is defined by Formula la, Ila, or Illa:
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxxso-Xxxsi (la), (SEQ ID NO: 46);
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (Ila), (SEQ ID NO: 47);
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (Illa), (SEQ ID NO: 48); wherein Xxxs is Thr or absent, Xxxe is Phe or L-3-phenyllactic acid (Pla), Xxx is Tyr or Lys, Xxxn is Ser or Lys, Xxxi4 is Met, Leu, lie or Nle, Xxxis is His, Arg, Orn, homoArg, XXX21 is Asp, Glu or homoGlu, Xxxso is Lys or absent, and Xxxsi is Gly or absent; provided that when Xxx6 is Pla, then Xxx5 is absent; wherein said GIP analogue of Formula la or Ila comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula Illa comprises a modified lysine in position 43, said modified lysine comprises the modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine in one of positions 11 , 10 or 43, the modifying group being defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present; or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof. In another aspect, the invention concerns the GIP derivative according to the invention for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
It has been found that the compounds of the invention provide for an even better effect on weight loss in combination with a GLP-1 receptor agonist. Accordingly, in still another aspect, the invention concerns a dosage form comprising a combination of a GIP derivative according to the invention and a GLP-1 receptor agonist.
In yet another aspect, the invention concerns a kit-of-parts comprising a first dosage form comprising a GIP derivative according to the invention and a second dosage form comprising a GLP-1 receptor agonist.
BRIEF DESCRIPTION OF DRAWINGS
Fig. 1 shows percent change in body weight over 26 days of treatment of DIO mice when administering vehicle (open circles), semaglutide 2 nmol/kg (closed squares), compound 7 500 nmol/kg (open triangles-up), compound 7 1500 nmol/kg (open triangles-down), semaglutide 2 nmol/kg + compound 7 500 nmol/kg (closed triangles-up), and semaglutide 2 nmol/kg + compound 7 1500 nmol/kg (closed triangles-down).
Fig. 2 shows cumulative food intake over 26 days of treatment of DIO mice when administering vehicle (open circles), semaglutide 2 nmol/kg (closed squares), compound 7 500 nmol/kg (open triangles-up), compound 7 1500 nmol/kg (open triangles-down), semaglutide 2 nmol/kg + compound 7 500 nmol/kg (closed triangles-up), and semaglutide 2 nmol/kg + compound 7 1500 nmol/kg (closed triangles-down).
DETAILED DESCRIPTION OF THE INVENTION
Definitions
In what follows, Greek letters may be represented by their symbol or the corresponding written name, for example: a = alpha; p = beta; s = epsilon; y = gamma; co = omega; etc. Also, the Greek letter of p may be represented by "u", e.g. in pl=ul, or in pM=uM.
Unless otherwise indicated in the specification, terms presented in singular form generally also include the plural situation.
Also described herein are derivatives of GIP analogues, pharmaceutical compositions and uses thereof in which open ended terms like “comprises” and ’’comprising” are replaced with closed terms such as “consists of”, “consisting of”, and the like. GIP receptor antagonists
A receptor antagonist may be defined as a compound that binds to a receptor and is capable of inhibiting or reducing a response typical of the natural ligand. As described herein, a “GIP antagonist” may be described as a compound capable of binding to the GIP receptor without activating the GIP receptor or reducing the activation compared to the native ligand.
GIP analogues
The term “hGIP(1-42)” as used herein refers to the human glucose-dependent insulinotropic polypeptide, the sequence of which is included in the sequence listing as SEQ ID NO: 5. The peptide having the sequence of SEQ ID NO: 5 may also be designated native hGIP or hGIP.
The term “hGIP(1-31)” as used herein refers to a truncated version of hGIP(1-42), comprising amino acids 1-31 of hGIP(1-42), the sequence of hGIP(1-31) is included in the sequence listings as SEQ ID NO: 4.
The term “hGIP(3-31)” as used herein refers to a truncated version of hGIP(1-42) comprising amino acids 3-31 of hGIP(1-42), and is a GIP antagonist. The sequence of hGIP(3-31) is included in the sequence listings a SEQ ID NO: 6.
The term “hGIP(3-42)” as used herein refers to truncated version of hGIP(1-42) comprising amino acids 3-42 of hGIP(1-42), and is a GIP antagonist. The sequence of hGIP(3-42) is included in the sequence listings as SEQ ID NO: 7.
The term “GIP analogue” as used herein refers to a peptide, or a compound, which is a variant of hGIP(1-31) or hGIP(1-42). The term “variant” is used for peptides comprising at least one amino acid substitution as compared to hGIP(1-31) or hGIP(1-42) and is capable of binding to the GIP receptor.
In the context of the present invention, the term “amino acid modification” refers to the substitution of an amino acid for another, the deletion of an amino acid, or the addition of an amino acid.
The term “substitution” as used herein refers to one amino acid being replaced by another in the backbone of the peptide. The GIP derivatives of the invention may comprise substitutions of one or more unnatural and/or non-amino acids, e.g., amino acid mimetics, into the sequence of the GIP derivative. As an example, replacing the serine in position 11 of hGIP(1-31) with lysine is considered a “substitution”.
The term “deletion” as used herein refers to one amino acid in the backbone of the peptide being removed without replacing it with another moiety. As an example, deleting the first four amino acids of hGIP(1-31) to prepare hGIP(5-31) (SEQ ID NO: 8) is considered a “deletion” of each of the four amino acids.
The term “addition” as used herein refers to adding one amino acid, or another moiety that may form part of the peptide backbone, into the backbone of the peptide. As an example, adding an amino acid at the C-terminal end of hGIP(1-31) to prepare (a modified) hGIP(1-32) is considered an “addition”.
GIP analogues of the derivatives of the invention may be described by reference to i) the number of the amino acid residue in hGIP(1-31) or hGIP(1-42) which corresponds to the amino acid residue which is changed (i.e. the corresponding position in hGIP(1-31) or hGIP(1-42), and to ii) the actual change. For example, [M14L]-hGIP(1-31) refers to a GIP analogue in which the methionine of position 14 of hGIP(1-31) has been replaced by a leucine.
The numbering of positions in the GIP analogues of the present invention is made with reference to hGIP(1-31) or hGIP(1-42), even if some of the amino acids in the N- terminal end have been deleted. As an example, the first position of hGIP(5-31) is referred to as position 5, even though it is the first amino acid of the sequence.
In one aspect, the GIP analogues of the derivatives of the invention comprises Formula la, Ila, or Illa (SEQ ID NOs: 46, 47, 48):
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxxso-Xxxsi (la), (SEQ ID NO: 46);
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (Ila), (SEQ ID NO: 47);
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Xxx2o-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (Illa), (SEQ ID NO: 48); wherein Xxxs is Thr or absent, Xxxe is Phe or L-3-phenyllactic acid (Pla), Xxx is Tyr or Lys, Xxxn is Ser or Lys, Xxxi4 is Met, Leu, lie or Nle, Xxxis is His, Arg, Orn, homoArg, XXX21 is Asp, Glu or homoGlu, Xxxso is Lys or absent, and Xxxsi is Gly or absent; provided that when Xxxe is Pla, then Xxxs is absent, wherein said GIP analogue of Formula la or Ila comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula Illa comprises a modified lysine in position 43, or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
In an alternative aspect, the GIP analogues of the derivatives of the invention comprises Formula I, II, or III (SEQ ID NOs: 1, 2, 3):
Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- As n-Trp- Le u- Le u- A I a- G I n-Xxxso-Xxxsi ( I ) , Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (II), Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (III), wherein Xxxs is Glu or absent, Xxx4 is Gly or absent, Xxxs is Thr or absent, Xxxso is Lys or absent, and Xxxsi is Gly or absent; wherein said GIP analogue of Formula I or II comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula III comprises a modified lysine in position 43; wherein the GIP analogue has a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31) (SEQ ID NO: 4), or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
The position of the modified lysine of the GIP analogue of the derivative of the invention depends on whether the GIP derivative of the invention comprises a GIP analogue of Formula I, II, III, la, Ila, or Illa. If the GIP derivative of the invention comprises a GIP analogue of Formula I, II, la or Ila, the modified lysine is in one of positions 11 or 10. If the GIP derivative of the invention comprises a GIP analogue of Formula III or Illa, the modified lysine is in position 43.
In one embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula la or Ila. In a further embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula la. In another embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula Illa.
In one embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula I or II. In a further embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula I. In another embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula III.
In still a further embodiment, the GIP derivative of the invention comprises a GIP analogue of Formula I, II, la or Ila, wherein the modified lysine is in one of positions 11 and 10. In yet a further embodiment, the modified lysine is in position 11 .
The GIP analogues of the derivatives of the present invention do not have any amino acids in positions 1 and 2 corresponding to positions in hGIP(1-31). In one embodiment, the derivatives do not have any amino acids in position 1 , 2, 3 and 4 corresponding to hGIP(1-31). In one embodiment, Xxx5 is Thr and Xxx6 is Phe. In a further embodiment, an additional deletion may be present in position 5. Accordingly, in one embodiment, Xxxs is absent. In another embodiment, Xxxs is absent and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
The GIP analogue of the derivative of the invention may have a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31) (i.e. compared to hGIP(3-31)), some of which may be substitutions. Accordingly, in one embodiment, Met in position 14 is substituted by Leu. In a further embodiment, His in position 18 is substituted by Arg. In still a further embodiment, Asp in position 21 is substituted by Glu. In yet a further embodiment, the GIP analogue of the derivative of the invention has a maximum of 8 amino acid modifications as compared to hGIP(3-31). In another embodiment, the GIP analogue of the derivative of the invention has a maximum of 7 amino acid modifications as compared to hGIP(3-31). In still another embodiment, the GIP analogue of the derivative of the invention has a maximum of 6 amino acid modifications as compared to hGIP(3-31). In one embodiment, Xxxi4 is Leu, Xxxis is Arg and XXX21 is Glu.
The GIP analogue of the derivatives of the present invention may have a combination of the features defined above. Accordingly, in one embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxx5 is Thr, Xxx6 is Phe, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a-acetylate. In another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la or Ila, wherein the modified lysine is in position 10 (Xxx is Lys), Xxxs is Thr, Xxxe is Phe, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a-acetylate.
In an alternative embodiment, the GIP analogue of the derivatives of the present invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx3 and Xxx4 are absent, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a-acetylate. In another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula I or II, wherein the modified lysine is in position 10, Xxx3 and Xxx4 are absent, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a- acetylate.
In still another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula Illa, wherein the modified lysine is in position 43 and the N-terminal end is substituted with an a-acetylate. In yet another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxxs is absent, and Xxx3 is L-3-phenyllactic acid (Pla). In yet another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la, wherein the modified lysine is in position 11 (Xxxn is Lys), Xxxs is absent, Xxx3o is Lys or absent, Xxx3i is Gly or absent, and Xxx3 is L-3-phenyllactic acid (Pla). In a further embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula la or Ila, wherein the modified lysine is in position 10 (Xxx is Lys), Xxxs is absent, Xxx3o is Lys, Xxx3i is Gly, and Xxx3 is L-3-phenyllactic acid (Pla).
In still another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula III, wherein the modified lysine is in position 43, Xxx3 and Xxx4 are absent, and the N-terminal end is substituted with an a-acetylate. In yet another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx3, Xxx4 and Xxxs are absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla). In yet another embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula I, wherein the modified lysine is in position 11 , Xxx3, Xxx4, and Xxxs are absent, Xxx3o is Lys or absent, Xxx3i is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla). In a further embodiment, the GIP analogue of the derivative of the invention is a GIP analogue of Formula I or II, wherein the modified lysine is in position 10, Xxx3, Xxx4, and Xxxs are absent, Xxx3o is Lys, Xxx3i is Gly, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
In some embodiment, the GIP analogues of the derivatives of the invention may comprise a substitution selected from the group consisting of: M14L, H18R, D21 E as compared to hGIP(1-31). In some embodiment, the GIP analogue of the derivative of the invention comprises all of substitutions M14L, H18R and D21 E as compared to hGIP(1-31). In one embodiment, the GIP analogue of the derivatives of the invention comprises the backbone of [S11 K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 9). Also or alternatively, in one embodiment, the GIP analogue of the derivatives of the invention comprises the backbone of [F6Pla, S11 K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 10). Also or alternatively, in one embodiment, the GIP analogue of the derivatives of the invention comprises the backbone of [Y10K, M14L, H18R, D21 E]-hGIP(6-29) (SEQ ID NO: 11).
In some embodiment, further substitutions may be present in additional positions. In one embodiment, the amino acid in position 13 is selected from Ala or Lys. In one embodiment, position 16 is selected from Lys or Aib. In one embodiment, the amino acid in position 17 is selected from lie or Lys. In some embodiment, position 20 is selected from Gin or Aib. In one embodiment, the amino acid in position 24 is selected from Asn, Gin or Lys. In one embodiment, the amino acid in position 30 is selected from Lys, Arg or is absent. In one embodiment, the amino acid in position 31 is selected from Gly, Pro or is absent.
Backbone of the GIP analogues of the derivative of the invention are included in the sequence listing. In some embodiment, the GIP analogue of the derivative of the invention is selected from SEQ ID NO: 12-45, such as 17-45. In some embodiment, the GIP analogue of the derivative of the invention is be selected from any one of SEQ ID NO: 17-31 and 43-45, such as SEQ ID NO: 17-26, 28-31 and 43-44. In one embodiment, the GIP analogue of the derivative of the invention is selected from the group consisting of SEQ ID NO: 17-23, 31, 43- 44 such as 17-18, 21-23 or such as 19-20. In some embodiment, the GIP analogue of the derivative of the invention is selected from the group consisting of SEQ ID NO: 24-25, 28-30, such as 24-25, 30 or such as 28-30. In some embodiment the GIP analogue of the derivative of the invention is SEQ ID NO: 26.
Analogues “comprising” certain specified changes may comprise further changes, when compared to the respective formula. In a particular embodiment, the analogue "has" the specified changes.
As is apparent from the above examples, amino acid residues may be identified by their full name, their one-letter code, and/or their three-letter code. These three ways are fully equivalent.
GIP derivatives
The term “GIP derivative” as used herein refers to a chemically modified GIP analogue, in which one or more substituents have been covalently attached to the peptide backbone.
In one aspect of the invention, the substituent may be an N-terminal substituent. Also or alternatively, in one aspect, the substituent may be a modifying group or, alternatively, referred to as a protracting moiety or albumin binding moiety.
The term “N-terminal substituent” or “modifying group” as used herein, means a chemical moiety or group replacing a hydrogen atom. In one aspect, the derivative of a GIP analogue comprises a substituent covalently attached to the alpha-amino group of the amino acid residue in the N-terminus of the analogue. In one aspect, the N-terminal substituent is an alkanoyl or acyl group. In a particular aspect, the N-terminal substituent is an acetyl group or glutaric acid. As an example of an N-terminal substituted amino acid is Ac-Thr at position 5. Such N-acetylation would not count as a substitution in the peptide backbone compared with hGI P(1-31 ), because the amino acid in the GIP analogue is the native Thr, e.g. N“-Ac- [Lys11 ,Met14,Arg18,Glu21]-hGIP(5-31) comprises 4 substitutions as compared to hGIP(5- 31).
Also or alternatively, in one aspect, the GIP analogue comprises a modifying group covalently attached to a lysine residue at one of positions 11, 10, 21 or 43. In a further aspect, the modifying group is capable of forming non-covalent conjugates with proteins, e.g. albumin, thereby promoting the circulation of the derivative with the blood stream, and also having the effect of protracting the time of action of the derivative, due to the fact that the conjugate of the GIP derivative and albumin is only slowly disintegrated to release the active pharmaceutical ingredient.
The modifying group may be covalently attached to a lysine residue of the GIP analogue by acylation, i.e. via an amide bond formed between a carboxylic acid group of the modifying group and the epsilon amino group of said lysine group.
In one aspect, the modifying group is covalently attached to a lysine residue at one of positions 11 , 10, 21 or 43 by acylation, i.e. via an amide bond formed between a carboxylic acid group of the modifying group and the epsilon amino group of the lysine residue.
In one aspect, the modifying group is defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present.
It has been found that A- provides increased hGIP receptor antagonistic activity. It has in particular been found that antagonistic activity of comparative compounds, wherein an additional carboxylic acid group, has been added to A- to prepare a diacid derivative instead of a mono fatty acid derivative, is lower. Accordingly, in one embodiment, A- is a mono fatty acid. In a further embodiment, the mono fatty acid is a straight-chain mono fatty acid. In still a further embodiment, the straight-chain mono fatty acid is a C14 to C18 fatty acid, such as a C16 fatty acid. In yet a further embodiment, the fatty acid is a saturated fatty acid or an unsaturated fatty acid, such as a mono-unsaturated acid. In another embodiment, the fatty acid is a saturated fatty acid. In a more particular embodiment, the fatty acid is palmitic acid (C16). In the context of the present invention, the term mono fatty acid refers to a monoacid having one carboxylic acid group attached to the lysine in the GIP analogue. A mono fatty acid may be straight or branched and it may be saturated or unsaturated. A mono fatty acid typically has an even number of carbon atoms from 4 to 28, such as from 14 to 18 carbon atoms. A straight chain mono fatty acid may also be described by formula -C=(O)- (CH2)n-CH3, where n is an integer between 2-26, preferably n is 12, 14 of 16. The monoacids could use abbreviation such as C14, C16 and C18, herein C16 refer to palmitic acid.
The type of linker represented by B-C- in the GIP derivatives of the present invention is known in the art and may vary. In a typical variation, B is an amino acid. Accordingly, in one embodiment, B is an amino acid linker. In a further embodiment, B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp, wherein y-Glu is gamma-glutamic acid represented by Chem. 1 :
Figure imgf000012_0001
Chem. 1 :
In still a further embodiment, B is selected from y-Glu and y-Glu-y-Glu. In yet a further embodiment, B is y-Glu. In another embodiment, B is absent.
B may be attached directly to the peptide backbone of the GIP derivatives of the invention, or the spacer C may be inserted in-between. Accordingly, in one embodiment, C is absent or selected from AEEA2 and AEEA, wherein AEEA is 8-amino-3,6-dioxaoctanoic acid represented by Chem. 2:
Figure imgf000012_0002
Chem. 2:
In another embodiment, C is absent or AEEA2. In still another embodiment, C is absent. In yet another embodiment, B-C is y-Glu. In a further embodiment, B-C is y-Glu and A is a C16 fatty acid, such as palmitic acid and as represented by Chem. 3:
Chem.
Figure imgf000012_0003
A number of GIP derivatives according to the present invention has been tested for its hGIP and mGIP receptors antagonistic activity. Accordingly, in one embodiment, the GIP derivative of the invention is selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 19, compound No. 17, compound No. 21, compound No. 22, compound No. 49, compound No. 50, compound No. 52 and compound No. 53 of example 1 herein.
Pharmaceutical indications
Inhibitors of the human GIP receptor are known as useful candidates for treating or preventing obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome. Accordingly, one aspect of the invention concerns the GIP derivative according to the invention for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
Other compounds known for their usefulness in these indications are GLP-1 receptor agonists. Hence, in a further aspect of the invention it concerns the GIP derivative according to the invention in combination with a GLP-1 receptor agonist for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome. In one embodiment, the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide. In a further embodiment, the GLP-1 receptor agonist is semaglutide.
Pharmaceutical compositions
Pharmaceutical compositions comprising a derivative of the invention or a pharmaceutically acceptable salt, or amide thereof, and a pharmaceutically acceptable excipient may be prepared as is known in the art. The term "excipient" broadly refers to any component other than the active therapeutic ingredient(s). The excipient may be an inert substance, an inactive substance, and/or a not medicinally active substance.
The formulation of pharmaceutically active ingredients with various excipients is known in the art, see e.g. Remington: The Science and Practice of Pharmacy (e.g. 19th edition (1995), and any later editions).
The pharmaceutical composition comprising the derivative of the invention may be of several dosage forms, e.g. a solution, a suspension, a tablet, and a capsule. The pharmaceutical composition comprising the derivative of the invention may be administered to a patient in need thereof at several sites, e.g. at topical sites such as skin or mucosal sites; at sites which bypass absorption such as in an artery, in a vein, or in the heart; and at sites which involve absorption, such as in the skin, under the skin, in a muscle, orally, or in the abdomen.
In one embodiment, the composition comprising the derivative of the invention is an injectable composition comprising GIP derivatives of the present invention can be prepared using the conventional techniques of the pharmaceutical industry which involve dissolving and mixing the ingredients as appropriate to give the desired end product. Thus, according to one procedure, a GIP derivative of this invention is dissolved in a suitable buffer at a suitable pH so precipitation is minimised or avoided. The injectable composition is made sterile, for example, by sterile filtration.
In one embodiment, the composition comprising the derivative of the invention is a solid formulation, e.g. a freeze-dried or spray-dried composition, which may be used as is, or whereto the physician or the patient adds solvents, and/or diluents prior to use. In one embodiment, the pharmaceutical composition is in the form of a tablet.
A composition may be a stabilised formulation. The term “stabilised formulation” refers to a formulation with increased physical and/or chemical stability, preferably both. In general, a formulation must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
In view of the combination use discussed above, a pharmaceutical composition may also be a dosage form combining a GIP derivative of the invention and a GLP-1 receptor agonist. Accordingly, in one aspect the present invention concerns a dosage form comprising a combination of a GIP derivative according to the invention and a GLP-1 receptor agonist, in terms of loose-dose combination and fixed-dose combination therapy of the GIP derivatives and the GLP-1 receptor agonists. In one embodiment, the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide. In a further embodiment, the GLP-1 receptor agonist is semaglutide.
The two active components may also be in separate dosage forms. Accordingly, in another aspect, the invention concerns a kit-of-parts comprising a first dosage form comprising a GIP derivative according to the invention and a second dosage form comprising a GLP-1 receptor agonist. In one embodiment, the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide. In a further embodiment, the GLP-1 receptor agonist is semaglutide. Pharmaceutically acceptable salts
In some embodiments, the derivatives as described herein are in the form of a pharmaceutically acceptable salt. Salts are e.g. formed by a chemical reaction between a base and an acid, e.g.: 2NH3 + H2SO4 — > (NH^SC . The salt may be a basic salt, an acid salt, or it may be neither (i.e. a neutral salt). Basic salts produce hydroxide ions and acid salts hydronium ions in water. The salts of the derivatives may be formed with added cations or anions between anionic or cationic groups, respectively. These groups may be situated in the peptide and/or in the substituent of the derivatives. Non-limiting examples of anionic groups include any free carboxylic acid groups in the substituent, if any, as well as in the peptide. The peptide may include a free carboxylic acid group at the C-terminus, if present, as well as any free carboxylic acid group of internal acidic amino acid residues such as aspartic acid and glutamic acid.
Non-limiting examples of cationic groups include any free amino groups in the substituent, if any, as well as in the peptide. The peptide may include a free amino group at the N-terminus, if present, as well as any free imidazole or amino group of internal basic amino acid residues such as histidine, arginine, and lysine.
Production processes
The production of peptides like hGIP(1-31) and hGIP(1-42) analogues is well known in the art.
The GIP derivatives of the invention (or fragments thereof) may for instance be produced by classical peptide synthesis, e.g., solid phase peptide synthesis using t-Boc or Fmoc chemistry or other well established techniques, see, e.g., Greene and Wuts, “Protective Groups in Organic Synthesis”, John Wiley & Sons, 1999, Florencio Zaragoza Dorwald, “Organic Synthesis on solid Phase”, Wiley-VCH Verlag GmbH, 2000, and moc Solid Phase Peptide Synthesis”, Edited by W.C. Chan and P.D. White, Oxford University Press, 2000.
Also, or alternatively, they may be produced by recombinant methods, viz. by culturing a host cell containing a DNA sequence encoding the analogue and capable of expressing the peptide in a suitable nutrient medium under conditions permitting the expression of the peptide. Non-limiting examples of host cells suitable for expression of these peptides are: Escherichia coli, Saccharomyces cerevisiae, as well as mammalian BHK or CHO cell lines.
Those derivatives of the invention which include non-natural amino acids and/or a covalently attached N-terminal mono- or dipeptide mimetic may e.g. be produced as described in the experimental part. Or see e.g., Hodgson et al: "The synthesis of peptides and proteins containing non-natural amino acids", Chemical Society Reviews, vol. 33, no. 7 (2004), p. 422-430.
Specific examples of methods of preparing a number of the derivatives of the invention are included in the experimental part.
PARTICULAR EMBODIMENTS
The invention may be further described by the following non-limiting embodiments:
Embodiment 1:
A glucose-dependent insulinotropic peptide (GIP) derivative comprising a GIP analogue comprising Formula I, II, or III:
Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxxso-Xxxsi (I), (SEQ ID NO: 1) Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (II), (SEQ ID NO: 2)
Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (III), (SEQ ID NO: 3) wherein Xxxs is Glu or absent, Xxx4 is Gly or absent, Xxxs is Thr or absent, Xxxso is Lys or absent, and Xxxsi is Gly or absent; wherein said GIP analogue of Formula I or II comprises a modified lysine in one of positions 10-32 and wherein said GIP analogue of Formula III comprises a modified lysine in position 43, said modified lysine comprising a modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine, the modifying group being defined by A-B-C-, wherein A- is a fatty acid and B-C- is a linker, which may be absent or present; and wherein the GIP analogue has a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGI P(1 -31 ) , or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
Embodiment 2: A glucose-dependent insulinotropic peptide (GIP) derivative comprising a GIP analogue comprising Formula I, II, or III:
Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxxso-Xxxsi (I), (SEQ ID NO: 1) Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (II), (SEQ ID NO: 2) Xxx3-Xxx4-Xxx5-Phe-lle-Ser-Asp-Tyr-Ser-lle-Ala-Met-Asp-Lys-lle-His-Gln-Gln-Asp-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (III), (SEQ ID NO: 3) wherein Xxxs is Glu or absent, Xxx4 is Gly or absent, Xxxs is Thr or absent, Xxxso is Lys or absent, and Xxxsi is Gly or absent; wherein said GIP analogue of Formula I or II comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula III comprises a modified lysine in position 43, said modified lysine comprising a modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine, the modifying group being defined by A-B-C-, wherein A- is a fatty acid and B-C- is a linker, which may be absent or present; and wherein the GIP analogue has a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGI P(1 -31 ) , or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
Embodiment 3:
The GIP derivative according to Embodiment 2, wherein the modified lysine of the GIP analogue is in one of positions 11 and 10.
Embodiment 4:
The GIP derivative according to any one of Embodiments 2-3, wherein the modified lysine is in position 11.
Embodiment 5:
The GIP derivative according to any one of Embodiments 2-3, wherein the modified lysine is in position 10. Embodiment 6:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I.
Embodiment 7:
The GIP derivative according to Embodiment 2, wherein the modified lysine of the GIP analogue is in position 43.
Embodiment 8:
The GIP derivative according to Embodiments 1-2 or 7, wherein the GIP analogue is of Formula III.
Embodiment 9:
The GIP derivative according to any one of the preceding Embodiments, wherein Xxxs is absent.
Embodiment 10:
The GIP derivative according to any one of the preceding Embodiments, wherein Xxxs and Xxx4 are absent, such as wherein Xxxs, Xxx4, and Xxxs are absent.
Embodiment 11 :
The GIP derivative according to any one of the preceding Embodiments, wherein Met in position 14 is substituted by Leu, lie or Nle.
Embodiment 12:
The GIP derivative according to any one of the preceding Embodiments, wherein Met in position 14 is substituted by Leu.
Embodiment 13:
The GIP derivative according to any one of the preceding Embodiments, wherein His in position 18 is substituted by Arg, Orn or homoArg.
Embodiment 14:
The GIP derivative according to any one of the preceding Embodiments, wherein His in position 18 is substituted by Arg. Embodiment 15:
The GIP derivative according to any one of the preceding Embodiments, wherein Asp in position 21 is substituted by Glu or homoGlu.
Embodiment 16:
The GIP derivative according to any one of the preceding Embodiments, wherein Asp in position 21 is substituted by Glu.
Embodiment 17:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of formula I, wherein the modified lysine is in position 11 , Xxx3 and Xxx4 are absent, Xxx3o is Lys, Xxxsi is Gly, and the N-terminal end is substituted with an a-acetylate.
Embodiment 18:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I or II, wherein the modified lysine is in position 10, Xxx3 and Xxx4 are absent, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a-acetylate.
Embodiment 19:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula III, wherein the modified lysine is in position 43, Xxx3 and Xxx4 are absent, and the N-terminal end is substituted with an a-acetylate.
Embodiment 20:
The GIP derivative according to any one of the preceding Embodiments, wherein Xxx3, Xxx4, and Xxxs are absent and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
Embodiment 21 :
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I, wherein the modified lysine is in position 11 , Xxx3, Xxx4, and Xxxs are absent, Xxx3o is Lys or absent, Xxx3i is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
Embodiment 22: The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula I or II, wherein the modified lysine is in position 10, Xxxs, Xxx4, and Xxxs are absent, Xxxso is Lys or absent, Xxxsi is Gly or absent, and Phe in position 6 is substituted by L-3-phenyllactic acid (Pla).
Embodiment 23:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue has a maximum of 8 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31).
Embodiment 24:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue has a maximum of 7 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31).
Embodiment 25:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue has a maximum of 6 amino acid modifications as compared to positions 3 to 31 of hGIP(1-31).
Embodiment 26:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a straight-chain fatty acid.
Embodiment 27:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a C14 to C18 fatty acid, such as a C16 fatty acid.
Embodiment 28:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a saturated fatty acid.
Embodiment 29:
The GIP derivative according to any one of the preceding Embodiments, wherein A is an unsaturated fatty acid. Embodiment 30:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a mono-unsaturated fatty acid.
Embodiment 31 :
The GIP derivative according to any one of the preceding Embodiments, wherein B is an amino acid linker.
Embodiment 32:
The GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp.
Embodiment 33:
The GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu and y-Glu-y-Glu.
Embodiment 34:
The GIP derivative according to any one of the preceding Embodiments, wherein B is y-Glu.
Embodiment 35:
The GIP derivative according to any one of the preceding Embodiments, wherein B is absent.
Embodiment 36:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent or selected from AEEA2 and AEEA.
Embodiment 37:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent or AEEA2.
Embodiment 38:
The GIP derivative according to any one of the preceding Embodiments, wherein C is AEEA2. Embodiment 39:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent.
Embodiment 40:
The GIP derivative according to any one of the preceding Embodiments, wherein B-C is y- Glu and A is a C16 fatty acid.
Embodiment 41 :
The GIP derivative according to any one of the preceding Embodiments, wherein A is palmitic acid.
Embodiment 42:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 6, compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, compound No. 21 , compound No. 22, compound No. 49 and compound No. 50 as shown in example 1 herein.
Embodiment 43:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, compound No. 21 , and compound No. 22 as shown in example 1 herein.
Embodiment 44:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, and compound No. 21 as shown in example 1 herein.
Embodiment 45:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17 and compound No. 19 as shown in example 1 herein.
Embodiment 46: The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, and compound No. 19 as shown in example 1 herein.
Embodiment 47:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, and compound No. 9 as shown in example 1 herein.
Embodiment 48:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7 and compound No. 8 as shown in example 1 herein.
Embodiment 49:
The GIP derivative according to any one of the preceding Embodiments which is compound No. 7 as shown in example 1 herein.
Embodiment 50:
The GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor.
Embodiment 51 :
The GIP derivative according to any one of the preceding Embodiments which binds to the human GIP receptor.
Embodiment 52:
A pharmaceutical composition comprising a derivative according to any one of the preceding embodiments, and at least one pharmaceutically acceptable excipient.
Embodiment 53:
A pharmaceutical composition comprising a derivative according to any one of embodiments 1-51 , a GLP-1 receptor agonist, and at least one pharmaceutically acceptable excipient.
Embodiment 54: The pharmaceutical composition according to embodiment 53, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 55:
The pharmaceutical composition according to embodiments 53-54, wherein the GLP-1 receptor agonist is semaglutide.
Embodiment 56:
The pharmaceutical composition according to any one of embodiments 52-55 for use as a medicament.
Embodiment 57:
The GIP derivative according to any one of Embodiments 1-51 for use as a medicament.
Embodiment 58:
The GIP derivative according to any one of Embodiments 1-51 for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
Embodiment 59:
The GIP derivative for use according to Embodiment 58 in combination with a GLP-1 receptor agonist.
Embodiment 60:
The GIP derivative for use according to Embodiment 59, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 61 :
The GIP derivative for use according to Embodiment 60, wherein the GLP-1 receptor agonist is semaglutide.
Embodiment 62: A dosage form comprising a combination of a GIP derivative according to any one of Embodiments 1 to 51 and a GLP-1 receptor agonist.
Embodiment 63:
The dosage form according to Embodiment 62, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 64:
The dosage form according to Embodiment 63, wherein the GLP-1 receptor agonist is semaglutide.
Embodiment 65:
A kit-of-parts comprising a first dosage form comprising a GIP derivative according to any one of Embodiments 1 to 51 and a second dosage form comprising a GLP-1 receptor agonist.
Embodiment 66:
The kits-of-parts according to Embodiment 67, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 67:
The kits-of-parts according to Embodiment 66, wherein the GLP-1 receptor agonist is semaglutide.
Embodiment 68:
Use of the GIP derivative according to any one of Embodiments 1-51 for the manufacture of a medicament for the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
Embodiment 69:
A method of prevention and/or treatment of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome comprising administering a pharmaceutically active amount of the derivative according to any one of embodiments 1-51.
Embodiment 70:
The method according to embodiment 69 administering the derivative according to any one of embodiments in combination with a pharmaceutically active amount of a GLP-1 receptor agonist.
Embodiment 71 :
The method according to embodiment 70, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 72:
The method according to embodiment 71, wherein the GLP-1 receptor agonist is semaglutide.
ADDITIONAL PARTICULAR EMBODIMENTS:
Embodiment 1:
A glucose-dependent insulinotropic peptide (GIP) derivative comprising of a GIP analogue comprising Formula la, Ila, or Illa and a modifying group, wherein Formula la, Ila, and Illa are defined by:
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxx3o-Xxx3i (la), (SEQ ID NO: 46) Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (Ila), (SEQ ID NO: 47)
Xxx5-Xxx6-lle-Ser-Asp-Tyr-Ser-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val-Asn- Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (Illa), (SEQ ID NO: 48) wherein Xxxs is Thr or absent; Xxxe is Phe or L-3-phenyllactic acid (Pla); Xxx is Tyr or Lys; Xxxn is Ser or Lys; Xxxi4 is Met, Leu, lie or Nle; Xxxis is His, Arg, Orn, homoArg; XXX21 is Asp, Glu, or homoGlu; Xxx3o is Lys or absent; and Xxx3i is Gly or absent; provided that when Xxxe is Pla, then Xxxs is absent; wherein said GIP analogue of Formula la or Ila comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula Illa comprises a modified lysine in position 43, said modified lysine comprises the modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine, the modifying group being defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present; and wherein the GIP analogue has a maximum of 10 amino acid modifications as compared to positions 3 to 31 of hGI P(1 -31 ) , such as maximum 8, 7 or 6 modifications compared to positions 3 to 31 of hGIP(1-31); or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
Embodiment 2:
A glucose-dependent insulinotropic peptide (GIP) derivative consisting of a GIP analogue of Formula la, Ila, or Illa and a modifying group, wherein Formula la, Ila, and Illa are defined by:
Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-Gln-Xxx3o-Xxx3i (la), (SEQ ID NO: 46) Xxx5-Xxx6-lle-Ser-Asp-Xxxio-Xxxii-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn (Ila), (SEQ ID NO: 47)
Xxx5-Xxx6-lle-Ser-Asp-Tyr-Ser-lle-Ala-Xxxi4-Asp-Lys-lle-Xxxi8-Gln-Gln-Xxx2i-Phe-Val-Asn- Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-lle-Thr-GIn-Lys (Illa), (SEQ ID NO: 48) wherein Xxxs is Thr or absent, Xxxe is Phe or L-3-phenyllactic acid (Pla), Xxx is Tyr or Lys; Xxxn is Ser or Lys; Xxxi4 is Met, Leu, lie or Nle; Xxxis is His, Arg, Orn, homoArg; XXX21 is Asp, Glu, or homoGlu; Xxx3o is Lys or absent, and Xxx3i is Gly or absent; provided that when Xxxe is Pla, then Xxxs is absent; wherein said GIP analogue of Formula la or Ila comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula Illa comprises a modified lysine in position 43, said modified lysine comprises the modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine, the modifying group being defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present; or a pharmaceutically acceptable salt, amide, or a-N acetylate thereof.
Embodiment 3:
The glucose-dependent insulinotropic peptide (GIP) derivative according to embodiments 1-2 wherein wherein Xxxs is Thr or absent, Xxxe is Phe or L-3-phenyllactic acid (Pla), Xxx is Tyr or Lys, Xxxn is Ser or Lys, Xxxi4 is Leu, lie or Nle, Xxxis is Arg, Orn, homoArg, XXX21 is Glu or homoGlu, Xxxso is Lys or absent, and Xxxsi is Gly or absent; provided that when Xxxe is Pla, then Xxxs is absent.
Embodiment 4:
The GIP derivative according to any one of the preceding embodiments wherein Xxxi4 is Leu.
Embodiment 5:
The GIP derivative according to any one of the preceding embodiments wherein Xxxis is His or Arg.
Embodiment 6:
The GIP derivative according to any one of the preceding embodiments wherein Xxxi8 is Arg.
Embodiment 7:
The GIP derivative according to any one of the preceding embodiments wherein Xxx2i is Glu.
Embodiment 8:
The GIP derivative according to any one of the preceding embodiments wherein Xxxi4 is Leu, Xxxis is Arg and Xxx2i is Glu.
Embodiment 9:
The GIP derivative according to any one of the preceding embodiments, wherein the modified lysine of the GIP analogue is in one of positions 11 and 10.
Embodiment 10: The GIP derivative according to any one of the preceding embodiments, wherein the GIP analogue is of Formula la or Ila.
Embodiment 11 :
The GIP derivative according to any one of the preceding Embodiment, wherein the modified lysine is in position 11.
Embodiment 12:
The GIP derivative according to any one of the preceding embodiments, wherein the GIP analogue is of Formula la or Ila, wherein Xxx is Tyr and Xxxn is Lys.
Embodiment 13:
The GIP derivative according to any one of Embodiments 1-10, wherein the modified lysine is in position 10.
Embodiment 14:
The GIP derivative according to any one of embodiments 1-10 or 13, wherein the GIP analogue is of Formula la or Ila, wherein Xxx is Lys and Xxxn is Ser.
Embodiment 15:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula la.
Embodiment 16:
The GIP derivative according to any one of Embodiments 1-8, wherein the modified lysine of the GIP analogue is in position 43.
Embodiment 17:
The GIP derivative according to any one of Embodiments 1-8 or 16, wherein the GIP analogue is of Formula Illa.
Embodiment 18:
The GIP derivative according to any one of the preceding Embodiments, wherein Xxxs is absent. Embodiment 19:
The GIP derivative according to any one of Embodiments 1-17, wherein Xxxs is Thr.
Embodiment 20:
The GIP derivative according to any one of the preceding Embodiments, wherein Xxxe is Phe.
Embodiment 21 :
The GIP derivative according to any one of Embodiments 1-18, wherein Xxx6 is L-3- phenyllactic acid (Pla).
Embodiment 22:
The GIP derivative according to any one of Embodiments 1-12, 15, 18-21 wherein Xxx is Tyr.
Embodiment 23:
The GIP derivative according to any one of Embodiments 1-10, 13-15, 18-21 , wherein Xxx is Lys.
Embodiment 24:
The GIP derivative according to any one of Embodiments 1-12, 15, 18-22, wherein Xxxn is Lys.
Embodiment 25:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of formula la, wherein the modified lysine is in position 11 , Xxx is Tyr, Xxxn is Lys, Xxxso is Lys, Xxxsi is Gly, and the N-terminal end is substituted with an a-acetylate.
Embodiment 26:
The GIP derivative according to embodiment 25, wherein Xxxi4 is Leu, Xxxis is Arg, and XXX21 is Glu.
Embodiment 27:
The GIP derivative according to any one of the Embodiments 1-24, wherein the GIP analogue is of Formula la or Ila, wherein the modified lysine is in position 10, Xxx is Lys, Xxxn is Ser, Xxx3o is Lys, Xxx3i is Gly, and the N-terminal end is substituted with an a- acetylate.
Embodiment 28:
The GIP derivative according to embodiment 27, wherein Xxxi4 is Leu, Xxxi8 is Arg, and Xxx2i is Glu.
Embodiment 29:
The GIP derivative according to any one of Embodiments 1-24, wherein the GIP analogue is of Formula Illa, wherein the modified lysine is in position 43, and the N-terminal end is substituted with an a-acetylate.
Embodiment 30:
The GIP derivative according to Embodiment 29, wherein Xxxi4 is Leu, Xxxis is Arg, and XXX21 is Glu.
Embodiment 31 :
The GIP derivative according to any one of the preceding Embodiments, wherein Xxx5 are absent and Xxx6 is L-3-phenyllactic acid (Pla).
Embodiment 32:
The GIP derivative according to any one of the preceding Embodiments, wherein the GIP analogue is of Formula la, wherein the modified lysine is in position 11 , Xxxs is absent, Xxx8 is L-3-phenyllactic acid (Pla), Xxx is Tyr, Xxxn is Lys, Xxx3o is Lys or absent, Xxx3i is Gly or absent.
Embodiment 33:
The GIP derivative according to any one of Embodiments 31-32, wherein Xxxi4 is Leu, Xxxis is Arg, and Xxx2i is Glu.
Embodiment 34:
The GIP derivative according to any one of Embodiments 1-31 , wherein the GIP analogue is of Formula la or Ila, wherein the modified lysine is in position 10, Xxxs is absent, Xxx8 is L-3- phenyllactic acid (Pla), Xxx is Lys, Xxxn is Ser, Xxx3o is Lys or absent, Xxx3i is Gly or absent. Embodiment 35:
The GIP derivative according to Embodiment 34, wherein Xxxi4 is Leu, Xxxis is Arg, and XXX21 is Glu.
Embodiment 36:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a straight-chain fatty acid of formula -C=(O)-(CH2)n-CH3, where n is an integer between 2-26, such as 8-20, preferably n is 12, 14 or 16.
Embodiment 37:
The GIP derivative according to embodiment 36, wherein n is 14.
Embodiment 38:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a C14 to C18 mono fatty acid, such as a C16 mono fatty acid.
Embodiment 39:
The GIP derivative according to any one of the preceding Embodiments, wherein A is a saturated fatty acid.
Embodiment 40:
The GIP derivative according to any one of the preceding Embodiments, wherein B is an amino acid linker.
Embodiment 41 :
The GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu-y-Glu, y-Glu, Glu, Lys, and Asp.
Embodiment 42:
The GIP derivative according to any one of the preceding Embodiments, wherein B is selected from y-Glu and y-Glu-y-Glu.
Embodiment 43:
The GIP derivative according to any one of the preceding Embodiments, wherein B is y-Glu. Embodiment 44:
The GIP derivative according to any one of the preceding Embodiments, wherein B is absent.
Embodiment 45:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent or selected from AEEA2 and AEEA.
Embodiment 46:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent or AEEA2.
Embodiment 47:
The GIP derivative according to any one of the preceding Embodiments, wherein C is AEEA2.
Embodiment 48:
The GIP derivative according to any one of the preceding Embodiments, wherein C is absent.
Embodiment 49:
The GIP derivative according to any one of the preceding Embodiments, wherein B-C is y- Glu and A is a C16 mono fatty acid.
Embodiment 50:
The GIP derivative according to any one of the preceding Embodiments, wherein A is palmitic acid.
Embodiment 51 :
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 6, compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, compound No. 21 , compound No. 22, compound No. 23, compound No. 25, compound No. 28, compound No. 49 and compound No. 50, compound No. 52, compound No. 53 as shown in example 1 herein. Embodiment 52:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, compound No. 21 , and compound No. 22 as shown in example 1 herein.
Embodiment 53:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17, compound No. 19, and compound No. 21 as shown in example 1 herein.
Embodiment 54:
The GIP derivative according to any one of the preceding Embodiments selected from the group consisting of: compound No. 7, compound No. 8, compound No. 9, compound No. 17 and compound No. 19 as shown in example 1 herein.
Embodiment 55:
The GIP derivative according to any one of the preceding Embodiments which is compound No. 7 as shown in example 1 herein.
Embodiment 56:
The GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor.
Embodiment 57:
The GIP derivative according to any one of the preceding Embodiments which is an antagonist at the human GIP receptor and an antagonist at the mouse GIP receptor.
Embodiment 58:
A pharmaceutical composition comprising a derivative according to any one of the preceding embodiments, and at least one pharmaceutically acceptable excipient.
Embodiment 59:
A pharmaceutical composition comprising a derivative according to any one of embodiments 1-57, a GLP-1 receptor agonist, and at least one pharmaceutically acceptable excipient. Embodiment 60:
The pharmaceutical composition according to embodiment 59, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 61 :
The pharmaceutical composition according to embodiments 59-60, wherein the GLP-1 receptor agonist is semaglutide.
Embodiment 62:
The pharmaceutical composition according to any one of embodiments 58-61 for use as a medicament.
Embodiment 63:
The GIP derivative according to any one of Embodiments 1-57 for use as a medicament.
Embodiment 64:
The GIP derivative according to any one of Embodiments 1-57 for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
Embodiment 65:
The GIP derivative for use according to Embodiment 64 in combination with a GLP-1 receptor agonist.
Embodiment 66:
The GIP derivative for use according to Embodiment 65, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 67:
The GIP derivative for use according to Embodiment 65-66, wherein the GLP-1 receptor agonist is semaglutide. Embodiment 68:
Use of the GIP derivative according to any one of Embodiments 1-574 for the manufacture of a medicament for the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome.
Embodiment 69:
A method of prevention and/or treatment of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome comprising administering a pharmaceutically active amount of the derivative according to any one of embodiments 1-57.
Embodiment 70:
The method according to embodiment 69 administering the derivative according to any one of embodiments in combination with a pharmaceutically active amount of a GLP-1 receptor agonist.
Embodiment 71 :
The method according to embodiment 70, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, semaglutide, exenatide, dulaglutide, lixisenatide, taspoglutide, and albiglutide.
Embodiment 72:
The method according to embodiment 70-71, wherein the GLP-1 receptor agonist is semaglutide.
EXAMPLES
This experimental part starts with a list of abbreviations and is followed by a section including general methods for synthesizing and characterising the GIP antagonists as described herein. Then follows a number of examples which relate to the preparation of the specific GIP antagonists of the invention and selected comparative compounds, and at the end a number of examples relating to the activity and properties of the GIP antagonists have been included.
Examples serve to illustrate the invention. List of Abbreviations yE: y-glutamic acid
Aib: a-aminoisobutyric acid
6-CI-HOBt: 6-chloro-1 -hydroxybenzotriazole
ACN: Acetonitrile
AEEA: 8-amino-3,6-dioxaoctanoic acid
BW: Body weight
C16: Palmitic acid
C18-OH: Octadecanedioic acid
DCM: Dichloromethane
DEPBT: Diethyl 3,4-dihydro-4-oxo-1 ,2,3-benzotriazin-3-yl phosphate
DIG: N, N-Diisopropylcarbodiimide
DIEA/DIPEA: N,N-Diisopropylethylamine
DIO: Diet-induced obese
DMF: Dimethylformamide
DODT : 3,6-dioxa-1 ,8-octanedithiol
Fmoc: Fluorenylmethyloxycarbonyl
GIP: Glucose-dependent insulinotropic peptide
GIPR: Glucose-dependent insulinotropic peptide receptor
GLP-1: Glucagon-like peptide 1
HFD: High fat diet
HFIP: Hexafluoroisopropanol hGIP: Human glucose-dependent insulinotropic peptide hGIPR: Human glucose-dependent insulinotropic peptide receptor
HPLC: High-performance liquid chromatography
IPGTT: Intra-peritoneal glucose tolerance test mGIP: Mouse glucose-dependent insulinotropic peptide mGIPR: Mouse glucose-dependent insulinotropic peptide receptor
Mtt: 4-Methyltrityl
OGTT: Oral glucose tolerance test
OxymaPure: ethyl 2-cyano-2-(hydroxyimino)acetate
Pla: L-(-)-3-phenyllactic acid I L-3-phenyllactic acid I S-3-phenyllactic acid tBu: tert-butyl
TFA: Trifluoroacetic acid
Tis: Triisopropylsilane General Methods of Peptide Synthesis
Generally, peptides were synthesized by automated Fmoc/tBu solid-phase methodology employing a Symphony peptide synthesizer (Gyros Protein Technologies, Tucson, AZ) and Applied Biosystems ABI 433A peptide synthesizer, starting with preloaded Wang resin (AAPPtec, Louisville, KY; Novabiochem, San Diego, CA) for acid C-terminal peptides, and H-Rink Amide ChemMatrix® (PCAS BioMatrix Inc, Saint-Jean-sur-Richelieu, Quebec, Canada J3B 8J8) for C-terminal amide peptides. All Fmoc-amino acids were coupling with 6-CI-HOBt/DIC or OxymaPure/DIC activations in DMF. All common Fmoc- amino acids, 6-CI-HOBt, OxymaPure and DIC were purchased from Midwest Biotech (Fisher, IN), AAPPtec and Gyros Protein Technologies. The Fmoc were removed by 20% piperidine in DMF. The N-terminal acetylation was performed on resin in the presence of tenfold excess of acetic anhydride/DIEA in DCM for 1 h. The N-terminal L-3-Phenyllactic acid (Pla) was coupled manually in 5-fold excess by DEPBT in DMF/DIEA. For the peptide with fatty acids, the residues were pre-incorporated with Fmoc-L-Lys(Mtt) - OH, after peptide assembling done and the N-terminal acetylation, the Mtt was deprotected with 1 - 2%TFA/5%Tis in DCM or with 30% HFIP/5% Tis in DCM.
The linkage amino acid of y-glutamic acid was coupled by Fmoc-Glu-OtBu; AEEA was coupled by Fmoc-8-amino-3,6-dioxaoctanoic acid/Fmoc-AEEA-OH (AAPPTec, Louisville, KY). Palmitic acid was coupled by 5-fold excess with DEPBT/DIEA in DMF for about 4h; the octadecanedioic acid acylation was done by coupling octadecanedioic acid mono-tert-butyl ester (Novo Nordisk, Malov, DK) with 5-fold excess DEPBT/DIEA in DMF for about 4h. Completed peptidyl resins were cleaved by standard TFA cleavage containing 5% Tis and 5% H2O, added 2.5% 2-Mercaptoethanol or the odourless thiol scavenger DODT (Sigma-Aldrich, St. Louis, MS) for the cysteine and methionine residue containing peptides for 2 h with agitation.
The cleaved peptides in TFA were precipitated with ether, centrifuged and washed twice with ether, the crude peptides were dissolved in 20% ACN containing 2% acetic acid and injected to a Luna 19 x 250nm/10 pm C8 column (Phenomenex, Torrance, CA) to purify with 0.1% TFA/ACN eluent solvents on the Waters 2545 preparative HPLC system.
General LC-MS Methods of Peptide Detection and Characterisation
Peptide molecular weights characterization were measured by liquid chromatography-mass spectrometry on an Agilent 1260 lnfinity/6120 Quadrupole instrument with a Kinetex C8 column. Eluent A is water with 0.05% TFA and eluent B is 10% water, 90% ACN and 0.05% TFA. Eluent gradient methods are Method A: a gradient of 10%-80% eluent B in 10 min with 1 ml/min flow rate; Method B: a gradient of 20%-100% eluent B in 10 min with 1 ml/min flow rate; Method C: a gradient of 30%-80% eluent B in 3.5 min with 4 ml/min flow rate; Method D: a gradient of 40%-100% eluent B in 3.5 min with 4 ml/min flow rate.
Pharmacological Methods
The utility of the derivatives of the present invention as pharmacologically active agents e.g. in the reduction of weight gain and treatment of obesity in mammals, such as humans may be demonstrated by the activity of the antagonists in conventional assays and in the in vitro and in vivo assays described below.
Such assays also provide a means whereby the activities of the antagonists of the invention can be compared with activities of known compounds
General methods of measuring in vitro receptor binding
The receptor binding assay may be performed at the desired receptors, herein the human GIP receptors (“hGIPR”) and mouse GIP receptors (“mGIPR”) are tested.
GIP receptors were inserted in pcDNA3 vectors under the control of a CMV promotor with Geneticin as the selectable marker. The pGL4.29[luc2P/CRE/Hygro] vector from Promega (USA, E8471) contains a cAMP response element (CRE), that drives the transcription of the luciferase (luc) reporter gene luc2P. Luc2P is a synthetically-derived luciferase sequence optimized for expression in mammalian cells. Upon transfection of Baby Hamster Kidney (BHK) cells with these plasmids stable clones expressing receptor as well as reporter gene were selected in culture media containing hygromycin and geneticin (G418). The clones used were: Human (hGIPR/BHK CRELuc2p clone#5), mouse (mGIPR/BHK CRELuc2p clone#3).
Binding of peptide antagonists to the full length GIP receptor was assessed in whole cells with competitive binding using radiolabelled human GIP (125l-NNC0090-0554, produced at Novo Nordisk A/S). BHK cells stably expressing the GIP receptor were seeded in poly-D- lysine coated white 96-well plates with clear bottom (Corning, cat. no. 354651). 5000-10000 cells are seeded pr. well in DMEM medium (cat. no 61965-026) supplemented with Geneticin (Gibco, cat. no 10131-027), Hygromycin B (Gibco cat no 10687-010) and 10% fetal bovine serum (FBS) (Gibco, cat. no 16140-071). The following day the medium was removed, and cells were rinsed twice with HBSS (Gibco, cat. no 14025), and the binding assay was performed. The assay buffer consists of HBSS (Gibco, cat no 14025), 10mM HEPES (Gibco, cat. no 15630), 0.1 % pluronic F-68 (Gibco, cat. no 2404,) and 0.1 % ovalbumin (SigmaAldrich, cat. no A5503), and pH is adjusted to 7.4. GIP receptor peptide antagonists were diluted in assay buffer and tested in 10-fold dilutions from 10 pM downwards. To each well was added 50pl assay buffer, 25 pl peptide (or assay buffer) and 25 pl radiolabelled GIP (equivalent of ~60pM). Compounds were tested in duplicates. The plate was sealed and incubated at 4°C overnight. The supernatant was discarded, and the plates were washed twice with ice cold PBS (Gibco, cat. no 14040-091). The cells were lysed with addition of 50pl 0.1M NaOH followed by 5 minutes shaking. 100 pl microscint-40 (Perkin Elmer, cat. no 6013641) was added to each well, and after incubation at room temperature for 30 minutes the plates were counted in a scintillation counter (Topcount® NXT™ HTS from Packard). Nonlinear regression analysis on the output files was performed in the Windows program GraphPad Prism 7 (GraphPad software, USA) using the equation “log(inhibitor) vs response (three parameters)”. Data were reported as geometric mean of IC50 values with 95% Cl.
General methods of measuring in vitro functional potency
The ability of peptides to activate or antagonize incretin receptors was studied in vitro using firefly luciferase reporter gene assay designed to indirectly measure cAMP production. BHK-21 cells (ATCC CCL-10) were transfected with human or mouse GLP-1 or GIP receptor and firefly luciferase under control of cAMP-response element. The cells were grown in Dulbecco’s modified Eagle medium (DMEM, Thermo Fisher 10566-016) supplemented with 10% Fetal Bovine Serum (Thermo Fisher 10082147), 100 lU/ml penicillin, 100 pg/mL streptomycin, and 10 mM HEPES at 37°C, 5% CO2 and 90% humidity for 2-3 days. Sixteen to twenty hours prior to the experiment the cells were plated at 5x104 cells per well in 96 well Isoplate (Perkin-Elmer 6005040, Waltham, MA). The plate was gently washed with warm Hank’s Balanced Salt Solution pH 7.4 (14025092, Invitrogen, Carlsbad, CA) and filled with 0.1 ml/well of serial dilutions of peptides prepared in sterile DMEM containing 1% Ovalbumin (A5503, Sigma-Aldrich). For the antagonism experiments the peptides were premixed with EC90 concentration- (75 - 95% maximal signal) of native ligand. After 3h incubation at 37°C and 5% CO2 in humidified atmosphere, the plates were washed with warm Hank’s Balanced Salt Solution pH 7.4 and 0.1 ml/well of Steady Lite HTS luminescence substrate reagent (Perkin-Elmer, Waltham, MA) was added to each well. The plate was sealed and shaken 10 min at 800 rpm. Luminescence signal was measured on Perkin-Elmer Envision plate reader. The luminescence data were plotted against peptide concentrations and EC50 or IC50 values were calculated by using logistic nonlinear 3 parameter regression analysis in GraphPad Prism 7 (GraphPad Software, La Jolla, CA). General methods for pharmacokinetic study in mice
C57BL/6J mice (58% HFD diet, male, body weight 61-74 g), were given subcutaneous injections of compounds 7, 9 and 22 at a single dose of 500 nmol/kg; each time point dosed 4 mice (n = 4). Plasma was collected at time points of 5min, 30min, 1 h, 2h, 6h, 8h, 24h, 48h, 72h, 96h, and 120h. The last time point of compound 9 is 48h. To measure plasma concentration, standards of each peptide were prepared in mouse plasma ranging from 0.5 to 4000 nM. A protein precipitation was performed by organic solvent extraction with a 14 - fold dilution with methanol. The samples were centrifuged for 20 min at 4°C at 13000g- force. Supernatants of samples were collected and diluted 3-fold with 0.1% formic acid in water. The diluted samples were then subjected to LC - MS analysis.
LC-MS analyses were carried out on a Thermo Q Exactive HF mass spectrometer interfaced with a Vanquish LIPLC. LC separations were performed on an Acquity LIPLC BEH C18 1.7 pm, 1.0 x 50 mm column. Mobile phase A was composed of 0.1% formic acid in water and mobile phase B was composed of 0.1% formic acid in ACN. The LC flow rate was set to 0.4 pL/min using a gradient elution from 10 to 95% B over the course of 4.0 min. Selected ion monitoring was used for all molecules, where the 3+ ion 1212.3491 m/z were chosen for compound 7, the 3+ ion 1102.9574 m/z were chosen for compound 9, and the 5+ ion 1017.1634 m/z for compound 22, respectively. Plasma data were analyzed by noncompartmental methods with sparse sampling using Phoenix WinNonlinTM 8.3 (Certara USA, Inc., Princeton, NJ).
General methods for pharmacodynamic study in mice
All mice studies were performed in accordance with Institutional Animal Care and Use Committee guidelines at University of Cincinnati. Male C57BL/6J mice (Jackson Laboratories) were housed 4 per cage under 12 h/12 h light-dark cycle at 22°C with ad libitum access to water and 58% fat, high-sugar diet (D12331 , Research Diets) for ~12 weeks. Mice achieved body weight (BW) greater than 55 g on average and were subsequently assigned to treatment groups (n = 8 per group) which were normalized for body weight. Treatment groups received either vehicle, semaglutide (2nmol/kg/d), GIPR antagonist (500 or 1500nmol/kg/d), semaglutide + GIPR antagonist (2 + 500 or 1500nmol/kg/d; co-formulated single injection), or semaglutide + GIPR antibody (2nmol/kg/d + 30mg/kg/w; separate injections). Test compounds (200pM) were dissolved in a vehicle (pH 7.4) containing 0.05% polysorbate-80, 50 mM sodium phosphate, and 70 mM sodium chloride; test compounds were administered once daily (semaglutide and GIPR antagonist) for 27d, subcutaneously during the light cycle at a volume of 3.9|jL/gram BW. Body weight and food intake were measured prior to dosing every other day; the percent change in body was calculated for each mouse based on its initial body weight. Food intake was measured on a per cage basis (n = 2 cages/group). Oral glucose tolerance tests (OGTT) were performed on day 0 (1h post-compound injection) and 22 (24h post-compound injection) and an intraperitoneal (i.p.) glucose tolerance test (IPGTT) was performed on d 27 (24h post-compound injection). For IPGTT and OGTT tests, animals were fasted for 4 hours prior to the test with access to water. Baseline tail-vein blood glucose was measured, then mice were injected i.p. or gavaged orally with 2.5 g/kg of 20% glucose solution (12uL/g). Tail blood glucose levels were measured 0, 15, 30, 60, 90, and 120 minutes following the glucose load. An additional measure of 6h fasting insulin (Crystal Chem 90080), resistin (R&D Systems MRSN00), CTX (Novus Biologicals NBP2-69074) and total GIP (Crystal Chem 81527) levels were taken at day 27.
Example 1 : Synthesis of compounds
The peptide analogues and derivative of the present invention were synthesized according to the General Methods of Peptide Synthesis as describe above. Name, structure, and properties are shown below for each compound. SEQ ID NO’s are included for the peptide backbone.
Compound No. 1 (reference cpd): hGIP(3-30) amide (SEQ ID NO: 12)
H— E G T F I S D Y S I A M D K I H Q Q D F V N W L L A Q K-N H2
Formula Weight: 3297.7; Exact Mass: 3295.63; Formula: Cd„H,,RN,sO..S LCMS: [M+3H]3+: 1100.0; [M+4H]4+: 825.2; RT = 6.5min (Method A)
Compound No. 2 (reference cpd):
[M14L, H18R, D21E]-hGIP(3-30) amide (SEQ ID NO: 13)
H— E G T F I S D Y S I A L D K I R Q Q E F V N W L L A Q K-NH2
Formula Weight: 3312.7; Exact Mass: 3310.74; Formula: Cd„H„RN,QO..
LCMS: [M+3H]3+: 1105.0; [M+4H]4+: 828.7; RT = 1.68min (Method C)
Compound No. 3 (reference cpd):
[M14L, H18R, D21E]-hGIP(5-31) (SEQ ID NO: 14) 210081WO01 42 Formula Weight: 3184.6; Exact Mass: 3182.68; Formula: C147H227N37O42 LCMS: [M+2H]2+: 1593.7; [M+3H]3+: 1062.2; [M+4H]4+: 797.0; RT = 1.97min (Method C) 5 Compound No.4 (reference cpd): [M14L, H18R, D21E]-hGIP(6-31) (SEQ ID NO: 15) Formula Weight: 3083.5; Exact Mass: 3081.63; Formula: C143H220N36O40 LCMS: [M+2H]2+: 1542.6; [M+3H]3+: 1028.6; [M+4H]4+: 771.7; RT = 1.54min (Method C) 10 Compound No.5 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) (SEQ ID NO: 16)
Figure imgf000043_0002
Formula Weight: 3084.5; Exact Mass: 3082.61; Formula: C143H219N35O41 15 LCMS: [M+2H]2+: 1542.9; [M+3H]3+: 1029.0; RT = 1.96min (Method C) Compound No.6: [M14L, H18R, D21E]-hGIP(3-30) - K11( ^E-C16) amide (SEQ ID NO: 17)
Figure imgf000043_0001
20 Formula Weight: 3721.3; Exact Mass: 3719.07; Formula: C176H279N41O47 LCMS: [M+2H]2+: 1861.5; [M+3H]3+: 1241.1; [M+4H]4+: 931.2; RT = 6.37min (Method B) Compound No.7: [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 18) 210081WO01 43
Figure imgf000044_0001
Formula Weight: 3635.2; Exact Mass: 3633.02; Formula: C173H273N39O46 LCMS: [M+2H]2+: 1817.9; [M+3H]3+: 1212.6; [M+4H]4+: 909.8; RT = 6.88min (Method B) 5 Compound No.8: [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K11( ^E-C16) (SEQ ID NO: 19)
Figure imgf000044_0002
Formula Weight: 3493.1; Exact Mass: 3490.95; Formula: C167H263N37O44 LCMS: [M+2H]2+: 1746.9; [M+3H]3+: 1165.1; RT = 7.19min (Method B) 10 Compound No.9: [F6Pla, M14L, H18R, D21E]-hGIP(6-29) - K11( ^E-C16) amide (SEQ ID NO: 20)
Figure imgf000044_0003
Formula Weight: 3306.9; Exact Mass: 3304.85; Formula: C159H249N35O41 15 LCMS: [M+2H]2+: 1653.9; [M+3H]3+: 1102.8; RT = 2.51min (Method D) Compound No.10 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-29) - K11(AEEA2- ^E-C18-OH) amide (SEQ ID NO: 20) 210081WO01 44
Figure imgf000045_0001
Formula Weight: 3655.2; Exact Mass: 3653.00; Formula: C173H273N37O49 LCMS: [M+2H]2+: 1828.5; [M+3H]3+: 1219.3; RT = 6.55min (Method B) 5 Compound No.11 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K11(AEEA2- ^E-C18-OH) (SEQ ID NO: 19)
Figure imgf000045_0002
Formula Weight: 3841.4; Exact Mass: 3839.10; Formula: C181H287N39O52 LCMS: [M+2H]2+: 1921.3; [M+3H]3+: 1281.2; [M+4H]4+: 961.3; RT = 2.78min (Method C) 10 Compound No.12 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K11(Lys2- ^E-C18-OH) (SEQ ID NO: 19)
Figure imgf000045_0003
Formula Weight: 3807.5; Exact Mass: 3805.14; Formula: C181H289N41O48 15 LCMS: [M+2H]2+: 1904.2; [M+3H]3+: 1270.1; [M+4H]4+: 952.7; RT = 2.28min (Method C) 210081WO01 45 Compound No.13 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(6-31) - K11(AEEA2- ^E-C18-OH) (SEQ ID NO: 21)
Figure imgf000046_0001
Formula Weight: 3882.5; Exact Mass: 3880.13; Formula: C183H290N40O52 5 LCMS: [M+2H]2+: 1942.1; [M+3H]3+: 1294.9; [M+4H]4+: 971.5; RT = 6.12min (Method B) Compound No.14 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K11(AEEA2- ^E-C18-OH) (SEQ ID NO: 18)
Figure imgf000046_0002
10 Formula Weight: 3983.6; Exact Mass: 3981.18; Formula: C187H297N41O54 LCMS: [M+2H]2+: 1992.6; [M+3H]3+: 1328.6; [M+4H]4+: 996.8; RT = 6.05min (Method B) Compound No.15 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K11(AEEA2-C18-OH) (SEQ ID NO: 18) 15
Figure imgf000046_0003
Formula Weight: 3854.5; Exact Mass: 3852.13; Formula: C182H290N40O51 LCMS: [M+2H]2+: 1927.8; [M+3H]3+: 1285.6; [M+4H]4+: 964.5; RT = 2.67min (Method C) 210081WO01 46 Compound No.16 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K11(-C18-OH) (SEQ ID NO: 18)
Figure imgf000047_0003
5 Formula Weight: 3564.2; Exact Mass: 3561.99; Formula: C170H268N38O45 LCMS: [M+2H]2+: 1782.7; [M+3H]3+: 1188.8; RT = 2.86min (Method C) Compound No.17: Nα -Ac - hGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 22) 10
Figure imgf000047_0001
Formula Weight: 3620.2; Exact Mass: 3617.92; Formula: C171H264N38O46S LCMS: [M+2H]2+: 1810.5; [M+3H]3+: 1207.4; RT = 2.22 min (Method C) Compound No.18 (reference cpd): 15 Nα -Ac - hGIP(5-31) - K11(AEEA2- ^E-C18-OH) (SEQ ID NO: 22)
Figure imgf000047_0002
Formula Weight: 3968.6; Exact Mass: 3966.07; Formula: C185H288N40O54S LCMS: [M+2H]2+: 1984.9; [M+3H]3+: 1323.4; [M+4H]4+: 992.7; RT = 1.74 min (Method C) 210081WO01 47 Compound No.19: Nα -Ac - mGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 23)
Figure imgf000048_0001
5 Formula Weight: 3667.3; Exact Mass: 3664.70; Formula: C171H269N41O46S LCMS: [M+2H]2+: 1834.8; [M+3H]3+: 1223.2; RT = 6.87 min (Method B) Compound No.20 (reference cpd): Nα -Ac - mGIP(5-31) - K11(AEEA2- ^E-C18-OH) (SEQ ID NO: 23) 10
Figure imgf000048_0002
Formula Weight: 4015.6; Exact Mass: 4013.12; Formula: C185H293N43O54S LCMS: [M+3H]3+: 1339.2; [M+4H]4+: 1004.7; RT = 6.83 min (Method B) 15 Compound No.21: [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K10( ^E-C16) (SEQ ID NO: 24)
Figure imgf000048_0003
Formula Weight: 3559.2; Exact Mass: 3557.00; Formula: C167H269N39O46 210081WO01 48 LCMS: [M+2H]2+: 1780.2; [M+3H]3+: 1187.2; [M+4H]4+: 890.7; RT = 2.12min (Method D) Compound No.22: [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K10( ^E ^E-C16) amide (SEQ ID NO: 25) 5
Figure imgf000049_0001
Formula Weight: 5080.8; Exact Mass: 5077.78; Formula: C234H373N61O65 LCMS: [M+3H]3+: 1694.2; [M+4H]4+: 1271.0; [M+5H]5+: 1017.0; RT = 2.37 min (Method C) Compound No.23: 10 [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43( ^E ^E-C16) amide (SEQ ID NO: 26)
Figure imgf000049_0002
Formula Weight: 5244.0; Exact Mass: 5240.84; Formula: C243H382N62O67 LCMS: [M+3H]3+: 1748.6; [M+4H]4+: 1311.8; [M+5H]5+: 1049.8; RT = 1.59 min (Method D) 15 Compound No.24 [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) amide (SEQ ID NO: 27)
Figure imgf000049_0003
Formula Weight: 4619.2; Exact Mass: 4616.43; Formula: C211H326N58O59 LCMS: [M+3H]3+: 1540.4; [M+4H]4+: 1155.6; RT = 1.55 min (Method C) 20 Compound No.25: 210081WO01 49 [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K10( ^E ^E-C16) (SEQ ID NO: 28)
Figure imgf000050_0001
Formula Weight: 3546.1; Exact Mass: 3543.96; Formula: C166H266N38O47 LCMS: [M+2H]2+: 1774.0; [M+3H]3+: 1182.8; [M+4H]4+: 887.4; RT = 2.86 min (Method C) 5 Compound No.26 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K10( ^E ^E-C18-OH) (SEQ ID NO: 28)
Figure imgf000050_0002
Formula Weight: 3604.2; Exact Mass: 3601.96; Formula: C168H268N38O49 10 LCMS: [M+2H]2+: 1802.0; [M+3H]3+: 1202.2; RT = 2.51 min (Method C) Compound No.27 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31)-K10(AEEA2- ^E-C18-OH) (SEQ ID NO: 28)
Figure imgf000050_0003
15 Formula Weight: 3765.3; Exact Mass: 3763.07; Formula: C175H283N39O52 LCMS: [M+2H]2+: 1883.4; [M+3H]3+: 1256.0; [M+4H]4+: 942.0; RT = 2.59 min (Method C) Compound No.28: 210081WO01 50 [F6Pla6, M14L, H18R, D21E]-hGIP(6-42) - K10( ^E ^E-C16) amide (SEQ ID NO: 29) Formula Weight: 4938.7; Exact Mass: 4935.70; Formula: C228H363N59O63 LCMS: [M+3H]3+: 1647.2; [M+4H]4+: 1235.5; [M+5H]5+: 988.9; RT = 2.51 min (Method C) 5 Compound No.29 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K10(AEEA2- ^E-C18-OH) amide (SEQ ID NO: 30) Formula Weight: 5300.1; Exact Mass: 5296.89; Formula: C243H390N62O70 10 LCMS: [M+3H]3+: 1767.8; [M+4H]4+: 1325.8; [M+5H]5+: 1060.9; RT = 2.13 min (Method C) Compound No.30 (reference cpd): [Nα-Ac, M14L, H18R, D21E]-hGIP(5-42) - K11(AEEA2- ^E-C18-OH) amide (SEQ ID NO: 31) 15 Formula Weight: 5376.2; Exact Mass: 5372.92; Formula: C249H394N62O70 LCMS: [M+3H]3+ : 1792.6; [M+4H]4+: 1345.1; RT = 2.29 min (Method C) Compound No.31 (reference cpd): 210081WO01 51 [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K13(AEEA2- ^E-C18-OH) (SEQ ID NO: 32)
Figure imgf000052_0001
Formula Weight: 3857.4; Exact Mass: 3855.10; Formula: C181H287N39O53 LCMS: [M+2H]2+: 1928.2; [M+3H]3+: 1286.8; [M+4H]4+: 965.3; RT = 2.55 min (Method C) 5 Compound No.32 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K13(AEEA2- ^E-C18-OH) amide (SEQ ID NO: 33)
Figure imgf000052_0002
Formula Weight: 5392.2; Exact Mass: 5388.91; Formula: C249H394N62O71 10 LCMS: [M+3H]3+: 1798.3; [M+4H]4+: 1348.8; [M+5H]5+: 1079.2; RT = 2.18 min (Method C) Compound No.33: [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K16( ^E ^E-C16) (SEQ ID NO: 16)
Figure imgf000052_0003
15 Formula Weight: 3581.1; Exact Mass: 3578.93; Formula: C169H263N37O48 LCMS: [M+2H]2+: 1791.5; [M+3H]3+: 1194.5; RT = 2.42 min (Method D) Compound No.34 (reference cpd): 210081WO01 52 [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K17(AEEA2- ^E-C18-OH) amide (SEQ ID NO: 34)
Figure imgf000053_0001
Formula Weight: 5350.1; Exact Mass: 5346.87; Formula: C246H388N62O71 LCMS: [M+3H]3+: 1784.1; [M+4H]4+: 13336.3; [M+5H]5+: 1070.8; RT = 2.1 min (Method C) 5 Compound No.35 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K17(AEEA2- ^E-C18-OH) (SEQ ID NO: 35)
Figure imgf000053_0002
Formula Weight: 3815.4; Exact Mass: 3813.05; Formula: C178H281N39O53 10 LCMS: [M+2H]2+: 1908.5; [M+3H]3+: 1272.5; [M+4H]4+: 954.7; RT = 2.48 min (Method C) Compound No.36 (reference cpd): [F6Pla, M14L, H18R]-hGIP(6-31) - K21( ^E ^E-C16) (SEQ ID NO: 36)
Figure imgf000053_0003
15 Formula Weight: 3580.2; Exact Mass: 3577.98; Formula: C170H268N38O46 LCMS: [M+2H]2+: 1790.2; [M+3H]3+: 1194.2; RT = 2.90 min (Method C) Compound No.37 (reference cpd): 210081WO01 53 [F6Pla, M14L, H18R]-hGIP(6-31) - K21( ^E ^E-C18-OH) (SEQ ID NO: 36) Formula Weight: 3638.2; Exact Mass: 3635.99; Formula: C172H270N38O48 LCMS: [M+2H]2+: 1820.3; [M+3H]3+: 1213.4; RT = 2.53 min (Method C) 5 Compound No.38 (reference cpd): [Nα -Ac, M14L, H18R]-hGIP(5-31) - K21( ^E ^E-C18-OH) (SEQ ID NO: 37) Formula Weight: 3780.4; Exact Mass: 3778.06; Formula: C178H280N40O50 10 LCMS: [M+2H]2+: 1891.0; [M+3H]3+: 1260.8; [M+4H]4+: 945.9; RT = 2.53 min (Method C) Compound No.39 (reference cpd): [Nα -Ac, M14L, H18R, G31P]-hGIP(5-31) - K21( ^E ^E ^E-C18-OH) amide (SEQ ID NO: 38) 15 Formula Weight: 3948.6; Exact Mass: 3946.15; Formula: C186H292N42O52 LCMS: [M+2H]2+: 1974.7; [M+3H]3+: 1317.1; [M+4H]4+: 988.2; RT = 2.45 min (Method C) Compound No.40 (reference cpd): [Nα -Ac, M14L, H18R]-hGIP(5-42) - K21( ^E ^E ^E-C18-OH) amide (SEQ ID NO: 39) 210081WO01 54 Formula Weight: 5303.0; Exact Mass: 5299.83; Formula: C245H384N62O69 LCMS: [M+3H]3+: 1768.7; [M+4H]4+: 1326.5; [M+5H]5+: 1061.4; RT = 2.08 min (Method C) 5 Compound No.41 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K24( ^E ^E-C16) (SEQ ID NO: 40) Formula Weight: 3595.2; Exact Mass: 3593.0; Formula: C171H269N37O47 10 LCMS: [M+2H]2+: 1798.5; [M+3H]3+: 1199.2; RT = 2.21 min (Method D) Compound No.42 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K24( ^E ^E-C16) amide (SEQ ID NO: 41) 15 Formula Weight: 5129.9; Exact Mass: 5126.80; Formula: C239H376N60O65 LCMS: [M+3H]3+: 1710.7; [M+4H]4+: 1283.3; [M+5H]5+: 1026.7; RT = 2.38 min (Method C) Compound No.43 (reference cpd): [F6Pla, M14L, H18R, D21E]-hGIP(6-31) - K30( ^E ^E-C16) (SEQ ID NO: 16)
Figure imgf000056_0001
Formula Weight: 3581.1; Exact Mass: 3578.93; Formula: C169H263N37O48 LCMS: [M+2H]2+: 1790.6; [M+3H]3+: 1194.1; RT = 2.34 min (Method D) 5 Compound No.44 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-42) - K30( ^E ^E-C16) amide (SEQ ID NO: 27)
Figure imgf000056_0002
Formula Weight: 5115.8; Exact Mass: 5112.74; Formula: C237H370N60O66 10 LCMS: [M+3H]3+: 1705.6; [M+4H]4+: 1279.7; [M+5H]5+: 1024.1; RT = 2.37 min (Method C) Compound No.45 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43( ^E ^E ^E-C18-OH) amide (SEQ ID NO: 26)
Figure imgf000056_0003
15 Formula Weight: 5431.2; Exact Mass: 5427.89; Formula: C250H391N63O72 LCMS: [M+3H]3+: 1811.1; [M+4H]4+: 1358.7; [M+5H]5+: 1087.2; RT = 1.29 min (Method D) Compound No.46 (reference cpd): 210081WO01 56 [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43( ^E ^E-C18-OH) amide (SEQ ID NO: 26)
Figure imgf000057_0001
Formula Weight: 5302.0; Exact Mass: 5298.84; Formula: C245H384N62O69 LCMS: [M+3H]3+: 1768.4; [M+4H]4+: 1326.3; [M+5H]5+: 1061.4; RT = 1.29 min (Method D) 5 Compound No.47 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43( ^E-C18-OH) amide (SEQ ID NO: 26)
Figure imgf000057_0002
Formula Weight: 5172.9; Exact Mass: 5169.80; Formula: C240H377N61O66 10 LCMS: [M+3H]3+: 1725.1; [M+4H]4+: 1294.0; [M+5H]5+: 1035.3; RT = 1.34 min (Method D) Compound No.48 (reference cpd): [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43(-C18-OH) amide (SEQ ID NO: 26)
Figure imgf000057_0003
15 Formula Weight: 5043.8; Exact Mass: 5040.76; Formula: C235H370N60O63 LCMS: [M+3H]3+: 1681.9; [M+4H]4+: 1261.8; [M+5H]5+: 1009.5; RT = 1.39 min (Method D) Compound No.49: 210081WO01 57 [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43(-C16) amide (SEQ ID NO: 26)
Figure imgf000058_0001
Formula Weight: 4985.8; Exact Mass: 4982.75; Formula: C233H368N60O61 LCMS: [M+3H]3+: 1662.6; [M+4H]4+: 1247.2; [M+5H]5+: 998.0; RT = 1.66 min (Method D) 5 Compound No.50: [Nα -Ac, M14L, H18R, D21E]-hGIP(5-43) - K43( ^E-C16) amide (SEQ ID NO: 26)
Figure imgf000058_0002
Formula Weight: 5114.9; Exact Mass: 5111.80; Formula: C238H375N61O64 10 LCMS: [M+3H]3+: 1705.8; [M+4H]4+: 1279.5; [M+5H]5+: 1023.8; RT = 1.61 min (Method D) Compound No.51 (reference cpd): [Nα -Ac, M14L, D21E] -hGIP(5-31) – K18( ^E-C16) (SEQ ID NO: 42)
Figure imgf000058_0003
15 Formula Weight : 3566.1 ; Exact Mass : 3564.0 ; Formula : C170H266N36O47 LCMS: [M+3H]3+ : 1189.6 ; [M+2H]2+ : 1783.5 ; RT = 7.18 min (Method B) Compound No.52: [Nα -Ac, M14L, Aib16, H18R, D21E] - hGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 43) 210081WO01 58
Figure imgf000059_0001
Formula Weight : 3592.2 ; Exact Mass : 3590.0 ; Formula : C171H268N38O46 LCMS: [M+2H]2+ : 1796.6 ; [M+3H]3+ : 1198.1 ; RT = 7.48min (Method B) 5 Compound No.53: [Nα -Ac, M14L, H18R, Aib20, D21E] - hGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 44)
Figure imgf000059_0002
Formula Weight : 3592.2 ; Exact Mass : 3590.0 ; Formula : C172H272N38O45 LCMS: [M+2H]2+ : 1796.6 ; [M+3H]3+ : 1198.1 ; RT = 7.15min (Method B) 10 Compound No.54 (reference cpd): [Nα -glutaric, M14L, H18R, D21E] - hGIP(5-31) - K11(OEG2- ^E-C18-OH) (SEQ ID NO: 45)
Figure imgf000059_0003
Formula Weight : 4055.7 ; Exact Mass : 4053.2 ; Formula : C190H301N41O56 15 LCMS: [M+3H]3+ : 1352.6 ; [M+4H]4+ : 1014.6 ; RT = 6.07min (Method B) Example 2: In vitro functional potency (CRE luciferase; whole cells) and binding
The purpose of this example is to test the activity, or potency, of the derivatives in vitro at the human or mouse GIP receptor.
The potencies and binding of the analogues and derivatives of Example 1 were determined as described under General methods of measuring in vitro receptor binding and General methods of measuring in vitro functional potency above at the human and mouse GIP receptor. All IC50 data are average ± SD of at least two independent experiments or average from one duplicate experiment. Percentage indicating the maximal inhibition, the inhibition is 95 ~ 100% for those without notice. Results are shown in Table 1.
Table 1 : Potency and binding of test compounds at hGIPR and mGIPR
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
inhibitions are above 95%. **: compound has di-fatty acid in side chain; nd: not determined or not available; Partial weak antagonism: with below 60% inhibition. All IC50 data are average ± SD of at least two independent experiments or average from one duplicate experiment.
From the compounds without an acyl group (1-5, 24) it is seen that going from hGIP (cpd 1) to 2-5, 24 all having M14L, H18R, D21 E substitutions especially antagonism at the mouse receptor is improved. When attaching a side chain with a mono fatty acid at Lys in position 11 (compounds 6-9, 17, 19, 52-53), potency is significantly improved on both receptors, especially on the truncated versions (compounds 7-9, 17, 19, 52-53), compared to attaching a di-fatty acid as present in all of compounds 10-16, 18, 20, 30, 54.
Generally, the above data shows that acylation with mono fatty acid acylation at one of positions 10, 11 , or 43, in particular 11 or 10, compared to fatty acid acylation at positions 13, 16, 17, 18, 21 , 24, or 30, result in very potent antagonists on both hGIPR and mGIPR, with mono fatty acid acylation at position 11 being the best antagonists on mGIPR. 210081WO01 62 Comparing the mono fatty acid acylation with diacid acylation, mono fatty acid acylated derivatives show better antagonism than comparative derivatives with diacid acylation (see e.g. compound 7 and 16). Example 3: In vivo pharmacokinetic study 5 The purpose of this study is to determine the half-life in vivo of the derivatives of the present invention after a single s.c. administration to mice, i.e. the prolongation of their time in the body and thereby their time of action. This is done in a pharmacokinetic (PK) study, as described under General methods for pharmacokinetic study in mice where the terminal half- life of the derivative in question is determined. By terminal half-life is generally meant the 10 period of time it takes to halve a certain plasma concentration, measured after the initial distribution phase. Results are shown in Table 2: Table 2: Maximal conc (Cmax), time taken to reach Cmax (Tmax), and half-life (t½)
Figure imgf000063_0001
15 Data shows that compounds 7 and 9 have long half-lives while compound 22 has shorter half-life. Example 4: In vivo pharmacodynamic study The purpose of this example is to assess the in vivo effect of the derivatives of the 20 present invention alone and in combination with a GLP-1 receptor agonist on food intake, body weight, and glucose tolerance in diet-induced obese (DIO) mice. The GLP-1 receptor agonist used for this example was semaglutide. The experiment was performed as described under General methods for pharmacodynamic study in mice. The results are shown below in Tables 3-6: 25 Table 3. Average (n=8) body weight changes in grams and percentage of body weight compared to day 0.
Figure imgf000063_0002
Figure imgf000064_0001
The data of Table 3 is also shown in Figure 1. It is seen that administration of compound No. 7 (500 nmol/kg) results in reduced body weight gain compared to vehicle and administration of compound No. 7 (1500 nmol/kg) results in a slight body weight reduction compared to day 0. Furthermore, combination of compound No. 7 with semaglutide significantly improves body weight lowering beyond that of semaglutide alone at both doses.
Table 4. Cumulative food intake in grams (average, n=8).
Figure imgf000064_0002
The data in Table 4 is also shown in Figure 2. It is seen that administration of the compound No. 7 at both 500 nmol/kg and 1500 nmol/kg results in reduced food intake compared to vehicle. Furthermore, when combined with semaglutide, food intake is lowered beyond that of semaglutide alone.
Table 5. Blood glucose levels glucose tolerance tests (AUC, average ±SEM, n=8)
Figure imgf000065_0001
On day 0, the OGTT shows that dosing compound No. 7 at 500 nmol/kg (‘low dose’) did not affect glucose level compared to vehicle. However, dosing compound No. 7 at 1500 nmol/kg (‘high dose’) increased glucose levels on day 0. This effect was overcome when combining both the low dose and high dose of compound No. 7 with GLP-1R agonist semaglutide 2 nmol/kg.
Results from day 22 OGTT and day 27 IPGTT shows that after chronical administration of compound No. 7 both alone and in combination with GLP-1R agonist semaglutide, the GIPR antagonist improves glucose control. Both high and low dose of compound No. 7 on day 22 (OGTT) and day 27 (IPGTT) improves glucose level compared to vehicle, and in combination with semaglutide showed significantly improved glucose levels compared to semaglutide alone on day 27 IPGTT.
Table 6. Insulin, GIP, resistin, and C-terminal telopeptide (CTX) levels on day 27 after 6h fasting (average ±SEM, n=8)
Figure imgf000065_0002
Figure imgf000066_0001
The data in Table 6 shows that compared to vehicle compound No. 7 and the combination of semaglutide and compound No. 7 improves insulin sensitivity and resistance (insulin), reduce the GIP response (GIP), does not significantly affect resistin level (resistin), and does not affect bone health (CTX).
While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims

CLAIMS Claim 1: A glucose-dependent insulinotropic peptide (GIP) derivative consisting of a GIP analogue of 5 Formula Ia, IIa, or IIIa and a modifying group, wherein Formulae Ia, IIa, and IIIa are defined by: Xxx5-Xxx6-lle-Ser-Asp-Xxx10-Xxx11-lle-Ala-Xxx14-Asp-Lys-lle-Xxx18-Gln-Gln-Xxx21-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Xxx30-Xxx31 (Ia), (SEQ ID NO: 46) 10 Xxx5-Xxx6-lle-Ser-Asp-Xxx10-Xxx11-lle-Ala-Xxx14-Asp-Lys-lle-Xxx18-Gln-Gln-Xxx21-Phe-Val- Asn-Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-Ile-Thr-Gln (IIa), (SEQ ID NO: 47) Xxx5-Xxx6-lle-Ser-Asp-Tyr-Ser-lle-Ala-Xxx14-Asp-Lys-lle-Xxx18-Gln-Gln-Xxx21-Phe-Val-Asn- Trp-Leu-Leu-Ala-GIn-Lys-Gly-Lys-Lys-Asn-Asp-Trp-Lys-His-Asn-Ile-Thr-Gln-Lys (IIIa), (SEQ 15 ID NO: 48) wherein Xxx5 is Thr or absent, Xxx6 is Phe or L-3-phenyllactic acid (Pla), Xxx10 is Tyr or Lys; Xxx11 is Ser or Lys; Xxx14 is Met, Leu, Ile or Nle; Xxx18 is His, Arg, Orn, homoArg; Xxx21 is Asp, Glu, or homoGlu; Xxx30 is Lys or absent, and Xxx31 is Gly or absent; provided that when 20 Xxx6 is Pla, then Xxx5 is absent; wherein said GIP analogue of Formula Ia or IIa comprises a modified lysine in one of positions 11 or 10 and wherein said GIP analogue of Formula IIIa comprises a modified lysine in position 43, said modified lysine comprises the modifying group that is covalently attached to the side chain of the epsilon amino group of the lysine, the modifying group being 25 defined by A-B-C-, wherein A- is a mono fatty acid and B-C- is a linker, which may be absent or present; or a pharmaceutically acceptable salt, amide, or ^-N acetylate thereof. Claim 2: The GIP derivative according to claim 1, wherein 30 Xxx5 is Thr or absent, Xxx6 is Phe or L-3-phenyllactic acid (Pla), Xxx10 is Tyr or Lys, Xxx11 is Ser or Lys, 35 Xxx14 is Leu, 210081WO01 67 Xxx18 is His or Arg, Xxx21 is Glu, Xxx30 is Lys or absent, and Xxx31 is Gly or absent; 5 provided that when Xxx6 is Pla, then Xxx5 is absent. Claim 3: The GIP derivative according to any of claims 1-2, wherein the Xxx18 is Arg. 10 Claim 4: The GIP derivative according to any one of claims 1-3, wherein the modified lysine is in position 11. Claim 5: 15 The GIP derivative according to any one of the preceding claims, wherein the GIP analogue is of Formula Ia. Claim 6: The GIP derivative according to any one of the preceding claims, wherein the GIP analogue 20 is of formula Ia, wherein the modified lysine is in position 11, Xxx10 is Tyr, Xxx11 is Lys, Xxx30 is Lys, Xxx31 is Gly, and the N-terminal end optionally substituted with an ^-acetylate. Claim 7: The GIP derivative according to any one of the preceding claims, wherein Xxx5 is Thr and 25 Xxx6 is Phe. Claim 8: The GIP derivative according to any one of claims 1-6, wherein Xxx5 is absent and Xxx6 is L- 3-phenyllactic acid (Pla). 30 Claim 9: The GIP derivative according to any one of the preceding claims, wherein B is selected from ^-Glu and ^-Glu- ^-Glu. 35 Claim 10: 210081WO01 68 The GIP derivative according to any one of the preceding claims, wherein B-C is ^-Glu and A is a C14 to C18 mono fatty acid, such as palmitic acid. Claim 11: 5 The GIP derivative according to any one of the preceding claims selected from the group consisting of: compound No.7, compound No.8, compound No.9, compound No.17, compound No.19, compound No.21, compound No.22, compound No.23, compound No. 25, compound No.28, compound No.49 and compound No.50 as shown in example 1 herein. 10 Claim 12: The GIP derivative according to any one of the preceding claims which is compound No.7: [Nα -Ac, M14L, H18R, D21E]-hGIP(5-31) - K11( ^E-C16) (SEQ ID NO: 18)
Figure imgf000069_0001
15 . Claim 13: A pharmaceutical composition comprising a derivative according to any one of the preceding claims, and at least one pharmaceutically acceptable excipient. 20 Claim 14: The GIP derivative according to any one of claims 1-13 for use in the treatment or prevention of obesity, diabetes, such as type II diabetes, hyperinsulinemia, such as congenital hyperinsulinemia, and Cushing’s syndrome. 25 Claim 15: The GIP derivative for use according to claim 14 in combination with a GLP-1 receptor agonist.
PCT/EP2023/051098 2022-01-18 2023-01-18 Long-acting gipr antagonists WO2023139106A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263300461P 2022-01-18 2022-01-18
US63/300,461 2022-01-18
EP22155553 2022-02-08
EP22155553.5 2022-02-08

Publications (2)

Publication Number Publication Date
WO2023139106A2 true WO2023139106A2 (en) 2023-07-27
WO2023139106A3 WO2023139106A3 (en) 2023-09-07

Family

ID=85076327

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/051098 WO2023139106A2 (en) 2022-01-18 2023-01-18 Long-acting gipr antagonists

Country Status (1)

Country Link
WO (1) WO2023139106A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200087373A1 (en) 2017-05-31 2020-03-19 University Of Copenhagen Long-acting gip peptide analogues
WO2020115048A1 (en) 2018-12-03 2020-06-11 Antag Therapeutics Aps Modified gip peptide analogues

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2023505441A (en) * 2019-12-03 2023-02-09 アンタグ セラピューティクス エーピーエス Optimized GIP peptide analogs
MX2022011816A (en) * 2020-03-25 2022-10-10 Takeda Pharmaceuticals Co Qd dosing of gip receptor agonist peptide compounds and uses thereof.
MX2022011729A (en) * 2020-03-25 2022-10-13 Takeda Pharmaceuticals Co Gif receptor agonist peptide compounds and uses thereof.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200087373A1 (en) 2017-05-31 2020-03-19 University Of Copenhagen Long-acting gip peptide analogues
WO2020115048A1 (en) 2018-12-03 2020-06-11 Antag Therapeutics Aps Modified gip peptide analogues

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
FLORENCIO ZARAGOZA DORWALD: "Fmoc Solid Phase Peptide Synthesis", 2000, OXFORD UNIVERSITY PRESS
GREENEWUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
HODGSON ET AL.: "The synthesis of peptides and proteins containing non-natural amino acids", CHEMICAL SOCIETY REVIEWS, vol. 33, no. 7, 2004, pages 422 - 430, XP008067398

Also Published As

Publication number Publication date
WO2023139106A3 (en) 2023-09-07

Similar Documents

Publication Publication Date Title
CN111491658B (en) Incretin analogues and uses thereof
KR102351313B1 (en) GIP/GLP1 co-agonist compounds
TWI767095B (en) Incretin analogs and uses thereof
KR102379958B1 (en) GIP derivatives and uses thereof
IL255781A (en) Selective pyy compounds and uses thereof
AU2014350197A1 (en) Selective PYY compounds and uses thereof
KR102667979B1 (en) Protein tyrosine-tyrosine analogues and methods of their use
KR20210102911A (en) Modified GIP peptide analogs
EP4345105A1 (en) Polypeptide derivative having effect of dual targeted activation of glp-1r and gipr, preparation method therefor, and use thereof
WO2016124687A1 (en) Selective pyy compounds and uses thereof
WO2023139106A2 (en) Long-acting gipr antagonists
TW202415675A (en) Incretin analogs and uses thereof
EA043950B1 (en) INCRETIN ANALOGUES AND THEIR APPLICATION
WO2023285334A1 (en) Novel fatty acid modified urocortin 2 derivatives and the uses thereof
KR20240093737A (en) Protein tyrosine-tyrosine analogs and methods of using the same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23701846

Country of ref document: EP

Kind code of ref document: A2