Long-acting GIP peptide analogs

文档序号:1631408 发布日期:2020-01-14 浏览:38次 中文

阅读说明:本技术 长效gip肽类似物 (Long-acting GIP peptide analogs ) 是由 M·M·罗森基尔德 J·J·霍尔斯特 L·S·加斯伯格 A·H·斯巴尔-乌尔里希 M·B·N 于 2018-05-31 设计创作,主要内容包括:公开了葡萄糖依赖性促胰岛素肽(GIP)衍生肽类似物GIP5-30和GIP3-30,所述类似物是GIP受体的拮抗剂,并包含至少一个脂肪酸分子以增加半衰期同时保持拮抗性质。(Glucose-dependent insulinotropic peptide (GIP) -derived peptide analogs GIP5-30 and GIP3-30 are disclosed that are antagonists of the GIP receptor and comprise at least one fatty acid molecule to increase half-life while maintaining antagonistic properties.)

1. A glucose-dependent insulinotropic peptide (GIP) analog having formula 1(hGIP5-30, SEQ ID NO: 1):

Figure FDA0002296622770000011

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

2. The GIP peptide analog of claim 1, wherein said peptide is TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1), optionally amidated at its C-terminus,

or a functional variant having at least 75% sequence identity to SEQ ID NO. 1, wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of SEQ ID NO. 1 or the functional variant having at least 75% sequence identity to SEQ ID NO. 1,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of SEQ ID NO 1 or a functional variant having at least 75% sequence identity to SEQ ID NO 1.

3. The GIP peptide analog of claim 1, wherein said peptide is EGTFISDYSIAMDKIHQQDFVNWLLAQK (hGIP3-30, SEQ ID NO:2), optionally amidated at its C-terminus,

or a functional variant having at least 75% sequence identity to SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of SEQ ID NO 2 or a functional variant having at least 75% sequence identity to SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of SEQ ID NO 2 or a functional variant having at least 75% sequence identity to SEQ ID NO 2.

4. The GIP peptide analog of claim 3, wherein the at least one fatty acid molecule is not attached to the N-terminal amino acid residue at position 3 of SEQ ID NO. 2, such as not attached to the N-terminal E at position 3 of SEQ ID NO. 2.

5. The GIP peptide analog of any one of claims 3 and 4, wherein E (Glu) at position 3 of SEQ ID NO 2 is substituted with pGlu (pyroglutamic acid).

6. The GIP peptide analogue of any one of the preceding claims, wherein the functional variant has at least 80% sequence identity, such as at least 85% sequence identity, such as at least 90% sequence identity, such as at least 95% sequence identity, with any one of SEQ ID No. 1 and SEQ ID No. 2.

7. The GIP peptide analogue of any one of the preceding claims, wherein the functional variant has 1 to 6 individual amino acid substitutions, such as 1 individual amino acid substitution, e.g. 2 individual amino acid substitutions, such as 3 individual amino acid substitutions, e.g. 4 individual amino acid substitutions, such as 5 individual amino acid substitutions, e.g. 6 individual amino acid substitutions, between the variant peptide and the corresponding native GIP peptide.

8. The GIP peptide analogue according to any one of the preceding claims, wherein K at position 30 of SEQ ID No. 1 or SEQ ID No. 2 or a functional variant thereof is substituted by any amino acid, such as a conservative amino acid substitution, such as by an amino acid selected from R, A and E.

9. The GIP peptide analogue according to any one of the preceding claims, wherein K at position 16 of SEQ ID No. 1 or SEQ ID No. 2 or a functional variant thereof is substituted by any amino acid, such as a conservative amino acid substitution, such as by an amino acid selected from R, A and E.

10. The GIP peptide analogue of any one of the preceding claims, wherein the K at position 16 of SEQ ID NO 1 or SEQ ID NO 2 or a functional variant thereof is substituted with any amino acid when the K at position 16 is not modified by attachment of a fatty acid molecule.

11. The GIP peptide analogue according to any one of the preceding claims, wherein K at position 16 and K at position 30 of SEQ ID No. 1 or SEQ ID No. 2 or a functional variant thereof are each substituted by any amino acid, such as a conservative amino acid substitution, such as by an amino acid selected from R, A and E.

12. The GIP peptide analogue according to any one of the preceding claims, wherein M at position 14 of SEQ ID NO 1 or SEQ ID NO 2 or a functional variant thereof is substituted by any amino acid, such as L, S, K or norleucine (Nle) or methoxyamino acid (Mox).

13. The GIP peptide analogue according to any one of the preceding claims, wherein D at position 15 of SEQ ID No. 1 or SEQ ID No. 2 or a functional variant thereof is substituted by any amino acid, such as by an amino acid selected from E, A, K and Orn.

14. The GIP peptide analogue according to any one of the preceding claims, wherein H at position 18 of SEQ ID No. 1 or SEQ ID No. 2 or a functional variant thereof is substituted by any amino acid, such as by an amino acid selected from A, R, K and Orn.

15. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the amino acid residue at any one of positions 5-29 of SEQ id No. 1 or a variant thereof; and/or wherein a fatty acid molecule is attached to an amino acid residue at any one of positions 4 to 29 of SEQ ID NO. 2 or a variant thereof.

16. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to the amino acid residue at position 5, 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, or 29 of SEQ id no 1 or a functional variant thereof.

17. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to the amino acid residue at position 4, 5, 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, or 29 of SEQ id no 2 or a functional variant thereof.

18. The GIP peptide analogue of any one of the preceding claims, wherein the at least one fatty acid molecule is attached to one or more amino acid residues of the middle region of either of SEQ ID No. 1 and SEQ ID No. 2, or a functional variant thereof; such as to one or more amino acid residues at any of positions 11 to 21 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

19. The GIP peptide analogue according to any one of the preceding claims, wherein the at least one fatty acid molecule is attached to one or more amino acid residues of the N-terminal region of either one of SEQ ID No. 1 and SEQ ID No. 2, or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 3 of SEQ ID No. 2; such as to one or more amino acid residues at any of positions 5 to 10 of SEQ ID NO. 1 or a functional variant thereof or such as to one or more amino acid residues at any of positions 4 to 10 of SEQ ID NO. 2 or a functional variant thereof.

20. The GIP peptide analogue according to any one of the preceding claims, wherein the at least one fatty acid molecule is attached to one or more amino acid residues of the C-terminal region of either one of SEQ ID No. 1 and SEQ ID No. 2, or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either one of SEQ ID No. 1 and SEQ ID No. 2; such as to one or more amino acid residues at any of positions 22 to 29 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

21. The GIP peptide analogue according to any one of the preceding claims, wherein a fatty acid molecule is attached to the epsilon-amino group of the K residue of either of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof comprising at least one K residue.

22. The GIP peptide analogue of any one of the preceding claims, wherein the fatty acid molecule is attached to the δ -amino group of the Orn residue of either of SEQ ID No. 1 and SEQ ID No. 2, or a variant thereof comprising at least one Orn residue.

23. The GIP peptide analogue of any one of the preceding claims, wherein the amino acid residue to which the fatty acid molecule is attached is the N-most terminal amino acid residue, such as the N-most terminal amino acid residue of SEQ ID No. 1 or a variant thereof.

24. The GIP peptide analogue of any one of the preceding claims, wherein the fatty acid molecule is attached to the alpha-amino group of the N-terminal amino acid residue, such as to the N-terminal amino acid residue at position 5 of SEQ ID NO:1 or a functional variant thereof, such as to T at position 5 of SEQ ID NO: 1.

25. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of the amino acid residue at position 16 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a functional variant thereof, such as to the K at position 16 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a functional variant thereof.

26. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue 18 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a variant thereof, wherein H at position 18 has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

27. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue 11 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a variant thereof, wherein S at position 11 has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

28. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue position 12 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a variant thereof, wherein position 12I has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

29. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue 13 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a variant thereof, wherein position 13A has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

30. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue 7 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a variant thereof, wherein position 7I has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

31. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue 9 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a variant thereof, wherein D at position 9 has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

32. The GIP peptide analog of any one of the preceding claims, wherein a fatty acid molecule is attached to the side chain amino group of amino acid residue position 21 of any one of SEQ ID No. 1 and SEQ ID No. 2, or a variant thereof, wherein position 21D has been substituted with K or Orn of any one of SEQ ID No. 1 and SEQ ID No. 2.

33. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to the side chain amino group of amino acid residue 5 of SEQ ID No. 2 or a variant thereof, wherein T at position 5 has been substituted with K or Orn of SEQ ID No. 2.

34. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to the side chain amino group of amino acid residue 15 of SEQ ID No. 2 or a variant thereof, wherein D at position 15 has been substituted with K or Orn of SEQ ID No. 2.

35. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to the side chain amino group of amino acid residue 20 of SEQ ID No. 2 or a variant thereof, wherein Q at position 20 has been substituted with K or Orn of SEQ ID No. 2.

36. The GIP peptide analog of any one of the preceding claims, wherein K at position 16 and/or K at position 30 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a functional variant thereof is individually substituted with any amino acid when a fatty acid molecule is attached to the amino acid residues at positions other than position 16 and position 30 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a functional variant thereof.

37. The GIP peptide analog of any one of the preceding claims, wherein the peptide is modified by attaching a fatty acid molecule at an amino acid residue of either one of SEQ ID NO 1 and SEQ ID NO 2, or a functional variant thereof.

38. The GIP peptide analog of any one of the preceding claims, wherein the peptide comprises no more than one K amino acid residue that is modified by attachment of a fatty acid molecule.

39. The GIP peptide analog of any one of the preceding claims, wherein the peptide comprises no more than one Orn amino acid residue modified by attachment of a fatty acid molecule.

40. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is a straight chain fatty acid.

41. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is a branched chain fatty acid.

42. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is a monoacyl fatty acid molecule comprising one fatty acid.

43. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is a diacyl fatty acid molecule.

44. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises the formula CH3(CH2)nAcyl radical of CO-A group wherein n is an integer from 4 to 24.

45. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises a moiety selected from CH3(CH2)6CO-、CH3(CH2)8CO-、CH3(CH2)10CO-、CH3(CH2)12CO-、CH3(CH2)14CO-、CH3(CH2)16CO-、CH3(CH2)18CO-、CH3(CH2)20CO-and CH3(CH2)22One or more acyl groups of CO-.

46. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises a moiety selected from CH3(CH2)10CO- (lauryl, C12), CH3(CH2)12CO- (myristoyl, C14), CH3(CH2)14CO- (palmitoyl, C16) and CH3(CH2)16Acyl radical of CO- (stearoyl, C18).

47. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises a single moiety selected from CH3(CH2)10CO- (lauryl, C12), CH3(CH2)12CO- (myristoyl, C14), CH3(CH2)14CO- (palmitoyl, C16) and CH3(CH2)16Two acyl groups of CO- (stearoyl, C18).

48. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises the formula COOH (CH)2)nAn acyl group of CO- (dicarboxylic acid), wherein n is an integer from 4 to 24.

49. According toThe GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule comprises a moiety selected from COOH (CH)2)14CO-、COOH(CH2)16CO-、COOH(CH2)18CO-and COOH (CH)2)20Acyl group of CO-.

50. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is directly attached to an amino acid residue.

51. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule is attached to an amino acid residue through a spacer.

52. The GIP peptide analog of any one of the preceding claims, wherein the fatty acid molecule can be attached to an amino acid residue through a spacer in a manner such that the carboxyl group of the spacer forms an amide bond with the amino group of the fatty acid molecule.

53. The GIP peptide analog of any one of the preceding claims, wherein the spacer comprises one or more moieties individually selected from the group consisting of:

a. one or more alpha, omega-amino acids,

b. one or more amino acids selected from the group consisting of succinic acid, Lys, Glu, Asp,

c.4-Abu,

d. gamma-aminobutyric acid

e. A dipeptide, such as a dipeptide, wherein the C-terminal amino acid residue is Lys, His or Trp, preferably Lys, and wherein the N-terminal amino acid residue is selected from Ala, Arg, Asp, Asn, Gly, Glu, Gln, Ile, Leu, Val, Phe and Pro, such as Gly-Lys,

f. one or more of gamma-aminobutyryl (gamma-aminobutyric acid), gamma-glutamyl (gamma-glutamic acid), beta-asparaginyl, beta-alanyl and glycyl, and

[ gamma-glutamic acid-8-amino ] -acetic acid-3, 6-dioxaoctanoic acid]n(γGlu-AEEAcn) Wherein n is an integer between 1 and 50, such as an integer between 1-2, 2-3, 3-4, 4-5, 5-6, 6-7, 7-8, 8-9, 9-10, 10-11, 11-12, 12-13, 13-14, 14-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45, 45-50.

54. The GIP peptide analog of any one of the preceding claims, wherein the peptide is TFISDYX11X12X13X1 4X15X16IX18QQDFVNWLLAQX30(SEQ ID NO:3), wherein

X11Selected from the group consisting of S, K and Orn,

X12selected from the group consisting of I, K and Orn,

X13selected from the group consisting of A, K and Orn,

X14selected from the group consisting of M, K, L, S, Nle and Mox,

X15selected from the group consisting of D, E, A, K and Orn,

X16selected from the group consisting of K, R, A and E,

X18selected from H, A, R, K and Orn, and

X30selected from the group consisting of K, R, A and E,

or a functional variant thereof having at least 75% sequence identity to said sequence,

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues of the sequence, with the proviso that the at least one fatty acid molecule is not attached to

X30The above.

55. The GIP peptide analog of any one of the preceding claims, wherein the peptide is selected from the group consisting of:

TFISDYSIAMDKIHQQDFVNWLLAQK(hGIP5-30,SEQ ID NO:1),TFISDYSIAMDRIKQQDFVNWLLAQR(hGIP(5-30)K16R H18K K30R;SEQ ID NO:5),

TFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(5-30)K16R K30R;SEQ ID NO:6),

TFISDYSIAMDKIAQQDFVNWLLAQK(hGIP(5-30)H18A;SEQ ID NO:7),

TFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(5-30)H18K;SEQ ID NO:8),

TFISDYSIAMEKIAQQDFVNWLLAQK(hGIP(5-30)D15E H18A;SEQ ID NO:9),

TFISDYSIAMDAIAQQDFVNWLLAQK(hGIP(5-30)K16A H18A;SEQ ID NO:10),

TFISDYSIAMEKIHQQDFVNWLLAQK(hGIP(5-30)D15E;SEQ ID NO:11),

TFISDYSIAMNKIHQQDFVNWLLAQK(hGIP(5-30)D15N;SEQ ID NO:12),

TFISDYSIAMDAIHQQDFVNWLLAQK(hGIP(5-30)K16A;SEQ ID NO:13),

TFISDYSIAMDHIHQQDFVNWLLAQK(hGIP(5-30)K16H;SEQ ID NO:14),

TFISDYSIAMDRIHQQDFVNWLLAQK(hGIP(5-30)K16R;SEQ ID NO:15),

TFISDYSIAMDKIFQQDFVNWLLAQK(hGIP(5-30)H18F;SEQ ID NO:16),

TFISDYSIAMDKIWQQDFVNWLLAQK(hGIP(5-30)H18W;SEQ ID NO:17),

TFISDYSIAMDKIHQQDFVNWLLAQR (hGIP (5-30) K30R; SEQ ID NO:18), and

TFISDYSIAMDKIHQQDFVNWLLAQH(hGIP(5-30)K30H;SEQ ID NO:19),

KFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)T5K,SEQ ID NO:20),

TKISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)F6K,SEQ ID NO:21),

TFKSDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)I7K,SEQ ID NO:22),

TFIKDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S8K,SEQ ID NO:23),

TFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)D9K,SEQ ID NO:24),

TFISDKSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)Y10K,SEQ ID NO:25),

TFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S11K,SEQ ID NO:26),

TFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(5-30)I12K,SEQ ID NO:27),

TFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(5-30)A13K,SEQ ID NO:28),

TFISDYSIAKDKIHQQDFVNWLLAQK(hGIP(5-30)M14K,SEQ ID NO:29),

TFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(5-30)D15K,SEQ ID NO:30),

TFISDYSIAMDKKHQQDFVNWLLAQK(hGIP(5-30)I17K,SEQ ID NO:31),

TFISDYSIAMDKIHKQDFVNWLLAQK(hGIP(5-30)Q19K,SEQ ID NO:32),

TFISDYSIAMDKIHQKDFVNWLLAQK(hGIP(5-30)Q20K,SEQ ID NO:33),

TFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(5-30)D21K,SEQ ID NO:34),

TFISDYSIAMDKIHQQDKVNWLLAQK(hGIP(5-30)F22K,SEQ ID NO:35),

TFISDYSIAMDKIHQQDFKNWLLAQK(hGIP(5-30)V23K,SEQ ID NO:36),

TFISDYSIAMDKIHQQDFVKWLLAQK(hGIP(5-30)N24K,SEQ ID NO:37),

TFISDYSIAMDKIHQQDFVNKLLAQK(hGIP(5-30)W25K,SEQ ID NO:38),

TFISDYSIAMDKIHQQDFVNWKLAQK(hGIP(5-30)L26K,SEQ ID NO:39),

TFISDYSIAMDKIHQQDFVNWLKAQK(hGIP(5-30)L27K,SEQ ID NO:40),

TFISDYSIAMDKIHQQDFVNWLLKQK(hGIP(5-30)A28K,SEQ ID NO:41),

TFISDYSIAMDKIHQQDFVNWLLAKK(hGIP(5-30)Q29K,SEQ ID NO:42),

TFISDYKIAMDKIAQQDFVNWLLAQK(hGIP(5-30)S11K H18A,SEQ ID NO:43),

TFISDYKIAMDKIRQQDFVNWLLAQK(hGIP(5-30)S11K H18R,SEQ ID NO:44),

TFISDYKIAMEKIHQQDFVNWLLAQK(hGIP(5-30)S11K D15E,SEQ ID NO:45),

TFISDYKIANleDKIHQQDFVNWLLAQK(hGIP(5-30)S11K M14Nle,SEQ ID NO:46),

TFISDYKIALDKIHQQDFVNWLLAQK(hGIP(5-30)S11K M14L,SEQ ID NO:47),

TFISDYSKAMDKIAQQDFVNWLLAQK(hGIP(5-30)I12K H18A,SEQ ID NO:48),

TFISDYSKAMDKIRQQDFVNWLLAQK(hGIP(5-30)I12K H18R,SEQ ID NO:49),

TFISDYSKAMEKIHQQDFVNWLLAQK(hGIP(5-30)I12K D15E,SEQ ID NO:50),

TFISDYSKANleDKIHQQDFVNWLLAQK(hGIP(5-30)I12K M14Nle,SEQ ID NO:51),

TFISDYSKALDKIHQQDFVNWLLAQK(hGIP(5-30)I12K M14L,SEQ ID NO:52),

TFISDYSIKMDKIAQQDFVNWLLAQK(hGIP(5-30)A13K H18A,SEQ ID NO:53),

TFISDYSIKMDKIRQQDFVNWLLAQK(hGIP(5-30)A13K H18R,SEQ ID NO:54),

TFISDYSIKMEKIHQQDFVNWLLAQK(hGIP(5-30)A13K D15E,SEQ ID NO:55),

TFISDYSIKNleDKIHQQDFVNWLLAQK(hGIP(5-30)A13K M14Nle,SEQ ID NO:56),

TFISDYSIKLDKIHQQDFVNWLLAQK(hGIP(5-30)A13K M14L,SEQ ID NO:57),

TFISDYSIAMEKIKQQDFVNWLLAQK(hGIP(5-30)D15E H18K;SEQ ID NO:58),

TFISDYSIANleDKIKQQDFVNWLLAQK(hGIP(5-30)M14Nle H18K;SEQ ID NO:59),

TFISDYSIALDKIKQQDFVNWLLAQK(hGIP(5-30)M14L H18K;SEQ ID NO:60),

TFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(5-30)D15Orn,SEQ ID NO:61),

TFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(5-30)H18Orn;SEQ ID NO:62),

TFISDYSIALDRIKQQDFVNWLLAQR(hGIP(5-30)M14L K16R H18K K30R;SEQ ID NO:63),

KFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(5-30)T5K K16R K30R,SEQ ID NO:64)

KFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)T5K M14L K16R K30R,SEQ ID NO:65),

TFISDYKIAMDRIHQQDFVNWLLAQR(hGIP(5-30)S11K K16R K30R,SEQ ID NO:66),

TFISDYKIALDRIHQQDFVNWLLAQR(hGIP(5-30)S11K M14L K16R K30R,SEQ ID NO:67),

TFISDYSKAMDRIHQQDFVNWLLAQR(hGIP(5-30)I12K K16R K30R,SEQ ID NO:68),

TFISDYSKALDRIHQQDFVNWLLAQR(hGIP(5-30)I12K M14L K16R K30R,SEQ ID NO:69),

TFISDYSIKMDRIHQQDFVNWLLAQR(hGIP(5-30)A13K K16R K30R,SEQ ID NO:70),

TFISDYSIKLDRIHQQDFVNWLLAQR (hGIP (5-30) A13K M14L K16R K30R, SEQ ID NO:71), and

TFISDYSIAMDRIHQQKFVNWLLAQR(SEQ ID NO:145),

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues, such as by attaching a fatty acid molecule at one amino acid residue, with the proviso that the at least one fatty acid molecule is not attached to the amino acid residue at position 30.

56. The GIP peptide analog of any one of the preceding claims, wherein the peptide is EGTFISDY X11X12X1 3X14X15X16IX18QQDFVNWLLAQX30(SEQ ID NO:4), wherein

X11Selected from the group consisting of S, K and Orn,

X12selected from the group consisting of I, K and Orn,

X13selected from the group consisting of A, K and Orn,

X14selected from the group consisting of M, K, L, S, Nle and Mox,

X15selected from the group consisting of D, E, A, K and Orn,

X16selected from the group consisting of K, R, A and E,

X18selected from H, A, R, K and Orn, and

X30selected from the group consisting of K, R, A and E,

or a functional variant thereof having at least 75% sequence identity to said sequence,

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues of the sequence, with the proviso that the at least one fatty acid molecule is not attached to X30The above.

57. The GIP peptide analog of any one of the preceding claims, wherein the peptide is selected from the group consisting of:

EGTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP3-30,SEQ ID NO:2)

EGTFISDYSIAMDRIKQQDFVNWLLAQR(hGIP(3-30)K16R H18K K30R;SEQ ID NO:72),

EGTFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(3-30)K16R K30R;SEQ ID NO:73),

EGTFISDYSIAMDKIAQQDFVNWLLAQK(hGIP(3-30)H18A;SEQ ID NO:74),

EGTFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(3-30)H18K;SEQ ID NO:75),

EGTFISDYSIAMEKIAQQDFVNWLLAQK(hGIP(3-30)D15E H18A;SEQ ID NO:76),

EGTFISDYSIAMDAIAQQDFVNWLLAQK(hGIP(3-30)K16A H18A;SEQ ID NO:77),

EGTFISDYSIAMEKIHQQDFVNWLLAQK(hGIP(3-30)D15E;SEQ ID NO:78),

EGTFISDYSIAMNKIHQQDFVNWLLAQK(hGIP(3-30)D15N;SEQ ID NO:79),

EGTFISDYSIAMDAIHQQDFVNWLLAQK(hGIP(3-30)K16A;SEQ ID NO:80),

EGTFISDYSIAMDHIHQQDFVNWLLAQK(hGIP(3-30)K16H;SEQ ID NO:81),

EGTFISDYSIAMDRIHQQDFVNWLLAQK(hGIP(3-30)K16R;SEQ ID NO:82),

EGTFISDYSIAMDKIFQQDFVNWLLAQK(hGIP(3-30)H18F;SEQ ID NO:83),

EGTFISDYSIAMDKIWQQDFVNWLLAQK(hGIP(3-30)H18W;SEQ ID NO:84),

EGTFISDYSIAMDKIHQQDFVNWLLAQR (hGIP (3-30) K30R; SEQ ID NO:85), and

EGTFISDYSIAMDKIHQQDFVNWLLAQH(hGIP(3-30)K30H;SEQ ID NO:86),

EKTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)G4K,SEQ ID NO:87),

EGKFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)T5K,SEQ ID NO:88),

EGTKISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)F6K,SEQ ID NO:89),

EGTFKSDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)I7K,SEQ ID NO:90),

EGTFIKDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S8K,SEQ ID NO:91),

EGTFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)D9K,SEQ ID NO:92),

EGTFISDKSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)Y10K,SEQ ID NO:93),

EGTFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S11K,SEQ ID NO:94),

EGTFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(3-30)I12K,SEQ ID NO:95),

EGTFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(3-30)A13K,SEQ ID NO:96),

EGTFISDYSIAKDKIHQQDFVNWLLAQK(hGIP(3-30)M14K,SEQ ID NO:97),

EGTFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(3-30)D15K,SEQ ID NO:98),

EGTFISDYSIAMDKKHQQDFVNWLLAQK(hGIP(3-30)I17K,SEQ ID NO:99),

EGTFISDYSIAMDKIHKQDFVNWLLAQK(hGIP(3-30)Q19K,SEQ ID NO:100),

EGTFISDYSIAMDKIHQKDFVNWLLAQK(hGIP(3-30)Q20K,SEQ ID NO:101),

EGTFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(3-30)D21K,SEQ ID NO:102),

EGTFISDYSIAMDKIHQQDKVNWLLAQK(hGIP(3-30)F22K,SEQ ID NO:103),

EGTFISDYSIAMDKIHQQDFKNWLLAQK(hGIP(3-30)V23K,SEQ ID NO:104),

EGTFISDYSIAMDKIHQQDFVKWLLAQK(hGIP(3-30)N24K,SEQ ID NO:105),

EGTFISDYSIAMDKIHQQDFVNKLLAQK(hGIP(3-30)W25K,SEQ ID NO:106),

EGTFISDYSIAMDKIHQQDFVNWKLAQK(hGIP(3-30)L26K,SEQ ID NO:107),

EGTFISDYSIAMDKIHQQDFVNWLKAQK(hGIP(3-30)L27K,SEQ ID NO:108),

EGTFISDYSIAMDKIHQQDFVNWLLKQK(hGIP(3-30)A28K,SEQ ID NO:109),

EGTFISDYSIAMDKIHQQDFVNWLLAKK(hGIP(3-30)Q29K,SEQ ID NO:110),

EGTFISDYKIAMDKIAQQDFVNWLLAQK(hGIP(3-30)S11K H18A,SEQ ID NO:111),

EGTFISDYKIAMDKIRQQDFVNWLLAQK(hGIP(3-30)S11K H18R,SEQ ID NO:112),

EGTFISDYKIAMEKIHQQDFVNWLLAQK(hGIP(3-30)S11K D15E,SEQ ID NO:113),

EGTFISDYKIANleDKIHQQDFVNWLLAQK(hGIP(3-30)S11K M14Nle,SEQ ID NO:114),

EGTFISDYKIALDKIHQQDFVNWLLAQK(hGIP(3-30)S11K M14L,SEQ ID NO:115),

EGTFISDYSKAMDKIAQQDFVNWLLAQK(hGIP(3-30)I12K H18A,SEQ ID NO:116),

EGTFISDYSKAMDKIRQQDFVNWLLAQK(hGIP(3-30)I12K H18R,SEQ ID NO:117),

EGTFISDYSKAMEKIHQQDFVNWLLAQK(hGIP(3-30)I12K D15E,SEQ ID NO:118),

EGTFISDYSKANleDKIHQQDFVNWLLAQK(hGIP(3-30)I12K M14Nle,SEQ ID NO:119),

EGTFISDYSKALDKIHQQDFVNWLLAQK(hGIP(3-30)I12K M14L,SEQ ID NO:120),

EGTFISDYSIKMDKIAQQDFVNWLLAQK(hGIP(3-30)A13K H18A,SEQ ID NO:121),

EGTFISDYSIKMDKIRQQDFVNWLLAQK(hGIP(3-30)A13K H18R,SEQ ID NO:122),

EGTFISDYSIKMEKIHQQDFVNWLLAQK(hGIP(3-30)A13K D15E,SEQ ID NO:123),

EGTFISDYSIKNleDKIHQQDFVNWLLAQK(hGIP(3-30)A13K M14Nle,SEQ ID NO:124),

EGTFISDYSIKLDKIHQQDFVNWLLAQK(hGIP(3-30)A13K M14L,SEQ ID NO:125),

EGTFISDYSIAMEKIKQQDFVNWLLAQK(hGIP(3-30)D15E H18K;SEQ ID NO:126),

EGTFISDYSIANleDKIKQQDFVNWLLAQK(hGIP(3-30)M14Nle H18K;SEQ ID NO:127),

EGTFISDYSIALDKIKQQDFVNWLLAQK(hGIP(3-30)M14L H18K;SEQ ID NO:128),

EGKFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(3-30)T5K K16R K30R,SEQ ID NO:129),

EGTFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(3-30)D15Orn,SEQ ID NO:130)

EGTFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(3-30)H18Orn;SEQ IDNO:131),

EGTFISDYSIALDRIKQQDFVNWLLAQR(hGIP(3-30)M14L K16R H18K K30R;SEQ ID NO:132),

EGWFISDYSIAMEKIAQQDFVNWLLAQK (SEQ ID NO:144), and

EGTFISDYSIAMDKIKQQDFVNWLLAQR(SEQ ID NO:146),

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues, such as by attaching a fatty acid molecule at one amino acid residue, with the proviso that the at least one fatty acid molecule is not attached to the amino acid residue at position 30.

58. The GIP peptide analog of any one of the preceding claims, selected from the group consisting of:

TFISDYSIAMDKIHQQDFVNWLLAQK-C12/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C12/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C14/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C16/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C16/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C18/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C18/K16

TFISDYSIAMDRIKQQDFVNWLLAQR-C16/T5

TFISDYSIAMDRIKQQDFVNWLLAQR-C16/K18

TFISDYSIAMDRIHQQDFVNWLLAQR-C16/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K16,

TFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K16,

TFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18,

TFISDYSIAMDKIKQQDFVNWLLAQK-C18-diacid/K18,

TFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18,

KFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/K5,

KFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K5,

KFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K5,

KFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K5,

KFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K5,

TFISDYKIAMDKIHQQDFVNWLLAQK-C16-diacid/K11,

TFISDYKIAMDRIHQQDFVNWLLAQR-C16-diacid/K11,

TFISDYKIAMDKIHQQDFVNWLLAQK-C18-diacid/K11,

TFISDYKIAMDRIHQQDFVNWLLAQR-C18-diacid/K11,

TFISDYSKAMDKIHQQDFVNWLLAQK-C16-diacid/K12,

TFISDYSKAMDRIHQQDFVNWLLAQR-C16-diacid/K12,

TFISDYSKAMDKIHQQDFVNWLLAQK-C18-diacid/K12,

TFISDYSKAMDRIHQQDFVNWLLAQR-C18-diacid/K12,

TFISDYSIKMDKIHQQDFVNWLLAQK-C16-diacid/K13,

TFISDYSIKMDRIHQQDFVNWLLAQR-C16-diacid/K13,

TFISDYSIKMDKIHQQDFVNWLLAQK-C18-diacid/K13,

TFISDYSIKMDRIHQQDFVNWLLAQR-C18-diacid/K13,

TFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K16,

TFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K16,

TFISDYSIAMDRIKQQDFVNWLLAQR-C18-diacid/K18,

TFISDYSIAMDKIHQQKFVNWLLAQK-C16-diacid/K21,

TFISDYSIAMDRIHQQKFVNWLLAQR-C16-diacid/K21,

TFISDYSIAMDKIHQQKFVNWLLAQK-C18-diacid/K21,

TFISDYSIAMDRIHQQKFVNWLLAQR-C18-diacid/K21,

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C12/K16,

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C16/K16,

EGWFISDYSIAMEKIAQQDFVNWLLAQK-C16/K16,

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C14/K16,

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C18/K16,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C12/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16/K18,

EGTFISDYSIAMEKIAQQDFVNWLLAQK-C16/K16,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C18-diacid/K18,

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQR-C18/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQR-C16/K18,

EGTFISDYSIALDKIKQQDFVNWLLAQK-C16/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C18/K18,

EGKFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K5,

EGKFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K5,

EGKFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K5,

EGKFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K5, and

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C18-diacid/K18,

or a functional variant thereof,

wherein the fatty acids are attached directly or via a linker/spacer as defined herein.

59. The GIP peptide analog of any one of the preceding claims, wherein the peptide is C-terminally amidated (-NH)2)。

60. The GIP peptide analog of any one of the preceding claims, wherein the peptide is an antagonist of the hGIP receptor.

61. The GIP peptide analogue of any one of the preceding claims, for use in a method of inhibiting or reducing one or more of: i) GIP-induced glucagon secretion, ii) GIP-induced insulin secretion, iii) GIP-induced somatostatin secretion, iv) GIP-induced glucose uptake, v) GIP-induced fatty acid synthesis and/or fatty acid incorporation, vi) higher or increased GIPR expression or activity, vii) postprandial GIP release, viii) serum levels of free fatty acids and/or triglycerides, ix) GIP-induced reduction in bone resorption.

62. The GIP peptide analogue of any one of the preceding claims, for use in a method of treating a disorder selected from: metabolic syndrome, obesity, overweight, obesity-related disorders, prediabetes (impaired fasting glucose), diabetes (types I and 2), diabetes-related disorders, insulin resistance, elevated fasting glucose (hyperglycemia), elevated fasting serum triglyceride (VLDL triglyceride) levels, low High Density Lipoprotein (HDL) levels, disorders of fatty acid metabolism, cardiovascular disease, elevated blood pressure, and atherosclerosis.

Technical Field

The present invention relates to glucose-dependent insulinotropic peptide (GIP) -derived peptide analogs that are antagonists of the GIP receptor, comprising a GIP peptide and at least one fatty acid molecule attached to increase half-life while maintaining antagonistic properties.

Background

GrapeSugar-dependent insulinotropic peptide (GIP) is a hormone secreted from intestinal K cells after meal1. Like the sister hormone glucagon-like peptide 1(GLP-1), GIP is a potent insulin secretagogue2. Contrary to the glucagon inhibitory effect of GLP-13,4GIP has been shown to exhibit glucagon release properties under certain conditions (3,5-13). The association between rodent GIPR (GIP receptor) and obesity has increased interest in understanding the biology of GIP14-21. In humans, although not yet clear, there is also evidence for a role of GIP in fat metabolism, demonstrating the expression of GIPR in adipose tissue22Correlation between high BMI and elevated GIP levels22,23Increased adipose tissue blood flow and TAG (triacylglycerol) deposition following GIP administration in high insulin and high glucose states24The basal and postprandial GIP level reductions observed with obese children diet25And elevated fasting GIP levels observed with a healthy young male on a high-fat diet26

Thus, in addition to the general needs of those researchers who have witnessed the progress of the GLP-1 study after the GLP-1 receptor antagonist exendin (9-39) was discovered27,28The potential of GLP-1 as an anti-obesity agent has also raised much interest in the development of effective GIPR antagonists. Many different strategies have been undertaken to antagonize GIP function, such as small molecule receptor antagonists29Immunization against GIP30-32Various truncations and mutations of GIP molecules with antagonistic properties33-39And more recently a potent antagonist antibody against GIPR40

Under physiological conditions, the 42 amino acid hormone, GIP, is degraded by dipeptidyl peptidase 4(DPP-4), which cleaves at the third position of the GIP molecule to yield GIP 3-42. The artificially synthesized porcine GIP3-42 showed no antagonist properties to porcine or perfused rat pancreas at physiological concentrations, but antagonized human GIPR in vitro41. Many peptide hormones undergo post-translational modifications to yield a variety of biological forms with varying lengths and amino acid modifications42,43. Thus, GIP1-30 has been shown to be a translationResult of post-processing44And it is an agonist of GIPR33,45. If GIP1-30 is secreted into the human circulatory system, DPP-4 catalyzed cleavage will yield GIP 3-30.

US 7,875,587 discloses GIP receptor antagonists derived from GIP (1-42) with enhanced resistance to degradation by DPP-4, and their use in the treatment of insulin resistance and obesity. In WO2004/067548, DPP-4 metabolites are modified by covalent coupling of pharmacophores to achieve longer half-lives associated with peptide metabolites and to retain the biological activity of cleaved peptides similar to native peptides, including GIP. WO 2012/055770 discloses GIP (3-42) as an endogenous metabolite which is easy to be cleared and has a GIPR antagonist effect, while GIP (2-30) is exemplified as a truncated GIP analog having a GIPR agonist activity. WO 1998/24464 discloses the antagonist GIP (7-30).

The antagonists GIP (3-30) and GIP (5-30) were disclosed in WO 2016/034186 and Hansen et al 2016. Pathak et al disclose in 2015 GIP (3-30) C-terminally modified with 9 amino acid Cex from Exendin (1-39) and a lysine residue modified with palmitoyl group.

A range of different approaches have been used to modify the structure of GLP-1 compounds to provide longer in vivo action times. These methods involve introducing lipophilic substituents to amino acid residues (WO 96/29342 and WO 98/08871) and acylated GLP-1 analogues (WO 00/34331). WO 02/46227 discloses GLP-1 and exendin-4 analogs fused to human serum albumin to increase half-lives in vivo.

Disclosure of Invention

The present inventors have identified GIP (3-30) and GIP (5-30) peptide analogs that are modified by fatty acylation to increase peptide half-life (T)1/2) While retaining their highly potent GIPR antagonistic properties. Surprisingly, acylation did not occur at the 30 th position of the C-terminus of GIP (3-30) and GIP (5-30) while maintaining GIPR antagonistic properties.

In one aspect, the invention relates to formula 1: TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1) or formula 2: EGTFISDYSIAMDKIHQQDFVNWLLAQK (hGIP3-30, SEQ ID NO:2) is a glucose-dependent insulinotropic peptide (GIP) analog,

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In another aspect, the invention relates to the use of such GIP peptide analogues as a medicament.

In yet another aspect, the present invention relates to the use of such GIP peptide analogues in a method of antagonizing the GIP receptor or treating metabolic disorders (or metabolic syndromes) such as obesity, overweight, diabetes, insulin resistance and disorders of fatty acid metabolism. In other aspects, the invention relates to methods of treating cancer. In other aspects, the invention relates to methods of treating bone volume/density disorders.

Drawings

Fig. 1.With GIP (5-30) NH2The C16-diacid from position 5 to position 30 coupled lysine (Lys) scan shows a highly promising lipidation site for the development of highly potent, long-acting GIP receptor antagonists.

GIP (5-30) NH coupled to C16-diacid on lysine scanning with Lys2After incubation together, cAMP accumulation in transiently transfected COS-7 cells expressing human GIP receptor was assessed.

Fig. 2.With GIP (3-30) NH2The C16-diacid from position 5 to position 30 coupled lysine (Lys) scan shows a highly promising lipidation site for the development of highly potent, long-acting GIP receptor antagonists.

GIP (3-30) NH coupled to C16-diacid on lysine scanning with Lys2After incubation together, cAMP accumulation in transiently transfected COS-7 cells expressing human GIP receptor was assessed.

Fig. 3.Addition of a linker (a molecule that links fatty acids to peptides) improves the antagonism curve.

cAMP accumulation in transiently transfected COS-7 cells expressing human GIP receptor was assessed after incubation with different lipidated analogs with linkers. Antagonist dose response curves were plotted by inhibiting constant amounts of native GIP (1-42) (corresponding to 50% -80% of maximal receptor activation) with increasing concentrations of lipidated analogs. The curves are shown as mean ± SEM, n ═ 2.

Fig. 4.Lipidation increases the elimination half-life (T) of GIP analogs1/2)。

Treating pig with lipidated GIP (3-30) NH2The analogues AT117 and non-lipidated GIP (3-30) NH2Subcutaneous administration was performed. Blood samples were then collected from the central venous catheter at the indicated time points. Measurement of GIP (3-30) NH Using an internal radioimmunoassay2And the amount of AT 117.

Definition of

The term "affinity" refers to the strength of binding between a receptor and its ligand or ligands. In this context, the affinity of a peptide antagonist for its binding site (Ki) will determine the duration of inhibition of agonist activity. The affinity of the antagonists can be determined experimentally using either the Schild regression of functional studies or by radioligand binding studies such as 1) competitive binding experiments using the Cheng-Prusoff equation, 2) saturation binding experiments using the Scatchard equation or 3) the rate of binding and dissociation (K, respectively)onAnd Koff) By kinetic studies.

The term "IC 50" refers to the half maximal inhibitory concentration (IC50), which is a measure of the effectiveness of a substance in inhibiting a particular biological or biochemical function. This quantitative measure indicates how much of a particular drug or other substance (e.g., antagonist) is needed to inhibit a given biological process (or component of a process, i.e., enzyme, cell receptor, or microorganism) by half. In pharmacological studies, it is commonly used as a measure of the potency of antagonists. IC50 represents the drug concentration required to inhibit 50% in vitro. In this context, the IC50 value may also refer to the concentration of drug transferred from the receptor by 50% of the radiolabeled ligand, which is characteristic of drug affinity in competitive binding experiments.

In the present context, the term "agonist" refers to a peptide capable of binding to and activating a receptor.

In the present context, the term "antagonist" refers to a GIP peptide analogue as defined herein, which is capable of binding to and blocking or reducing an agonist-mediated receptor response. Antagonists generally do not elicit a biological response by themselves upon binding to the receptor. Antagonists have affinity but no efficacy for their cognate receptor, and binding disrupts the interaction and inhibits the function of agonists or inverse agonists at the receptor. Antagonists mediate their effects by binding to the active (orthosteric) site or allosteric site of the receptor, or they may interact at unique binding sites not normally involved in the biological regulation of receptor activity. The activity of the antagonist may be reversible or irreversible, depending on the longevity of the antagonist-receptor complex, which in turn depends on the nature of the antagonist-receptor binding. Most drug antagonists typically achieve their efficacy by competing with endogenous ligands or substrates for structurally defined binding sites on the receptor. Antagonists may be competitive, non-competitive, silent antagonists, partial agonists or inverse agonists.

Competitive antagonists (also known as surmountable antagonists) reversibly bind to the receptor at the same binding site (i.e., active site) as endogenous ligands or agonists, but do not activate the receptor. Agonists and antagonists therefore "compete" for the same binding site on the receptor. Once bound, the antagonist will block agonist binding. The level of receptor activity is determined by the relative affinity of each molecule for the site and its relative concentration. A high concentration of the competitive antagonist will increase the proportion of receptor occupied by the antagonist; to obtain the same degree of binding site occupancy, higher concentrations of agonist would be required.

The term "non-competitive antagonism" (also known as non-surmountable or non-surmountable antagonism) describes two different phenomena with functionally similar results: one is the active site of antagonist binding to the receptor and the other is the allosteric site of antagonist binding to the receptor. Unlike competitive antagonists, which affect the amount of agonist necessary to achieve a maximal response, but do not affect the magnitude of the maximal response, noncompetitive antagonists decrease the magnitude of the maximal response that can be achieved by any amount of agonist.

The term "silent antagonist" refers to a competitive receptor antagonist that absolutely does not activate the intrinsic activity of the receptor.

The term "partial agonist" refers to agonists that may differ in the magnitude of the functional response elicited after maximal receptor occupancy in a given receptor. In the presence of a full agonist (or more potent agonist), a partial agonist may act as a competitive antagonist because it competes for receptor occupancy with a full agonist, resulting in a net reduction in receptor activation compared to that observed with a full agonist alone.

The term "inverse agonist" refers to an agonist that acts similarly to an antagonist but elicits a series of different downstream biological responses. A constitutively active receptor exhibiting intrinsic or basal activity may have an inverse agonist that not only blocks the effect of a bound agonist, as does a classical antagonist, but also inhibits the basal activity of the receptor.

The term "individual" refers to a vertebrate, i.e., a specific member of a mammalian species, preferably a primate including a human. As used herein, "subject" and "individual" may be used interchangeably.

An "isolated peptide" is a peptide that is isolated and/or recovered from a component of its natural environment (typically the cellular environment) and is substantially free of contamination by cellular components, such as carbohydrates, lipids, or other protein impurities associated with polypeptides in nature. Typically, a preparation of an isolated peptide comprises the peptide in a highly purified form (i.e., at least about 80% pure, at least about 90% pure, at least about 95% pure, greater than 95% pure, or greater than 99% pure). The term "isolated" does not exclude the presence of the same peptide in alternative physical forms, such as dimers, tetramers or alternatively glycosylated or derivatized forms.

An "amino acid residue" may be a natural or non-natural amino acid residue connected by a peptide bond or a bond other than a peptide bond. The amino acid residue may be in the D-configuration or the L-configuration. The amino acid residue comprises amino terminal portions (NH) separated by a central portion comprising a carbon atom or a chain of carbon atoms2) And a carboxyl terminal moiety (COOH), the amino terminal moiety (NH)2) And a carboxyl terminal moiety (COOH) comprising at least one side chain or functional group. NH (NH)2Refers to an amino group present at the amino terminus of an amino acid or peptide, and COOH refers to a carboxyl group present at the carboxyl terminus of an amino acid or peptide. The generic term amino acid encompasses both natural and unnatural amino acids. Natural amino acids of standard nomenclature as listed in j.biol.chem.,243:3552-59(1969) and employed in section 37c.f.r., 1.822(b) (2) belong to the amino acid group listed herein: y, G, F, M, A, S, I, L, T, V, P, K, H, Q, E, W, R, D, N and C. Unnatural amino acids are those not listed immediately above. Furthermore, non-natural amino acid residues include, but are not limited to, modified amino acid residues, L-amino acid residues, and stereoisomers of D-amino acid residues.

An "equivalent amino acid residue" refers to an amino acid residue that can be substituted for another amino acid residue in a polypeptide without substantially altering the structure and/or function of the polypeptide. Thus, equivalent amino acids have similar properties, such as the volume of the side chain, the polarity of the side chain (polar or non-polar), the hydrophobicity (hydrophobic or hydrophilic), the pH (acidic, neutral or basic), and the side chain organization of the carbon molecule (aromatic/aliphatic). Thus, an "equivalent amino acid residue" may be considered a "conservative amino acid substitution".

In one embodiment, within the meaning of the term "equivalent amino acid substitution" as used herein, one amino acid of the amino acid group shown below may be substituted with another amino acid:

i) amino acids having polar side chains (Asp, Glu, Lys, Arg, His, Asn, Gln, Ser, Thr, Tyr and Cys)

ii) amino acids having nonpolar side chains (Gly, Ala, Val, Leu, Ile, Phe, Trp, Pro and Met)

iii) amino acids having aliphatic side chains (Gly, Ala, Val, Leu, Ile)

iv) amino acids with a cyclic side chain (Phe, Tyr, Trp, His, Pro)

v) amino acids having aromatic side chains (Phe, Tyr, Trp)

vi) amino acids having acidic side chains (Asp, Glu)

vii) amino acids having basic side chains (Lys, Arg, His)

viii) amino acids having amide side chains (Asn, Gln)

ix) amino acids with hydroxyl side chains (Ser, Thr)

x) amino acids having sulfur-containing side chains (Cys, Met)

xi) neutral, weakly hydrophobic amino acids (Pro, Ala, Gly, Ser, Thr)

xii) hydrophilic, acidic amino acids (Gln, Asn, Glu, Asp), and

xiii) hydrophobic amino acids (Leu, Ile, Val)

In the case where the L or D form (optical isomer) is not specified, it will be understood that the amino acid in question has the native L form (see Pure & appl. chem. Vol.56 (5) pp.595-624 (1984)) or the D form, and that the peptide so formed may be composed of the L form, the D form or a series of amino acids in a mixed form of the L and D forms.

A "functional variant" of a peptide is a peptide that is capable of performing substantially the same function as the peptide that is the functional variant. In particular, a functional variant may bind essentially the same molecule as the peptide it is a functional variant.

A "bioactive agent" (i.e., a substance/agent that is biologically active) is any agent, drug, compound, composition of matter, or mixture that provides some pharmacological effect (typically a beneficial effect) that may be demonstrated in vivo or in vitro. It refers to GIP peptide analogs as defined herein, as well as compounds or compositions comprising the same. The term as used herein also includes any physiologically or pharmacologically active substance that produces a local or systemic effect in an individual.

The term "drug" or "agent" as used herein includes biologically, physiologically or pharmacologically active substances which act locally or systemically on the human or animal body.

The terms "treatment" and "treating" as used herein refer to the management and care of a patient for the purpose of combating a condition, disease or disorder. The term is intended to include the full spectrum of treatments for a given condition in a patient, and equally refers to curative, prophylactic (preventative) and palliative or palliative treatments, such as administration of the peptide or composition for the following purposes: alleviating or alleviating the symptoms or complications; delay of progression of the condition, partial arrest of the clinical manifestation, disease or disorder; cure or eliminate the condition, disease or disorder; reduction or alleviation, and regression (whether partial or total) of a condition or symptom, whether detectable or undetectable; and/or preventing or reducing the risk of acquiring a condition, disease or disorder, wherein "preventing" is understood to mean managing and caring for a patient for the purpose of impeding the development of the condition, disease or disorder and includes administration of an active compound to prevent or reduce the risk of onset of symptoms or complications. The term "alleviate" and variations thereof as used herein means a reduction or prolongation of the extent of a physiological condition or symptom and/or the time course of an undesirable manifestation and/or progression as compared to the absence of administration of a composition of the present invention.

The individual to be treated is preferably a mammal, in particular a human. However, animal (such as mouse, rat, dog, cat, cow, horse, sheep, and pig) treatments are also included herein.

An "individual in need thereof" refers to an individual who may benefit from the present disclosure. In one embodiment, the individual in need thereof is a diseased individual, wherein the disease may be a metabolic disease or disorder (such as obesity or diabetes), a bone density disorder, or cancer.

The treatment according to the invention may be prophylactic, palliative and/or curative.

A "pharmacologically effective amount," "pharmaceutically effective amount," or "physiologically effective amount" of a biologically active agent is the amount of active agent present in a pharmaceutical composition described herein that is required to provide a desired level of active agent in the bloodstream or site of action (e.g., lung, gastric system, colorectal system, prostate, etc.) of the individual being treated to produce the desired physiological response when the composition is administered. In this context, bioactive agents refer to GIP peptide analogs disclosed herein.

As used herein, "Co-administration" refers to administration of one or more GIP peptide analogs of the present invention and the most advanced pharmaceutical compositions. The at least two components may be administered separately, sequentially or simultaneously.

Detailed Description

GIP refers to a glucose-dependent insulinotropic polypeptide, also known as a gastric inhibitory peptide (or polypeptide). The abbreviation GIP or hGIP as used herein is human GIP (Uniprot accession number P09681). GIP is derived from a 153 amino acid preprotein and circulates as a biologically active 42 amino acid peptide. It is synthesized by the mucosa of the duodenum and by K cells of the jejunum of the gastrointestinal tract.

GIPR (or GIP receptor) refers to gastric inhibitory polypeptide receptor. These 7 transmembrane proteins are found at least in the beta cells of the pancreas. The abbreviation GIPR or hGIPR as used herein is human GIPR (Uniprot accession number P48546).

The present inventors have identified GIP peptides that are antagonists of GIPR and are acylated herein to increase half-life and in vivo stability, while retaining surprising antagonistic properties. This makes them potentially useful in a range of therapeutic applications.

GIP peptides

The present invention relates to GIP peptide analogues comprising a peptide fragment of GIP (native or variant) having GIPR antagonistic properties, and one or more fatty acids attached thereto to increase the half-life of said peptide while retaining GIPR antagonistic properties.

In one aspect, a glucose-dependent insulinotropic peptide (GIP) analog is provided having formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000081

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In another aspect, a glucose-dependent insulinotropic peptide (GIP) analog is provided, which is selected from formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000091

and formula 2(hGIP3-30, SEQ ID NO: 2):

Figure BDA0002296622780000092

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the peptide or GIP peptide analog is C-terminally amidated (-NH)2)。

In one embodiment, the at least one fatty acid molecule is not attached to the Lys (K) residue at position 30 of either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID No. 1 and SEQ ID No. 2, wherein said amino acid residue 30 has been substituted with an amino acid selected from the group consisting of R, A and E.

In one embodiment, a glucose-dependent insulinotropic peptide (GIP) analog is provided comprising

A peptide selected from TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1) and EGTFISDYSIAMDKIHQQDFVNWLLAQK (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

which is optionally amidated at the C-terminus (-NH)2) And an

-at least one fatty acid molecule attached to one or more amino acid residues of any one of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to any one of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, a glucose-dependent insulinotropic peptide (GIP) analog is provided comprising

A peptide consisting of TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1) or consisting of EGTFISDYSIAMDKIHQQDFVNWLLAQK (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2, and

-at least one fatty acid molecule attached to one or more amino acid residues of any one of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to any one of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In another aspect, a glucose-dependent insulinotropic peptide (GIP) analog is provided having formula 1(hGIP6-30, SEQ ID NO: 147):

Figure BDA0002296622780000101

wherein the peptide optionally further comprises

N-terminal peptide T (hGIP5-30, SEQ ID NO:1),

the N-terminal dipeptide G-T (hGIP4-30, SEQ ID NO:148),

the N-terminal tripeptide E-G-T (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of any one of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148 or a functional variant having at least 75% sequence identity to any one of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148 or a functional variant having at least 75% sequence identity to any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148.

Thus, in one embodiment, a glucose-dependent insulinotropic peptide (GIP) analog is disclosed, which is selected from the group consisting of:

TFISDYSIAMDKIHQQDFVNWLLAQK(hGIP5-30,SEQ ID NO:1),

EGTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP3-30,SEQ ID NO:2),

FISDYSIAMDKIHQQDFVNWLLAQK (hGIP6-30, SEQ ID NO:147), and

GTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP4-30,SEQ ID NO:148),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of any one of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148 or a functional variant having at least 75% sequence identity to any one of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148 or a functional variant having at least 75% sequence identity to any of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 147 or SEQ ID NO 148.

Functional variants-function

The glucose-dependent insulinotropic peptide (GIP) analogs defined throughout comprise a peptide sequence (SEQ ID NO:1 or SEQ ID NO:2, and variants thereof) and at least one fatty acid molecule.

The terms "peptide" and "isolated peptide" are used interchangeably herein. The terms "variant" and "functional variant" may be used interchangeably herein. A peptide as defined herein includes the native peptide sequence as well as functional variants of the defined amino acid sequence of said peptide.

In one embodiment, there is provided a glucose-dependent insulinotropic peptide (GIP) analog TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1),

wherein the peptide optionally further comprises the dipeptide EG at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2, and

wherein the peptide is capable of binding and/or antagonizing a GIPR.

In one embodiment, the GIP peptide analog is capable of binding to a GIPR. In one embodiment, the GIP peptide analog is capable of antagonizing GIPR. In one embodiment, the GIP peptide analog is capable of binding to and antagonizing GIPR. In some embodiments, the GIPR is human GIPR (Uniprot accession No. P48546), mouse GIPR (Uniprot accession No. Q0P543), rat GIPR (Uniprot accession No. P43219), dog GIPR (Uniprot accession No. E2RIK5), porcine GIPR (Uniprot accession No. I3LND8), and/or rhesus GIPR (Uniprot accession No. A0A1D5QDM0) (primates).

In one embodiment, the GIP peptide analogs disclosed herein are antagonists of the hGIP receptor.

In one embodiment, the GIP peptide analogs disclosed herein are competitive antagonists of the hGIP receptor.

When referring to a "peptide" or "GIP peptide analogue" herein, the term will include reference to both the peptide analogue itself and the peptide analogue used in the method of treatment as defined herein.

A functional variant of a peptide of the present GIP peptide analogue is a functional equivalent of the peptide sequence, i.e. the functional variant retains at least some of the effects associated with the native peptide sequence.

In one embodiment, a functional variant of a peptide selected from SEQ ID NO:1 or SEQ ID NO:2 (or SEQ ID NO:147 or SEQ ID NO:148) retains the same biological activity or ability as the native peptide or the peptide from which it is derived. In one embodiment, the peptides and functional variants thereof as defined herein are capable of one or more of: binding one or more GIPR; antagonizing one or more GIPRs; replacement of GIP1-42 and/or GIP1-30 from one or more GIPRs; an affinity for a given GIPR that is higher than, equal to, or lower than that for GIP1-42 and/or GIP 1-30; antagonizing somatostatin secretion induced by native GIP, GIP1-42 and/or GIP 1-30; antagonizing native GIP, GIP1-42 and/or GIP1-30 induced insulin secretion; and antagonizing glucagon secretion induced by native GIP, GIP1-42 and/or GIP 1-30.

In one embodiment, a peptide and functional variants thereof are capable of binding (or are bound to) one or more of: hGIPR (Uniprot accession No. P48546), rGIPR (Uniprot accession No. P43219), mGIPR (Uniprot accession No. Q0P543), dog GIPR (Uniprot accession No. E2RIK5), pig GIPR (Uniprot accession No. I3LND8), and rhesus GIPR (Uniprot accession No. A0A1D5QDM0) (primates).

In one embodiment, a peptide and functional variants thereof are capable of inhibiting (reducing, antagonizing) one or more of the following: i) GIP-induced glucagon secretion, ii) GIP-induced insulin secretion, iii) GIP-induced somatostatin secretion, iv) GIP-induced glucose uptake, v) GIP-induced fatty acid synthesis and/or fatty acid incorporation, vi) higher or increased GIPR expression or activity, and vii) postprandial GIP release (postprandial GIP release).

Functional variant-mutant

In one embodiment, the functional variants of SEQ ID No. 1 and SEQ ID No. 2 as defined herein have at least 75% sequence identity with any one of SEQ ID No. 1 and SEQ ID No. 2. "identity" and "sequence identity" may be used interchangeably herein.

In another embodiment, the functional variant has at least 80% sequence identity to any one of SEQ ID NO 1 and SEQ ID NO 2.

In yet another embodiment, the functional variant has at least 85% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the functional variant has at least 90% sequence identity to any one of SEQ ID NO 1 and SEQ ID NO 2.

In another embodiment, the functional variant has at least 95% sequence identity to any of SEQ ID NO 1 and SEQ ID NO 2.

In another embodiment, the functional variant has at least 80% sequence identity, at least 85% sequence identity, at least 90% sequence identity, or at least 95% sequence identity to any one of SEQ ID NO:147 and SEQ ID NO: 148.

In one embodiment, the functional variant has 75% to 80% sequence identity to any of SEQ ID No. 1 and SEQ ID No. 2, such as 80% to 85%, such as 85% to 90%, such as 90% to 95% sequence identity to any of SEQ ID No. 1 and SEQ ID No. 2.

In one embodiment, there is provided a glucose-dependent insulinotropic peptide (GIP) analog TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1),

wherein the peptide optionally further comprises the dipeptide EG at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 with 1-6 individual amino acid substitutions,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2.

As used herein, 1 individual amino acid substitution is understood to be an amino acid substitution at any particular position in any one of SEQ ID NO:1 and SEQ ID NO:2 that is substituted independently of any other feature (e.g., other amino acid substitutions or fatty acid modifications) in any one of SEQ ID NO:1 and SEQ ID NO: 2.

In one embodiment, the functional variant of any of SEQ ID No. 1 and SEQ ID No. 2 has from 1 to 6 individual amino acid substitutions, such as from 1 to 2, from 2 to 3, from 3 to 4, from 4 to 5 or from 5 to 6 individual amino acid substitutions, as compared to the corresponding portion or position of any of SEQ ID No. 1 and SEQ ID No. 2 (or between the variant and the corresponding native GIP peptide).

In one embodiment, the functional variant of any of SEQ ID NO:147 and SEQ ID NO:148 has from 1 to 6 individual amino acid substitutions, such as from 1 to 2, from 2 to 3, from 3 to 4, from 4 to 5 or from 5 to 6 individual amino acid substitutions, as compared to the corresponding portion or position of any of SEQ ID NO:147 and SEQ ID NO:148 (or between the variant and the corresponding native GIP peptide).

In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 1 individual amino acid substitution. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 1 individual amino acid substitution.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 2 individual amino acid substitutions. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 2 individual amino acid substitutions.

In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 3 individual amino acid substitutions. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 3 individual amino acid substitutions.

In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 4 individual amino acid substitutions. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 4 individual amino acid substitutions.

In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 5 individual amino acid substitutions. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 5 individual amino acid substitutions.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has 6 individual amino acid substitutions. In one embodiment, said functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 has NO more than 6 individual amino acid substitutions.

In one embodiment, one or more or all of the amino acid substitutions are conservative amino acid substitutions (or synonymous substitutions). Conservative substitutions are substitutions of amino acids whose side chains have similar biochemical properties and therefore do not affect the function of the peptide.

Among the common amino acids, for example, a "conservative amino acid substitution" can also be illustrated by substitutions between amino acids within the following groups: (1) glycine, alanine, valine, leucine and isoleucine, (2) phenylalanine, tyrosine and tryptophan, (3) serine and threonine, (4) aspartic acid and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine.

In one embodiment, the serine residue of the peptide of the invention is substituted with an amino acid selected from the group consisting of gin, Asn, and Thr (all amino acids having a polar uncharged side chain); and the glycine residue (Gly) is independently substituted with an amino acid selected from Ala, Val, Leu and Ile; and the arginine residue (Arg) is independently substituted with an amino acid selected from Lys and His, both having a positively charged side chain; and its lysine residue (Lys) is independently substituted with an amino acid selected from Arg and His; and the methionine residue (Met) thereof is independently substituted with an amino acid selected from Leu, Pro, Ile, Val, Phe, Tyr and Trp, each having a hydrophobic side chain; and its glutamine residue (Gln) is independently substituted with an amino acid selected from Asp, Glu and Asn; and the alanine residue (Ala) is independently substituted with an amino acid selected from the group consisting of Gly, Val, Leu and Ile.

Specific amino acid substitutions disclosed herein are K to R, E to D, L to M, Q to E, I to V, I to L, A to S, Y to W, K to Q, S to T, N to S, M to L and Q to R.

Identity between amino acid sequences can be calculated using well known algorithms (such as BLOSUM 30, BLOSUM 40, BLOSUM 45, BLOSUM 50, BLOSUM 55, BLOSUM 60, BLOSUM 62, BLOSUM 65, BLOSUM 70, BLOSUM75, BLOSUM 80, BLOSUM 85, or BLOSUM 90) or by simply comparing specific amino acids present at corresponding positions in the two peptide sequences to be compared.

Homology may be used as a synonym for identity/sequence identity.

In another embodiment, a functional variant as defined herein comprises the following sequence: wherein an alkyl amino acid replaces an alkyl amino acid, wherein an aromatic amino acid replaces an aromatic amino acid, wherein a sulfur amino acid replaces a sulfur amino acid, wherein a hydroxyl amino acid replaces a hydroxyl amino acid, wherein an acidic amino acid replaces an acidic amino acid, wherein a basic amino acid replaces a basic amino acid, and/or wherein a dibasic monocarboxylic amino acid replaces a dibasic monocarboxylic amino acid.

Conservative substitutions may be introduced at any one or more positions of a peptide selected from any one of SEQ ID NO. 1 and SEQ ID NO. 2, as long as the resulting variant remains functional. However, it may also be desirable to introduce non-conservative substitutions (non-synonymous substitutions) at one or more positions.

In one embodiment, the non-conservative substitutions that result in the formation of a variant of a peptide selected from any one of SEQ ID NO:1 and SEQ ID NO:2 comprise substitutions of amino acid residues: i) substantially different in polarity, e.g., a residue with a non-polar side chain (Ala, Leu, Pro, Trp, Val, Ile, Leu, Phe, or Met) is substituted for a residue with a polar side chain (such as Gly, Ser, Thr, Cys, Tyr, Asn, or Gln or a charged amino acid (such as Asp, Glu, Arg, or Lys), or a charged or polar residue is substituted for a non-polar residue; and/or ii) its effect on the orientation of the peptide backbone is substantially different, such as Pro or Gly being substituted by another amino acid or another amino acid for Pro or Gly; and/or iii) substantially different charge, e.g., a negatively charged residue (such as Glu or Asp) in place of a positively charged residue (such as Lys, His or Arg) (and vice versa); and/or iv) substantially different in steric bulk, e.g., substitution of a bulky residue (such as His, Trp, Phe, or Tyr) for a residue with a smaller side chain (e.g., Ala, Gly, or Ser) (and vice versa).

In one embodiment, amino acid substitutions may be made based on their hydrophobicity and hydrophilicity values, as well as the relative similarity (including charge, size, etc.) of the amino acid side-chain substituents.

A peptide or functional variant counterpart thereof as defined herein comprises a proprotein or a natural amino acid, i.e. 22 amino acids naturally incorporated in a polypeptide. Of these, 20 are encoded by the universal genetic code, and the remaining 2 (selenocysteine (Sec, U) and pyrrolysine (Pyl, O)) are incorporated into proteins by unique synthetic mechanisms.

In one embodiment, a peptide as defined herein comprises one or more non-naturally occurring amino acid residues (non-natural, non-proteinogenic or non-standard amino acids). Non-naturally occurring amino acids include, for example, but are not limited to, beta-2-naphthyl-alanine, trans-3-methylproline, 2, 4-methylproline, cis-4-hydroxyproline, ornithine (Orn), trans-4-hydroxyproline, N-methylglucamine, allothreonine, methylthreonine, hydroxyethylcysteine, hydroxyethylhomocysteine, nitroglutamic acid, homoglutamic acid, pipecolic acid, thiazolidinecarboxylic acid, dehydroproline, 3-and 4-methylproline, 3-dimethylproline, tert-leucine, norleucine (Nle), methoxyamino acid (moxine, Mox), pent-aine, 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine and 4-fluorophenylalanine.

In one embodiment, the amino acid Met is substituted with an antioxidant amino acid analog, such as norleucine (Nle), which retains the length of the amino acid side chain important for hydrophobic interactions, but does not retain its hydrogen bonding properties; or a methoxyamino acid (Mox), an atypical amino acid that more closely resembles the electronic properties of Met than Nle.

Any amino acid as defined herein may be in the L or D configuration. If not specified, L-isomeric forms are preferred.

The standard and/or non-standard amino acids may be linked by peptide bonds (to form a linear peptide chain) or by non-peptide bonds (e.g. by variable side chains of amino acids). Preferably, the amino acids of the peptides defined herein are linked by peptide bonds.

The term peptide also includes post-translational modifications introduced by chemical or enzymatic reactions, as known in the art. These include acetylation, phosphorylation, methylation, glycosylation, glycation, amidation, hydroxylation, deimidation, carbamylation, and sulfation of one or more amino acid residues, as well as proteolytic modifications by known proteases, including lysosomal cathepsins, as well as calpains, secretases, and matrix-metalloproteinases.

In a preferred embodiment, the peptide selected from SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof is amidated, such as by C-terminal amidation (-NH)2). In an exemplary embodiment thereof, the peptide is hGIP (5-30) -NH2Or hGIP (3-30) -NH2Or a variant thereof, further comprising a fatty acid molecule.

In one embodiment, the peptide selected from SEQ ID NO:1 and SEQ ID NO:2, or a functional variant thereof, is acetylated, such as N-terminally acetylated.

Likewise, functional equivalents of the peptides may comprise chemical modifications such as ubiquitination, labeling (e.g., with radionuclides, various enzymes, etc.), pegylation (derivatization with polyethylene glycol), or substitution by insertion (or by chemical synthesis) of amino acids (such as ornithine) that are not normally found (non-proteinogenic) in human proteins.

Spatially similar compounds can be formulated to mimic key portions of peptide structure. This can be achieved by modeling and chemical design techniques known to those skilled in the art. For example, esterification and other alkylation may be employed to modify, for example, the amino terminus of the di-arginine peptide backbone to mimic the tetrapeptide structure. It is understood that all such spatially similar constructs fall within the scope of the present invention. Peptides having N-terminal and C-terminal alkylation and esterification are also included in the present invention.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein an amino acid selected from N, Q and T is substituted with an amino acid selected from D, E and S. In one embodiment, such amino acid substitutions increase the solubility of the resulting peptide.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 30 is substituted with any amino acid.

Any amino acid as used herein refers to both naturally occurring and non-naturally occurring amino acids as defined herein.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 30 is substituted with a conservative amino acid substitution.

In one embodiment, the functional variant of any of SEQ ID NO. 1 and SEQ ID NO. 2 is a variant wherein a hydrophobic amino acid is substituted with a hydrophilic amino acid.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 30 is substituted with a hydrophilic amino acid.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 30 is substituted with an amino acid selected from D, E, S, R and A, such as an amino acid selected from R, A and E.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 30 is substituted with any amino acid when K at position 30 is not modified by attaching a fatty acid molecule.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 16 is substituted with any amino acid.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 16 is substituted with a conservative amino acid substitution.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 16 is substituted with an amino acid selected from D, E, S, R and A, such as an amino acid selected from R, A and E.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein K at position 16 is substituted with any amino acid when K at position 16 is not modified by attaching a fatty acid molecule.

In one embodiment, the functional variant of any of SEQ ID No. 1 and SEQ ID No. 2 is a variant wherein both K at position 16 and K at position 30 are substituted with any amino acid, such as a conservative amino acid substitution, such as with an amino acid selected from D, E, S, R and a, such as with an amino acid selected from R, A and E.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein M at position 14 is substituted with any amino acid, such as L, S, K, norleucine (Nle) or a methoxyamino acid (Mox).

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein D at position 15 is substituted with any amino acid.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein D at position 15 is substituted with a conservative amino acid substitution.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein D at position 15 is substituted with an amino acid selected from the group consisting of E, A, Orn and K.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein H at position 18 is substituted with any amino acid.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein the H at position 18 is replaced with a conservative amino acid substitution.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein H at position 18 is substituted with an amino acid selected from the group consisting of A, R, Orn and K.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein S at position 11 is substituted with an amino acid selected from Orn and K.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein position 12I is substituted with an amino acid selected from Orn and K.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein position 13A is substituted with an amino acid selected from Orn and K.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant having (comprising) amino acid substitutions, including conservative and non-conservative amino acid substitutions, at one or more positions of 11(Ser), 12(Ile), 13(Ala), 14(Met), 15(Asp), 16(Lys), 18(His) and 30 (Lys).

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein one or more amino acid residues are substituted with ornithine. In one embodiment, a variant mentioned as substituted with a Lys residue as defined herein may likewise be substituted with an Orn residue. Both Lys and Orn may be modified at their side chain amino groups (. epsilon. -or. delta. -amino groups, respectively).

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein one or more amino acid residues are substituted with ornithine to which a fatty acid molecule is attached.

In one embodiment, the functional variant of any of SEQ ID NO 1 and SEQ ID NO 2 is a variant wherein one of the amino acid residues is substituted with ornithine.

In one embodiment, the functional variant of SEQ ID NO. 2 is a variant wherein E (Glu) at position 3 is substituted with pGlu (pyroglutamic acid).

In one embodiment, the peptide is non-naturally occurring.

In one embodiment, the peptide is synthetic.

In one embodiment, the peptide is an isolated peptide.

GIP (5-30) peptide

In one embodiment, a GIP peptide analog is provided, the peptide having the sequence TFISDYX11X12X13X14X1 5X16IX18QQDFVNWLLAQX30(SEQ ID NO:3), wherein

X11Selected from the group consisting of S, K and Orn,

X12selected from the group consisting of I, K and Orn,

X13selected from the group consisting of A, K and Orn,

X14selected from the group consisting of M, K, L, S, Nle and Mox,

X15selected from the group consisting of D, E, A, K and Orn,

X16selected from the group consisting of K, R, A and E,

X18selected from H, A, R, K and Orn, and

X30selected from the group consisting of K, R, A and E,

or a functional variant thereof having at least 75% sequence identity to said sequence,

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues of the sequence, with the proviso that the at least one fatty acid molecule is not attached to X30The above.

In one embodiment, the peptide of the present GIP peptide analog is selected from the group consisting of:

TFISDYSIAMDKIHQQDFVNWLLAQK(hGIP5-30,SEQ ID NO:1),

TFISDYSIAMDRIKQQDFVNWLLAQR(hGIP(5-30)K16R H18K K30R;SEQ ID NO:5),

TFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(5-30)K16R K30R;SEQ ID NO:6),

TFISDYSIAMDKIAQQDFVNWLLAQK(hGIP(5-30)H18A;SEQ ID NO:7),

TFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(5-30)H18K;SEQ ID NO:8),

TFISDYSIAMEKIAQQDFVNWLLAQK(hGIP(5-30)D15E H18A;SEQ ID NO:9),

TFISDYSIAMDAIAQQDFVNWLLAQK(hGIP(5-30)K16A H18A;SEQ ID NO:10),

TFISDYSIAMEKIHQQDFVNWLLAQK(hGIP(5-30)D15E;SEQ ID NO:11),

TFISDYSIAMNKIHQQDFVNWLLAQK(hGIP(5-30)D15N;SEQ ID NO:12),

TFISDYSIAMDAIHQQDFVNWLLAQK(hGIP(5-30)K16A;SEQ ID NO:13),

TFISDYSIAMDHIHQQDFVNWLLAQK(hGIP(5-30)K16H;SEQ ID NO:14),

TFISDYSIAMDRIHQQDFVNWLLAQK(hGIP(5-30)K16R;SEQ ID NO:15),

TFISDYSIAMDKIFQQDFVNWLLAQK(hGIP(5-30)H18F;SEQ ID NO:16),

TFISDYSIAMDKIWQQDFVNWLLAQK(hGIP(5-30)H18W;SEQ ID NO:17),

TFISDYSIAMDKIHQQDFVNWLLAQR(hGIP(5-30)K30R;SEQ ID NO:18),

TFISDYSIAMDKIHQQDFVNWLLAQH(hGIP(5-30)K30H;SEQ ID NO:19),

KFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)T5K,SEQ ID NO:20),

TKISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)F6K,SEQ ID NO:21),

TFKSDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)I7K,SEQ ID NO:22),

TFIKDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S8K,SEQ ID NO:23),

TFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)D9K,SEQ ID NO:24),

TFISDKSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)Y10K,SEQ ID NO:25),

TFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S11K,SEQ ID NO:26),

TFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(5-30)I12K,SEQ ID NO:27),

TFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(5-30)A13K,SEQ ID NO:28),

TFISDYSIAKDKIHQQDFVNWLLAQK(hGIP(5-30)M14K,SEQ ID NO:29),

TFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(5-30)D15K,SEQ ID NO:30),

TFISDYSIAMDKKHQQDFVNWLLAQK(hGIP(5-30)I17K,SEQ ID NO:31),

TFISDYSIAMDKIHKQDFVNWLLAQK(hGIP(5-30)Q19K,SEQ ID NO:32),

TFISDYSIAMDKIHQKDFVNWLLAQK(hGIP(5-30)Q20K,SEQ ID NO:33),

TFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(5-30)D21K,SEQ ID NO:34),

TFISDYSIAMDKIHQQDKVNWLLAQK(hGIP(5-30)F22K,SEQ ID NO:35),

TFISDYSIAMDKIHQQDFKNWLLAQK(hGIP(5-30)V23K,SEQ ID NO:36),

TFISDYSIAMDKIHQQDFVKWLLAQK(hGIP(5-30)N24K,SEQ ID NO:37),

TFISDYSIAMDKIHQQDFVNKLLAQK(hGIP(5-30)W25K,SEQ ID NO:38),

TFISDYSIAMDKIHQQDFVNWKLAQK(hGIP(5-30)L26K,SEQ ID NO:39),

TFISDYSIAMDKIHQQDFVNWLKAQK(hGIP(5-30)L27K,SEQ ID NO:40),

TFISDYSIAMDKIHQQDFVNWLLKQK(hGIP(5-30)A28K,SEQ ID NO:41),

TFISDYSIAMDKIHQQDFVNWLLAKK(hGIP(5-30)Q29K,SEQ ID NO:42),

TFISDYKIAMDKIAQQDFVNWLLAQK(hGIP(5-30)S11K H18A,SEQ ID NO:43),

TFISDYKIAMDKIRQQDFVNWLLAQK(hGIP(5-30)S11K H18R,SEQ ID NO:44),

TFISDYKIAMEKIHQQDFVNWLLAQK(hGIP(5-30)S11K D15E,SEQ ID NO:45),

TFISDYKIANleDKIHQQDFVNWLLAQK(hGIP(5-30)S11K M14Nle,SEQ ID NO:46),

TFISDYKIALDKIHQQDFVNWLLAQK(hGIP(5-30)S11K M14L,SEQ ID NO:47),

TFISDYSKAMDKIAQQDFVNWLLAQK(hGIP(5-30)I12K H18A,SEQ ID NO:48),

TFISDYSKAMDKIRQQDFVNWLLAQK(hGIP(5-30)I12K H18R,SEQ ID NO:49),

TFISDYSKAMEKIHQQDFVNWLLAQK(hGIP(5-30)I12K D15E,SEQ ID NO:50),

TFISDYSKANleDKIHQQDFVNWLLAQK(hGIP(5-30)I12K M14Nle,SEQ ID NO:51),

TFISDYSKALDKIHQQDFVNWLLAQK(hGIP(5-30)I12K M14L,SEQ ID NO:52),

TFISDYSIKMDKIAQQDFVNWLLAQK(hGIP(5-30)A13K H18A,SEQ ID NO:53),

TFISDYSIKMDKIRQQDFVNWLLAQK(hGIP(5-30)A13K H18R,SEQ ID NO:54),

TFISDYSIKMEKIHQQDFVNWLLAQK(hGIP(5-30)A13K D15E,SEQ ID NO:55),

TFISDYSIKNleDKIHQQDFVNWLLAQK(hGIP(5-30)A13K M14Nle,SEQ ID NO:56),

TFISDYSIKLDKIHQQDFVNWLLAQK(hGIP(5-30)A13K M14L,SEQ ID NO:57),

TFISDYSIAMEKIKQQDFVNWLLAQK(hGIP(5-30)D15E H18K;SEQ ID NO:58),

TFISDYSIANleDKIKQQDFVNWLLAQK(hGIP(5-30)M14Nle H18K;SEQ ID NO:59),

TFISDYSIALDKIKQQDFVNWLLAQK(hGIP(5-30)M14L H18K;SEQ ID NO:60),

TFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(5-30)D15Orn,SEQ ID NO:61),

TFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(5-30)H18Orn;SEQ ID NO:62),

TFISDYSIALDRIKQQDFVNWLLAQR(hGIP(5-30)M14L K16R H18K K30R;SEQ ID NO:63),

KFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(5-30)T5K K16R K30R,SEQ ID NO:64)

KFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)T5K M14L K16R K30R,SEQ ID NO:65),

TFISDYKIAMDRIHQQDFVNWLLAQR(hGIP(5-30)S11K K16R K30R,SEQ ID NO:66),

TFISDYKIALDRIHQQDFVNWLLAQR(hGIP(5-30)S11K M14L K16R K30R,SEQ ID NO:67),

TFISDYSKAMDRIHQQDFVNWLLAQR(hGIP(5-30)I12K K16R K30R,SEQ ID NO:68),

TFISDYSKALDRIHQQDFVNWLLAQR(hGIP(5-30)I12K M14L K16R K30R,SEQ ID NO:69),

TFISDYSIKMDRIHQQDFVNWLLAQR(hGIP(5-30)A13K K16R K30R,SEQ ID NO:70),

TFISDYSIKLDRIHQQDFVNWLLAQR (hGIP (5-30) A13K M14L K16R K30R, SEQ ID NO:71), and

TFISDYSIAMDRIHQQKFVNWLLAQR(SEQ ID NO:145),

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues, such as by attaching a fatty acid molecule at one amino acid residue, with the proviso that the at least one fatty acid molecule is not attached to an amino acid residue at position 30, such as to amino acid residue K or R at position 30

In one embodiment, the fatty acid molecule is attached to the K residues within positions 5-29 of SEQ ID NO 1 and variants thereof. In one embodiment, the fatty acid molecule is attached to the N-terminal amino acid residue, i.e., the T residue at position 5 of SEQ ID NO 1.

In one embodiment, the peptide is C-terminally amidated (-NH)2)。

GIP (3-30) peptide

In one embodiment, a GIP peptide analog is provided, the peptide having the sequence EGTFISDY X11X12X13X1 4X15X16IX18QQDFVNWLLAQX30(SEQ ID NO:4), wherein

X11Selected from the group consisting of S, K and Orn,

X12selected from the group consisting of I, K and Orn,

X13selected from the group consisting of A, K and Orn,

X14selected from the group consisting of M, K, L, S, Nle and Mox,

X15selected from the group consisting of D, E, A, K and Orn,

X16selected from the group consisting of K, R, A and E,

X18selected from H, A, R, K and Orn, and

X30selected from the group consisting of K, R, A and E,

or a functional variant thereof having at least 75% sequence identity to said sequence,

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues of the sequence, with the proviso that the at least one fatty acid molecule is not attached to X30The above.

In one embodiment, the peptide of the present GIP peptide analog is selected from the group consisting of:

EGTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP3-30,SEQ ID NO:2),

EGTFISDYSIAMDRIKQQDFVNWLLAQR(hGIP(3-30)K16R H18K K30R;SEQ ID NO:72),

EGTFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(3-30)K16R K30R;SEQ ID NO:73),

EGTFISDYSIAMDKIAQQDFVNWLLAQK(hGIP(3-30)H18A;SEQ ID NO:74),

EGTFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(3-30)H18K;SEQ ID NO:75),

EGTFISDYSIAMEKIAQQDFVNWLLAQK(hGIP(3-30)D15E H18A;SEQ ID NO:76),

EGTFISDYSIAMDAIAQQDFVNWLLAQK(hGIP(3-30)K16A H18A;SEQ ID NO:77),

EGTFISDYSIAMEKIHQQDFVNWLLAQK(hGIP(3-30)D15E;SEQ ID NO:78),

EGTFISDYSIAMNKIHQQDFVNWLLAQK(hGIP(3-30)D15N;SEQ ID NO:79),

EGTFISDYSIAMDAIHQQDFVNWLLAQK(hGIP(3-30)K16A;SEQ ID NO:80),

EGTFISDYSIAMDHIHQQDFVNWLLAQK(hGIP(3-30)K16H;SEQ ID NO:81),

EGTFISDYSIAMDRIHQQDFVNWLLAQK(hGIP(3-30)K16R;SEQ ID NO:82),

EGTFISDYSIAMDKIFQQDFVNWLLAQK(hGIP(3-30)H18F;SEQ ID NO:83),

EGTFISDYSIAMDKIWQQDFVNWLLAQK(hGIP(3-30)H18W;SEQ ID NO:84),

EGTFISDYSIAMDKIHQQDFVNWLLAQR(hGIP(3-30)K30R;SEQ ID NO:85),

EGTFISDYSIAMDKIHQQDFVNWLLAQH(hGIP(3-30)K30H;SEQ ID NO:86),

EKTFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)G4K,SEQ ID NO:87),

EGKFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)T5K,SEQ ID NO:88),

EGTKISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)F6K,SEQ ID NO:89),

EGTFKSDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)I7K,SEQ ID NO:90),

EGTFIKDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S8K,SEQ ID NO:91),

EGTFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)D9K,SEQ ID NO:92),

EGTFISDKSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)Y10K,SEQ ID NO:93),

EGTFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S11K,SEQ ID NO:94),

EGTFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(3-30)I12K,SEQ ID NO:95),

EGTFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(3-30)A13K,SEQ ID NO:96),

EGTFISDYSIAKDKIHQQDFVNWLLAQK(hGIP(3-30)M14K,SEQ ID NO:97),

EGTFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(3-30)D15K,SEQ ID NO:98),

EGTFISDYSIAMDKKHQQDFVNWLLAQK(hGIP(3-30)I17K,SEQ ID NO:99),

EGTFISDYSIAMDKIHKQDFVNWLLAQK(hGIP(3-30)Q19K,SEQ ID NO:100),

EGTFISDYSIAMDKIHQKDFVNWLLAQK(hGIP(3-30)Q20K,SEQ ID NO:101),

EGTFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(3-30)D21K,SEQ ID NO:102),

EGTFISDYSIAMDKIHQQDKVNWLLAQK(hGIP(3-30)F22K,SEQ ID NO:103),

EGTFISDYSIAMDKIHQQDFKNWLLAQK(hGIP(3-30)V23K,SEQ ID NO:104),

EGTFISDYSIAMDKIHQQDFVKWLLAQK(hGIP(3-30)N24K,SEQ ID NO:105),

EGTFISDYSIAMDKIHQQDFVNKLLAQK(hGIP(3-30)W25K,SEQ ID NO:106),

EGTFISDYSIAMDKIHQQDFVNWKLAQK(hGIP(3-30)L26K,SEQ ID NO:107),

EGTFISDYSIAMDKIHQQDFVNWLKAQK(hGIP(3-30)L27K,SEQ ID NO:108),

EGTFISDYSIAMDKIHQQDFVNWLLKQK(hGIP(3-30)A28K,SEQ ID NO:109),

EGTFISDYSIAMDKIHQQDFVNWLLAKK(hGIP(3-30)Q29K,SEQ ID NO:110),

EGTFISDYKIAMDKIAQQDFVNWLLAQK(hGIP(3-30)S11K H18A,SEQ ID NO:111),

EGTFISDYKIAMDKIRQQDFVNWLLAQK(hGIP(3-30)S11K H18R,SEQ ID NO:112),

EGTFISDYKIAMEKIHQQDFVNWLLAQK(hGIP(3-30)S11K D15E,SEQ ID NO:113),

EGTFISDYKIANleDKIHQQDFVNWLLAQK(hGIP(3-30)S11K M14Nle,SEQ ID NO:114),

EGTFISDYKIALDKIHQQDFVNWLLAQK(hGIP(3-30)S11K M14L,SEQ ID NO:115),

EGTFISDYSKAMDKIAQQDFVNWLLAQK(hGIP(3-30)I12K H18A,SEQ ID NO:116),

EGTFISDYSKAMDKIRQQDFVNWLLAQK(hGIP(3-30)I12K H18R,SEQ ID NO:117),

EGTFISDYSKAMEKIHQQDFVNWLLAQK(hGIP(3-30)I12K D15E,SEQ ID NO:118),

EGTFISDYSKANleDKIHQQDFVNWLLAQK(hGIP(3-30)I12K M14Nle,SEQ ID NO:119),

EGTFISDYSKALDKIHQQDFVNWLLAQK(hGIP(3-30)I12K M14L,SEQ ID NO:120),

EGTFISDYSIKMDKIAQQDFVNWLLAQK(hGIP(3-30)A13K H18A,SEQ ID NO:121),

EGTFISDYSIKMDKIRQQDFVNWLLAQK(hGIP(3-30)A13K H18R,SEQ ID NO:122),

EGTFISDYSIKMEKIHQQDFVNWLLAQK(hGIP(3-30)A13K D15E,SEQ ID NO:123),

EGTFISDYSIKNleDKIHQQDFVNWLLAQK(hGIP(3-30)A13K M14Nle,SEQ ID NO:124),

EGTFISDYSIKLDKIHQQDFVNWLLAQK(hGIP(3-30)A13K M14L,SEQ ID NO:125),

EGTFISDYSIAMEKIKQQDFVNWLLAQK(hGIP(3-30)D15E H18K;SEQ ID NO:126),

EGTFISDYSIANleDKIKQQDFVNWLLAQK(hGIP(3-30)M14Nle H18K;SEQ ID NO:127),

EGTFISDYSIALDKIKQQDFVNWLLAQK(hGIP(3-30)M14L H18K;SEQ ID NO:128),

EGKFISDYSIAMDRIHQQDFVNWLLAQR(hGIP(3-30)T5K K16R K30R,SEQ ID NO:129),

EGTFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(3-30)D15Orn,SEQ ID NO:130),

EGTFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(3-30)H18Orn;SEQ ID NO:131),

EGTFISDYSIALDRIKQQDFVNWLLAQR(hGIP(3-30)M14L K16R H18K K30R;SEQ ID NO:132),

EGWFISDYSIAMEKIAQQDFVNWLLAQK (SEQ ID NO:144), and

EGTFISDYSIAMDKIKQQDFVNWLLAQR(SEQ ID NO:146),

wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues, such as by attaching one fatty acid molecule at one amino acid residue, with the proviso that the at least one fatty acid molecule is not attached to an amino acid residue at position 30, such as to amino acid residue K or R at position 30.

In one embodiment, the fatty acid molecule is attached to the K residues within positions 4-29 of SEQ ID NO 2 and variants thereof.

In one embodiment, the peptide is C-terminally amidated (-NH)2)。

Attachment of fatty acid molecules

In one embodiment, a glucose-dependent insulinotropic peptide (GIP) analog is provided comprising or consisting of peptide sequence TFISDYSIAMDKIHQQDFVNWLLAQK (hGIP5-30, SEQ ID NO:1) (optionally amidated at the C-terminus) or a functional variant having at least 75% sequence identity to SEQ ID NO:1,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of SEQ ID NO 1 or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of SEQ ID NO 1 or to amino acid residue 30 of a functional variant having at least 75% sequence identity to SEQ ID NO 1.

In another embodiment, a glucose-dependent insulinotropic peptide (GIP) analog is provided comprising or consisting of peptide sequence EGTFISDYSIAMDKIHQQDFVNWLLAQK (hGIP3-30, SEQ ID NO:2) (optionally amidated at the C-terminus) or a functional variant having at least 75% sequence identity to SEQ ID NO: 2.

Wherein the peptide is modified by attaching at least one fatty acid molecule to one or more of the amino acid residues of SEQ ID NO. 2 or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of SEQ ID NO. 2 or to amino acid residue 30 of a functional variant having at least 75% sequence identity to SEQ ID NO. 2.

In one embodiment, the at least one fatty acid molecule is not attached to the N-terminal amino acid residue at position 3 of SEQ ID No. 2, such as not attached to the N-terminal E at position 3 of SEQ ID No. 2.

In one embodiment, the fatty acid molecule is attached to an amino acid residue at any of positions 5 to 29 of SEQ ID NO 1 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to an amino acid residue at any one of positions 4 to 29 of SEQ ID NO 2 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to the amino acid residue at any one of positions 6 to 29 of SEQ ID NO:147 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to the amino acid residue at any one of positions 4 to 29 of SEQ ID NO 148 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to the amino acid residue at position 5, 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29 of SEQ ID No. 1 or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the amino acid residue at position 4, 5, 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29 of SEQ ID No. 2 or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the amino acid residue at position 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29 of SEQ ID NO 147 or a functional variant thereof.

In one embodiment, at least one fatty acid molecule is attached to one or more amino acid residues in the middle region of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

In one embodiment, at least one fatty acid molecule is attached to one or more amino acid residues at any of positions 11 to 21 of any of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

In one embodiment, at least one fatty acid molecule is attached to one or more amino acid residues at any of positions 11 to 12, 12 to 13, 13 to 14, 14 to 15, 15 to 16, 16 to 17, 17 to 18, 18 to 19, 19 to 20 or 20 to 21 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a functional variant thereof.

In one embodiment, at least one fatty acid molecule is attached to one or more amino acid residues at any of positions 11, 12 and 13 of any of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues of the N-terminal region of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 3 of SEQ ID NO 2.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 5 to 10 of SEQ ID No. 1 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 5 to 6, 6 to 7, 7 to 8, 8 to 9 or 9 to 10 of SEQ ID No. 1 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 4 to 10 of SEQ ID No. 2 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 4 to 5,5 to 6, 6 to 7, 6 to 8, 8 to 9 or 9 to 10 of SEQ ID No. 2 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues of the C-terminal region of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 22 to 29 of any one of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

In one embodiment, the at least one fatty acid molecule is attached to one or more amino acid residues at any one of positions 22 to 23, 23 to 24, 24 to 25, 25 to 26, 26 to 27, 27 to 28 or 28 to 29 of any one of SEQ ID No. 1 and SEQ ID No. 2 or a functional variant thereof.

In one embodiment, the lipidAttachment of fatty acid molecules to amino-alkyl groups (-C) with side chainsnH2nNH2) At one or more amino acid residues.

In one embodiment, the fatty acid molecule is attached to a linker having a pendant amino group (NH)2) At one or more amino acid residues.

In one embodiment, the fatty acid molecule is attached to a side chain amino group of an amino acid residue.

In one embodiment, the fatty acid molecule is attached to the epsilon side chain amino group of a lysine residue (Lys, K).

In one embodiment, the fatty acid molecule is attached to the delta side chain amino group of an ornithine residue (Orn).

In one embodiment, the amino acid residue to which the fatty acid molecule is attached is selected from Lys and Orn.

In one embodiment, the amino acid residue to which the fatty acid molecule is attached is Lys.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of the Orn residue of the functional variant comprising an Orn amino acid residue of either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the fatty acid molecule is attached to the epsilon amino group of the K residue of either of SEQ ID NO 1 and SEQ ID NO 2 or a variant thereof.

In one embodiment, the amino acid residue to which the fatty acid molecule is attached is the N-most terminal amino acid residue, such as the N-most terminal amino acid residue of SEQ ID No. 1 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to the alpha-amino group of the N-terminal amino acid residue. In one embodiment, the fatty acid molecule is attached to the N-terminal amino acid residue at position 5 of SEQ ID NO 1 or a functional variant thereof. In one embodiment, the fatty acid molecule is attached to T at position 5 of SEQ ID NO. 1 or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 16 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to K-16 of any one of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof, such as to the epsilon amino group of K-16 of any one of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 18 of any one of SEQ ID NO:1 and SEQ ID NO:2 or a variant thereof, wherein H at position 18 of any one of SEQ ID NO:1 and SEQ ID NO:2 has been substituted with K (or Orn).

In one embodiment, the fatty acid molecule is attached to the epsilon amino group at K position 18 of either of SEQ ID NO 1 and SEQ ID NO 2 or a variant thereof.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 18 of a peptide selected from the group consisting of:

TFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(5-30)H18K;SEQ ID NO:8),

EGTFISDYSIAMDKIKQQDFVNWLLAQK(hGIP(3-30)H18K;SEQ ID NO:75),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of the K residue at any of the positions of SEQ ID NO 1 and SEQ ID NO 2 or functional variants thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 18 of a peptide selected from:

TFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(5-30)H18Orn;SEQ ID NO:62),

EGTFISDYSIAMDKIOrnQQDFVNWLLAQK(hGIP(3-30)H18Orn;SEQ ID NO:131),

or a functional variant thereof.

In one embodiment, when the fatty acid molecule is attached to an amino acid residue at a position other than positions 16 and 30 of any one of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof, the K at position 16 and/or the K at position 30 of any one of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof is individually substituted with any amino acid.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 15 of a peptide selected from the group consisting of:

TFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(5-30)D15K,SEQ ID NO:30),

EGTFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(3-30)D15K,SEQ ID NO:98),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group at position 15 Orn of a peptide selected from:

TFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(5-30)D15Orn,SEQ ID NO:61),

EGTFISDYSIAMOrnKIHQQDFVNWLLAQK(hGIP(3-30)D15Orn,SEQ ID NO:130),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 11 of a peptide selected from the group consisting of:

TFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S11K,SEQ ID NO:26),

EGTFISDYKIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S11K,SEQ ID NO:94),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 11 of a peptide selected from the group consisting of:

TFISDYOrnIAMDKIHQQDFVNWLLAQK(hGIP(5-30)S11Orn,SEQ ID NO:133),

EGTFISDYOrnIAMDKIHQQDFVNWLLAQK(hGIP(3-30)S11Orn,SEQ ID NO:134),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 12 of a peptide selected from the group consisting of:

TFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(5-30)I12K,SEQ ID NO:27),

EGTFISDYSKAMDKIHQQDFVNWLLAQK(hGIP(3-30)I12K,SEQ ID NO:95),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 12 of a peptide selected from the group consisting of:

TFISDYSOrnAMDKIHQQDFVNWLLAQK(hGIP(5-30)I12Orn,SEQ ID NO:135),

EGTFISDYSOrnAMDKIHQQDFVNWLLAQK(hGIP(3-30)I12Orn,SEQ ID NO:136),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 13 of a peptide selected from the group consisting of:

TFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(5-30)A13K,SEQ ID NO:28),

EGTFISDYSIKMDKIHQQDFVNWLLAQK(hGIP(3-30)A13K,SEQ ID NO:96),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 13 of a peptide selected from the group consisting of:

TFISDYSIOrnMDKIHQQDFVNWLLAQK(hGIP(5-30)A13Orn,SEQ ID NO:137),

EGTFISDYSIOrnMDKIHQQDFVNWLLAQK(hGIP(3-30)A13Orn,SEQ ID NO:138),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 9 of any one of SEQ ID NO 1 and SEQ ID NO 2 or a variant thereof, wherein D at position 9 of any one of SEQ ID NO 1 and SEQ ID NO 2 has been substituted with K or Orn.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 9 of a peptide selected from the group consisting of:

TFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)D9K,SEQ ID NO:24),

EGTFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)D9K,SEQ ID NO:92),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 9 of a peptide selected from:

TFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(5-30)D9Orn,SEQ ID NO:24),

EGTFISKYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)D9Orn,SEQ ID NO:92),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 21 of any one of SEQ ID NO 1 and SEQ ID NO 2 or a variant thereof, wherein D21 of any one of SEQ ID NO 1 and SEQ ID NO 2 has been substituted with K or Orn.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 21 of a peptide selected from the group consisting of:

TFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(5-30)D21K,SEQ ID NO:34),

EGTFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(3-30)D21K,SEQ ID NO:102),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 21 of a peptide selected from:

TFISDYSIAMDKIHQQOrnFVNWLLAQK(hGIP(5-30)D21Orn,SEQ ID NO:139),

EGTFISDYSIAMDKIHQQOrnFVNWLLAQK(hGIP(3-30)D21Orn,SEQ ID NO:140),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 5 of SEQ ID NO 2 or a variant thereof, wherein T at position 5 has been substituted by K or Orn in SEQ ID NO 2.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group at K position 5 of a peptide having the sequence:

EGKFISDYSIAMDKIHQQDFVNWLLAQK(hGIP(3-30)T5K,SEQ ID NO:88),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 5 of a peptide having the sequence:

EGOrnFISDYSIAMDKIHQQKFVNWLLAQK(hGIP(3-30)T5Orn,SEQ ID NO:141),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 15 of SEQ ID NO 2 or a variant thereof, wherein D at position 15 has been substituted by K or Orn in SEQ ID NO 2.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group at K position 15 of a peptide having the sequence:

EGTFISDYSIAMKKIHQQDFVNWLLAQK(hGIP(3-30)D15K,SEQ ID NO:98),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group at Orn 15 of a peptide having the sequence:

EGTFISDYSIAMOrnKIHQQKFVNWLLAQK(hGIP(3-30)D15Orn,SEQ ID NO:142),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to amino acid residue 20 of SEQ ID NO 2 or a variant thereof, wherein Q at position 20 has been substituted by K or Orn in SEQ ID NO 2.

In one embodiment, the fatty acid molecule is attached to the epsilon-amino group of K at position 20 of a peptide having the sequence:

EGTFISDYSIAMDKIHQKDFVNWLLAQK(hGIP(3-30)Q20K,SEQ ID NO:101),

or a functional variant thereof.

In one embodiment, the fatty acid molecule is attached to the delta-amino group of Orn at position 20 of a peptide having the sequence:

EGTFISDYSIAMDKIHQOrnKFVNWLLAQK(hGIP(3-30)Q20Orn,SEQ ID NO:143),

or a functional variant thereof.

In one embodiment, the peptide is modified by attaching a (one) fatty acid molecule at one (single) amino acid residue of either of SEQ ID NO:1 and SEQ ID NO:2 or a functional variant thereof.

In one embodiment, the peptide of the GIP peptide analog comprises no more than one K amino acid residue modified by the attachment of a fatty acid molecule to the epsilon amino group of K.

In one embodiment, the fatty acid molecule according to the present disclosure is a straight chain fatty acid.

In one embodiment, the fatty acid molecule according to the present disclosure is a branched chain fatty acid.

In one embodiment, the fatty acid molecule according to the present disclosure is a monoacyl fatty acid molecule comprising one fatty acid.

In one embodiment, the fatty acid molecule according to the present disclosure is a diacyl fatty acid molecule.

In one embodiment, the fatty acid molecule according to the present disclosure is a diacyl fatty acid molecule comprising two fatty acids.

In one embodiment, the fatty acid molecule according to the present disclosure is a diacyl fatty acid molecule containing two carboxyl functional groups.

In one embodiment, a fatty acid molecule according to the present disclosure comprises the formula CH3(CH2)nAn acyl group of CO-, wherein n is an integer of 4 to 24.

In one embodiment, the fatty acid molecule comprises a moiety selected from CH3(CH2)6CO-、CH3(CH2)8CO-、CH3(CH2)10CO-、CH3(CH2)12CO-、CH3(CH2)14CO-、CH3(CH2)16CO-、CH3(CH2)18CO-、CH3(CH2)20CO-and CH3(CH2)22Acyl group of CO-.

In one embodiment, the fatty acid molecule comprises a moiety selected from CH3(CH2)10CO- (lauryl, C12), CH3(CH2)12CO- (myristoyl, C14), CH3(CH2)14CO- (palmitoyl, C16) and CH3(CH2)16Acyl radical of CO- (stearoyl, C18).

In one embodiment, the fatty acid molecule is selected from CH3(CH2)10CO- (lauryl, C12), CH3(CH2)12CO- (myristoyl, C14), CH3(CH2)14CO- (palmitoyl, C16) and CH3(CH2)16Monoacyl fatty acids of CO- (stearoyl, C18).

In one embodiment, the fatty acid molecule comprises a moiety selected from the group consisting of CH3(CH2)10CO- (lauryl, C12), CH3(CH2)12CO- (myristoyl, C14), CH3(CH2)14CO- (palmitoyl, C16) and CH3(CH2)16Two fatty acids of CO- (stearoyl, C18).

In one embodiment, the fatty acid molecule comprises the formula COOH (CH)2)nAn acyl group of CO- (dicarboxylic acid), wherein n is an integer from 4 to 24.

In one embodiment, the fatty acid molecule comprises a compound selected from the group consisting of COOH (CH)2)14CO-、COOH(CH2)16CO-、COOH(CH2)18CO-and COOH (CH)2)20Acyl group of CO-.

In one embodiment, the fatty acid molecule is selected from C12, C14, C16, and C18.

In one embodiment, the fatty acid molecule is selected from the group consisting of C14 diacid, C16 diacid, and C18 diacid.

In one embodiment, the fatty acid molecule is palmitoyl.

In one embodiment, the fatty acid molecule is 1, 16-hexadecanedioic acid/hexadecanedioic acid.

In one embodiment, the fatty acid molecule is stearoyl.

In one embodiment, the fatty acid molecule is 1, 18-octadecanedioic acid/octadecanedioic acid.

A fatty acid molecule may be attached to an amino acid residue in such a way that the carboxyl group of the fatty acid molecule forms an amide bond with the amino group of the amino acid residue.

The fatty acid molecule and the peptide herein may be attached directly or indirectly (i.e., via a linker or spacer).

In one embodiment, the fatty acid molecule according to the present disclosure is directly attached to an amino acid residue.

In one embodiment, the fatty acid molecule according to the present disclosure is directly attached to the alpha-amino group of an amino acid residue, wherein said amino acid residue is the N-terminal amino acid residue.

In one embodiment, the fatty acid molecule according to the present disclosure is attached directly to the epsilon-amino group of the Lys residue.

In one embodiment, the fatty acid molecule according to the present disclosure is attached directly to the δ -amino group of the Orn residue.

In one embodiment, the fatty acid molecule according to the present disclosure is attached to the amino acid residue by a linker or spacer.

In one embodiment, the fatty acid molecule according to the present disclosure is attached to the alpha-amino group of an amino acid residue by a linker or spacer, wherein the amino acid residue is the N-terminal amino acid residue.

In one embodiment, the fatty acid molecule according to the present disclosure is attached to the epsilon-amino group of the Lys residue via a linker or spacer.

In one embodiment, the fatty acid molecule according to the present disclosure is attached to the δ -amino group of the Orn residue via a linker or spacer.

In one embodiment, the fatty acid molecule is attached to an amino acid residue through a spacer (or linker) in such a way that the carboxyl group of the spacer forms an amide bond with the amino group of the fatty acid molecule.

In one embodiment, the spacer is an α, ω -amino acid. Examples of suitable spacers are succinic acid, Lys, Glu or Asp, or dipeptides such as Gly-Lys. When the spacer is succinic acid, one of its carboxyl groups may form an amide bond with the amino group of the amino acid residue, and the other carboxyl group thereof may form an amide bond with the amino group of the fatty acid molecule. When the spacer is Lys, Glu or Asp, its carboxyl group may form an amide bond with the amino group of the amino acid residue, and its amino group may form an amide bond with the carboxyl group of the fatty acid molecule. When Lys is used as a spacer, in some cases, another spacer may be inserted between the epsilon-amino group of Lys and the fatty acid molecule. In one embodiment, the further spacer is succinic acid, which forms amide bonds with the epsilon-amino group of Lys and with the amino group present in the fatty acid molecule. Other spacers are N epsilon- (gamma-L-glutamyl), N epsilon- (beta-L-asparaginyl), N epsilon-glycyl and N epsilon- (alpha- (gamma-aminobutyryl).

In one embodiment, the spacer is a hydrophilic linker. In one embodiment, the spacer is a non-natural amino acid hydrophilic linker.

In one embodiment, the spacer is selected from the group consisting of gamma-aminobutyryl (gamma-aminobutyric acid), gamma-glutamyl (gamma-glutamic acid), beta-asparaginyl, beta-alanyl and glycyl. In one embodiment, the spacer comprises one or more of gamma-aminobutyryl (gamma-aminobutyric acid), gamma-glutamyl (gamma-glutamic acid), beta-asparaginyl, beta-alanyl, and glycyl.

In one embodiment, the spacer is a repetition of a separate spacer portion. In one embodiment, the spacer is a repetition of the same spacer sub-portion. In one embodiment, the spacer is a repetition of different spacer portions.

In one embodiment, the spacer is γ -glutamic acid-8-amino-3, 6-dioxaoctanoic acid (γ -Glu) -AEEAc or a repeat thereof.

In one embodiment, the spacer comprises gamma-glutamic acid-8-amino-3, 6-dioxaoctanoic acid (gamma-Glu-AEEAc)n) One or more repetitions of (a).

In one embodiment, the spacer is [ gamma-glutamic acid-8-amino-3, 6-dioxaoctanoic acid]n(γ-Glu-AEEAcn) Wherein n is between 1 and 50Is an integer of (1).

In one embodiment, the spacer is [ gamma-glutamic acid-8-amino-3, 6-dioxaoctanoic acid]n(γ-Glu-AEEAcn) Wherein n is an integer between 1 and 50, such as an integer between 1-2, 2-3, 3-4, 4-5, 5-6, 6-7, 7-8, 8-9, 9-10, 10-11, 11-12, 12-13, 13-14, 14-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45, 45-50.

In one embodiment, the spacer is [ gamma-glutamic acid-8-amino-3, 6-dioxaoctanoic acid]n(γ-Glu-AEEAcn) Wherein n is an integer selected from 1, 2, 3, 4, 5, 6, 7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 and 50.

In one embodiment, the spacer is an amino acid residue other than Cys. In one embodiment, the spacer is 4-Abu. In one embodiment, the spacer is gamma-aminobutyric acid.

In another embodiment, the spacer is a dipeptide, such as a dipeptide wherein the C-terminal amino acid residue is Lys, His or Trp, preferably Lys, and wherein the N-terminal amino acid residue is selected from Ala, Arg, Asp, Asn, Gly, Glu, gin, Ile, Leu, Val, Phe and Pro. In one embodiment, the dipeptide spacer is Gly-Lys.

In one embodiment, the spacer comprises one or more moieties selected from the group consisting of γ -aminobutyryl (γ -aminobutyric acid), γ -glutamyl (γ -glutamic acid), β -asparaginyl, β -alanyl and glycyl. In one embodiment, the spacer comprises γ -aminobutyryl (γ -aminobutyric acid), γ -glutamyl (γ -glutamic acid), β -asparaginyl, β -alanyl, glycyl, γ -glutamic acid-8-amino-3, 6-dioxaoctanoic acid (γ -Glu-AEEAc)nWhere n is an integer between 1 and 50), an amino acid residue other than Cys, 4-Abu, gamma-aminobutyric acid, and a dipeptide.

In another embodiment, the spacer is a non-branched alkane α, ω -dicarboxylic acid group having 1 to 7 methylene groups (preferably two methylene groups), said spacer forming a bridge between the amino group of the parent peptide and the amino group of the fatty acid molecule.

GIP (5-30) peptide having fatty acid

In one embodiment, the GIP analogue as defined herein is selected from:

TFISDYSIAMDKIHQQDFVNWLLAQK-C12/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C12/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C14/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C16/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C16/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C18/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C18/K16

TFISDYSIAMDRIKQQDFVNWLLAQR-C16/T5

TFISDYSIAMDRIKQQDFVNWLLAQR-C16/K18

TFISDYSIAMDRIHQQDFVNWLLAQR-C16/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/T5

TFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/K16

TFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K16,

TFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K16,

TFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18,

TFISDYSIAMDKIKQQDFVNWLLAQK-C18-diacid/K18,

TFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18,

KFISDYSIAMDKIHQQDFVNWLLAQK-C14-diacid/K5,

KFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K5,

KFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K5,

KFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K5,

KFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K5,

TFISDYKIAMDKIHQQDFVNWLLAQK-C16-diacid/K11,

TFISDYKIAMDRIHQQDFVNWLLAQR-C16-diacid/K11,

TFISDYKIAMDKIHQQDFVNWLLAQK-C18-diacid/K11,

TFISDYKIAMDRIHQQDFVNWLLAQR-C18-diacid/K11,

TFISDYSKAMDKIHQQDFVNWLLAQK-C16-diacid/K12,

TFISDYSKAMDRIHQQDFVNWLLAQR-C16-diacid/K12,

TFISDYSKAMDKIHQQDFVNWLLAQK-C18-diacid/K12,

TFISDYSKAMDRIHQQDFVNWLLAQR-C18-diacid/K12,

TFISDYSIKMDKIHQQDFVNWLLAQK-C16-diacid/K13,

TFISDYSIKMDRIHQQDFVNWLLAQR-C16-diacid/K13,

TFISDYSIKMDKIHQQDFVNWLLAQK-C18-diacid/K13,

TFISDYSIKMDRIHQQDFVNWLLAQR-C18-diacid/K13,

TFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K16,

TFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K16,

TFISDYSIAMDRIKQQDFVNWLLAQR-C18-diacid/K18,

TFISDYSIAMDKIHQQKFVNWLLAQK-C16-diacid/K21,

TFISDYSIAMDRIHQQKFVNWLLAQR-C16-diacid/K21,

TFISDYSIAMDKIHQQKFVNWLLAQK-C18-diacid/K21, and

TFISDYSIAMDRIHQQKFVNWLLAQR-C18-diacid/K21,

or a functional variant thereof,

wherein the fatty acids are attached directly or through a linker/spacer as defined herein.

This gave C12 as the fatty acid CH3(CH2)10CO- (lauryl); c14 is a fatty acid CH3(CH2)12CO- (myristoyl); c16 is a fatty acid CH3(CH2)14CO- (palmitoyl) and C18 is a fatty acid CH3(CH2)16CO- (stearoyl). The suffix "-diacid" means that the fatty acid molecule is a diacyl fatty acid molecule. The absence of such a suffix refers to the monoacyl fatty acid molecule.

In one embodiment, the peptide is C-terminally amidated (-NH)2)。

In one embodiment, the GIP analog is selected from the group consisting of:

hGIP (5-30) NH2[ H18K ] -C16/K18+ gamma-glutamic acid,

hGIP (5-30) NH2[ H18K ] -C16/K18+ gamma-aminobutyric acid,

hGIP (5-30) NH2[ H18K ] -C16/K18+ beta-alanine, and

one or more repeats of hGIP (5-30) NH2[ H18K ] -C16/K18+ gamma-glutamic acid + 8-amino-3, 6-dioxaoctanoic acid.

In one embodiment, the GIP analog is selected from the group consisting of:

hGIP (5-30) NH2[ H18K ] -C16-diacid/K18 + gamma-glutamic acid,

hGIP (5-30) NH2[ H18K ] -C16-diacid/K18 + gamma-aminobutyric acid,

hGIP (5-30) NH2[ H18K ] -C16-diacid/K18 + beta-alanine, and

one or more repeats of hGIP (5-30) NH2[ H18K ] -C16-diacid/K18 + gamma-glutamic acid + 8-amino-3, 6-dioxaoctanoic acid.

GIP (3-30) peptide having fatty acid

In one embodiment, the GIP analogue as defined herein is selected from:

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C12/K16

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C16/K16

EGWFISDYSIAMEKIAQQDFVNWLLAQK-C16/K16

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C14/K16

EGTFISDYSIAMDKIHQQDFVNWLLAQK-C18/K16

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C12/K18

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16/K18,

EGTFISDYSIAMEKIAQQDFVNWLLAQK-C16/K16,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C18-diacid/K18,

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQR-C18/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQR-C16/K18,

EGTFISDYSIALDKIKQQDFVNWLLAQK-C16/K18,

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C16-diacid/K18

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C16-diacid/K18, and

EGTFISDYSIAMDKIKQQDFVNWLLAQK-C18/K18,

EGKFISDYSIAMDKIHQQDFVNWLLAQK-C16-diacid/K5,

EGKFISDYSIAMDRIHQQDFVNWLLAQR-C16-diacid/K5,

EGKFISDYSIAMDKIHQQDFVNWLLAQK-C18-diacid/K5,

EGKFISDYSIAMDRIHQQDFVNWLLAQR-C18-diacid/K5, and

EGTFISDYSIAMDRIKQQDFVNWLLAQR-C18-diacid/K18,

or a functional variant thereof,

wherein the fatty acids are attached directly or through a linker/spacer as defined herein.

In one embodiment, the peptide is C-terminally amidated (-NH)2)。

In one embodiment, the GIP analog is selected from the group consisting of:

AT164[ hGIP (3-30) NH2[ H18K ] -C16/K18+ gamma-glutamic acid ],

AT165[ hGIP (3-30) NH2[ H18K ] -C16/K18+ gamma-aminobutyric acid ],

AT166[ hGIP (3-30) NH2[ H18K ] -C16/K18+ beta-alanine ], and

AT167[ hGIP (3-30) NH2[ H18K ] -C16/K18+ gamma-glutamic acid + one or more repeats of 8-amino-3, 6-dioxaoctanoic acid ].

In one embodiment, the GIP analog is selected from the group consisting of:

[ hGIP (3-30) NH2[ H18K ] -C16-diacid/K18 + gamma-glutamic acid ],

[ hGIP (3-30) NH2[ H18K ] -C16-diacid/K18 + gamma-aminobutyric acid ],

[ hGIP (3-30) NH2[ H18K ] -C16-diacid/K18 + -alanine ], and

one or more repeats of [ hGIP (3-30) NH2[ H18K ] -C16-diacid/K18 + gamma-glutamic acid + 8-amino-3, 6-dioxaoctanoic acid.

Compound (I)

In another aspect, there is provided a compound comprising or consisting of a peptide as defined herein. In one embodiment, the compound is formulated as a peptide monomer (i.e., comprising 1 copy of the peptide), while in another embodiment, the compound is formulated as a peptide multimer.

Polymeric compounds

In one embodiment, the peptides according to the present disclosure are formulated as multimers. Multimers are proteins that comprise or consist of multiple peptide monomers. Multimers are aggregates of multiple molecules that are typically held together by non-covalent bonds. This definition distinguishes multimers from polymers, which are a series of monomers held together by covalent bonds.

In one embodiment, a peptide sequence of the present disclosure is linked to another (same or different) peptide sequence of the present disclosure by a chemical bond or by a linker group. In some embodiments, the peptides of the disclosure are formulated as oligomers or multimers of monomers, wherein each monomer is a peptide sequence defined according to the disclosure.

Thus, in one embodiment, a multimeric compound according to the present disclosure is a polymer comprising two or more peptide sequences of the present disclosure, the peptide sequences being the same or different, wherein at least one of the two or more peptide sequences is a peptide according to the present disclosure. Preferably, both peptide sequences are peptides according to the present disclosure.

In one embodiment, the multimeric compound is a dimer comprising two peptides according to the present disclosure, the two peptides being the same or different relative to each other.

In another embodiment, the multimeric compound is a trimer comprising three peptides according to the disclosure, the peptides being the same or different with respect to each other.

In another embodiment, the multimeric compound is a tetramer comprising four peptides according to the present disclosure, the peptides being the same or different relative to each other.

In one embodiment, the polymeric compound is a dendrimer, such as a tetrameric or octameric dendrimer. Dendrimers are repeatedly branched, generally spherical macromolecules, typically symmetric about a core, often adopting a spherical three-dimensional morphology.

Dendrimers according to the present disclosure may comprise 4 peptides, 8 peptides, 16 peptides, or 32 peptides. In a particular embodiment, the dendrimer comprises four peptides (i.e., a tetrameric dendrimer) or eight peptides (an octameric dendrimer).

In some particular embodiments, the multimeric compound comprises two identical amino acid sequences of the present invention (dimers) or the compound comprises four identical copies of an amino acid sequence of the present disclosure (tetrameric dendrimers).

In one embodiment, multimers according to the present disclosure are prepared by linking two or more peptide monomers by peptide bonds or linker groups. In one embodiment, they are attached to a lysine backbone such as lysine residues (each peptide chain is attached to a single lysine residue), or are coupled to a polymeric carrier, e.g., a protein carrier. In one embodiment, the linker group comprises a plurality of lysine residues, such as a core moiety having a plurality of lysine residues, such as found in lysine-based dendritic structures comprising three, seven, fifteen, and more lysine residues. However, any other linkage of peptide monomers known to the person skilled in the art is conceivable.

In one embodiment, the linkage occurs at the N-terminus and/or C-terminus of the peptide monomers.

In one embodiment, a polymeric compound is provided that consists of: A) one or more glucose-dependent insulinotropic peptide (GIP) analogs of formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000471

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2, and

B) optionally one or more linker groups.

Determination of antagonist Properties and affinities

To determine whether a peptide is an antagonist of a GIPR, methods known in the art can be employed, for example, by determining the IC50 of the peptide. This can be done by constructing a dose response curve and examining the effect of different concentrations of peptide on the inverse agonist activity. The agonist may be GIP1-42, such as hGIP-1-42 or hGIP 1-30. The GIPR may be hGIPR, rGIPR, mGIPR, dog GIPR, pig GIPR or rhesus GIPR. IC50 values for a given antagonist can be calculated by determining the concentration that inhibits half of the maximal biological response of the agonist. A method of determining whether a peptide is an antagonist is described in example 4, but other methods known in the art may also be used. For example, as the concentration of GIP-derived peptides increases, Schild plot analysis can be performed on hGIP1-42 cAMP dose response curves. In this way, the type of antagonist activity can also be determined.

Heterologous competitive binding experiments can be performed to measure the affinity of a peptide for GIPR, i.e. how efficiently the peptide replaces a given GIP1-42, e.g. hGIP 1-42. These competitive binding experiments can be performed by methods known in the art. For example, GIP1-42 can be radiolabeled, e.g., with 125I. Other suitable isotopes are known to those skilled in the art.

Method of treatment

In one aspect, there is provided a peptide as defined herein or a composition comprising said peptide for use as a medicament.

In one embodiment, a glucose-dependent insulinotropic peptide (GIP) analog of formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000481

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

for use as a medicament.

In another embodiment, there is provided a glucose-dependent insulinotropic peptide (GIP) analog of formula 1(hGIP5-30, SEQ ID NO: 1):

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

in a method for inhibiting or reducing one or more of: i) GIP-induced glucagon secretion, ii) GIP-induced insulin secretion, iii) GIP-induced somatostatin secretion, iv) GIP-induced glucose uptake, v) GIP-induced fatty acid synthesis and/or fatty acid incorporation, vi) higher or increased GIPR expression or activity, vii) postprandial GIP release, viii) serum levels of free fatty acids and/or triglycerides, ix) GIP-induced reduction in bone resorption.

In another embodiment, there is provided a glucose-dependent insulinotropic peptide (GIP) analog of formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000491

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

in a method for treating a condition selected from the group consisting of: metabolic syndrome, obesity, overweight, obesity-related disorders, prediabetes (impaired fasting glucose), diabetes (types I and 2), diabetes-related disorders, insulin resistance, elevated fasting glucose (hyperglycemia), elevated fasting serum triglyceride (VLDL triglyceride) levels, low High Density Lipoprotein (HDL) levels, disorders of fatty acid metabolism, cardiovascular disease, hypertension and atherosclerosis.

In a particular embodiment, there is provided a peptide as defined herein for use in a method of treating obesity.

In a particular embodiment, there is provided a peptide as defined herein for use in a method of treating diabetes (including type I and type II diabetes).

In a particular embodiment, there is provided a peptide as defined herein for use in a method of treating insulin resistance.

In another embodiment, there is provided a glucose-dependent insulinotropic peptide (GIP) analog of formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000501

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

for use in a method of inducing weight loss.

In another aspect, there is provided a peptide as defined herein for use in a method of treating cancer.

In one embodiment, the cancer is selected from colon cancer, neuroendocrine cancer, and adrenal adenoma.

In another aspect, there is provided a peptide as defined herein for use in a method of treating a bone density disorder (or bone volume disorder).

In one embodiment, there is provided a peptide as defined herein for use in a method of inhibiting bone cell activity. In one embodiment, there is provided a peptide as defined herein for use in a method of inhibiting (or antagonizing) GIP-induced postprandial reduction in bone resorption. In one embodiment, there is provided a peptide as defined herein for use in a method of treating bone cancer.

In one embodiment, the bone density (or volume) disorder is selected from the group consisting of osteoporosis, disorders characterized by low bone density and/or reduced bone volume, disorders characterized by high bone density and/or increased bone volume, and osteoporosis.

In another aspect, there is provided a GIP peptide analogue as defined herein for use in a method of characterising or examining aspects of a disorder and/or aspects of human physiology associated with a disorder, wherein in one embodiment the disorder is selected from a metabolic disorder or syndrome, such as obesity, diabetes, insulin resistance or a disorder of fatty acid metabolism. In other aspects, the invention relates to methods of treating cancer (such as colon cancer or adrenal adenoma). In other aspects, the invention relates to methods of treating bone density disorders characterized by high bone density and/or increased bone volume or osteoporosis. In other aspects, the invention relates to methods of treating atherosclerosis.

In another embodiment, there is provided a use of a glucose-dependent insulinotropic peptide (GIP) analog of formula 1(hGIP5-30, SEQ ID NO: 1):

Figure BDA0002296622780000511

wherein the peptide optionally further comprises the dipeptide E-G at the N-terminus (hGIP3-30, SEQ ID NO:2),

or a functional variant having at least 75% sequence identity to any of SEQ ID NO. 1 and SEQ ID NO. 2,

wherein the peptide is modified by attaching at least one fatty acid molecule to one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2,

manufacture of a medicament for:

-treating a condition selected from: metabolic syndrome, obesity, overweight, obesity-related disorders, prediabetes (impaired fasting glucose), diabetes (types I and 2), diabetes-related disorders, insulin resistance, elevated fasting glucose (hyperglycemia), elevated fasting serum triglyceride (VLDL triglyceride) levels, low High Density Lipoprotein (HDL) levels, disorders of fatty acid metabolism, cardiovascular disease, hypertension and atherosclerosis, or

-inducing weight loss, or

-treating cancer, including but not limited to colon cancer, neuroendocrine cancer and adrenal adenoma, or

-treating bone density disorders including, but not limited to, osteoporosis, disorders characterized by low bone density and/or reduced bone volume, disorders characterized by high bone density and/or increased bone volume and osteoporosis.

Also provided is a method for treating metabolic syndrome (such as obesity, overweight, diabetes, insulin resistance or fatty acid metabolism disorder); cancer (such as colon cancer or adrenal adenoma); bone density disorders (such as those characterized by high bone density and/or increased bone volume); or atherosclerosis; the method comprises the step of administering to an individual in need thereof an effective amount of a peptide as defined herein.

Reference herein to an individual in need thereof is an individual that may benefit from administration of a peptide or pharmaceutical composition according to the present disclosure. Such individuals may suffer from, or be at risk of developing, metabolic disorders such as obesity, overweight, diabetes, insulin resistance or fatty acid metabolic disorders, cancer such as colon cancer or adrenal adenoma, bone density disorders. The individual may be any person, male or female, infant, middle aged or elderly. The disorder to be treated or prevented in an individual may be related to the age of the individual, the general health of the individual, the drugs used to treat the individual and the past history of whether the individual has a disease or disorder that may or has induced a metabolic disorder (such as obesity, overweight, diabetes, insulin resistance or fatty acid metabolic disorder), cancer (such as colon cancer or adrenal adenoma), atherosclerosis, bone density disorder. In some embodiments, the disorder to be treated is associated with GIP-induced glucagon secretion, GIP-induced insulin secretion, GIP-induced somatostatin secretion, GIP-induced glucose uptake, GIP-induced fatty acid synthesis and/or fatty acid incorporation, higher GIPR expression or activity, postprandial GIP release; wherein the term "high" is to be interpreted as referring to a level higher than the corresponding level observed in an individual not requiring treatment.

Preparation method (peptide)

The peptides according to the present disclosure may be prepared by any method known in the art. Thus, GIP-derived peptides can be prepared by standard peptide preparation techniques, such as solution synthesis or Merrifield-type solid phase synthesis.

In one embodiment, a peptide as defined herein is a non-naturally occurring peptide; native GIP, such as GIP1-42, is derived from a naturally occurring protein.

In one embodiment, a peptide according to the present disclosure is purified from its naturally occurring source (such as serum). Protein purification is a series of processes aimed at separating a single type of protein from a complex mixture. The starting material is typically biological tissue. Various steps in the purification process may release the protein from the matrix in which it is defined, separate the proteinaceous and non-proteinaceous parts of the mixture, and finally separate the desired protein from all other proteins. The separation step may utilize differences in, for example, protein size, physicochemical properties, binding affinity, and biological activity.

In one embodiment, the peptides according to the present disclosure are synthetically prepared or produced.

Methods for synthetically producing peptides are well known in the art. Details and practical suggestions for producing Synthetic Peptides can be found in Synthetic Peptides, A User's Guide (Advances in Molecular Biology), Grant G.A. eds, Oxford University Press,2002, or in Pharmaceutical Formulation, Development of Peptides and Proteins, Frokjaer and Hovgaard eds, Taylor and Francis, 1999.

In one embodiment, one or more peptide sequences of the invention are synthetic, in particular by a Sequence Assisted Peptide Synthesis (SAPS) method, solution synthesis, Solid Phase Peptide Synthesis (SPPS) such as solid phase synthesis of the Merrifield type, recombinant techniques (produced by a host cell comprising a first nucleic acid sequence encoding a peptide operably bound to a second nucleic acid capable of directing expression in said host cell) or enzymatic synthesis. These are well known to those skilled in the art.

Peptides can be synthesized batchwise on a fully automated peptide synthesizer using 9-fluorenylmethyloxycarbonyl (Fmoc) or tert-butyloxycarbonyl (Boc) as the N-alpha-amino protecting group and the appropriate common side chain functional group protecting group.

After purification, such as by reverse phase HPLC, the peptide may be further processed to obtain, for example, a circular or C-or N-terminally modified isoform. Methods for cyclization and end modification are well known in the art.

The peptides of the invention may be synthesized as monomers or multimers (such as dimers or tetramers).

Pharmaceutical compositions and formulations

Although the bioactive agents of the present disclosure may be administered as chemical starting materials (peptides), it is sometimes preferred to present them in the form of pharmaceutical formulations. Such pharmaceutical formulations may be referred to as pharmaceutical compositions, pharmaceutically acceptable compositions, or pharmaceutically safe compositions.

Thus, there is further provided a pharmaceutical formulation comprising a biologically active agent of the invention, or a pharmaceutically acceptable salt or ester thereof, and a pharmaceutically acceptable carrier, excipient and/or diluent. Such pharmaceutical formulations may be prepared by conventional techniques, for example as described in Remington: The Science and Practice of Pharmacy 2005, Lippincott, Williams & Wilkins.

The invention is also intended to include pharmaceutically acceptable salts of the instant peptide compounds that can be prepared. These salts will be acceptable salts when they are used for pharmaceutical purposes. This means that the salt will retain the biological activity of the parent compound and that the salt will not produce adverse or deleterious effects in its use and use for the treatment of disease.

Pharmaceutically acceptable salts are prepared in a standard manner. If the parent compound is a base, it is treated with an excess of an organic or inorganic acid in a suitable solvent. If the parent compound is an acid, it is treated with an inorganic or organic base in a suitable solvent.

The peptide compounds disclosed herein may be administered in an effective amount in the form of their alkali or alkaline earth metal salts concurrently, simultaneously or together with a pharmaceutically acceptable carrier or diluent, especially and preferably in the form of their pharmaceutical compositions, whether by oral, rectal or parenteral (including subcutaneous) routes.

For example, examples of pharmaceutically acceptable acid addition salts for use in the pharmaceutical compositions of the present invention include acid addition salts derived from inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, metaphosphoric acid, nitric acid, and sulfuric acid, and organic acids such as tartaric acid, acetic acid, citric acid, malic acid, lactic acid, fumaric acid, benzoic acid, glycolic acid, gluconic acid, succinic acid, p-toluenesulfonic acid, and arylsulfonic acid.

In particular embodiments, peptides according to the present disclosure are formulated as acetate or TFA (trifluoroacetic acid) salts.

Administration and dosage

In accordance with the present disclosure, a peptide as defined herein or a composition comprising a peptide as defined herein is administered to an individual in need of treatment at a pharmaceutically effective dose or a therapeutically effective amount. The dosage requirements will vary with the particular pharmaceutical composition used, the route of administration, and the particular subject being treated, depending on the severity and type of the disorder, as well as the weight and general condition of the subject. Those skilled in the art will also recognize that the optimal number and spacing of individual doses of a peptide compound will depend on the nature and extent of the condition being treated, the form, route and site of administration and the particular patient being treated, and that such optimal values may be determined using routine techniques. It will also be appreciated by those skilled in the art that routine course of treatment determination tests can be used to determine the optimal course of treatment, i.e., the dose of compound administered per day over a defined number of days.

In one embodiment, the bioactive agent is administered at least once daily, such as twice daily, such as three times daily, such as four times daily, such as five times daily.

The dose may also be administered at intermittent intervals or intervals, so that the dose is not administered every day. But may be administered in one or more doses every second day, every third day, every fourth day, every fifth day, every sixth day, every week, every second week, every third week, every fourth week, every fifth week, every sixth week, or at intervals within these ranges, such as every 2 to 4 weeks or 4 to 6 weeks.

In one embodiment, the bioactive agent is administered at a dose of at least 30000 pmol/kg/day, such as at least 60000 pmol/kg/day, such as at least 72000 pmol/kg/day, such as at least 90000 pmol/kg/day, such as at least 120000 pmol/kg/day, such as at least 150000 pmol/kg/day, such as at least 30000 pmol/kg/day, preferably such as at least 60000 pmol/kg/day. In a particular embodiment, the bioactive agent is administered at a dose of 72000 pmol/kg/day.

In one embodiment, the biologically active agent is administered at a daily dose of 30000 to 40000pmol/kg, such as 40000 to 50000, such as 50000 to 60000, such as 60000 to 70000, such as 70000 to 80000, such as 80000 to 90000, such as 90000 to 100000, such as 100000 to 110000, such as 110000 to 120000 pmol/kg. In particular embodiments, the bioactive agent is a peptide and is administered at a daily dose of 60000pmol/kg or 72000 pmol/kg.

In one embodiment, the bioactive agent is administered by infusion. In one embodiment, the biologically active agent is a peptide and the infusion duration is at least 15 minutes, such as at least 20 minutes, such as at least 30 minutes, such as at least 40 minutes, such as at least 50 minutes, such as at least 60 minutes, such as at least 90 minutes, such as at least 120 minutes, preferably such as 60 minutes.

In one embodiment, the bioactive agent is administered over a period of between 15 and 120 minutes, such as between 15 and 20 minutes, such as between 20 and 30 minutes, such as between 30 and 40 minutes, such as between 40 and 50 minutes, such as between 50 and 60 minutes, such as between 60 and 90 minutes, such as between 90 and 120 minutes.

In one embodiment, the bioactive agent is administered once daily for a duration of 60 minutes, or twice daily for a duration of 30 minutes, or three times daily for a duration of 20 minutes, or four times daily for a duration of 15 minutes, or five times daily for a duration of 12 minutes, wherein the duration is the duration of each individual administration.

In one embodiment, the bioactive agent is administered at a dose of at least 500pmol/kg/min, such as at least 1000pmol/kg/min, such as at least 1200pmol/kg/min, such as at least 1500pmol/kg/min, such as at least 2000pmol/kg/min, such as at least 2500pmol/kg/min, such as at least 5000 pmol/kg/min.

One skilled in the art will appreciate that if the number of daily administrations is increased, the dose administered in each administration can be correspondingly decreased. Likewise, if the duration of each administration is decreased, the dosage may be increased accordingly.

The bioactive agent to be administered is a peptide according to the present disclosure. In a preferred embodiment, the peptide is SEQ ID NO 1 or SEQ ID NO 2, or a functional variant having at least 75% sequence identity to either of SEQ ID NO 1 and SEQ ID NO 2, wherein the peptide is modified by attaching at least one fatty acid molecule at one or more amino acid residues of either of SEQ ID NO 1 and SEQ ID NO 2 or a functional variant thereof, with the proviso that the at least one fatty acid molecule is not attached to amino acid residue 30 of either of SEQ ID NO 1 and SEQ ID NO 2.

In one embodiment, the bioactive agent is administered with one or more other active ingredients. These other ingredients may be pharmaceutically active. In some embodiments, the bioactive agent is a peptide as defined above and the other ingredient is hGIP1-42 or a variant thereof.

Route of administration

It will be appreciated that the preferred route of administration will depend upon the general condition and age of the subject to be treated, the nature of the condition to be treated, the location of the tissue to be treated in the body and the active ingredient selected.

Systemic treatment

For systemic treatment according to the present disclosure, the route of administration is capable of introducing the bioactive agent into the bloodstream to ultimately target the desired site of action.

These routes of administration are any suitable route, such as enteral (including oral, rectal, nasal, pulmonary, buccal, sublingual, transdermal, intracisternal and intraperitoneal) and/or parenteral (including subcutaneous, intramuscular, intrathecal, intracerebral, intravenous and intradermal) routes.

Parenteral administration

Parenteral administration refers to any route of administration that is not oral/enteral, whereby the agent avoids prior degradation in the liver. Thus, parenteral administration includes any injection and infusion, for example a bolus or continuous infusion, such as intravenous, intramuscular or subcutaneous administration. In addition, parenteral administration includes inhalation and topical administration.

Thus, the bioactive agent can be administered topically to cross any mucosal membrane of the animal to which the bioactive substance is to be administered, for example, in the nose, vagina, eye, mouth, reproductive tract, lung, gastrointestinal tract, or rectum, preferably the nasal or oral mucosa, and thus, parenteral administration can also include buccal, sublingual, nasal, rectal, vaginal, and intraperitoneal administration, as well as pulmonary and bronchial administration by inhalation or mounting. In addition, the agent may also be administered topically to penetrate the skin.

Topical treatment

In one embodiment, the bioactive agents of the present invention may be used as a topical treatment, i.e., introduced directly to one or more sites of action. Thus, the bioactive agent may be administered directly to the skin or mucosa, or the bioactive agent may be injected to the site of action, for example, into the diseased tissue or directly into the terminal artery leading through the diseased tissue. These administration forms preferably avoid the blood brain barrier.

Multi-component kit

The present disclosure also relates to a kit of parts comprising one or more of the above bioactive agents and at least one other or additional component, such as one or more second active ingredients.

Reference to the literature

1.Baggio LL,Drucker DJ.Biology of Incretins:GLP-1andGIP.Gastroenterology 2007;132(6):2131-2157.

2.Holst JJ.On the Physiology of GIP and GLP-1.Horm Metab Res2004;36(11/12):747-754.

3.Heer J,Rasmussen C,Coy DH,Holst JJ.Glucagon-like peptide-1,but notglucose-dependent insulinotropic peptide,inhibits glucagon secretion viasomatostatin(receptor subtype 2)in the perfused ratpancreas.Diabetologia2008;51(12):2263-2270.

4.Gutniak M,

Figure BDA0002296622780000571

C,Holst JJ,B,

Figure BDA0002296622780000573

S.AntidiabetogenicEffect of Glucagon-like Peptide-1amide in Normal Subjects andPatients with Diabetes Mellitus.N Engl J Med1992;326(20):1316-1322.

5.Christensen M,Vedtofte L,Holst JJ,Vilsboell T,Knop FK.Glucose-Dependent Insulinotropic Polypeptide:A Bifunctional Glucose-DependentRegulator of Glucagon and Insulin Secretion in Humans.Diabetes 2011;60(12):3103-3109.

6.Pederson R,Brown J.Interaction of Gastric Inhibitory Polypeptide,Glucose,and Arginine on Insulin and Glucagon Secretion from the Perfused RatPancreas.Endocrinology 1978;103(2):610-615.

7.Adrian TE,Bloom SR,Hermansen K,Iversen J.Pancreatic polypeptide,glucagon and insulin secretion from the isolated perfused caninepancreas.Diabetologia 1978;14(6):413-417.

8.Brunicardi FC,Druck P,Seymour NE,Sun YS,Elahi D,AndersenDK.Selective neurohormonal interactions in islet cell secretion in theisolated perfused human pancreas.Journal of Surgical Research 1990;48(4):273-278.

9.Dupre J,Caussignac Y,McDonald TJ,Van Vliet S.Stimulation ofGlucagon Secretion by Gastric Inhibitory Polypeptide in Patients with HepaticCirrhosis and Hyperglucagonemia.The Journal of Clinical Endocrinology&Metabolism 1991;72(1):125-129.

10.Ding WG,Renstrom E,Rorsman P,Buschard K,Gromada J.Glucagon-likepeptide I and glucose-dependent insulinotropic polypeptide stimulate Ca2+-induced secretion in rat alpha-cells by a protein kinase A-mediatedmechanism.Diabetes 1997;46(5):792-800.

11.Meier JJ,Gallwitz B,Siepmann N et al.Gastric inhibitorypolypeptide(GIP)dose-dependently stimulates glucagon secretion in healthyhuman subjects at euglycaemia.Diabetologia 2003;46(6):798-801.

12.Christensen MB,Calanna S,Holst JJ,Vilsboell T,Knop FK.Glucose-dependent Insulinotropic Polypeptide:Blood Glucose Stabilizing Effects inPatients With Type 2 Diabetes.The Journal of Clinical Endocrinology&Metabolism 2013;99(3):E418-E426.

13.Christensen M,Calanna S,Sparre-Ulrich AH et al.Glucose-DependentInsulinotropic Polypeptide Augments Glucagon Responses to Hypoglycemia inType 1 Diabetes.Diabetes 2014.

14.Song DH,

Figure BDA0002296622780000581

Kaushik L,Tseng E,Simon J,Corkey BE,WolfeMM.Glucose-Dependent Insulinotropic Polypeptide Enhances AdipocyteDevelopment and Glucose Uptake in Part Through AktActivation.Gastroenterology 2007;133(6):1796-1805.

15.Miyawaki K,Yamada Y,Ban N et al.Inhibition of gastric inhibitorypolypeptide signaling prevents obesity.Nat Med 2002;8(7):738-742.

16.Starich GH,Bar RS,Mazzaferri EL.GIP increases insulin receptoraffinity and cellular sensitivity in adipocytes.Am J Physiol 1985;249(6 Pt1):E603-E607.

17.Getty-Kaushik L,Song DH,Boylan MO,Corkey BE,Wolfe MM.Glucose-Dependent Insulinotropic Polypeptide Modulates Adipocyte Lipolysis andReesterification.Obesity 2006;14(7):1124-1131.

18.Hauner H,Glatting G,Kaminska D,Pfeiffer EF.Effects of gastricinhibitory polypeptide on glucose and lipid metabolism of isolated ratadipocytes.Ann Nutr Metab 1988;32(5-6):282-288.

19.Kim SJ,Nian C,Karunakaran S,Clee SM,Isales CM,McIntosh CHS.GIP-Overexpressing Mice Demonstrate Reduced Diet-Induced Obesity and Steatosis,and Improved Glucose Homeostasis.PLoS ONE 2012;7(7):e40156.

20.Nasteska D,Harada N,Suzuki K et al.Chronic Reduction of GIPSecretion Alleviates Obesity and Insulin Resistance Under High-Fat DietConditions.Diabetes 2014;63(7):2332-2343.

21.Miyawaki K,Yamada Y,Yano H et al.Glucose intolerance caused by adefect in the entero-insular axis:A study in gastric inhibitory polypeptidereceptor knockout mice.Proceedings of the National Academy of Sciences1999;96(26):14843-14847.

22.Ahlqvist E,Osmark P,Kuulasmaa T et al.Link Between GIP andOsteopontin in Adipose Tissue and Insulin Resistance.Diabetes2013;62(6):2088-2094.

23.Calanna S,Christensen M,Holst JJ et al.Secretion of Glucose-Dependent Insulinotropic Polypeptide in Patients With Type 2 Diabetes:Systematic review and meta-analysis of clinical studies.Diabetes Care2013;36(10):3346-3352.

24.Asmar M,Simonsen L,Madsbad S,Stallknecht B,Holst JJ,B++lowJ.Glucose-Dependent Insulinotropic Polypeptide May Enhance Fatty Acid Re-esterification in Subcutaneous Abdominal Adipose Tissue in LeanHumans.Diabetes 2010;59(9):2160-2163.

25.Deschamps I,Heptner W,Desjeux JF,Baltakse V,Machinot S,LestradetH.Effects of diet on insulin and gastric inhibitory polypeptide levels inobese children.Pediatr Res 1980;14(4 Pt 1):300-303.

26.C,Jensen CB,Storgaard H et al.Impact of short-term high-fatfeeding on glucose and insulin metabolism in young healthy men.The Journal ofPhysiology 2009;587(10):2387-2397.

27.Raufman JP,Singh L,Eng J.Exendin-3,a novel peptide from Helodermahorridum venom,interacts with vasoactive intestinal peptide receptors and anewly described receptor on dispersed acini from guinea pigpancreas.Description of exendin-3(9-39)amide,a specific exendin receptorantagonist.Journal of Biological Chemistry 1991;266(5):2897-2902.

28.

Figure BDA0002296622780000601

NB,Dirksen C,Bojsen-

Figure BDA0002296622780000602

KN et al.Exaggerated Glucagon-Like Peptide 1 Response Is Important for Improved+

Figure BDA0002296622780000604

-Cell Function and GlucoseTolerance After Roux-en-Y Gastric Bypass in Patients With Type2Diabetes.Diabetes 2013;62(9):3044-3052.

29.Nakamura T,Tanimoto H,Mizuno Y,Tsubamoto Y,Noda H.Biological andfunctional characteristics of a novel

Figure BDA0002296622780000603

weight antagonist ofglucose-dependent insulinotropic polypeptide receptor,SKL-14959,in vitro andin vivo.Diabetes,Obesity and Metabolism 2012;14(6):511-517.

30.Ebert R,Illmer K,Creutzfeldt W.Release of gastric inhibitorypolypeptide(GIP)by intraduodenal acidification in rats and humans andabolishment of the incretin effect of acid by GIP-antiserum inrats.Gastroenterology 1979;76(3):515-523.

31.Fulurija A,Lutz TA,Sladko K et al.Vaccination against GIP fortheTreatment of Obesity.PLoS ONE 2008;3(9):e3163.

32.Irwin N,McClean PL,Patterson S,Hunter K,Flatt PR.Activeimmunisation against gastric inhibitory polypeptide(GIP)improves bloodglucose control in an animal model of obesity-diabetes.BiologicalChemistry.bchm 390,75.2009.16-7-2014.

33.Hinke SA,Manhart S,Pamir N et al.Identification of a bioactivedomain in the amino-terminus of glucose-dependent insulinotropic polypeptide(GIP).Biochimica et Biophysica Acta(BBA)-Protein Structure and MolecularEnzymology 2001;1547(1):143-155.

34.Tseng CC,Kieffer TJ,Jarboe LA,Usdin TB,Wolfe MM.Postprandialstimulation of insulin release by glucose-dependent insulinotropicpolypeptide(GIP).Effect of a specific glucose-dependent insulinotropicpolypeptide receptor antagonist in the rat.J Clin Invest 1996;98(11):2440-2445.

35.Irwin N,Green BD,Parker JC,Gault VA,O'Harte FPM,FlattPR.Biological activity and antidiabetic potential of synthetic fragmentpeptides of glucose-dependent insulinotropic polypeptide,GIP(1-16)and(Pro3)GIP(1-16).Regulatory Peptides 2006;135

Figure BDA0002296622780000611

:45-53.

36.Kerr BD,Flatt AJS,Flatt PR,Gault VA.Characterization andbiological actions of N-terminal truncated forms of glucose-dependentinsulinotropic polypeptide.Biochemical and Biophysical ResearchCommunications2011;404(3):870-876.

37.Gelling RW,Coy DH,Pederson RA et al.GIP(6-30amide)contains thehigh affinity binding region of GIP and is a potent inhibitor of GIP1-42action in vitro.Regulatory Peptides 1997;69(3):151-154.

38.Deacon CFP.GIP-(3-42)does not antagonize insulinotropic effects ofGIP at physiological concentrations.American Journal of Physiology-Endocrinology and Metabolism 2006;291(3):E468-E475.

39.Gault VA,O'Harte FPM,Harriott P,Flatt PR.Characterization of theCellular and Metabolic Effects of a Novel Enzyme-Resistant Antagonist ofGlucose-Dependent Insulinotropic Polypeptide.Biochemical and BiophysicalResearch Communications 2002;290(5):1420-1426.

40.Ravn P,Madhurantakam C,Kunze S et al.Structural andPharmacological Characterization of Novel Potent and Selective MonoclonalAntibody Antagonists of Glucose-dependent Insulinotropic PolypeptideReceptor.Journal of Biological Chemistry 2013;288(27):19760-19772.

41.Deacon CF,Plamboeck A,Rosenkilde MM,de Heer J,Holst JJ.GIP-(3-42)does not antagonize insulinotropic effects of GIP at physiologicalconcentrations.American Journal of Physiology-Endocrinology andMetabolism2006;291(3):E468-E475.

42.Goetze JP,Hunter I,Lippert SK,Bardram L,Rehfeld JF.Processing-independent analysis of peptide hormones and prohormones in plasma.FrontBiosci 2012;17:1804-1815.

43.Goetze JP,Rehfeld JF.Peptide hormones and their prohormones asbiomarkers.Biomarkers Med 2009;3(4):335-338.

44.Fujita Y,Asadi A,Yang GK,Kwok YN,Kieffer TJ.Differentialprocessing of pro-glucose-dependent insulinotropic polypeptide ingut.American Journal of Physiology-Gastrointestinal and Liver Physiology2010;298(5):G608-G614.

45.Widenmaier SB,Kim SJ,Yang GK et al.A GIP Receptor Agonist Exhibitsbeta-Cell Anti-Apoptotic Actions in Rat Models of Diabetes Resulting inImproved beta-Cell Function and Glycemic Control.PLoS ONE2010;5(3):e9590.

46.Graham FL,van der Eb AJ.A new technique for the assay ofinfectivity of human adenovirus 5 DNA.Virology 1973;52(2):456-467.

47.Kissow H,Hartmann B,Holst JJ et al.Glucagon-like peptide-1(GLP-1)receptor agonism or DPP-4 inhibition does not accelerate neoplasia incarcinogen treated mice.Regulatory Peptides 2012;179

Figure BDA0002296622780000621

:91-100.

48.Hoejberg PV,Vilsboell T,Raboel R et al.Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-likepeptide-1 and glucose-dependent insulinotropic polypeptide in patients withtype 2 diabetes.Diabetologia 2009;52(2):199-207.

DEBLASI,A.,O'REILLY,K.&MOTULSKY,H.J.1989.Calculating receptor numberfrom binding experiments using same compound as radioligand andcompetitor.Trends in Pharmacological Sciences,10,227-229.

LAZARENO,S.&BIRDSALL,N.J.1993.Estimation of competitive antagonistaffinity from functional inhibition curves using the Gaddum,Schild and Cheng-Prusoff equations.Br J Pharmacol,109,1110-9.

ROSENKILDE,M.M.,CAHIR,M.,GETHER,U.,HJORTH,S.A.&SCHWARTZ,T.W.1994.Mutations along transmembrane segment II of the NK-1 receptor affectsubstance P competition with non-peptide antagonists but not substance Pbinding.J Biol Chem,269,28160-4.

HOLST,J.J.&BERSANI,M.1991.1-Assays for Peptide Products ofSomatostatin Gene Expression.In:CONN,P.M.(ed.)Methods inNeurosciences.Academic Press.

PATHAK,V.,GAULT,V.A.,FLATT,P.R.&IRWIN,N.2015.Antagonism of gastricinhibitory polypeptide(GIP)by palmitoylation of GIP analogues with N-and C-terminal modifications improves obesity and metabolic control in high fat fedmice.Mol Cell Endocrinol,401,120-9.

HANSEN LS,SPARRE-ULRICH AH,CHRISTENSEN M,KNOP FK,HARTMANN B,HOLST JJ&ROSENKILDE M.N-terminally and C-terminally truncated forms of glucose-dependent insulinotropic polypeptide are high-affinity competitiveantagonists of the human GIP receptor.British Journal of Pharmacology(2016)173,826-838.

139页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:新型抗CD3抗体

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!