ACYLATED GLP-1/GLP-2 DUAL AGONISTS

A compound having agonist activity at the GLP-1 (glucagon-like-peptide 1) and GLP-2 (glucagon-like peptide 2) receptors, and a pharmaceutical composition containing the compound or a pharmaceutically acceptable salt or solvate thereof in admixture with a pharmaceutically acceptable carrier, an excipient or a vehicle are provided. The compound can be used, inter alia, in the prophylaxis or treatment of intestinal damage and dysfunction, regulation of body weight, and prophylaxis or treatment of metabolic dysfunction.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
INCORPORATION BY REFERENCE TO ANY PRIORITY APPLICATIONS

Any and all applications for which a foreign or domestic priority claim is identified in the Application Data Sheet as filed with the present application are hereby incorporated by reference under 37 CFR 1.57.

SEQUENCE LISTING IN ELECTRONIC FORMAT

The present application is being filed along with an Electronic Sequence Listing as an ASCII text file via EFS-Web. The Electronic Sequence Listing is provided as a file entitled 29418620_1.txt created and last saved on Nov. 13, 2018, which is approximately 113 kilobytes in size. The information in the Electronic Sequence Listing is incorporated herein by reference in its entirety in accordance with 35 U.S.C. § 1.52(e).

BACKGROUND OF THE INVENTION Field of the Invention

The present invention relates to acylated compounds having agonist activity at the GLP-1 (glucagon-like-peptide 1) and GLP-2 (glucagon-like peptide 2) receptors. The compounds find use, inter alia, in the prophylaxis or treatment of intestinal damage and dysfunction, regulation of body weight, and prophylaxis or treatment of metabolic dysfunction.

Description of the Related Art

Intestinal tissue is responsible for the production of both human glucagon-like peptide 1 (GLP-1(7-36)) and human glucagon-like peptide 2 (GLP-2 (1-33)) as they are produced by the same cells. Human GLP-2 is a 33-amino-acid peptide with the following sequence: Hy-His-Ala-Asp-Gly-Ser-Phe-Ser-Asp-Glu-Met-Asn-Thr-Ile-Leu-Asp-Asn-Leu-Ala-Ala-Arg-Asp-Phe-Ile-Asn-Trp-Leu-Ile-Gln-Thr-Lys-Ile-Thr-Asp-OH (SEQ ID NO 1). It is derived from specific posttranslational processing of proglucagon in the enteroendocrine L cells of the intestine and in specific regions of the brainstem. GLP-2 binds to a single G-protein-coupled receptor belonging to the class II glucagon secretin family. GLP-2 is co-secreted with GLP-1, oxyntomodulin and glicentin, in response to nutrient ingestion. Human GLP-1 is produced as a 30-amino acid peptide with the following sequence: Hy-His-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Arg- Gly-NH2 (SEQ ID NO 2).

GLP-2 has been reported to induce significant growth of the small intestinal mucosal epithelium via the stimulation of stem cell proliferation in the crypts, and by inhibition of apoptosis in the villi (Drucker et al., 1996, Proc. Natl. Acad. Sci. USA 93: 7911-7916). GLP-2 also has growth effects on the colon. Furthermore, GLP-2 inhibits gastric emptying and gastric acid secretion (Wojdemann et al., 1999, J. Clin. Endocrinol. Metab. 84: 2513-2517), enhances intestinal barrier function (Benjamin et al., 2000, Gut 47: 112-119), stimulates intestinal hexose transport via the upregulation of glucose transporters (Cheeseman, 1997, Am. J. Physiol. R1965-71), and increases intestinal blood flow (Guan et al., 2003, Gastroenterology, 125: 136-147).

GLP-1 has been described as a physiological incretin hormone and has thus been mostly reported to augment an insulin response after an oral intake of glucose or fat. It is, however, generally understood that GLP-1 lowers glucagon concentrations, has beneficial effects on inhibition of fast bowel movements (Tolessa et al., 1998, Dig. Dis. Sci. 43(10): 2284-90), and slows gastric emptying.

WO2013/164484 discloses GLP-2 analogues which comprise one or more substitutions compared to h[Gly2]GLP-2 and which may have the property of an altered GLP-1 activity, and their medical use.

WO2016/066818 describes peptides having dual agonist activity at the GLP-1 and GLP-2 receptors, and proposes medical uses thereof. However, there remains a need for further compounds which combine effective agonist activities at both receptors with acceptable levels of stability.

SUMMARY OF THE INVENTION

Broadly, the present invention relates to compounds which have agonist activity at the GLP-1 (glucagon-like peptide 1) and GLP-2 (glucagon-like peptide 2) receptors, e.g. as assessed in in vitro potency assays. Such compounds are referred to in this specification as “GLP-1/GLP-2 dual agonists”, or simply “dual agonists”. Thus, the compounds of the present invention have activities of both GLP-1 (7-36) and GLP-2 (1-33).

In a first aspect there is provided a GLP-1/GLP-2 dual agonist represented by the formula:


R1—X*—U—R2

wherein:

R1 is hydrogen (Hy), 01-4 alkyl (e.g. methyl), acetyl, formyl, benzoyl or trifluoroacetyl;

R2 is NH2 or OH;

X* is a peptide of formula I:


H—X2-EG-X5-F—X7-X8-E-X10-X11-TIL-X15-X16-X17-A-X19-X20-X21-FI—X24-WL-X27-X28-X29-KIT-X33  (I)

wherein:

X2 is Aib or G

X5 is T or S;

X7 is T or S;

X8 is S, E or D;

X10 is L, M, V or ψ;

X11 is A, N or S;

X15 is D or E;

X16 is G, E, A or ψ;

X17 is Q, E, K, L or ψ;

X19 is A, V or S;

X20 is R, K or ψ;

X21 is D, L or E;

X24 is A, N or S;

X27 is I, Q, K, H or Y;

X28 is Q, E, A, H, Y, L, K, R or S;

X29 is H, Y, K or Q;

X33 is D or E;

U is absent or a sequence of 1-15 residues each independently selected from K, k, E, A, T, I, L and ψ;

the molecule contains one and only one ψ, wherein ψ is a residue of K, k, R, Orn, Dap or Dab in which the side chain is conjugated to a substituent having the formula Z1— or Z1—Z2—, wherein

Z1— is CH3—(CH2)10-22—(CO)— or HOOC—(CH2)10-22—(CO)—; and

—Z2— is selected from —ZS1—, —ZS1—ZS2, —ZS2—ZS1—, —ZS2—, —ZS3—, —ZS1ZS3—, —ZS2ZS3—, —ZS3ZS1—, —ZS3ZS2—, —ZS1ZS2ZS3—, —ZS1ZS3ZS2—, —ZS2ZS1ZS3—, —ZS2ZS3ZS1—, —ZS3ZS1ZS2—, —ZS3ZS2ZS1—, —ZS2ZS3ZS2— wherein

—ZS3— is isoGlu, β-Ala, isoLys, or 4-aminobutanoyl;

ZS2 is —(Peg3)m-where m is 1, 2, or 3; and

—ZS3— is a peptide sequence of 1-6 amino acid units independently selected from the group consisting of A, L, S, T, Y, Q, D, E, K, k, R, H, F and G;

and wherein at least one of X5 and X7 is T;

or a pharmaceutically acceptable salt or solvate thereof.

The various amino acid positions in peptide X* of the formulae provided here are numbered according to their linear position from N- to C-terminus in the amino acid chain.

In the present context, β-Ala and 3-Aminopropanoyl are used interchangeably.

Dual agonists having aspartic acid (Asp, D) at position 3 and glycine (Gly) in position 4 can be very potent agonists at the GLP-1 and GLP-2 receptors. However, this combination of substitutions results in compounds which are unstable and may not be suitable for long term storage in aqueous solution. Without wishing to be bound by theory, it is believed that the Asp at position 3 may isomerise to iso-Asp via a cyclic intermediate formed between the carboxylic acid functional group of its side chain and the backbone nitrogen atom of the residue at position 4.

It has now been found that molecules having glutamic acid (Glu, E) at position 3 instead of Asp are much less susceptible to such reactions and hence may be considerably more stable when stored in aqueous solution. However, replacement of Asp with Glu at position 3 in molecules having a lipophilic substituent in the middle portion of the peptide (e.g. at or near to positions 16 and 17) tends to reduce the potency at one or both of the GLP-2 receptor and the GLP-1 receptor, even though Glu is present at position 3 of the native GLP-1 molecule. Simultaneously incorporating a Thr residue at one or both of positions 5 and 7 appears to compensate for some or all of the lost potency. It is believed that further improvements in potency are also provided by incorporation of His (H), Tyr (Y), Lys (K) or Gln (Q) at position 29 instead of the Gly (G) and Thr (T) residues present in wild type human GLP-1 and 2 respectively.

In some embodiments of formula I:

X2 is Aib or G

X5 is T or S;

X7 is T or S;

X8 is S;

X10 is L or ψ;

X11 is A or S;

X15 is D or E;

X16 is G, E, A or ψ;

X17 is Q, E, K, L or ψ;

X19 is A or S;

X20 is R or ψ;

X21 is D, L or E;

X24 is A;

X27 is I, Q, K, or Y;

X28 is Q, E, A, H, Y, L, K, R or S;

X29 is H, Y or Q; and

X33 is D or E.

Where ψ is not at X16 or X17, it may be desirable that X16 is E and X17 is Q.

In some embodiments, X11 is A and X15 is D. In other embodiments, X11 is S and X15 is E. In further embodiments, X11 is A and X15 is E.

In some embodiments, X27 is I.

In some embodiments, X29 is H. In certain of these embodiments, X28 is A and X29 is H, or X28 is E and X29 is H.

In some embodiments, X29 is Q and optionally X27 is Q.

In some embodiments, the residues at X27-X29 have a sequence selected from:

IQH;

IEH

IAH;

IHH;

IYH;

ILH;

IKH;

IRH;

ISH;

QQH;

YQH;

KQH;

IQQ;

IQY;

IQT; and

IAY.

In some embodiments, X* is a peptide of formula II:


H—X2-EG-X5-F—X7-SELATILD-X16-X17-AAR—X21-FIAWLI—X28-X29-KITD  (II)

wherein:

X2 is Aib or G

X5 is T or S;

X7 is T or S;

X16 is G or ψ;

X17 is Q, E, K, L or ψ;

X21 is D or L;

X28 is Q, E, A, H, Y, L, K, R or S;

X29 is H, Y or Q;

In some embodiments of Formula I or Formula II, X16 is ψ and X17 is Q, E, K or L. For example, X17 may be Q, or X17 may be selected from E, K and L. In other embodiments, X16 is G and X17 is ψ.

It may be desirable that X21 is D.

X28 may be selected from Q, E and A, e.g. it may be Q or E. In some residue combinations, Q may be preferred. In others, E may be preferred, including but not limited to when X16 is G and X17 is ψ. Alternatively, X28 may be selected from A, H, Y, L, K, R and S.

X* may be a peptide of formula III:


H[Aib]EG-X5-F—X7-SE-X10-ATILD-X16-X17-AA-X20-X21-FIAWLI—X28-X29-KITD  (III)

wherein:

X5 is T or S;

X7 is T or S;

X10 is L or ψ;

X16 is G, E, A or ψ;

X17 is Q, E, K, L or ψ;

X20 is R or ψ;

X21 is D or L;

X28 is E, A or Q;

X29 is H, Y or Q;

and at least one of X5 and X7 is T.

X* may be a peptide of formula IV:


H[Aib]EG-X5-F—X7-SELATILD-X16-X17-AAR—X21-FIAWLI—X28-X29-KITD  (IV)

wherein:

X5 is T or S;

X7 is T or S;

X16 is G or ψ;

X17 is E, K, L or ψ;

X21 is D or L;

X28 is E or A;

X29 is H, Y or Q;

and at least one of X5 and X7 is T.

In some embodiments of any of formulae I to IV, X16 is ψ and X17 is E, K or L.

In other embodiments of formula I to IV, X16 is G and X17 is ψ.

In either case, the following combinations of residues may also be included:

X21 is D and X28 is E;

X21 is D and X28 is A;

X21 is L and X28 is E;

X21 is L and X28 is A.

X* may be a peptide of formula V:


H[Aib]EG-X5-F—X7-SELATILD-ψ-QAARDFIAWLI—X28-X29-KITD  (V)

wherein

X5 is T or S;

X7 is T or S;

X28 is Q, E, A, H, Y, L, K, R or S, e.g. Q, E, A, H, Y or L;

X29 is H, Y or Q;

and at least one of X5 and X7 is T.

In some embodiments of formula III, X28 is Q or E. In some embodiments of formula III, X28 is Q.

In other embodiments, X28 is A, H, Y, L, K, R or S, e.g. A, H, Y or L.

In any of the formulae or embodiments described above, the dual agonist contains one of the following combinations of residues:

X5 is S and X7 is T;

X5 is T and X7 is S;

X5 is T and X7 is T.

It may be preferred that X5 is S and X7 is T, or X5 is T and X7 is T.

In any of the formulae or embodiments described above, it may be desirable that X29 is H.

In some embodiments, ψ is a Lys residue whose side chain is conjugated to the substituent Z1— or Z1—Z2—.

In some embodiments, Z1—, alone or in combination with —Z2—, is dodecanoyl, tetradecanoyl, hexadecanoyl, octadecanoyl or eicosanoyl.

In some embodiments, Z1—, alone or in combination with —Z2—, is:

13-carboxytridecanoyl, i.e. HOOC—(CH2)12—(CO)—;

15-carboxypentadecanoyl, i.e. HOOC—(CH2)14—(CO)—;

17-carboxyheptadecanoyl, i.e. HOOC—(CH2)16—(CO)—;

19-carboxynonadecanoyl, i.e. HOOC—(CH2)18—(CO)—; or

21-carboxyheneicosanoyl, i.e. HOOC—(CH2)20—(CO)—.

In some embodiments Z2 is absent.

In some embodiments, Z2 comprises ZS1 alone or in combination with ZS2 and/or ZS3.

In such embodiments:

—ZS1— is isoGlu, β-Ala, isoLys, or 4-aminobutanoyl;

—ZS2—, when present, is -(Peg3)m-where m is 1, 2, or 3; and

—ZS3— is a peptide sequence of 1-6 amino acid units independently selected from the group consisting of A, L, S, T, Y, Q, D, E, K, k, R, H, F and G, such as the peptide sequence KEK.

Z2 may have the formula —ZS1—ZS3—ZS2—, where ZS1 is bonded to Z1 and ZS2 is bonded to the side chain of the amino acid component of ψ.

Thus, in some embodiments, —Z2— is:

isoGlu(Peg 3)0-3;

β-Ala(Peg 3)0-3;

isoLys(Peg3)0-3; or

4-aminobutanoyl(Peg3)0-3.

In further embodiments, —Z2— is:

isoGlu-KEK-(Peg3)0-3.

Specific examples of the substituent Z1—Z2— are set out below. In some embodiments, Z1—Z2— is [17-carboxy-heptadecanoyl]-isoGlu. For example, ψ may be K([17-carboxy-heptadecanoyl]-isoGlu). In some embodiments, Z1—Z2— is:

[17-Carboxy-heptadecanoyl]-isoGlu-KEK-Peg3-;

[17-carboxy-heptadecanoyl]-isoGlu-Peg3-;

[19-Carboxy-nonadecanoyl]-isoGlu-;

[19-Carboxy-nonadecanoyl]-isoGlu-KEK—;

[19-Carboxy-nonadecanoyl]-isoGlu-KEK-Peg3-;

[19-carboxy-nonadecanoyl]-isoGlu-KEK-Peg3-Peg3-;

[19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3-;

[19-carboxy-nonadecanoyl]-isoLys-Peg3-Peg3-Peg3-;

[Hexadecanoyl]-βAla-;

[Hexadecanoyl]-isoGlu-; or

Octadecanoyl-.

For example, ψ may be:

K([17-Carboxy-heptadecanoyl]-isoGlu-KEK-Peg3);

K([17-carboxy-heptadecanoyl]-isoGlu-Peg3);

K([19-Carboxy-nonadecanoyl]-isoGlu);

K([19-Carboxy-nonadecanoyl]-isoGlu-KEK);

K([19-Carboxy-nonadecanoyl]-isoGlu-KEK-Peg3);

K([19-carboxy-nonadecanoyl]-isoGlu-KEK-Peg3-Peg3);

K([19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3);

K([19-carboxy-nonadecanoyl]-isoLys-Peg3-Peg3-Peg3);

K([Hexadecanoyl]-βAla-;

K([Hexadecanoyl]-isoGlu); or

K(Octadecanoyl).

When present, U represents a peptide sequence of 1-15 residues each independently selected from K (i.e. L-lysine), k (i.e. D-lysine) E (Glu), A (Ala), T (Thr), I (Ile), L (Leu) and ψ. For example, U may be 1-10 amino acids in length, 1-7 amino acids in length, 3-7 amino acids in length, 1-6 amino acids in length, or 3-6 amino acids in length.

Typically U includes at least one charged amino acid (K, k or E) and preferably two or more charged amino acids. In some embodiments it includes at least 2 positively charged amino acids (K or k), or at least 1 positively charged amino acid (K or k) and at least one negatively charged amino acid (E).

In some embodiments, all amino acid residues of U (except for ψ, if present) are charged. For example, U may be a chain of alternately positively and negatively charged amino acids.

In certain embodiments, U comprises residues selected only from K, k, E and ψ.

In certain embodiments, U comprises residues selected only from K, k, and ψ.

When U comprises only lysine residues (whether K or k), all residues may have an L-configuration or all may have a D-configuration. Examples include K1-15, K1-10 and K1-7, e.g., K3, K4, K5, K6 and K7, especially K5 and K6. Further examples include k1-15, k1-10 and k1-7, e.g. k3, k4, k5, k6 and k7, especially k5 and k6.

Further examples of peptide sequences U include KEK, EKEKEK, EkEkEk, AKAAEK, AKEKEK and ATILEK.

In any case, one of those residues may be exchanged for ψ. Where the sequence U contains a residue ψ, it may be desirable that the C-terminal residue of U is ψ. Thus, further examples of sequences U include K1-14-ψ, K1-9-ψ and K1-6-ψ, e.g., K2-ψ, K3-ψ, K4-ψ, K5-ψ and K6-ψ, especially K4-ψ and K5-ψ. Yet further examples include k1-14-ψ, k1-9-ψ, and k1-6-ψ, e.g. k2-ψ, k3-ψ, k4-ψ, k5-ψ and k6-ψ especially k4-ψ and k5-ψ. Yet further examples include KEψ, EKEKEψ, EkEkEψ AKAAEψ, AKEKEψ and ATILEψ.

In some embodiments, U is absent.

In some embodiments, R1 is Hy and/or R2 is OH.

The peptide X* or the peptide X*—U may have the sequence:

H[Aib]EGTFSSELATILDΨEAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDΨEAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDΨEAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDΨKAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDΨKAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDΨKAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDGΨAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDGΨAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDGΨAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDΨLAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDΨLAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDΨLAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDΨLAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILDΨLAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILDΨLAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILDΨEAARLFIAWLIEHKITD; H[Aib]EGTFSSELATILDΨQAARDFIAWLIQHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIQHKITD; H[Aib]EGTFTSELATILDΨQAARDFIAWLIQHKITD; H[Aib]EGTFSSELATILDΨQAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDΨQAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILDΨQAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDΨQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIHHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIYHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLILHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIKHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIRHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLISHKITD H[Aib]EGSFTSELATILDΨQAARDFIAWLQQHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLYQHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLKQHKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIQQKITD; H[Aib]EGSFTSELATILDΨQAARDFIAWLIQYKITD; H[Aib]EGTFSSELSTILEΨQASREFIAWLIAYKITE; H[Aib]EGTFSSELATILDEQAARDFIAWLIAHKITDkkkkkΨ; H[Aib]EGTFTSELATILDEQAARDFIAWLIAHKITDkkkkkΨ; H[Aib]EGSFTSELATILDEQAARDFIAWLIEHKITDkkkkkΨ; H[Aib]EGSFTSEΨATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILEGΨAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDEQAAΨDFIAWLIEHKITD; H[Aib]EGTFTSELATILDEQAAΨDFIAWLIEHKITD; H[Aib]EGTFTSEΨATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDAΨAARDFIAWLIEHKITD; or H[Aib]EGSFTSELATILDAKAAΨDFIAWLIEHKITD.

The peptide X* or the peptide X*—U may have the sequence:

H[Aib]EGTFSSELATILD[K*]EAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K*]EAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K*]EAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K*]KAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K*]KAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K*]KAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDG[K*]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDG[K*]AARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDG[K*]AARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K*]LAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K*]LAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K*]LAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K*]LAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K*]LAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K*]LAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K*]EAARLFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K*]QAARDFIAWLIQHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIQHKITD; H[Aib]EGTFTSELATILD[K*]QAARDFIAWLIQHKITD; H[Aib]EGTFSSELATILD[K*]QAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K*]QAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K*]QAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K*]QAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIHHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIYHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLILHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIKHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIRHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLISHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLQQHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLYQHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLKQHKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIQQKITD; H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIQYKITD; H[Aib]EGTFSSELSTILE[K*]QASREFIAWLIAYKITE; H[Aib]EGTFSSELATILDEQAARDFIAWLIAHKITDkkkkk[k*]; H[Aib]EGTFTSELATILDEQAARDFIAWLIAHKITDkkkkk[k*]; H[Aib]EGSFTSELATILDEQAARDFIAWLIEHKITDkkkkk[k*]; H[Aib]EGSFTSE[K*]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILEG[K*]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDEQAA[K*]DFIAWLIEHKITD; H[Aib]EGTFTSELATILDEQAA[K*]DFIAWLIEHKITD; H[Aib]EGTFTSE[K*]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDA[K*]AARDFIAWLIEHKITD; or H[Aib]EGSFTSELATILDAKAA[K*]DFIAWLIEHKITD;

wherein K* or k* indicates an L or D lysine residue respectively in which the side chain is conjugated to the substituent Z1— or Z1Z2—.

For example, the peptide X* or the peptide X*—U may have the sequence:

H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]EAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]EAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]EAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]KAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]KAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]KAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILDG[K([17-carboxy-heptadecanoyl]- isoGlu)]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDG[K([17-carboxy-heptadecanoyl]- isoGlu)]AARDFIAWLIEHKITD; H[Aib]EGTFTSELATILDG[K([17-carboxy-heptadecanoyl]- isoGlu)]AARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]LAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]EAARLFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIQHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIQHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIQHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIAHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIHHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIYHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLILHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIKHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLIRHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl]- isoGlu)]QAARDFIAWLISHKITD; H[Aib]EGSFTSELATILD[K([Hexadecanoyl]-βAla)] QAARDFIAWLQQHKITD; H[Aib]EGSFTSELATILD[K([17-carboxy-heptadecanoyl] iso-Glu-Peg3)]QAARDFIAWLYQHKITD; H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Glu-Peg3-Peg3)]QAARDFIAWLKOHKITD; H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Lys-Peg3-Peg3-Peg3)]QAARDFIAWLIQQKITD; H[Aib]EGSFTSELATILD[K(Octadecanoyl)] QAARDFIAWLIQYKITD; H[Aib]EGTFSSELSTILE[K(Hexadecanoyl-isoGlu)] QASREFIAWLIAYKITE; H[Aib]EGTFSSELATILDEQAARDFIAWLIAHKITDkkkkkk([17- carboxy-Heptadecanoyl]-isoGlu)]; H[Aib]EGTFTSELATILDEQAARDFIAWLIAHKITDkkkkkk([17- carboxy-Heptadecanoyl]-isoGlu)]; H[Aib]EGSFTSELATILDEQAARDFIAWLIEHKITDkkkkkk([17- carboxy-Heptadecanoyl]-isoGlu)]; H[Aib]EGTFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu)]QAARDFIAWLIQHKITD; H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl]iso-Glu- Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Glu-Peg3-Peg3)]KAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILEG[K([19-carboxy-nonadecanoyl] iso-Glu-Peg3-Peg3)]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)]DFIAWLIEHKITD; H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)]DFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([17-Carboxy-heptadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD; H[Aib]EGTFSSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD; H[Aib]EGTFSSELATILD[K([17-Carboxy-heptadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD; H[Aib]EGTFSSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK)]QAARDFIAWLIQHKITD; H[Aib]EGTFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD; H[Aib]EGSFTSE[K([19-Carboxy-nonadecanoyl]-isoGlu- KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGTFTSE[K([19-Carboxy-nonadecanoyl]-isoGlu- KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl]iso-Glu- KEK-Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD; H[Aib]EGTFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]QAARDFIAWLIAHKITD; H[Aib]EGSFTSELATILD[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]KAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Glu-KEK-Peg3-Peg3)]QAARDFIAWLIEHKITD; H[Aib]EGSFTSELATILEG[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDA[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDA[K([19-carboxy-nonadecanoyl] iso-Glu-KEK-Peg3-Peg3)]AARDFIAWLIEHKITD; H[Aib]EGSFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD; H[Aib]EGTFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD; H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)]DFIAWLIEHKITD; H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)]DFIAWLIEHKITD; or H[Aib]EGSFTSELATILDAKAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD.

The dual agonist may be:

Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH (Compound 1); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH (Compound 2); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH (Compound 3); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH (Compound 4); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH (Compound 5); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH (Compound 6); Hy-H[Aib]EGTFSSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH (Compound 7); Hy-H[Aib]EGSFTSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH (Compound 8); Hy-H[Aib]EGTFTSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH (Compound 9); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH (Compound 10); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH (Compound 11); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH (Compound 12); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH (Compound 13); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH (Compound 14); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH (Compound 15); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARLFIAWLIEHKITD-OH (Compound 16); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH (Compound 17); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH (Compound 18); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH (Compound 19); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH (Compound 20); Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH (Compound 21); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH (Compound 22); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH (Compound 23); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH (Compound 24); Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH (Compound 25); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIHHKITD-OH (Compound 26); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIYHKITD-OH (Compound 27); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLILHKITD-OH (Compound 28); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIKHKITD-OH (Compound 29); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIRHKITD-OH (Compound 30); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLISHKITD-OH (Compound 31). Hy-H[Aib]EGSFTSELATILD[K([Hexadecanoyl]-βAla] QAARDFIAWLQQHKITD-OH (Compound 32); Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]iso-Glu-Peg3)]QAARDFIAWLYQHKITD-OH (Compound 33); Hy-H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Glu-Peg3-Peg3)]QAARDFIAWLKQHKITD-OH (Compound 34); Hy-H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Lys-Peg3-Peg3-Peg3)]QAARDFIAWLIQQKITD-OH (Compound 35); Hy-H[Aib]EGSFTSELATILD[K(Octadecanoyl)] QAARDFIAWLIQYKITD-OH (Compound 36); Hy-H[Aib]EGTFSSELSTILE[K(Hexadecanoyl-isoGlu)] QASREFIAWLIAYKITE-OH (Compound 37); Hy-H[Aib]EGTFSSELATILDEQAARDFIAWLIAHKITDkkkkkk ([17-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] (Compound 38); Hy-H[Aib]EGTFTSELATILDEQAARDFIAWLIAHKITDkkkkkk ([17-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] (Compound 39); Hy-H[Aib]EGSFTSELATILDEQAARDFIAWLIEHKITDkkkkkk ([17-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] (Compound 40); Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH (Compound 41); Hy-H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl]iso- Glu-Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH (Compound 42); Hy-H[Aib]EGSFTSELATILD[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)] KAARDFIAWLIEHKITD-OH (Compound 43); Hy-H[Aib]EGSFTSELATILEG[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)] AARDFIAWLIEHKITD-OH (Compound 44); Hy-H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)]DFIAWLIEHKITD-OH (Compound 45); Hy-H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-Peg3-Peg3)]DFIAWLIEHKITD-OH (Compound 46). Hy-H[Aib]EGTFSSELATILD[K([17-Carboxy- heptadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD- OH (Compound 47); Hy-H[Aib]EGTFSSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD- OH (Compound 48); Hy-H[Aib]EGTFSSELATILD[K([17-Carboxy- heptadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD- OH (Compound 49); Hy-H[Aib]EGTFSSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD- OH (Compound 50); Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK)]QAARDFIAWLIQHKITD-OH (Compound 51); Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIQHKITD- OH (Compound 52); Hy-H[Aib]EGSFTSE[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH (Compound 53); Hy-H[Aib]EGTFTSE[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH (Compound 54); Hy-H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl]iso- Glu-KEK-Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH (Compound 55); Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD- OH (Compound 56); Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIEHKITD- OH (Compound 57); Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]QAARDFIAWLIAHKITD- OH (Compound 58); Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]KAARDFIAWLIEHKITD- OH (Compound 59); Hy-H[Aib]EGSFTSELATILD[K([19-carboxy-nonadecanoyl] iso-Glu-KEK-Peg3-Peg3)]QAARDFIAWLIEHKITD-OH (Compound 60); Hy-H[Aib]EGSFTSELATILEG[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]AARDFIAWLIEHKITD-OH (Compound 61); Hy-H[Aib]EGSFTSELATILDA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]AARDFIAWLIEHKITD-OH (Compound 62); Hy-H[Aib]EGSFTSELATILDA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)] AARDFIAWLIEHKITD-OH (Compound 63); Hy-H[Aib]EGSFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD-OH (Compound 64); Hy-H[Aib]EGTFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD-OH (Compound 65); Hy-H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)]DFIAWLIEHKITD- OH (Compound 66); Hy-H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)]DFIAWLIEHKITD- OH (Compound 67); or Hy-H[Aib]EGSFTSELATILDAKAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD-OH (Compound 68).

The dual agonist may be in the form of a pharmaceutically acceptable salt or solvate, such as a pharmaceutically acceptable acid addition salt.

The invention also provides a composition comprising a dual agonist of the invention, or a pharmaceutically acceptable salt or solvate thereof, together with a carrier, excipient or vehicle. The carrier may be a pharmaceutically acceptable carrier.

The composition may be a pharmaceutical composition. The pharmaceutical composition may be formulated as a liquid suitable for administration by injection or infusion. It may be formulated to achieve slow release of the dual agonist.

The present invention further provides a dual agonist of the invention for use in therapy. In yet another aspect there is provided a dual agonist of the present invention for use as a medicament. Also provided is a dual agonist of the invention for use in a method of medical treatment.

The invention also provides a dual agonist of the invention for use in a method of increasing intestinal mass, improving intestinal function (especially intestinal barrier function), increasing intestinal blood flow, or repairing intestinal damage or dysfunction, e.g. damage to the intestinal epithelium.

The invention also provides a dual agonist of the invention for use in a method of prophylaxis or treatment of malabsorption, ulcers (e.g. peptic ulcers, Zollinger-Ellison Syndrome, drug-induced ulcers, and ulcers related to infections or other pathogens), short-bowel syndrome, cul-de-sac syndrome, inflammatory bowel disease (Crohns disease and ulcerative colitis), irritable bowel syndrome (IBS), pouchitis, celiac sprue (for example arising from gluten induced enteropathy or celiac disease), tropical sprue, hypogammaglobulinemic sprue, mucositis induced by chemotherapy or radiation therapy, diarrhea induced by chemotherapy or radiation therapy, low grade inflammation, metabolic endotoxemia, necrotising enterocolitis, primary biliary cirrhosis, hepatitis, fatty liver disease (including parental nutrition associated gut atrophy, PNALD (Parenteral Nutrition-Associated Liver Disease), NAFLD (Non-Alcoholic Fatty Liver Disease) and NASH (Non-Alcoholic Steatohepatitis)), or gastrointestinal side-effects of inflammatory conditions such as pancreatitis or graft versus host disease (GVHD).

The invention also provides a dual agonist of the invention for use in a method of reducing or inhibiting weight gain, reducing gastric emptying or intestinal transit, reducing food intake, reducing appetite, or promoting weight loss.

The invention also provides a dual agonist of the invention for use in a method of prophylaxis or treatment of obesity, morbid obesity, obesity-linked gallbladder disease, obesity-induced sleep apnea, inadequate glucose control, glucose tolerance, dyslipidaemia (e.g. elevated LDL levels or reduced HDL/LDL ratio), diabetes (e.g. Type 2 diabetes, gestational diabetes), pre-diabetes, metabolic syndrome or hypertension.

The invention also provides a method of increasing intestinal mass, improving intestinal function (especially intestinal barrier function), increasing intestinal blood flow, or repairing intestinal damage or dysfunction in a subject in need thereof, the method comprising administering a dual agonist of the invention to the subject.

The invention also provides a method of prophylaxis or treatment of malabsorption, ulcers (e.g. peptic ulcers, Zollinger-Ellison Syndrome, drug-induced ulcers, and ulcers related to infections or other pathogens), short-bowel syndrome, cul-de-sac syndrome, inflammatory bowel disease (Crohns disease and ulcerative colitis), irritable bowel syndrome (IBS), pouchitis, celiac sprue (for example arising from gluten induced enteropathy or celiac disease), tropical sprue, hypogammaglobulinemic sprue, mucositis induced by chemotherapy or radiation therapy, diarrhea induced by chemotherapy or radiation therapy, low grade inflammation, metabolic endotoxemia, necrotising enterocolitis, primary biliary cirrhosis, hepatitis, fatty liver disease (including parental nutrition associated gut atrophy, PNALD (Parenteral Nutrition-Associated Liver Disease), NAFLD (Non-Alcoholic Fatty Liver Disease) and NASH (Non-Alcoholic Steatohepatitis)), or gastrointestinal side-effects of inflammatory conditions such as pancreatitis or graft versus host disease (GVHD) in a subject in need thereof, the method comprising administering a dual agonist of the invention to the subject.

The invention also provides a method of reducing or inhibiting weight gain, reducing gastric emptying or intestinal transit, reducing food intake, reducing appetite, or promoting weight loss in a subject in need thereof, the method comprising administering a dual agonist of the invention to the subject.

The invention also provides a method of prophylaxis or treatment of obesity, morbid obesity, obesity-linked gallbladder disease, obesity-induced sleep apnea, inadequate glucose control, glucose tolerance, dyslipidaemia (e.g. elevated LDL levels or reduced HDL/LDL ratio), diabetes (e.g. Type 2 diabetes, gestational diabetes), pre-diabetes, metabolic syndrome or hypertension in a subject in need thereof, the method comprising administering a dual agonist of the invention to the subject.

The invention also provides the use of a dual agonist of the invention in the preparation of a medicament for increasing intestinal mass, improving intestinal function (especially intestinal barrier function), increasing intestinal blood flow, or repairing intestinal damage or dysfunction, e.g. damage to the intestinal epithelium.

The invention also provides the use of a dual agonist of the invention in the preparation of a medicament for prophylaxis or treatment of malabsorption, ulcers (e.g. peptic ulcers, Zollinger-Ellison Syndrome, drug-induced ulcers, and ulcers related to infections or other pathogens), short-bowel syndrome, cul-de-sac syndrome, inflammatory bowel disease (Crohns disease and ulcerative colitis), irritable bowel syndrome (IBS), pouchitis, celiac sprue (for example arising from gluten induced enteropathy or celiac disease), tropical sprue, hypogammaglobulinemic sprue, mucositis induced by chemotherapy or radiation therapy, diarrhea induced by chemotherapy or radiation therapy, low grade inflammation, metabolic endotoxemia, necrotising enterocolitis, primary biliary cirrhosis, hepatitis, fatty liver disease (including parental nutrition associated gut atrophy, PNALD (Parenteral Nutrition-Associated Liver Disease), NAFLD (Non-Alcoholic Fatty Liver Disease) and NASH (Non-Alcoholic Steatohepatitis)), or gastrointestinal side-effects of inflammatory conditions such as pancreatitis or graft versus host disease (GVHD).

The invention also provides the use of a dual agonist of the invention in the preparation of a medicament for reducing or inhibiting weight gain, reducing gastric emptying or intestinal transit, reducing food intake, reducing appetite, or promoting weight loss.

The invention also provides the use of a dual agonist of the invention in the preparation of a medicament for prophylaxis or treatment of obesity, morbid obesity, obesity-linked gallbladder disease, obesity-induced sleep apnea, inadequate glucose control, glucose tolerance, dyslipidaemia (e.g. elevated LDL levels or reduced HDL/LDL ratio), diabetes (e.g. Type 2 diabetes, gestational diabetes), pre-diabetes, metabolic syndrome or hypertension.

A further aspect provides a therapeutic kit comprising a dual agonist, or a pharmaceutically acceptable salt or solvate thereof, according to the invention.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT

Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, chemistry, molecular biology, cell and cancer biology, immunology, microbiology, pharmacology, and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art.

All patents, published patent applications and non-patent publications referred to in this application are specifically incorporated by reference herein. In case of conflict, the present specification, including its specific definitions, will control.

Each embodiment of the invention described herein may be taken alone or in combination with one or more other embodiments of the invention.

Definitions

Unless specified otherwise, the following definitions are provided for specific terms which are used in the present written description.

Throughout this specification, the word “comprise”, and grammatical variants thereof, such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated integer or component, or group of integers or components, but not the exclusion of any other integer or component, or group of integers or components.

The singular forms “a,” “an,” and “the” include the plurals unless the context clearly dictates otherwise.

The term “including” is used to mean “including but not limited to”. “Including” and “including but not limited to” may be used interchangeably.

The terms “patient”, “subject” and “individual” may be used interchangeably and refer to either a human or a non-human animal. These terms include mammals such as humans, primates, livestock animals (e.g., bovines and porcines), companion animals (e.g., canines and felines) and rodents (e.g., mice and rats).

The term “solvate” in the context of the present invention refers to a complex of defined stoichiometry formed between a solute (in casu, a peptide or pharmaceutically acceptable salt thereof according to the invention) and a solvent. The solvent in this connection may, for example, be water, ethanol or another pharmaceutically acceptable, typically small-molecular organic species, such as, but not limited to, acetic acid or lactic acid. When the solvent in question is water, such a solvate is normally referred to as a hydrate.

The term “agonist” as employed in the context of the invention refers to a substance (ligand) that activates the receptor type in question.

Throughout the present description and claims the conventional three-letter and one-letter codes for naturally occurring amino acids are used, i.e.

A (Ala), G (Gly), L (Leu), I (Ile), V (Val), F (Phe), W (Trp), S (Ser), T (Thr), Y (Tyr), N (Asn), Q (Gln), D (Asp), E (Glu), K (Lys), R (Arg), H (His), M (Met), C (Cys) and P (Pro);

as well as generally accepted three-letter codes for other α-amino acids, such as sarcosine (Sar), norleucine (Nle), α-aminoisobutyric acid (Aib), 2,3-diaminopropanoic acid (Dap), 2,4-diaminobutanoic acid (Dab) and 2,5-diaminopentanoic acid (ornithine; Orn). Such other α-amino acids may be shown in square brackets “[ ]” (e.g. “[Aib]”) when used in a general formula or sequence in the present specification, especially when the rest of the formula or sequence is shown using the single letter code. Unless otherwise specified, amino acid residues in peptides of the invention are of the L-configuration. However, D-configuration amino acids may be incorporated. In the present context, an amino acid code written with a small letter represents the D-configuration of said amino acid, e.g. “k” represents the D-configuration of lysine (K).

Among sequences disclosed herein are sequences incorporating a “Hy-” moiety at the amino terminus (N-terminus) of the sequence, and either an “—OH” moiety or an “—NH2” moiety at the carboxy terminus (C-terminus) of the sequence. In such cases, and unless otherwise indicated, a “Hy-” moiety at the N-terminus of the sequence in question indicates a hydrogen atom [i.e. R1=hydrogen=Hy in the general formulas; corresponding to the presence of a free primary or secondary amino group at the N-terminus], while an “—OH” or an “—NH2” moiety at the C-terminus of the sequence indicates a hydroxy group [e.g. R2═OH in general formulas; corresponding to the presence of a carboxy (COOH) group at the C-terminus] or an amino group [e.g. R2═[NH2] in the general formulas; corresponding to the presence of an amido (CONH2) group at the C-terminus], respectively. In each sequence of the invention, a C-terminal “—OH” moiety may be substituted for a C-terminal “—NH2” moiety, and vice-versa.

“Percent (%) amino acid sequence identity” with respect to the GLP-2 polypeptide sequences is defined as the percentage of amino acid residues in a candidate sequence that are identical to the amino acid residues in the wild-type (human) GLP-2 sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Sequence alignment can be carried out by the skilled person using techniques well known in the art, for example using publicly available software such as BLAST, BLAST2 or Align software. For examples, see Altschul et al., Methods in Enzymology 266: 460-480 (1996) or Pearson et al., Genomics 46: 24-36, 1997.

The percentage sequence identities used herein in the context of the present invention may be determined using these programs with their default settings. More generally, the skilled worker can readily determine appropriate parameters for determining alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.

Dual Agonist Compounds

In accordance with the present invention, the dual agonist has at least one GLP-1 and at least one GLP-2 biological activity. Exemplary GLP-1 physiological activities include reducing rate of intestinal transit, reducing rate of gastric emptying, reducing appetite, food intake or body weight, and improving glucose control and glucose tolerance. Exemplary GLP-2 physiological activities include causing an increase in intestinal mass (e.g. of small intestine or colon), intestinal repair, and improving intestinal barrier function (i.e. reducing permeability of the intestine). These parameters can be assessed in in vivo assays in which the mass and the permeability of the intestine, or a portion thereof, is determined after a test animal has been treated with a dual agonist.

The dual agonists have agonist activity at the GLP-1 and GLP-2 receptors, e.g. the human GLP-1 and GLP-2 receptors. EC50 values for in vitro receptor agonist activity may be used as a numerical measure of agonist potency at a given receptor. An EC50 value is a measure of the concentration (e.g. mol/L) of a compound required to achieve half of that compound's maximal activity in a particular assay. A compound having a numerical EC50 at a particular receptor which is lower than the EC50 of a reference compound in the same assay may be considered to have higher potency at that receptor than the reference compound.

GLP-1 Activity In some embodiments, the dual agonist has an EC50 at the GLP-1 receptor (e.g. the human GLP-1 receptor) which is below 2.0 nM, below 1.5 nM, below 1.0 nM, below 0.9 nM, below 0.8 nM, below 0.7 nM, below 0.6 nM, below 0.5 nM, below 0.4 nM, below 0.3 nM, below 0.2 nM, below 0.1 nM, below 0.09 nM, below 0.08 nM, below 0.07 nM, below 0.06 nM, below 0.05 nM, below 0.04 nM, e.g. when assessed using the GLP-1 receptor potency assay described in the Examples below.

In some embodiments, the dual agonist has an EC50 at the GLP-1 receptor which is between 0.005 and 2.5 nM, between 0.01 nM and 2.5 nM, between 0.025 and 2.5 nM, between 0.005 and 2.0 nM, between 0.01 nM and 2.0 nM, between 0.025 and 2.0 nM, between 0.005 and 1.5 nM, between 0.01 nM and 1.5 nM, between 0.025 and 1.5 nM, between 0.005 and 1.0 nM, between 0.01 nM and 1.0 nM, between 0.025 and 1.0 nM, between 0.005 and 0.5 nM, between 0.01 nM and 0.5 nM, between 0.025 and 0.5 nM, between 0.005 and 0.25 nM, between 0.01 nM and 0.25 nM, between 0.025 and 0.25 nM, e.g. when assessed using the GLP-1 receptor potency assay described in the Examples below.

An alternative measure of GLP-1 agonist activity may be derived by comparing the potency of a dual agonist with the potency of a known (or reference) GLP-1 agonist when both are measured in the same assay. Thus the relative potency at the GLP-1 receptor may be defined as:


[EC50(reference agonist)]/[EC50(dual agonist)].

Thus a value of 1 indicates that the dual agonist and reference agonist have equal potency, a value of >1 indicates that the dual agonist has higher potency (i.e. lower EC50) than the reference agonist, and a value of <1 indicates that the dual agonist has lower potency (i.e. higher EC50) than the reference agonist.

The reference GLP-1 agonist may, for example, be human GLP-1 (7-37), liraglutide (NN2211; Victoza), or Exendin-4, but is preferably liraglutide.

Typically the relative potency will be between 0.001 and 100, e.g.

between 0.001 and 10, between 0.001 and 5, between 0.001 and 1, between 0.001 and 0.5, between 0.001 and 0.1, between 0.001 and 0.05, or between 0.001 and 0.01;

between 0.01 and 10, between 0.01 and 5, between 0.01 and 1, between 0.01 and 0.5, between 0.01 and 0.1, or between 0.01 and 0.05;

between 0.05 and 10, between 0.05 and 5, between 0.05 and 1, between 0.05 and 0.5, or between 0.05 and 0.1;

between 0.1 and 10, between 0.1 and 5, between 0.1 and 1, or between 0.1 and 0.5;

between 0.5 and 10, between 0.5 and 5, or between 0.5 and 1;

between 1 and 10, or between 1 and 5;

or between 5 and 10.

The dual agonists described in the examples below have slightly lower GLP-1 potency than liraglutide and so may, for example, have a relative potency between 0.01 and 1, between 0.01 and 0.5 or between 0.01 and 0.1.

By contrast, the dual agonists of the invention have higher potency at the GLP-1 receptor (e.g. the human GLP-1 receptor) than wild type human GLP-2 (hGLP-2 (1-33)) or [Gly2]-hGLP-2 (1-33) (i.e. human GLP-2 having glycine at position 2, also known as teduglutide). Thus, the relative potency of the dual agonists at the GLP-1 receptor compared to hGLP-2 (1-33) or teduglutide is greater than 1, typically greater than 5 or greater than 10, and may be up to 100, up to 500, or even higher.

GLP-2 Activity In some embodiments, the dual agonist has an EC50 at the GLP-2 receptor (e.g. the human GLP-2 receptor) which is below 2.0 nM, below 1.5 nM, below 1.0 nM, below 0.9 nM, below 0.8 nM, below 0.7 nM, below 0.6 nM, below 0.5 nM, below 0.4 nM, below 0.3 nM, below 0.2 nM, below 0.1 nM, below 0.09 nM, below 0.08 nM, below 0.07 nM, below 0.06 nM, below 0.05 nM, below 0.04 nM, below 0.03 nM, below 0.02 nM, or below 0.01 nM, e.g. when assessed using the GLP-1 receptor potency assay described in the Examples below.

In some embodiments, the dual agonist has an EC50 at the GLP-2 receptor which is between 0.005 and 2.0 nM, between 0.01 nM and 2.0 nM, between 0.025 and 2.0 nM, between 0.005 and 1.5 nM, between 0.01 nM and 1.5 nM, between 0.025 and 1.5 nM, between 0.005 and 1.0 nM, between 0.01 nM and 1.0 nM, between 0.025 and 1.0 nM, between 0.005 and 0.5 nM, between 0.01 nM and 0.5 nM, between 0.025 and 0.5 nM, between 0.005 and 0.25 nM, between 0.01 nM and 0.25 nM, between 0.025 and 0.25 nM, e.g. when assessed using the GLP-2 receptor potency assay described in the Examples below.

An alternative measure of GLP-2 agonist activity may be derived by comparing the potency of a dual agonist with the potency of a known (or reference) GLP-2 agonist when both are measured in the same assay. Thus the relative potency at the GLP-2 receptor may be defined as:


[EC50(reference agonist)]/[EC50(dual agonist)].

Thus a value of 1 indicates that the dual agonist and reference agonist have equal potency, a value of >1 indicates that the dual agonist has higher potency (i.e. lower EC50) than the reference agonist, and a value of <1 indicates that the dual agonist has lower potency (i.e. higher EC50) than the reference agonist.

The reference GLP-2 agonist may, for example, be human GLP-2 (1-33) or teduglutide ([Gly2]-hGLP-2 (1-33)), but is preferably teduglutide. Typically the relative potency will be between 0.001 and 100, e.g.

between 0.001 and 10, between 0.001 and 5, between 0.001 and 1, between 0.001 and 0.5, between 0.001 and 0.1, between 0.001 and 0.05, or between 0.001 and 0.01;

between 0.01 and 10, between 0.01 and 5, between 0.01 and 1, between 0.01 and 0.5, between 0.01 and 0.1, or between 0.01 and 0.05;

between 0.05 and 10, between 0.05 and 5, between 0.05 and 1, between 0.05 and 0.5, or between 0.05 and 0.1;

between 0.1 and 10, between 0.1 and 5, between 0.1 and 1, or between 0.1 and 0.5;

between 0.5 and 10, between 0.5 and 5, or between 0.5 and 1;

between 1 and 10, or between 1 and 5;

or between 5 and 10.

The dual agonists described in the examples below have slightly lower GLP-2 potency than teduglutide and so may, for example, have a relative potency between 0.01 and 1, between 0.01 and 0.5, or between 0.01 and 0.1.

By contrast, the dual agonists of the invention have higher potency at the GLP-2 receptor (e.g. the human GLP-2 receptor) than human GLP-1 (7-37), liraglutide (NN2211; Victoza), or Exendin-4. Thus, the relative potency of the dual agonists at the GLP-2 receptor compared to human GLP-1 (7-37), liraglutide (NN2211; Victoza), or Exendin-4 is greater than 1, typically greater than 5 or greater than 10, and may be up to 100, up to 500, or even higher (if the reference GLP-1 agonist even exerts detectable activity at the GLP-2 receptor).

It will be understood that the absolute potencies of the dual agonists at each receptor are much less important than the balance between the GLP-1 and GLP-2 agonist activities. Thus it is perfectly acceptable for the absolute GLP-1 or GLP-2 potency to be lower than that of known agonists at those receptors, as long as the dual agonist compound exerts acceptable relative levels of potency at both receptors. Any apparent deficiency in absolute potency can be compensated by an increased dose if required.

Substituents

The dual agonist of the present invention contains a residue ψ which comprises a residue of Lys, Arg, Orn, Dap or Dab in which the side chain is conjugated to a substituent Z1— or Z1—Z2— wherein Z1 represents a moiety CH3—(CH2)10-22—(CO)— or HOOC—(CH2)10-22—(CO)— and Z2 when present represents a spacer.

The spacer Z2 is selected from —ZS1—, —ZS1—ZS2—, —ZS2—ZS1—, —ZS2—, —ZS3—, —ZS1ZS3—, —ZS2ZS3—, —ZS3Z1—, —ZS3ZS2—, —ZS1ZS2ZS3—, —ZS1ZS3ZS2—, —ZS2ZS1ZS3—, —ZS2ZS3Z1—, —ZS3Z1ZS2—, —ZS3ZS2ZS1—, ZS2ZS3ZS2— wherein

ZS1 is isoGlu, β-Ala, isoLys, or 4-aminobutanoyl;

ZS2 is -(Peg3)m-where m is 1, 2, or 3; and

ZS3— is a peptide sequence of 1-6 amino acid units selected from the group consisting of A, L, S, T, Y, Q, D, E, K, k, R, H, F and G.

In some embodiments, Z2 is a spacer of the formula —ZS1—, —ZS1—ZS2—, —ZS2—Zs1, or ZS2, where —ZS1— is isoGlu, β-Ala, isoLys, or 4-aminobutanoyl; and —ZS2— is -(Peg3)m-where m is 1, 2, or 3.

Without wishing to be bound by theory, it is believed that the hydrocarbon chain of Z1 binds albumin in the blood stream, thus shielding the dual agonists of the present invention from enzymatic degradation, which can enhance the half-life of the dual agonists.

The substituent may also modulate the potency of the dual agonists, with respect to the GLP-2 receptor and/or the GLP-1 receptor.

The substituent Z1— or Z1—Z2— is conjugated to the functional group at the distal end of the side-chain from the alpha-carbon of the relevant amino acid residue. The normal ability of the amino acid (Lys, Arg, Orn, Dab, Dap) side-chain in question to participate in interactions mediated by that functional group (e.g. intra- and inter-molecular interactions) may therefore be reduced or completely eliminated by the presence of the substituent. Thus, the overall properties of the dual agonist may be relatively insensitive to changes in the actual amino acid conjugated to the substituent. Consequently, it is believed that any of the residues Lys, Arg, Orn, Dab, or Dap may be present at any position where ψ is permitted. However, in certain embodiments, it may be advantageous that the amino acid to which the substituent is conjugated is Lys or Orn.

The moiety Z1 may be covalently bonded to the functional group in the amino acid side-chain, or alternatively may be conjugated to the amino acid side-chain functional group via a spacer Z2.

The term “conjugated” is used here to describe the covalent attachment of one identifiable chemical moiety to another, and the structural relationship between such moieties. It should not be taken to imply any particular method of synthesis.

The bonds between Z1, ZS1, ZS2, ZS3 and the amino acid side chain to which the substituent is bound (collectively referred to herein as ψ) are peptidic. In other words, the units may be joined by amide condensation reactions.

Z1 comprises a hydrocarbon chain having from 10 to 24 carbon (C) atoms, such as from 10 to 22 C atoms, e.g. from 10 to 20 C atoms. Preferably, it has at least 10 or at least 11 C atoms, and preferably it has 20 C atoms or fewer, e.g. 18 C atoms or fewer. For example, the hydrocarbon chain may contain 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbon atoms. For example, it may contain 18 or 20 carbon atoms.

In some embodiments, Z1 is a group selected from dodecanoyl, tetradecanoyl, hexadecanoyl, octadecanoyl and eicosanoyl, preferably hexadecanoyl, octadecanoyl or eicosanoyl, more preferably octadecanoyl or eicosanoyl.

Alternative Z1 groups are derived from long-chain saturated α,ω-dicarboxylic acids of formula HOOC—(CH2)12-22—COOH, preferably from long-chain saturated α,ω-dicarboxylic acids having an even number of carbon atoms in the aliphatic chain. For example, Z1 may be:

13-carboxytridecanoyl, i.e. HOOC—(CH2)12—(CO)—;

15-carboxypentadecanoyl, i.e. HOOC—(CH2)14—(CO)—;

17-carboxyheptadecanoyl, i.e. HOOC—(CH2)16—(CO)—;

19-carboxynonadecanoyl, i.e. HOOC—(CH2)18—(CO)—; or

21-carboxyheneicosanoyl, i.e. HOOC—(CH2)20—(CO)—.

As mentioned above, Z1 may be conjugated to the amino acid side-chain by a spacer Z2. When present, the spacer is attached to Z1 and to the amino acid side-chain.

The spacer Z2 has the —ZS1—, —ZS1—ZS2—, —ZS2—Z1—, —ZS2—, —ZS3—, —ZS1ZS3—, —ZS2ZS3—, —ZS3Z1—, ZS3ZS2, —ZS1ZS2ZS3—, —ZS1ZS3ZS2—, —ZS2ZS1ZS3—, —ZS2ZS3ZS1—, —ZS3ZS1ZS2—, —ZS3ZS2ZS1—, ZS2ZS3ZS2; where

—ZS1— is isoGlu, 1-Ala, isoLys, or 4-aminobutanoyl;

—ZS2— is -(Peg3)m-where m is 1, 2, or 3; and

—ZS3— is a peptide sequence of 1-6 amino acid units independently selected from the group consisting of A (Ala), L (Leu), S (Ser), T (Thr), Y (Tyr), Q (GIn), D (Asp), E (Glu), K (L-Lys), k (D-Lys), R (Arg), H (His), F (Phe) and G (Gly).

The terms “isoGlu” and “isoLys” indicate residues of amino acids which participate in bonds via their side chain carboxyl or amine functional groups. Thus isoGlu participates in bonds via its alpha amino and side chain carboxyl group, while isoLys participates via its carboxyl and side chain amino groups. In the context of the present specification, the terms “γ-Glu” and “isoGlu” are used interchangeably.

The term Peg3 is used to refer to an 8-amino-3,6-dioxaoctanoyl group.

ZS3 may, for example, be 3 to 6 amino acids in length, i.e. 3, 4, 5 or 6 amino acids in length.

In some embodiments, the amino acids of ZS3 are independently selected from K, k, E, A, T, I and L, e.g. from K, k, E and A, e.g. from K, k and E.

Typically ZS3 includes at least one charged amino acid (K, k, R or E, e.g. K, k or E) and preferably two or more charged amino acids. In some embodiments it includes at least 2 positively charged amino acids (K, k or R, especially K or k), or at least 1 positively charged amino acid (K, k or R, especially K or k) and at least one negatively charged amino acid (E). In some embodiments, all amino acid residues of ZS3 are charged. For example, ZS3 may be a chain of alternately positively and negatively charged amino acids.

Examples of ZS3 moieties include KEK, EKEKEK, kkkkkk, EkEkEk, AKAAEK, AKEKEK and ATILEK.

Without being bound by theory, it is believed that the incorporation of ZS3 into the linker between the fatty acid chain and the peptide backbone may increase the half-life of the dual agonist by enhancing its affinity for serum albumin.

In some embodiments, —Z2— is —ZS1— or —ZS1—ZS2—; in other words, —Z2— is selected from:

isoGlu(Peg3)0-3;

β-Ala(Peg3)0-3;

isoLys(Peg3)0-3; and

4-aminobutanoyl(Peg3)0-3.

Thus, certain examples of substituents Z1— include

[Dodecanoyl], [Tetradecanoyl], [Hexadecanoyl], [Octadecanoyl], [Eicosanoyl], [13-Carboxy-tridecanoyl], [15-Carboxy-pentadecanoyl], [17-Carboxy-heptadecanoyl], [19-Carboxy-nonadecanoyl], [21-carboxy-heneicosanoyl].

More broadly, —Z2— may be —ZS1—, —ZS1—ZS2—, —ZS3—ZS1—, —ZS1—ZS3—, —ZS1—ZS3—ZS2—, —ZS3—ZS2—ZS1— or ZS3—.

Thus, —Z2— may be selected from the group consisting of:

isoGlu(Peg3)0-3;

β-Ala(Peg3)0-3;

isoLys(Peg3)0-3;

4-aminobutanoyl(Peg3)0-3;

isoGlu(KEK)(Peg3)0-3;

β-Ala(KEK)(Peg3)0-3;

isoLys(KEK)(Peg3)0-3;

4-aminobutanoyl(KEK)(Peg3)0-3;

KEK(isoGlu);

KEK(β-Ala);

KEK(isoLys);

KEK(4-aminobutanoyl);

isoGlu(KEK);

β-Ala(KEK);

isoLys(KEK);

4-aminobutanoyl(KEK);

KEK(isoGlu)(Peg3)0-3;

KEK(β-Ala)(Prg3)0-3;

KEK(isoLys)(Peg3)0-3; and

KEK(4-aminobutanoyl)(Peg3)0-3;

Certain examples of substituents Z1—Z2— include:

[Dodecanoyl]-isoGlu, [Tetradecanoyl]-isoGlu, [Hexadecanoyl]-isoGlu, [Octadecanoyl]-isoGlu, [Eicosanoyl]-isoGlu,

[Hexadecanoyl]-βAla, [Octadecanoyl]-βAla, [Eicosanoyl]-βAla, [Tetradecanoyl]-βAla, [Dodecanoyl]-βAla,

[Dodecanoyl]-isoGlu-Peg3, [Tetradecanoyl]-isoGlu-Peg3, [Hexadecanoyl]-isoGlu-Peg3, [Octadecanoyl]-isoGlu-Peg3, [Eicosanoyl]-isoGlu-Peg3,

[Dodecanoyl]-βAla-Peg3, [Tetradecanoyl]-βAla-Peg3, [Hexadecanoyl]-βAla-Peg3, [Octadecanoyl]-βAla-Peg3, [Eicosanoyl]-βAla-Peg3,

[Dodecanoyl]-isoGlu-Peg3-Peg3, [Tetradecanoyl]-isoGlu-Peg3-Peg3, [Hexadecanoyl]-isoGlu-Peg3-Peg3, [Octadecanoyl]-isoGlu-Peg3-Peg3, [Eicosanoyl]-isoGlu-Peg3-Peg3,

[Dodecanoyl]-βAla-Peg3-Peg3, [Tetradecanoyl]-βAla-Peg3-Peg3, [Hexadecanoyl]-βAla-Peg3-Peg3, [Octadecanoyl]-βAla-Peg3-Peg3, [Eicosanoyl]-βAla-Peg3-Peg3,

[Dodecanoyl]-isoGlu-Peg3-Peg3-Peg3, [Tetradecanoyl]-isoGlu-Peg3-Peg3-Peg3, [Hexadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [Octadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [Eicosanoyl]-isoGlu-Peg3-Peg3-Peg3,

[Dodecanoyl]-βAla-Peg3-Peg3-Peg3, [Tetradecanoyl]-βAla-Peg3-Peg3-Peg3, [Hexadecanoyl]-βAla-Peg3-Peg3-Peg3, [Octadecanoyl]-βAla-Peg3-Peg3-Peg3, [Eicosanoyl]-βAla-Peg3-Peg3-Peg3,

[Dodecanoyl]-isoLys, [Tetradecanoyl]-isoLys, [Hexadecanoyl]-isoLys, [Octadecanoyl]-isoLys, [Eicosanoyl]-isoLys,

[Hexadecanoyl]-[4-aminobutanoyl], [Octadecanoyl]-[4-aminobutanoyl], [Eicosanoyl]-[4-aminobutanoyl], [Tetradecanoyl]-[4-aminobutanoyl], [Dodecanoyl]-[4-aminobutanoyl],

[Dodecanoyl]-isoLys-Peg3, [Tetradecanoyl]-isoLys-Peg3, [Hexadecanoyl]-isoLys-Peg3, [Octadecanoyl]-isoLys-Peg3, [Eicosanoyl]-isoLys-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3,

[Octadecanoyl]-[4-aminobutanoyl]-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3,

[Dodecanoyl]-isoLys-Peg3-Peg3, [Tetradecanoyl]-isoLys-Peg3-Peg3, [Hexadecanoyl]-isoLys-Peg3-Peg3, [Octadecanoyl]-isoLys-Peg3-Peg3, [Eicosanoyl]-isoLys-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[Dodecanoyl]-isoLys-Peg3-Peg3-Peg3, [Tetradecanoyl]-isoLys-Peg3-Peg3-Peg3, [Hexadecanoyl]-isoLys-Peg3-Peg3-Peg3, [Octadecanoyl]-isoLys-Peg3-Peg3-Peg3, [Eicosanoyl]-isoLys-Peg3-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu, [15-carboxy-Pentadecanoyl]-isoGlu, [17-carboxy-Heptadecanoyl]-isoGlu, [19-carboxy-Nonadecanoyl]-isoGlu, [21-carboxy-heneicosanoyl]-isoGlu,

[17-carboxy-Heptadecanoyl]-βAla, [19-carboxy-Nonadecanoyl]-Ala, [21-carboxy-heneicosanoyl]-βAla, [15-carboxy-Pentadecanoyl]-βAla, [13-carboxy-tridecanoyl]-βAla,

[13-carboxy-tridecanoyl]-isoGlu-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys, [15-carboxy-Pentadecanoyl]-isoLys, [17-carboxy-Heptadecanoyl]-isoLys, [19-carboxy-Nonadecanoyl]-isoLys, [21-carboxy-heneicosanoyl]-isoLys,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl], [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl], [21-carboxy-heneicosanoyl]-[4-aminobutanoyl], [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl], [13-carboxy-tridecanoyl]-[4-aminobutanoyl],

[13-carboxy-tridecanoyl]-isoLys-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3,

[19-carboxy-Nonadecanoyl]-βAla-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3,

[13-carboxy-tridecanoyl]-isoLys-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3 and [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3.

Further examples of substituents Z1—Z2— include:

[Dodecanoyl]-isoLys, [Tetradecanoyl]-isoLys, [Hexadecanoyl]-isoLys, [Octadecanoyl]-isoLys, [Eicosanoyl]-isoLys,

[Hexadecanoyl]-[4-aminobutanoyl], [Octadecanoyl]-[4-aminobutanoyl], [Eicosanoyl]-[4-aminobutanoyl], [Tetradecanoyl]-[4-aminobutanoyl], [Dodecanoyl]-[4-aminobutanoyl],

[Hexadecanoyl]-KEK, [Octadecanoyl]-KEK, [Eicosanoyl]-KEK, [Tetradecanoyl]-KEK, [Dodecanoyl]-KEK,

[Dodecanoyl]-Peg3, [Tetradecanoyl]-Peg3, [Hexadecanoyl]-Peg3, [Octadecanoyl]-Peg3, [Eicosanoyl]-Peg3,

[Dodecanoyl]-Peg3-Peg3, [Tetradecanoyl]-Peg3-Peg3, [Hexadecanoyl]-Peg3-Peg3, [Octadecanoyl]-Peg3-Peg3, [Eicosanoyl]-Peg3-Peg3,

[Dodecanoyl]-Peg3-Peg3-Peg3, [Tetradecanoyl]-Peg3-Peg3-Peg3, [Hexadecanoyl]-Peg3-Peg3-Peg3, [Octadecanoyl]-Peg3-Peg3-Peg3, [Eicosanoyl]-Peg3-Peg3-Peg3,

[Dodecanoyl]-isoLys-Peg3, [Tetradecanoyl]-isoLys-Peg3, [Hexadecanoyl]-isoLys-Peg3, [Octadecanoyl]-isoLys-Peg3, [Eicosanoyl]-isoLys-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3,

[Dodecanoyl]-KEK-Peg3, [Tetradecanoyl]-KEK-Peg3, [Hexadecanoyl]-KEK-Peg3, [Octadecanoyl]-KEK-Peg3, [Eicosanoyl]-KEK-Peg3,

[Dodecanoyl]-isoLys-Peg3-Peg3, [Tetradecanoyl]-isoLys-Peg3-Peg3, [Hexadecanoyl]-isoLys-Peg3-Peg3, [Octadecanoyl]-isoLys-Peg3-Peg3, [Eicosanoyl]-isoLys-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[Dodecanoyl]-KEK-Peg3-Peg3, [Tetradecanoyl]-KEK-Peg3-Peg3, [Hexadecanoyl]-KEK-Peg3-Peg3, [Octadecanoyl]-KEK-Peg3-Peg3, [Eicosanoyl]-KEK-Peg3-Peg3,

[Dodecanoyl]-isoLys-Peg3-Peg3-Peg3, [Tetradecanoyl]-isoLys-Peg3-Peg3-Peg3, [Hexadecanoyl]-isoLys-Peg3-Peg3-Peg3, [Octadecanoyl]-isoLys-Peg3-Peg3-Peg3, [Eicosanoyl]-isoLys-Peg3-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[Dodecanoyl]-KEK-Peg3-Peg3-Peg3, [Tetradecanoyl]-KEK-Peg3-Peg3-Peg3, [Hexadecanoyl]-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-KEK-Peg3-Peg3-Peg3, [Eicosanoyl]-KEK-Peg3-Peg3-Peg3,

[Dodecanoyl]-isoGlu-KEK-Peg3, [Tetradecanoyl]-isoGlu-KEK-Peg3, [Hexadecanoyl]-isoGlu-KEK-Peg3, [Octadecanoyl]-isoGlu-KEK-Peg3, [Eicosanoyl]-isoGlu-KEK-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-KEK-Peg3,

[Dodecanoyl]-isoLys-KEK-Peg3, [Tetradecanoyl]-isoLys-KEK-Peg3, [Hexadecanoyl]-isoLys-KEK-Peg3, [Octadecanoyl]-isoLys-KEK-Peg3, [Eicosanoyl]-isoLys-KEK-Peg3,

[Dodecanoyl]-βAla-KEK-Peg3, [Tetradecanoyl]-βAla-KEK-Peg3, [Hexadecanoyl]-βAla-KEK-Peg3, [Octadecanoyl]-βAla-KEK-Peg3, [Eicosanoyl]-βAla-KEK-Peg3,

[Dodecanoyl]-isoGlu-KEK-Peg3-Peg3, [Tetradecanoyl]-isoGlu-KEK-Peg3-Peg3, [Hexadecanoyl]-isoGlu-KEK-Peg3-Peg3, [Octadecanoyl]-isoGlu-KEK-Peg3-Peg3, [Eicosanoyl]-isoGlu-KEK-Peg3-Peg3,

[Dodecanoyl]-βAla-KEK-Peg3-Peg3, [Tetradecanoyl]-βAla-KEK-Peg3-Peg3, [Hexadecanoyl]-βAla-KEK-Peg3-Peg3, [Octadecanoyl]-βAla-KEK-Peg3-Peg3, [Eicosanoyl]-βAla-KEK-Peg3-Peg3,

[Dodecanoyl]-isoLys-KEK-Peg3-Peg3, [Tetradecanoyl]-isoLys-KEK-Peg3-Peg3, [Hexadecanoyl]-isoLys-KEK-Peg3-Peg3, [Octadecanoyl]-isoLys-KEK-Peg3-Peg3, [Eicosanoyl]-isoLys-KEK-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[Dodecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [Tetradecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [Hexadecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [Eicosanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3,

[Dodecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [Tetradecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [Hexadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [Eicosanoyl]-βAla-KEK-Peg3-Peg3-Peg3,

[Dodecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [Tetradecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [Hexadecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [Eicosanoyl]-isoLys-KEK-Peg3-Peg3-Peg3,

[Dodecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [Tetradecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [Hexadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [Eicosanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3,

[Dodecanoyl]-KEK-isoGlu-Peg3, [Tetradecanoyl]-KEK-isoGlu-Peg3, [Hexadecanoyl]-KEK-isoGlu-Peg3, [Octadecanoyl]-KEK-isoGlu-Peg3, [Eicosanoyl]-KEK-isoGlu-Peg3,

[Dodecanoyl]-KEK-βAla-Peg3, [Tetradecanoyl]-KEK-βAla-Peg3, [Hexadecanoyl]-KEK-βAla-Peg3, [Octadecanoyl]-KEK-βAla-Peg3, [Eicosanoyl]-KEK-βAla-Peg3,

[Dodecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [Tetradecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [Hexadecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [Octadecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [Eicosanoyl]-KEK-[4-aminobutanoyl]-Peg3,

[Dodecanoyl]-KEK-isoLys-Peg3, [Tetradecanoyl]-KEK-isoLys-Peg3, [Hexadecanoyl]-KEK-isoLys-Peg3, [Octadecanoyl]-KEK-isoLys-Peg3, [Eicosanoyl]-KEK-isoLys-Peg3,

[Dodecanoyl]-KEK-isoGlu-Peg3-Peg3, [Tetradecanoyl]-KEK-isoGlu-Peg3-Peg3, [Hexadecanoyl]-KEK-isoGlu-Peg3-Peg3, [Octadecanoyl]-KEK-isoGlu-Peg3-Peg3, [Eicosanoyl]-KEK-isoGlu-Peg3-Peg3,

[Dodecanoyl]-KEK-βAla-Peg3-Peg3, [Tetradecanoyl]-KEK-βAla-Peg3-Peg3, [Hexadecanoyl]-KEK-βAla-Peg3-Peg3, [Octadecanoyl]-KEK-βAla-Peg3-Peg3, [Eicosanoyl]-βAla-KEK-Peg3-Peg3,

[Dodecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [Tetradecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [Hexadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [Octadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [Eicosanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3,

[Dodecanoyl]-KEK-isoLys-Peg3-Peg3, [Tetradecanoyl]-KEK-isoLys-Peg3-Peg3, [Hexadecanoyl]-KEK-isoLys-Peg3-Peg3, [Octadecanoyl]-KEK-isoLys-Peg3-Peg3, [Eicosanoyl]-KEK-isoLys-Peg3-Peg3,

[Dodecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [Tetradecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [Hexadecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [Octadecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [Eicosanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3,

[Dodecanoyl]-KEK-βAla-Peg3-Peg3-Peg3, [Tetradecanoyl]KEK-βAla-Peg3-Peg3-Peg3, [Hexadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [Octadecanoyl]-KEK-βAla-Peg3-Peg3-Peg3, [Eicosanoyl]-KEK-βAla-Peg3-Peg3-Peg3,

[Dodecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Tetradecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Hexadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Octadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [Eicosanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[Dodecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [Tetradecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [Hexadecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [Octadecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [Eicosanoyl]-KEK-isoLys-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu, [15-carboxy-Pentadecanoyl]-isoGlu, [17-carboxy-Heptadecanoyl]-isoGlu, [19-carboxy-Nonadecanoyl]-isoGlu, [21-carboxy-hen21-carboxy-heneicosanoyl]-isoGlu,

[17-carboxy-Heptadecanoyl]-βAla, [19-carboxy-Nonadecanoyl]-Ala, [21-carboxy-heneicosanoyl]-βAla, [15-carboxy-Pentadecanoyl]-βAla, [13-carboxy-tridecanoyl]-βAla,

[13-carboxy-tridecanoyl]-isoLys, [15-carboxy-Pentadecanoyl]-isoLys, [17-carboxy-Heptadecanoyl]-isoLys, [19-carboxy-Nonadecanoyl]-isoLys, [21-carboxy-heneicosanoyl]-isoLys,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl], [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl], [21-carboxy-heneicosanoyl]-[4-aminobutanoyl], [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl], [13-carboxy-tridecanoyl]-[4-aminobutanoyl],

[17-carboxy-Heptadecanoyl]-KEK, [19-carboxy-Nonadecanoyl]-KEK, [21-carboxy-heneicosanoyl]-KEK, [15-carboxy-Pentadecanoyl]-KEK, [13-carboxy-tridecanoyl]-KEK,

[13-carboxy-tridecanoyl]-Peg3, [15-carboxy-Pentadecanoyl]-Peg3, [17-carboxy-Heptadecanoyl]-Peg3, [19-carboxy-Nonadecanoyl]-Peg3, [21-carboxy-heneicosanoyl]-Peg3,

[13-carboxy-tridecanoyl]-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-Peg3-Peg3, [21-carboxy-heneicosanoyl]-Peg3-Peg3,

[13-carboxy-tridecanoyl]-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3,

[13-carboxy-tridecanoyl]-isoLys-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-Peg3,

[13-carboxy-tridecanoyl]-KEK-Peg3, [15-carboxy-Pentadecanoyl]-KEK-Peg3, [17-carboxy-Heptadecanoyl]-KEK-Peg3, [19-carboxy-Nonadecanoyl]-KEK-Peg3, [21-carboxy-heneicosanoyl]-KEK-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-KEK-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-KEK-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-KEK-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-KEK-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-KEK-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-KEK-Peg3,

[13-carboxy-tridecanoyl]-isoLys-KEK-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-KEK-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-KEK-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-KEK-Peg3, [21-carboxy-heneicosanoyl]-isoLys-KEK-Peg3,

[13-carboxy-tridecanoyl]-βAla-KEK-Peg3, [15-carboxy-Pentadecanoyl]-βAla-KEK-Peg3, [17-carboxy-Heptadecanoyl]-3βAla-KEK-Peg3, [19-carboxy-Nonadecanoyl]-βAla-KEK-Peg3, [21-carboxy-heneicosanoyl]-βAla-KEK-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-KEK-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-KEK-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-KEK-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-KEK-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-KEK-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-KEK-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-KEK-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-KEK-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys-KEK-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-KEK-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-KEK-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-KEK-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoGlu-KEK-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-KEK-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-isoLys-KEK-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-isoLys-KEK-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoGlu-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoGlu-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoGlu-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoGlu-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoGlu-Peg3,

[13-carboxy-tridecanoyl]-KEK-βAla-Peg3, [15-carboxy-Pentadecanoyl]-KEK-βAla-Peg3, [17-carboxy-Heptadecanoyl]-KEK-βAla-Peg3, [19-carboxy-Nonadecanoyl]-KEK-βAla-Peg3, [21-carboxy-heneicosanoyl]-KEK-βAla-Peg3,

[13-carboxy-tridecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [15-carboxy-Pentadecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [17-carboxy-Heptadecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [19-carboxy-Nonadecanoyl]-KEK-[4-aminobutanoyl]-Peg3, [21-carboxy-heneicosanoyl]-KEK-[4-aminobutanoyl]-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoLys-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoLys-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoLys-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoLys-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoLys-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoGlu-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoGlu-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoGlu-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoGlu-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoGlu-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-βAla-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-βAla-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-βAla-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-βAla-Peg3-Peg3, [21-carboxy-heneicosanoyl]-βAla-KEK-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoLys-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoLys-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoLys-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoLys-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoLys-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoGlu-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoGlu-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-βAla-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]KEK-βAla-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-βAla-KEK-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-βAla-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-βAla-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[13-carboxy-tridecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [15-carboxy-Pentadecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [17-carboxy-Heptadecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [19-carboxy-Nonadecanoyl]-KEK-isoLys-Peg3-Peg3-Peg3, [21-carboxy-heneicosanoyl]-KEK-isoLys-Peg3-Peg3-Peg3.

Certain preferred substituents Z1— and Z1—Z2— include:

[Hexadecanoyl], [Octadecanoyl], [17-Carboxy-heptadecanoyl], [19-Carboxy-nonadecanoyl],

[Hexadecanoyl]-isoGlu, [Octadecanoyl]-isoGlu,

[Hexadecanoyl]-βAla, [Octadecanoyl]-βAla,

[Hexadecanoyl]-isoGlu-Peg3,

[Hexadecanoyl]-βAla-Peg3,

[Hexadecanoyl]-isoGlu-Peg3-Peg3,

[Hexadecanoyl]-βAla-Peg3-Peg3,

[Hexadecanoyl]-βAla-Peg3-Peg3-Peg3,

[Hexadecanoyl]-isoLys,

[Hexadecanoyl]-[4-aminobutanoyl],

[Hexadecanoyl]-isoLys-Peg3,

[Hexadecanoyl]-[4-aminobutanoyl]-Peg3,

[Hexadecanoyl]-isoLys-Peg3-Peg3,

[Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[Hexadecanoyl]-isoLys-Peg3-Peg3-Peg3,

[Hexadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu,

[19-carboxy-Nonadecanoyl]-isoGlu,

[17-carboxy-Heptadecanoyl]-βAla,

[19-carboxy-Nonadecanoyl]-βAla,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3,

[17-carboxy-Heptadecanoyl]-βAla-Peg3,

[19-carboxy-Nonadecanoyl]-βAla-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-βAla-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-βAla-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys,

[19-carboxy-Nonadecanoyl]-isoLys,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl],

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl],

[17-carboxy-Heptadecanoyl]-isoLys-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-Peg3-Peg3-Peg3.

More preferred substituents Z1—Z2— include:

[Hexadecanoyl]-isoGlu,

[Hexadecanoyl]-βAla,

[Hexadecanoyl]-isoGlu-Peg3,

[Hexadecanoyl]-βAla-Peg3,

[Hexadecanoyl]-isoGlu-Peg3-Peg3,

[Hexadecanoyl]-isoLys,

[Hexadecanoyl]-isoLys-Peg3,

[Hexadecanoyl]-isoLys-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu,

[19-carboxy-Nonadecanoyl]-isoGlu,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys,

[19-carboxy-Nonadecanoyl]-isoLys,

[17-carboxy-Heptadecanoyl]-isoLys-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoLys-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoLys-Peg3-Peg3-Peg3.

Yet further preferred substituents Z1—Z2— include:

[Hexadecanoyl]-KEK, [Octadecanoyl]-KEK,

[Hexadecanoyl]-βAla-Peg3,

[Hexadecanoyl]-KEK-Peg3,

[Hexadecanoyl]-KEK-Peg3-Peg3,

[Hexadecanoyl]-KEK-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-KEK,

[19-carboxy-Nonadecanoyl]-KEK,

[17-carboxy-Heptadecanoyl]-KEK-Peg3,

[19-carboxy-Nonadecanoyl]-KEK-Peg3,

[17-carboxy-Heptadecanoyl]-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-KEK-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-KEK

[19-carboxy-Nonadecanoyl]-isoGlu-KEK,

[17-carboxy-Heptadecanoyl]-isoLys-KEK

[19-carboxy-Nonadecanoyl]-isoLys-KEK,

[17-carboxy-Heptadecanoyl]-βAla-KEK

[19-carboxy-Nonadecanoyl]-βAla-KEK, [17-carboxy-Heptadecanoyl]-KEK-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-KEK-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[Hexadecanoyl]-isoGlu-KEK-Peg3,

[Hexadecanoyl]-isoGlu-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-KEK,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-[4-aminobutanoyl]-KEK-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-KEK-Peg3-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-KEK-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-KEK-Peg3,

[17-carboxy-Heptadecanoyl]-isoGlu-KEK-Peg3-Peg3,

[19-carboxy-Nonadecanoyl]-isoGlu-KEK-Peg3-Peg3.

Examples of ψ comprising different substituents (fatty acids, FA), conjugated to the amino acid side-chain, optionally by a spacer, are illustrated below:

Furthermore, the substituent [Hexadecanoyl]-isoGlu, conjugated to the side chain of a lysine residue, is illustrated below:

Thus, the side chain of the Lys residue is covalently attached to the side-chain carboxyl group of the isoGlu spacer —Z2— (—ZS1—) via an amide linkage. A hexadecanoyl group (Z1) is covalently attached to the amino group of the isoGlu spacer via an amide linkage.

The substituent [Hexadecanoyl]-[4-aminobutanoyl]- conjugated to the side chain of a lysine residue, is illustrated below

The substituent [(Hexadecanoyl)iso-Lys]- conjugated to the side chain of a lysine residue, is illustrated below

The substituent [(Hexadecanoyl)β-Ala]- conjugated to the side chain of a lysine residue, is illustrated below

Some further specific examples of —Z2—Z1 combinations are illustrated below. In each case, --- indicates the point of attachment to the side chain of the amino acid component of P:

The skilled person will be well aware of suitable techniques for preparing the substituents employed in the context of the invention and conjugating them to the side chain of the appropriate amino acid in the dual agonist peptide. For examples of suitable chemistry, see WO98/08871, WO00/55184, WO00/55119, Madsen et al., J. Med. Chem. 50:6126-32 (2007), and Knudsen et al., J. Med Chem. 43:1664-1669 (2000), incorporated herein by reference.

Synthesis of Dual Agonists

It is preferred to synthesize dual agonists of the invention by means of solid-phase or liquid-phase peptide synthesis methodology. In this context, reference may be made to WO 98/11125 and, among many others, Fields, G. B. et al., 2002, “Principles and practice of solid-phase peptide synthesis”. In: Synthetic Peptides (2nd Edition), and the Examples herein.

In accordance with the present invention, a dual agonist of the invention may be synthesized or produced in a number of ways, including for example, a method which comprises

(a) synthesizing the dual agonist by means of solid-phase or liquid-phase peptide synthesis methodology and recovering the synthesized dual agonist thus obtained; or

(b) expressing a precursor peptide sequence from a nucleic acid construct that encodes the precursor peptide, recovering the expression product, and modifying the precursor peptide to yield a compound of the invention.

The precursor peptide may be modified by introduction of one or more non-proteinogenic amino acids, e.g. Aib, Orn, Dap, or Dab, introduction of a lipophilic substituent Z1 or Z1—Z2— at a residue ψ, introduction of the appropriate terminal groups R1 and R2, etc.

Expression is typically performed from a nucleic acid encoding the precursor peptide, which may be performed in a cell or a cell-free expression system comprising such a nucleic acid.

It is preferred to synthesize the analogues of the invention by means of solid-phase or liquid-phase peptide synthesis. In this context, reference is made to WO 98/11125 and, among many others, Fields, G B et al., 2002, “Principles and practice of solid-phase peptide synthesis”. In: Synthetic Peptides (2nd Edition), and the Examples herein.

For recombinant expression, the nucleic acid fragments encoding the precursor peptide will normally be inserted in suitable vectors to form cloning or expression vectors. The vectors can, depending on purpose and type of application, be in the form of plasmids, phages, cosmids, mini-chromosomes, or virus, but also naked DNA which is only expressed transiently in certain cells is an important vector. Preferred cloning and expression vectors (plasmid vectors) are capable of autonomous replication, thereby enabling high copy-numbers for the purposes of high-level expression or high-level replication for subsequent cloning.

In general outline, an expression vector comprises the following features in the 5′→3′ direction and in operable linkage: a promoter for driving expression of the nucleic acid fragment, optionally a nucleic acid sequence encoding a leader peptide enabling secretion (to the extracellular phase or, where applicable, into the periplasma), the nucleic acid fragment encoding the precursor peptide, and optionally a nucleic acid sequence encoding a terminator. They may comprise additional features such as selectable markers and origins of replication. When operating with expression vectors in producer strains or cell lines it may be preferred that the vector is capable of integrating into the host cell genome. The skilled person is very familiar with suitable vectors and is able to design one according to their specific requirements.

The vectors of the invention are used to transform host cells to produce the precursor peptide. Such transformed cells can be cultured cells or cell lines used for propagation of the nucleic acid fragments and vectors, and/or used for recombinant production of the precursor peptides.

Preferred transformed cells are micro-organisms such as bacteria [such as the species Escherichia (e.g. E. coli), Bacillus (e.g. Bacillus subtilis), Salmonella, or Mycobacterium (preferably non-pathogenic, e.g. M. bovis BCG), yeasts (e.g., Saccharomyces cerevisiae and Pichia pastoris), and protozoans. Alternatively, the transformed cells may be derived from a multicellular organism, i.e. it may be fungal cell, an insect cell, an algal cell, a plant cell, or an animal cell such as a mammalian cell. For the purposes of cloning and/or optimised expression it is preferred that the transformed cell is capable of replicating the nucleic acid fragment of the invention. Cells expressing the nucleic fragment can be used for small-scale or large-scale preparation of the peptides of the invention.

When producing the precursor peptide by means of transformed cells, it is convenient, although far from essential, that the expression product is secreted into the culture medium.

Pharmaceutical Compositions and Administration

An aspect of the present invention relates to a composition comprising a dual agonist according to the invention, or a pharmaceutically acceptable salt or solvate thereof, together with a carrier. In one embodiment of the invention, the composition is a pharmaceutical composition and the carrier is a pharmaceutically acceptable carrier. The present invention also relates to a pharmaceutical composition comprising a dual agonist according to the invention, or a salt or solvate thereof, together with a carrier, excipient or vehicle. Accordingly, the dual agonist of the present invention, or salts or solvates thereof, especially pharmaceutically acceptable salts or solvates thereof, may be formulated as compositions or pharmaceutical compositions prepared for storage or administration, and which comprise a therapeutically effective amount of a dual agonist of the present invention, or a salt or solvate thereof.

Suitable salts formed with bases include metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts; ammonia salts and organic amine salts, such as those formed with morpholine, thiomorpholine, piperidine, pyrrolidine, a lower mono-, di- or tri-alkylamine (e.g., ethyl-tert-butyl-, diethyl-, diisopropyl-, triethyl-, tributyl- or dimethylpropylamine), or a lower mono-, di- or tri-(hydroxyalkyl)amine (e.g., mono-, di- or triethanolamine). Internal salts may also be formed. Similarly, when a compound of the present invention contains a basic moiety, salts can be formed using organic or inorganic acids. For example, salts can be formed from the following acids: formic, acetic, propionic, butyric, valeric, caproic, oxalic, lactic, citric, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, phthalic, hydrochloric, hydrobromic, phosphoric, nitric, sulphuric, benzoic, carbonic, uric, methanesulphonic, naphthalenesulphonic, benzenesulphonic, toluenesulphonic, p-toluenesulphonic (i.e. 4-methylbenzene-sulphonic), camphorsulphonic, 2-aminoethanesulphonic, aminomethylphosphonic and trifluoromethanesulphonic acid (the latter also being denoted triflic acid), as well as other known pharmaceutically acceptable acids. Amino acid addition salts can also be formed with amino acids, such as lysine, glycine, or phenylalanine.

In one embodiment, a pharmaceutical composition of the invention is one wherein the dual agonist is in the form of a pharmaceutically acceptable acid addition salt.

As will be apparent to one skilled in the medical art, a “therapeutically effective amount” of a dual agonist compound or pharmaceutical composition thereof of the present invention will vary depending upon, inter alia, the age, weight and/or gender of the subject (patient) to be treated. Other factors that may be of relevance include the physical characteristics of the specific patient under consideration, the patient's diet, the nature of any concurrent medication, the particular compound(s) employed, the particular mode of administration, the desired pharmacological effect(s) and the particular therapeutic indication. Because these factors and their relationship in determining this amount are well known in the medical arts, the determination of therapeutically effective dosage levels, the amount necessary to achieve the desired result of treating and/or preventing and/or remedying malabsorption and/or low-grade inflammation described herein, as well as other medical indications disclosed herein, will be within the ambit of the skilled person.

As used herein, the term “a therapeutically effective amount” refers to an amount which reduces symptoms of a given condition or pathology, and preferably which normalizes physiological responses in an individual with that condition or pathology. Reduction of symptoms or normalization of physiological responses can be determined using methods routine in the art and may vary with a given condition or pathology. In one aspect, a therapeutically effective amount of one or more dual agonists, or pharmaceutical compositions thereof, is an amount which restores a measurable physiological parameter to substantially the same value (preferably to within 30%, more preferably to within 20%, and still more preferably to within 10% of the value) of the parameter in an individual without the condition or pathology in question.

In one embodiment of the invention, administration of a compound or pharmaceutical composition of the present invention is commenced at lower dosage levels, with dosage levels being increased until the desired effect of preventing/treating the relevant medical indication is achieved. This would define a therapeutically effective amount. For the dual agonists of the present invention, alone or as part of a pharmaceutical composition, such human doses of the active dual agonist may be between about 0.01 pmol/kg and 500 μmol/kg body weight, between about 0.01 pmol/kg and 300 μmol/kg body weight, between 0.01 pmol/kg and 100 μmol/kg body weight, between 0.1 pmol/kg and 50 μmol/kg body weight, between 1 pmol/kg and 10 μmol/kg body weight, between 5 pmol/kg and 5 μmol/kg body weight, between 10 pmol/kg and 1 μmol/kg body weight, between 50 pmol/kg and 0.1 μmol/kg body weight, between 100 pmol/kg and 0.01 μmol/kg body weight, between 0.001 μmol/kg and 0.5 μmol/kg body weight, between 0.05 μmol/kg and 0.1 μmol/kg body weight.

The therapeutic dosing and regimen most appropriate for patient treatment will of course vary with the disease or condition to be treated, and according to the patient's weight and other parameters. Without wishing to be bound by any particular theory, it is expected that doses, in the g/kg range, and shorter or longer duration or frequency of treatment may produce therapeutically useful results, such as a statistically significant increase particularly in small bowel mass. In some instances, the therapeutic regimen may include the administration of maintenance doses appropriate for preventing tissue regression that occurs following cessation of initial treatment. The dosage sizes and dosing regimen most appropriate for human use may be guided by the results obtained by the present invention, and may be confirmed in properly designed clinical trials.

An effective dosage and treatment protocol may be determined by conventional means, starting with a low dose in laboratory animals and then increasing the dosage while monitoring the effects, and systematically varying the dosage regimen as well. Numerous factors may be taken into consideration by a clinician when determining an optimal dosage for a given subject. Such considerations are known to the skilled person.

Medical Conditions

In a broad aspect, the present invention provides a dual agonist of the invention for use as a medicament.

In a further aspect, the present invention relates to a dual agonist of the invention for use in therapy.

The dual agonists described in this specification have biological activities of both GLP-1 and GLP-2.

GLP-2 induces significant growth of the small intestinal mucosal epithelium via the stimulation of stem cell proliferation in the crypts and inhibition of apoptosis on the villi (Drucker et al. Proc Natl Acad Sci USA. 1996, 93:7911-6). GLP-2 also has growth effects on the colon. GLP-2 also inhibits gastric emptying and gastric acid secretion (Wojdemann et al. J Clin Endocrinol Metab. 1999, 84:2513-7), enhances intestinal barrier function (Benjamin et al. Gut. 2000, 47:112-9.), stimulates intestinal hexose transport via the upregulation of glucose transporters (Cheeseman, Am J Physiol. 1997, R1965-71), and increases intestinal blood flow (Guan et al. Gastroenterology. 2003, 125, 136-47).

The beneficial effects of GLP-2 in the small intestine have raised considerable interest as to the use of GLP-2 in the treatment of intestinal disease or injury (Sinclair and Drucker, Physiology 2005: 357-65). Furthermore, GLP-2 has been shown to prevent or reduce mucosal epithelial damage in a wide number of preclinical models of gut injury, including chemotherapy-induced enteritis, ischemia-reperfusion injury, dextran sulfate-induced colitis and genetic models of inflammatory bowel disease (Sinclair and Drucker Physiology 2005: 357-65). The GLP-2 analogue teduglutide (Gly2-hGLP-2) is approved for treatment of short bowel syndrome under the trade names Gattex and Revestive.

GLP-1 is a peptide hormone known for its important role in glucose homeostasis. When secreted from the gastrointestinal tract in response to nutrient ingestion, GLP-1 potentiates glucose-stimulated insulin secretion from the 1-cells (Kim and Egan, 2008, Pharmacol. Rev. 470-512).

Furthermore, GLP-1 or it analogues has been shown to increase somatostatin secretion and suppress glucagon secretion (Holst J J, 2007, Physiol Rev. 1409-1439).

Besides the primary actions of GLP-1 on glucose-stimulated insulin secretion, GLP-1 is also known as a key regulator of appetite, food intake, and body weight. Moreover, GLP-1 can inhibit gastric emptying and gastrointestinal motility in both rodents and humans, most likely through GLP-1 receptors present in the gastrointestinal tract (Holst J J, 2007, Physiol Rev. 1409-1439; Hellstrom et al., 2008, Neurogastroenterol Motil. June; 20(6):649-659). In addition, GLP-1 seems to have insulin-like effects in major extrapancreatic tissues, participating in glucose homeostasis and lipid metabolism in tissues such as muscle, liver, and adipose tissues (Kim and Egan, 2008, Pharmacol. Rev. 470-512).

Thus the dual agonist compounds of the present invention may be used to increase intestinal mass, improve intestinal function (especially intestinal barrier function), increase intestinal blood flow, or repair intestinal damage or dysfunction (whether structural or functional), e.g. damage to the intestinal epithelium. They may also be used in the prophylaxis or treatment of conditions which may be ameliorated by these effects, and in reducing the morbidity related to gastrointestinal damage.

The dual agonists therefore find use in many gastrointestinal disorders. The term “gastrointestinal” is used here to include the entire gastrointestinal tract, including oesophagus, stomach, small intestine (duodenum, jejunum, ileum) and large intestine (cecum, colon, rectum), but especially the small intestine and colon.

Thus, conditions in which the dual agonists may be of benefit include malabsorption, ulcers (which may be of any aetiology, e.g., peptic ulcers, Zollinger-Ellison Syndrome, drug-induced ulcers, and ulcers related to infections or other pathogens), short-bowel syndrome, cul-de-sac syndrome, inflammatory bowel disease (Crohn's disease and ulcerative colitis), irritable bowel syndrome (IBS), pouchitis, celiac sprue (for example arising from gluten induced enteropathy or celiac disease), tropical sprue, hypogammaglobulinemic sprue, and mucositis or diarrhea induced by chemotherapy or radiation therapy.

The dual agonists may also find use in certain conditions which do not primarily affect gastrointestinal tissue but which may be caused or exacerbated by factors arising from intestinal dysfunction. For example, impaired intestinal barrier function (which may be referred to as “leakiness” of the intestine or gut) can lead to transit of materials from the lumen of the gut directly into the bloodstream and thus to the kidney, lung and/or liver. These materials may include food molecules such as fats, which contribute to hepatitis and/or fatty liver diseases, including parenteral nutrition associated gut atrophy, PNALD (Parenteral Nutrition-Associated Liver Disease), NAFLD (Non-Alcoholic Fatty Liver Disease) and NASH (Non-Alcoholic Steatohepatitis). The materials crossing into the bloodstream may also include pathogens such as bacteria, and toxins such as bacterial lipopolysaccharide (LPS), which may contribute to systemic inflammation (e.g. vascular inflammation). Such inflammation is often referred to as “low grade inflammation” and is a contributing factor to the pathogenesis of metabolic endotoxemia (a condition seen in both diabetes and obesity, discussed further below), primary biliary cirrhosis and hepatitis. Entry of pathogens to the bloodstream may also result in conditions such as necrotising enterocolitis.

Low grade inflammation is not characterised by the normal symptoms of acute inflammation such as pain, fever and redness, but can be detected via the presence of inflammatory markers in the blood, such as C-reactive protein and pro-inflammatory cytokines including TNF-alpha (tumour necrosis factor alpha).

The dual agonists may also find use in conditions which primarily affect other tissues but have gastrointestinal side-effects. For example, inflammatory conditions such as pancreatitis result in elevated levels of circulating inflammatory mediators which may in turn induce intestinal damage or intestinal dysfunction, such as impairment of barrier function. In some circumstances, this may lead to more severe systemic inflammatory conditions such as sepsis, or to surgical procedures or mechanical injuries (volvulus) where blood supply to the intestine is interrupted, ultimately leading to ischaemia-reperfusion injuries.

Similarly, graft versus host disease (GVHD) may result in substantial tissue damage to the gastrointestinal tract, resulting in impaired barrier function and other side effects such as diarrhea. Thus, the dual agonists described may be useful for the prophylaxis or treatment of intestinal dysfunction or damage caused by or associated with GVHD, as well as prophylaxis or treatment of side effects such as diarrhea caused by or associated with GVHD.

The dual agonist compounds described herein also find use, inter alia, in reducing or inhibiting weight gain, reducing rate of gastric emptying or intestinal transit, reducing food intake, reducing appetite, or promoting weight loss. The effect on body weight may be mediated in part or wholly via reducing food intake, appetite or intestinal transit.

Thus the dual agonists of the invention can be used for the prophylaxis or treatment of obesity, morbid obesity, obesity-linked gallbladder disease and obesity-induced sleep apnea.

Independently of their effect on body weight, the dual agonists of the invention may have a beneficial effect on glucose tolerance and/or glucose control. They may also be used to modulate (e.g. improve) circulating cholesterol levels, being capable of lowering circulating triglyceride or LDL levels, and increasing HDL/LDL ratio.

Thus, they may be used for the prophylaxis or treatment of inadequate glucose control, glucose tolerance or dyslipidaemia (e.g. elevated LDL levels or reduced HDL/LDL ratio) and associated conditions, including diabetes (e.g. Type 2 diabetes, gestational diabetes), pre-diabetes, metabolic syndrome and hypertension.

Many of these conditions are also associated with obesity or overweight. The effects of the dual agonists on these conditions may therefore follow from their effect on body weight, in whole or in part, or may be independent thereof.

Effects on body weight may be therapeutic or cosmetic.

The dual agonist activity of the compounds described herein may be particularly beneficial in many of the conditions described, as the two activities may complement one another.

For example, malabsorption is a condition arising from abnormality in the absorption of water and/or food nutrients, such as amino acids, sugars, fats, vitamins or minerals, via the gastrointestinal (GI) tract, leading to malnutrition and/or dehydration. Malabsorption may be a result of physical (e.g. traumatic) or chemical damage to the intestinal tract. Dual agonists as described in this specification may be capable of improving intestinal barrier function, reducing gastric empting, and increasing intestinal absorption while at the same time normalising intestinal transit time. This would not only help patients to increase the absorption of nutrients and liquid, but would also alleviate patients' social problems related to meal-stimulated bowel movements.

Furthermore, intestinal function and metabolic disorders may be closely inter-related, with each contributing to the development or symptoms of the other.

As mentioned above, obesity is linked with low grade inflammation (sometimes designated “obesity-linked inflammation”). It is also generally recognised that obesity (along with other syndromes) causes an increased vascular permeability which allows pathogens and toxins such as LPS to enter the cell wall of the intestinal tract and thereby initiate inflammation. The changes that result from the inflammatory response are essentially the same regardless of the cause and regardless of where the insult arises. The inflammatory response may be acute (short lived) or chronic (longer lasting).

It has been demonstrated that, e.g., obese mice (ob/ob and db/db mice) have a disrupted mucosal barrier function and exhibit increased low-grade inflammation (Brun et al., 2007, Am. J. Physiol. Gastrointest. Liver Physiol., 292: G518-G525, Epub 5 Oct. 2006). These observations were further extended to C57BL6/J mice maintained on a high-fat diet (Cani et al., 2008, Diabetes, vol. 57, 1470-1481) and to non-obese diabetic mice (Hadjiyanni et al., 2009, Endocrinology, 150(2): 592-599).

Cani and colleagues (Gut; 2009, 58:1091-1103,) reported that in ob/ob mice, the modulation of the gut microbiota resulted in decreased intestinal barrier dysfunction and reduced systemic inflammation via a GLP-2 dependent pathway. Further, the increased intestinal permeability observed in obese and diabetic patients is likely to play a more vital role in the disease progression than previously anticipated. Increased intestinal permeability leads to increased bacterial lipopolysaccharide (LPS) transport across the intestinal barrier. This increased LPS activates immune cells, such as circulating macrophages and macrophages residing in organs in the body, causing low-grade chronic inflammation that may be involved in the pathogenesis of many diseases. This phenomenon is called metabolic endotoxemia (ME).

The inflammatory process may also play a role in causing metabolic dysfunction in obese individuals, such as insulin resistance and other metabolic disturbances.

Thus the dual agonist compounds of the invention may be particularly useful for prophylaxis or treatment of low grade inflammation, especially in obese or overweight individuals, exerting beneficial effects via the GLP-1 agonist component of their activity and/or the GLP-2 component of their activity.

The therapeutic efficacy of treatment with a dual agonist of the invention may be monitored by enteric biopsy to examine the villus morphology, by biochemical assessment of nutrient absorption, by non-invasive determination of intestinal permeability, by patient weight gain, or by amelioration of the symptoms associated with these conditions.

In a further aspect there is provided a therapeutic kit comprising a dual agonist of the invention, or a pharmaceutically acceptable salt or solvate thereof.

The following examples are provided to illustrate preferred aspects of the invention and are not intended to limit the scope of the invention in any way.

EXAMPLES

The following examples are provided to illustrate preferred aspects of the invention and are not intended to limit the scope of the invention.

Materials and Methods

General Peptide Synthesis

List of abbreviations and suppliers are provided in the table below

List of abbreviations and suppliers Abbreviation Name Brand/Supplyer Resins TentaGel ™ PHB AA(Proct)- Rapp Polymere Fmoc TentaGel ™ SRAM Rapp Polymere Amino Pseudoprolines (E.g. QT, Jupiter Bioscience Ltd. acids AT, FS) Fmoc-L-AA-OH Senn Chemicals AG Coupling COMU (1-Cyano-2-ethoxy-2- Watson International Ltd. reagents oxoethylidenaminooxy)dimethylamino- morpholino- carbenium hexafluorophosphate DIC Diisopropylcarbodiimide Fluka/Sigma Aldrich Co. HATU N-[(dimethylamino)-1H- ChemPep Inc. 1,2,3-triazol[4,5-b]pyridine- 1-ylmethylene]-N- methylmethanaminium hexafluorophosphate N- oxide HOBt Hydroxybenzotriazole Sigma-Aldrich Co. Solvents Boc2O Di-tert-butyl pyrocarbonate Advanced ChemTech reagents DCM Dichloromethane Prolabo (VWR) DIPEA Diisopropylethylamine Fluka/Sigma Aldrich Co. DMF N,N-dimethylformamide Taminco DODT 3,6-dioxa-1,8-octanedithiol Sigma-Aldrich Co. Et2O Diethyl ether Prolabo (VWR) EtOH Ethanol CCS Healthcare AB Formic acid (HPLC) Sigma-Aldrich Co. H2O Water, Milli-Q water Millipore MeCN Acetonitrile (HPLC) Sigma-Aldrich Co. NMP N-methylpyrrolidone Sigma-Aldrich Co. Piperidine Jubliant Life Sciences Ltd. TFA Trifluoroacetic acid (HPLC) Chemicals Raw Materials Ltd. TIS Triisopropylsilane Sigma-Aldrich Co. MeOH Methanol Sigma-Aldrich Co.

Apparatus and Synthetic Strategy

Peptides were synthesized batchwise on a peptide synthezier, such as a CEM Liberty Peptide Synthesizer or a Symphony X Synthesizer, according to solid phase peptide synthetic procedures using 9-fluorenylmethyloxycarbonyl (Fmoc) as N-α-amino protecting group and suitable common protection groups for side-chain functionalities.

As polymeric support based resins, such as e.g. TentaGel™, was used. The synthesizer was loaded with resin that prior to usage was swelled in DMF.

Coupling

CEM Liberty Peptide Synthesizer

A solution of Fmoc-protected amino acid (4 equiv.) was added to the resin together with a coupling reagent solution (4 equiv.) and a solution of base (8 equiv.). The mixture was either heated by the microwave unit to 70-75° C. and coupled for 5 minutes or coupled with no heat for 60 minutes. During the coupling nitrogen was bubbled through the mixture.

Symphony X Synthesizer

The coupling solutions were transferred to the reaction vessels in the following order: amino acid (4 equiv.), HATU (4 equiv.) and DIPEA (8 equiv.). The coupling time was 10 min at room temperature (RT) unless otherwise stated. The resin was washed with DMF (5×0.5 min). In case of repeated couplings the coupling time was in all cases 45 min at RT.

Deprotection

CEM Liberty Peptide Synthesizer

The Fmoc group was deprotected using piperidine in DMF or other suitable solvents. The deprotection solution was added to the reaction vessel and the mixture was heated for 30 sec. reaching approx. 40° C. The reaction vessel was drained and fresh deprotection solution was added and subsequently heated to 70-75° C. for 3 min. After draining the reaction vessel the resin was washed with DMF or other suitable solvents.

Symphony X Synthesizer

Fmoc deprotection was performed for 2.5 minutes using 40% piperidine in DMF and repeated using the same conditions. The resin was washed with DMF (5×0.5 min).

Side-Chain Acylation

A suitable trifunctional amino acid with an orthogonal side chain protecting group according to Fmoc methodology is introduced at the position of the acylation. The N-terminal of the growing peptide chain is then Boc-protected using Boc2O or alternatively by using an N-α-Boc-protected amino acid in the last coupling. While the peptide is still attached to the resin, the orthogonal side chain protecting group is selectively cleaved using a suitable deprotection reagent. The lipophilic moiety is then coupled directly to the free sidechain functionality or alternatively via a linker in between according to suitable coupling protocols.

Cleavage

The dried peptide resin was treated with TFA and suitable scavengers for approximately 2 hours. The volume of the filtrate was reduced and the crude peptide was precipitated after addition of diethylether. The crude peptide precipitate was washed several times with diethylether and finally dried.

HPLC Purification of the Crude Peptide

The crude peptide was purified by preparative reverse phase HPLC using a conventional HPLC apperatus, such as a Gilson GX-281 with 331/332 pump combination’, for binary gradient application equipped with a column, such as 5×25 cm Gemini NX 5 u C18 110 A column, and a fraction collector using a flow 20-40 ml/min with a suitable gradient of buffer A (0.1% Fomic acid, aq.) or A (0.1% TFA, aq.) and buffer B (0.1% Formic acid, 90% MeCN, aq.) or B (0.1% TFA, 90% MeCN, aq.). Fractions were analyzed by analytical HPLC and MS and selected fractions were pooled and lyophilized. The final product was characterized by HPLC and MS.

Analytical HPLC

Final purities were determined by analytic HPLC (Agilent 1100/1200 series) equipped with auto sampler, degasser, 20 μl flow cell and Chromeleon software. The HPLC was operated with a flow of 1.2 ml/min at 40° C. using an analytical column, such as Kinetex 2.6-μm XB-C18 100 A 100×4.6 mm column. The compound was detected and quantified at 215 nm. Buffers A (0.1% TFA, aq.) and buffer B (0.1% TFA, 90% MeCN, aq.).

Mass Spectroscopy

Final MS analysis were determined on a conventionial mass spectroscopy, e.g. Waters Xevo G2 Tof, equipped with electrospray detector with lock-mass calibration and MassLynx software. It was operated in positive mode using direct injection and a cone voltage of 15V (1 TOF), 30 V (2 TOF) or 45 V (3 TOF) as specified on the chromatogram. Precision was 5 ppm with a typical resolution of 15,000-20,000.

GLP-1 and GLP-2 Receptor Efficacy Assays

Peptides of this invention function as both GLP-1 and GLP-2 agonists and thus activate the GLP-1 receptor and GLP-2 receptor, respectively. One useful in vitro assay for measuring GLP-1 and GLP-2 receptor activity is quantitation of cAMP, i.e. 3′-5′-cyclic adenosine monophosphate, which is a second messenger essential in many biological processes, and one of the most ubiquitous mechanisms for regulating cellular functions. An example is the cAMP AlphaScreen® assay from Perkin Elmer which has been used to quantitate the cAMP response upon GLP-1 and GLP-2 receptor activation in HEK293 cells stably expressing GLP-1 R or GLP-2 R. Test compounds eliciting an increase in the intracellular level of cAMP can be tested in these assays, and the response normalized relative to a positive and negative control (vehicle) to calculate the EC50 and maximal response from the concentration response curve using the 4-parameter logistic (4PL) nonlinear model for curve fitting.

Example 1: Synthesis of the Compounds

Compounds Synthesised

The following compounds of Table 1 were synthesized using the above techniques.

TABLE 1 Compounds synthesized  1 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH  2 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH  3 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARDFIAWLIEHKITD-OH  4 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH  5 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH  6 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH  7 Hy-H[Aib]EGTFSSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH  8 Hy-H[Aib]EGSFTSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH  9 Hy-H[Aib]EGTFTSELATILDG[K([17-carboxy- heptadecanoyl]-isoGlu)]AARDFIAWLIEHKITD-OH 10 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH 11 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH 12 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIEHKITD-OH 13 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH 14 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH 15 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]LAARDFIAWLIAHKITD-OH 16 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]EAARLFIAWLIEHKITD-OH 17 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH 18 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH 19 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH 20 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH 21 Hy-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH 22 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH 23 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIAHKITD-OH 24 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH 25 Hy-H[Aib]EGTFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIEHKITD-OH 26 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIHHKITD-OH 27 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIYHKITD-OH 28 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLILHKITD-OH 29 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIKHKITD-OH 30 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIRHKITD-OH 31 Hy-H[Aib]EGSFTSELATILD[K([17-Carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLISHKITD-OH 32 Hy-H[Aib]EGSFTSELATILD[K([Hexadecanoyl]-βAla)] QAARDFIAWLQQHKITD-OH 33 Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu-Peg3)]QAARDFIAWLYQHKITD- OH 34 Hy-H[Aib]EGSFTSELATILD[K([19-carboxy- nonadecanoyl]-isoGlu-Peg3-Peg3)] QAARDFIAWLKQHKITD-OH 35 Hy-H[Aib]EGSFTSELATILD[K([19-carboxy- nonadecanoyl]iso-Lys-Peg3-Peg3-Peg3)] QAARDFIAWLIQQKITD-OH 36 Hy-H[Aib]EGSFTSELATILD[K(Octadecanoyl)] QAARDFIAWLIQYKITD-OH 37 Hy-H[Aib]EGTFSSELSTILE[K(Hexadecanoyl-isoGlu)] QASREFIAWLIAYKITE-OH 38 Hy-H[Aib]EGTFSSELATILDEQAARDFIAWLIAHKITDkkkkkk ([17-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] 39 Hy-H[Aib]EGTFTSELATILDEQAARDFIAWLIAHKITDkkkkkk ([l7-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] 40 Hy-H[Aib]EGSFTSELATILDEQAARDFIAWLIEHKITDkkkkkk ([17-carboxy-Heptadecanoyl]-isoGlu)]-[NH2] 41 Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH 42 Hy-H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH 43 Hy-H[Aib]EGSFTSELATILD[K([19-carboxy- nonadecanoyl]-isoGlu-Peg3-Peg3)] KAARDFIAWLIEHKITD-OH 44 Hy-H[Aib]EGSFTSELATILEG[K([19-carboxy- nonadecanoyl]-isoGlu-Peg3-Peg3)] AARDFIAWLIEHKITD-OH 45 Hy-H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]-isoGlu-Peg3-Peg3)]DFIAWLIEHKITD- OH 46 Hy-H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]-isoGlu-Peg3-Peg3)]DFIAWLIEHKITD- OH 47 Hy-H[Aib]EGTFSSELATILD[K([l7-Carboxy- heptadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIQHKITD-OH 48 Hy-H[Aib]EGTFSSELATILD[K([l9-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIQHKITD-OH 49 Hy-H[Aib]EGTFSSELATILD[K([l7-Carboxy- heptadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIEHKITD-OH 50 Hy-H[Aib]EGTFSSELATILD[K([l9-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIEHKITD-OH 51 Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK)]QAARDFIAWLIQHKITD-OH 52 Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIQHKITD-OH 53 Hy-H[Aib]EGSFTSE[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH 54 Hy-H[Aib]EGTFTSE[K([19-Carboxy-nonadecanoyl]- isoGlu-KEK-Peg3)]ATILDEQAARDFIAWLIEHKITD-OH 55 Hy-H[Aib]EGSFTSE[K([19-carboxy-nonadecanoyl] iso-Glu-KEK-Peg3-Peg3)]ATILDEQAARDFIAWLIEHKITD- OH 56 Hy-H[Aib]EGTFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIEHKITD-OH 57 Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIEHKITD-OH 58 Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] QAARDFIAWLIAHKITD-OH 59 Hy-H[Aib]EGSFTSELATILD[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] KAARDFIAWLIEHKITD-OH 60 Hy-H[Aib]EGSFTSELATILD[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)] QAARDFIAWLIEHKITD-OH 61 Hy-H[Aib]EGSFTSELATILEG[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] AARDFIAWLIEHKITD-OH 62 Hy-H[Aib]EGSFTSELATILDA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] AARDFIAWLIEHKITD-OH 63 Hy-H[Aib]EGSFTSELATILDA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)] ARDFIAWLIEHKITD-OH 64 Hy-H[Aib]EGSFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD-OH 65 Hy-H[Aib]EGTFTSELATILDEQAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)]DFIAWLIEHKITD-OH 66 Hy-H[Aib]EGSFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)] DFIAWLIEHKITD-OH 67 Hy-H[Aib]EGTFTSELATILDEQAA[K([19-carboxy- nonadecanoyl]iso-Glu-KEK-Peg3-Peg3)] DFIAWLIEHKITD-OH 68 Hy-H[Aib]EGSFTSELATILDAKAA[K([19-Carboxy- nonadecanoyl]-isoGlu-KEK-Peg3)] DFIAWLIEHKITD-OH

The following reference compounds A and B were also synthesised:

A Hy-H[Aib]DGSFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH B Hy-H[Aib]EGSFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH

For illustration purposes only, the synthesis of two selected compounds is described in detail below.

Synthesis of Compound 17


H—H[Aib]EGTFSSELATILD[K([17-carboxy-heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH

Solid phase peptide synthesis was performed on a Symphony X Synthesizer using standard Fmoc chemistry. TentaGel S PHB Asp(tBu)Fmoc (1.15 g; 0.23 mmol/g) was swelled in DMF (10 ml) prior to use and the Fmoc-group was deprotected according to the procedure described above.

Coupling

Suitable protected Fmoc-amino acids according to the sequence were coupled as described above using HATU as coupling reagent. All couplings were performed at R.T. In order to facilitate the synthesis, a pseudoproline were used: in position 6 and 7 Fmoc-Phe-Ser(psi Me,Mepro)-OH.

Acylation in position 16 was obtained according to the side-chain acylation procedure described above. The pseudoproline was coupled according to the standard procedure described above for Fmoc-amino acids.

Deprotection

Fmoc deprotection was performed according to the procedure described above.

Cleavage of the Peptide from the Solid Support

The peptide-resin was washed with EtOH (3×10 ml) and Et2O (3×10 ml) and dried to constant weight at room temperature (r.t.). The peptide was cleaved from the resin by treatment with TFA/TIS/H2O (95/2,5/2,5; 40 ml, 2 h; r.t.). The volume of the filtrate was reduced and the crude peptide was precipitated after addition of diethylether. The crude peptide precipitate was washed several times with diethylether and finally dried to constant weight at room temperature yield 1100 mg crude peptide product (purity ˜40%).

HPLC Purification of the Crude Peptide

The crude peptide was purified by preparative reverse phase HPLC using a Gilson GX-281 with 331/332 pump combination for binary gradient application equipped with a 5×25 cm Gemini NX 5 u C18 110 A, column and a fraction collector and run at 35 ml/min with a gradient of buffer A (0.1% TFA, aq.) and buffer B (0.1% TFA, 90% MeCN, aq.) gradient from 25% B to 60% B in 47 min. Fractions were analyzed by analytical HPLC and MS and relevant fractions were pooled and lyophilized to yield 105.7 mg, with a purity of 91% as characterized by HPLC and MS as described above. Calculated monoisotopic MW=4164.21, found 4164.23.

Synthesis of Compound 4

H-H[Aib]EGTFSSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]KAARDFIAWLIEHKITD-OH

Solid phase peptide synthesis was performed on a Symphony X Synthesizer using standard Fmoc chemistry. TentaGel S PHB Asp(tBu)Fmoc (1.20 g; 0.23 mmol/g) was swelled in DMF (10 ml) prior to use and the Fmoc-group was deprotected according to the procedure described above.

Coupling

Suitable protected Fmoc-amino acids according to the sequence were coupled as described above using HATU as coupling reagent. All couplings were performed at R.T. In order to facilitate the synthesis, a pseudoproline were used: in position 6 and 7 Fmoc-Phe-Ser(psi Me,Mepro)-OH. Acylation in position 16 was obtained according to the side-chain acylation procedure described above. The pseudoproline was coupled according to the standard procedure described above for Fmoc-amino acids.

Deprotection

Fmoc deprotection was performed according to the procedure described above.

Cleavage of the Peptide from the Solid Support

The peptide-resin was washed with EtOH (3×10 ml) and Et2O (3×10 ml) and dried to constant weight at room temperature (r.t.). The peptide was cleaved from the resin by treatment with TFA/TIS/H2O (95/2,5/2,5; 40 ml, 2 h; r.t.). The volume of the filtrate was reduced and the crude peptide was precipitated after addition of diethylether. The crude peptide precipitate was washed several times with diethylether and finally dried to constant weight at room temperature yield 900 mg crude peptide product (purity ˜35%).

HPLC Purification of the Crude Peptide

The crude peptide was purified by preparative reverse phase HPLC using a Gilson GX-281 with 331/332 pump combination for binary gradient application equipped with a 5×25 cm Gemini NX 5 u C18 110 A, column and a fraction collector and run at 35 ml/min with a gradient of buffer A (0.1% TFA, aq.) and buffer B (0.1% TFA, 90% MeCN, aq.) gradient from 30% B to 65% B in 47 min. Fractions were analyzed by analytical HPLC and MS and relevant fractions were pooled and lyophilized to yield 100.7 mg, with a purity of 91% as characterized by HPLC and MS as described above. Calculated monoisotopic MW=4165.23, found 4165.26.

Example 2: GLP-1R and GLP-2R EC50 Measurements

Generation of Cell Line Expressing Human GLP-1 Receptors

The cDNA encoding the human glucagon-like peptide 1 receptor (GLP-1R) (primary accession number P43220) was cloned from the cDNA BC112126 (MGC:138331/IMAGE:8327594). The DNA encoding the GLP-1-R was amplified by PCR using primers encoding terminal restriction sites for subcloning. The 5′-end primers additionally encoded a near Kozak consensus sequence to ensure efficient translation. The fidelity of the DNA encoding the GLP-1-R was confirmed by DNA sequencing. The PCR products encoding the GLP-1-R were subcloned into a mammalian expression vector containing a neomycin (G418) resistance marker. The mammalian expression vectors encoding the GLP-1-R were transfected into HEK293 cells by a standard calcium phosphate transfection method. 48 hours post-transfection, cells were seeded for limited dilution cloning and selected with 1 mg/ml G418 in the culture medium. Following 3 weeks in G418 selection clones were picked and tested in a functional GLP-1 receptor potency assay as described below. One clone was selected for use in compound profiling.

Generation of Cell Line Expressing Human GLP-2 Receptors

The hGLP2-R was purchased from MRC-geneservice, Babraham, Cambridge as an Image clone: 5363415 (11924-117). For subcloning into a mammalian expression vector, primers for subcloning were obtained from DNA-Technology, Risskov, Denmark. The 5′ and 3′ primers used for the PCR reaction include terminal restriction sites for cloning and the context of the 5′ primer is modified to a Kozak consensus without changing the sequence of the product encoded by the ORF. A standard PCR reaction was run using Image clone 5363415 (11924-117) as a template with the above mentioned primers and Polymerase Herculase II Fusion in a total vol. of 50μl. The generated PCR product was purified using GFX PCR and Gel band purification kit, digested with restriction enzymes and cloned into the mammalian expression vector using Rapid DNA Ligation Kit. Ligation reaction was transformed to XL10 Gold Ultracompetent cells and colonies were picked for DNA production using Endofree Plasmid maxi kit. Subsequent sequence analysis was conducted by MWG Eurofins, Germany. The clone was confirmed to be the hGLP-2 (1-33) receptor, splice variant rs17681684.

HEK293 cells were transfected using the Lipofectamine PLUS transfection method. The day before transfection, HEK293 cells were seeded in two T75 flasks at a density of 2×106 cells/T75 flask in cell culturing medium without antibiotics. On the day of transfection, cells were washed with 1×DPBS and medium was replaced with Optimem to a volume of 5 mL/T75 flask before addition of Lipofectamine-plasmid complexes were added gently and drop wise to the cells in T75 flasks and replaced with growth medium after 3 hours and again to growth medium supplemented with 500 μg/mL G418 after 24 hours. Following 4 weeks in G418 selection, clones were picked and tested in a functional GLP-2 receptor potency assay as described below. One clone was selected for use in compound profiling.

GLP-1R and GLP-2 Receptor Potency Assays

The cAMP AlphaScreen® assay from Perkin Elmer was used to quantitate the cAMP response to activation of the GLP1 and GLP2 receptor, respectively. Exendin-4 was used as reference compound for GLP1 receptor activation and Teduglutide as reference compound for GLP2 receptor activation. Data from test compounds eliciting an increase in the intracellular level of cAMP were normalized relative to the positive and negative control (vehicle) to calculate the EC50 and maximal response from the concentration response curve. The results are listed in Table 2.

TABLE 2 EC50 measurements Compound EC50 GLP-1 (nM) EC50 GLP-2 (nM) Teduglutide 39 0.027 Liraglutide 0.029 N/A A 0.490 0.083 B 3.900 0.280 1 0.630 0.350 2 0.130 0.250 3 0.042 0.330 4 0.660 0.087 5 0.170 0.063 6 0.058 0.120 7 0.920 0.019 8 0.220 0.039 9 0.056 0.056 10 1.800 0.087 11 0.320 0.085 12 0.140 0.110 13 2.200 0.099 14 0.570 0.086 15 0.250 0.160 16 0.073 0.680 17 0.900 0.330 18 0.190 0.210 19 0.066 0.230 20 0.550 0.370 21 1.800 0.270 22 0.230 0.200 23 0.130 0.240 24 0.210 0.170 25 0.094 0.330 26 0.290 0.590 27 0.450 1.100 28 0.360 0.510 29 0.310 0.290 30 0.310 0.380 31 0.270 0.240 32 0.380 0.460 33 0.850 0.072 34 0.280 0.130 35 0.099 0.300 36 0.320 3.200 38 0.250 0.890 39 0.044 0.980 40 0.074 0.500 41 0.048 0.620 42 0.067 0.330 43 0.096 0.150 44 0.063 0.140 45 1.400 0.360 46 0.260 0.380 47 0.440 0.048 48 0.470 0.054 49 0.270 0.044 50 0.310 0.056 51 0.020 0.180 52 0.020 0.075 53 0.076 0.240 54 0.034 0.990 55 0.110 0.780 56 0.033 0.076 57 0.093 0.083 58 0.089 0.090 59 0.088 0.110 60 0.097 0.074 61 0.130 0.200 62 0.270 0.150 63 0.310 0.170 64 0.490 0.200 65 0.130 0.350 66 0.650 0.180 67 0.160 0.220 68 0.084 0.100 N/A = no detectable activity

Example 3: Solubility Assessment

A stock solution of the test peptide (2 mg/ml; determined from the weighed amount of peptide) in demineralized water adjusted to pH 2.5 with HCl was prepared, and aliquots were diluted 1:1 in 100 mM acetate buffer (pH 4.0 and pH 5.0), 100 mM histidine buffer (pH 6.0 and pH 7.0) and 100 mM phosphate buffer (pH 6.0, pH 7.0 and pH7.5), respectively, and loaded in a standard flat-bottom, non-sterile 96-well UV Microplate. The absorbance of samples (single samples, n=1) at 280 and 325 nm was measured in an absorbance-based plate reader, which was preheated to ambient temperature (typically 25° C.). The turbidity absorbance criterion for a peptide solubility of >1 mg/ml was an absorbance at 325 nm of <0.025 absorbance units (which is 5 to 6 times the standard deviation of 8 buffer samples in a plate). Solubility data for peptides of the invention are shown in Table 3, below.

TABLE 3 Solubility data. Acetate Acetate Histidine Histidine Phosphate Phosphate Phosphate buffer buffer buffer buffer buffer buffer buffer Cdp. pH 4 pH 5 pH 6 pH 7 pH 6 pH 7 pH 7.5 Tedu- II II II SS II II SS glutide 17 II II SS SS SS SS SS 18 II II SS SS SS SS SS 19 II II SS SS SS SS SS 20 II II SS SS SS SS SS 22 II II SS SS SS SS SS 23 II II SS SS SS SS SS 24 II II SS SS SS SS SS 25 II II SS SS SS SS SS 26 SS II II SS II SS SS 27 II II SS SS SS SS SS 28 II II SS SS SS SS SS *SS indicates solubility ≥ 1 mg/ml **II indicates solubility < 1 mg/ml

Example 4: Chemical Stability

Samples of each test peptide were dissolved in MilliQ™ water, and the pH of the solution was adjusted to pH 6, 7, 7.5 or 9 using either HCl or NaOH. The final peptide concentration was 0.2 mg/ml. Samples were placed in glass vials and incubated at 40° C. The samples were analyzed by RP-HPLC on a C18 column with gradient elution using an acetonitrile/TFA/water eluent system. The area-percentage (area-%) of the main peak after incubation time T=t (relative to time T=0) was determined by UV spectroscopy at 220 nm.

The purity was first determined as follows:


Purity(area-%)=(area of main peak/total area of all peaks)×100.

The purity was then normalized between time points by setting purity at time 0 (T=0) to 100 for each pH value for a given peptide, as follows:


Normalised area-% at time t(T=t)=[area-%(T=t)/area-%(T=0)]×100.

The chemical stability assessment results after 14 day incubation (in the form of normalized purity values) are summarized in Table 4.

TABLE 4 Chemical stability data. pH 6 pH 7 normalised normalised Compound stability stability Teduglutide A C B B A A 18 A A 19 A A 22 A B 23 A B 24 B A 25 A 26 A 28 A 29 A B 30 A 31 A A 2 A A 5 A A 11 A 14 A 53 A 42 A 55 A 58 A 67 A 68 A 32 A 33 A 34 A Key: A ->90% normalised stability; B ->80% stability; C -<80% normalized stability.

Example 5: Effect on Fasting Glucose and Intestinal Weight in Normal Mice

Normal chow-fed C57BL/6J male mice were used. The mice were kept in standard housing conditions, light-, temperature-, and humidity-controlled room (12:12 h light-dark cycle, with lights on at 06.00-18.00 h; 20-22° C.; 50-80% relative humidity). Each dosing group consisted of 6 animals. Mice were dosed once daily with 100 nmol/kg with the test compounds or vehicle for 4 days via subcutaneous administration.

On day 0 mice were fasted and blood glucose levels measured after a single s.c. injection with peptides. Animals were sacrificed 24 hours after final dosing on day 3, and small intestinal wet weights were measured.

All test compounds (100 nmol/kg) reduced fasting blood glucose levels compared to vehicle group (Table 5).

All test compounds (100 nmol/kg) increased small intestine wet weight as compared to the vehicle-treated mice (Table 5).

TABLE 5 Effects on fasting blood glucose levels and small intestinal weight. Fasting blood glucose Small intestinal wet Treatment (mM) weight (g) Vehicle 8.99 0.80 Cpd. 18 5.26 1.37 Cpd. 48 5.08 1.41 Cpd. 50 5.57 1.43 Cpd. 5 4.60 1.35 Cpd. 8 5.28 1.27 Cpd. 9 4.98 1.11 Cpd. 52 4.69 1.09

Claims

1. A dual agonist or pharmaceutically acceptable salt or solvate thereof represented by the formula:

R1—X*—R2
wherein:
R1 is hydrogen (Hy), C1-4 alkyl (e.g. methyl), acetyl, formyl, benzoyl or trifluoroacetyl;
R2 is NH2 or OH;
X* is a peptide H[Aib]EGSFTSELATILDψQAARDFIAWLIQHKITD;
ψ is an L or D Lys residue whose side chain is conjugated to a substituent of formula Z1—Z2—;
and Z1—Z2— is [17-carboxy-heptadecanoyl]-isoGlu.

2. The dual agonist or pharmaceutically acceptable salt or solvate thereof according to claim 1, wherein X* has the sequence: H[Aib]EGSFTSELATILD[K*]QAARDFIAWLIQHKITD;

wherein K* indicates an L lysine residue in which the side chain is conjugated to the substituent Z1—Z2—.

3. The dual agonist or pharmaceutically acceptable salt or solvate thereof according to claim 2, which is: Hy-H[Aib]EGSFTSELATILD[K([17-carboxy- heptadecanoyl]-isoGlu)]QAARDFIAWLIQHKITD-OH.

4. A pharmaceutical composition comprising the dual agonist or pharmaceutically acceptable salt or solvate thereof according to claim 1, in admixture with a pharmaceutically acceptable carrier, an excipient or a vehicle.

Patent History
Publication number: 20190142904
Type: Application
Filed: Nov 19, 2018
Publication Date: May 16, 2019
Inventors: Bjarne DUE LARSEN (Roskilde), Jonathan GRIFFIN (Lund), Lise GIEHM (Frederiksberg), Alistair Vincent Gordon EDWARDS (Copenhagen S)
Application Number: 16/195,752
Classifications
International Classification: A61K 38/26 (20060101); A61P 3/04 (20060101); A61P 3/10 (20060101);